Download as pdf or txt
Download as pdf or txt
You are on page 1of 254

2 VIRAL

DISEASES
W. L. Mengeling, Editor
Adenovirus
4 J. B. Derbyshire

The first isolation of a porcine adenovirus was made reported from secondary pig thyroid cell cultures by Dea
from a rectal swab from a piglet with diarrhea by Haig et and Elazhary (1984a). In unstained cultures the cyto-
al. (1964). A second isolation was made by Kasza (1966) pathic effects (CPE) are characterized by enlargement
from the brain of a pig with encephalitis. Subsequently, and rounding of the cells, followed by detachment; in
porcine adenoviruses were isolated from a variety of stained monolayers characteristic nuclear inclusion bod-
sources, including stocks of hog cholera virus, swine fe- ies are seen. Ultrastructural studies of infected cells
ces, and kidney tissue used for routine cell culture pro- showed that the virus was intranuclear, sometimes in
duction. Serologic surveys have indicated that aden- crystalline arrays (Chandler 1965).
ovirus infections are widespread and probably
ubiquitous in swine (Bibrack 1974). Strains of serotype 4 Serologic Classification
appear to be the most widely distributed, both in Europe The porcine adenoviruses contain the mammalian aden-
and North America. While the majority of infections are ovirus group-specific antigen, detectable by immunodif-
asymptomatic, these viruses have been associated with fusion or complement fixation. Virus-neutralization
encephalitis, pneumonia, kidney lesions, and diarrhea. (VN) tests have failed to reveal relationships between
Porcine adenoviruses are not known to be infectious for porcine adenoviruses and those of other species. Four
other species, although pigs can be infected with certain serotypes of porcine adenovirus are currently recognized
human adenoviruses (Betts et al. 1962), and some DNA (Table 4.1) on the basis of VN tests, and there is evidence
homology has been demonstrated between porcine and that additional serotypes may exist (Derbyshire et al.
bovine subgroup 1 adenoviruses (Benko et al. 1990). 1975).

ETIOLOGY EPIDEMIOLOGY

Physicochemical Characteristics Strains of porcine adenovirus type 4 appear to be the


The basic characteristics of porcine adenoviruses resem- most widely distributed infection; this virus has been
ble those of other members of the Adenoviridae. The identified either serologically or by virus isolation in Aus-
roughly spherical, nonenveloped virions of about 75 nm tralia, Belgium, Bulgaria, Canada, Denmark, Germany,
diameter contain a genome of double-stranded deoxyri- Hungary, Japan, the Netherlands, the United Kingdom,
bonucleic acid (DNA) surrounded by an icosahedral cap- and the United States. Transmission of porcine aden-
sid of 252 capsomeres. Strains of porcine adenovirus oviruses is probably by the fecal-oral route. Inhalation of
type 4 hemagglutinate various species of red blood cells. infectious aerosols may also occur. Virus is excreted in
The porcine adenoviruses are stable at pH 4 or when the feces, most frequently by pigs in the postweaning pe-
treated with chloroform or ether. They are relatively riod (Derbyshire et al. 1966). Adults rarely excrete virus,
heat-resistant viruses, surviving for more than 10 days at but they frequently have high serum antibody levels.
room temperature (Kasza 1966). Effective chemical dis- Nursing piglets are probably protected by antibodies in
infectants for porcine adenoviruses include sodium the milk of the sow.
hypochlorite, formaldehyde, phenolic compounds, ethyl
alcohol, and sodium hydroxide (Derbyshire and Arkell
1971). In liquid manure, porcine adenoviruses are inacti-
vated by aeration or treatment with calcium hydroxide Table 4.1. Serologic classification of porcine
adenoviruses
(Derbyshire and Brown 1979).
Serotype No. WHO Reference Strain
Laboratory Cultivation 1 25R
Porcine adenoviruses can be cultivated fairly readily in 2 A47
primary pig kidney (PK) cell cultures and in certain es- 3 6618
tablished porcine cell lines. Relatively high yields were 4 F618

89
90 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

CLINICAL SIGNS LESIONS

The reference strain of porcine adenovirus type 4 was Although most of the descriptions of lesions associated
isolated from the brain of a pig that showed anorexia, en- with porcine adenoviruses are based on experimental in-
teritis, incoordination, muscle twitching, and frequent fections, there are several reports of adenovirus-associat-
recumbency (Kasza 1966); other strains of the same ed lesions in field material. Porcine adenovirus type 4
serotype have been isolated from pigs with respiratory or was isolated from the brain of a pig that showed lesions
gastrointestinal disease (Genov and Bodon 1976). Other of encephalitis, including perivascular cellular infiltra-
serotypes have been isolated from piglets with diarrhea tion and microglial nodule formation (Kasza 1966).
(Coussement et al. 1981), but adenoviruses have also Rondhuis (1972) isolated a strain of the same serotype
been isolated fairly frequently from the feces of clinically from pneumonic lung. Pavlov (1977) described kidney
normal piglets. The only consistent clinical sign in lesions in pigs naturally infected with adenovirus. The le-
piglets infected experimentally with porcine adenovirus sions involved dystrophy of the kidney tubules and cap-
type 4 was diarrhea (Shadduck et al. 1967), and infec- illary dilatation, which gave the gross appearance of pe-
tions with other strains have also produced diarrhea ex- techiation. Nuclear inclusion bodies have been found in
perimentally (Coussement et al. 1981). Although experi- the enterocytes of the terminal jejunum and ileum in
mentally infected piglets may develop lesions in the natural infections (Ducatelle et al. 1982; Sanford and
brain, lungs, and kidneys, clinical signs associated with Hoover 1983). Transplacentally infected piglets showed
these lesions have not been described. In a study of two vascular lesions, with nuclear inclusions in the endothe-
herds with respiratory disease, Watt (1978) concluded lial cells (Narita et al. 1985).
that porcine adenovirus type 4 may have been directly as- In experimentally infected piglets, gross lesions have
sociated with the disease, and Kirkbride and McAdaragh been described only in the lungs. These consisted of ar-
(1978) reported the isolation of adenovirus from a small eas of atelectasis of variable extent (Shadduck et al.
proportion of abortions in swine. Narita et al. (1985) 1967). Microscopically, the lungs showed interstitial
found skin cyanosis and subcutaneous edema in new- pneumonia characterized by thickened alveolar septa
born piglets with natural transplacental adenovirus in- due to proliferated septal cells, some of which contained
fections. inclusion bodies, infiltrating lymphocytes, plasma cells,
and histiocytes. The histologic lesion produced most
PATHOGENESIS consistently with porcine adenovirus type 4 was a severe
peritubular infiltration in the kidney, although menin-
Natural infection with porcine adenoviruses is acquired goencephalitis, characterized by focal accumulations of
by ingestion or inhalation. Experimentally, intranasal in- microglia and oligodendrocytes within the brain sub-
oculation of virus was the most effective route of infec- stance and perivascular lymphocytic cuffing, was also
tion in terms of the widespread distribution of virus and produced by this virus (Edington et al. 1972). Brack et al.
production of lesions (Shadduck et al. 1967). Experi- (1969) produced meningoencephalitis experimentally
mental pathogenesis studies with various strains of ade- with porcine adenovirus types 1 and 3 and also described
novirus (Sharpe and Jessett 1967; Shadduck et al. 1968; chronic inflammatory changes in the liver, heart, pan-
Ducatelle et al. 1982) suggest that the primary sites of vi- creas, and adrenal. In a sequential study of the enteric le-
ral replication are the tonsil and lower small intestine. sions produced by porcine adenovirus type 3, Ducatelle
Spread of the infection to a variety of tissues, including et al. (1982) described stunting of the villi in the lower je-
the central nervous system, lung, heart, liver, kidney, and junum and ileum and demonstrated infected enterocytes
spleen, was recorded. Viremia was detected in one piglet. by histology, immunoperoxidase staining, and electron
Viral excretion in the feces continued for several weeks microscopy.
after infection, and there was also evidence of persis-
tence of the virus in the kidney and other tissues. Some DIAGNOSIS
limited findings suggest that adenoviruses may play a
role in the pathogenesis of bacterial infections of the res- Clinical signs and gross lesions are minimal or absent in
piratory tract. Thus Kasza et al. (1969) found that porcine adenovirus infections. Histologically, nuclear in-
porcine adenovirus type 4 and Mycoplasma hyopneumo- clusion bodies in the lung, kidney, or intestinal epitheli-
niae produced a more severe experimental pneumonia um may be suggestive of adenovirus infection, but they
when inoculated together than either agent did alone. are not diagnostic unless viral antigen is demonstrated
However, Smith et al. (1973) found that the experimental by immunofluorescence or immunoperoxidase staining.
pneumonia produced by porcine adenovirus type 4 was A virologic diagnosis can best be made by isolation of
not enhanced by infection with Pasteurella septica. the virus from infected tissue, which can be fairly readily
CHAPTER 4 ADENOVIRUS Derbyshire 91

accomplished in porcine cell cultures. Some strains of Derbyshire, J. B., and Arkell, S. 1971. The activity of
the virus require several blind passages in cell culture be- some chemical disinfectants against Talfan virus and
fore CPE are evident. Stained coverslip preparations of porcine adenovirus type 2. Br Vet J 127:137–142.
the infected cells show nuclear inclusion bodies, and the Derbyshire, J. B., and Brown, E. G. 1979. The inactivation
virions may be demonstrated by negative staining of of viruses in cattle and pig slurry by aeration or treat-
lysates of the infected cells. The virus can also be identi- ment with calcium hydroxide. J Hyg 82:293–299.
fied by specific immunofluorescence of the infected cells Derbyshire, J. B., and Collins, A. P. 1971. Virological
(Dea and Elazhary 1984b). Serologic typing of the isolat- studies on an experimental minimal disease herd of
ed virus may be attempted by VN if reference antisera pigs. Br Vet J 127:436–441.
are available. Serologic diagnosis of suspected aden- Derbyshire, J. B.; Clarke, M. C.; and Jessett, D. M. 1966.
ovirus infections in swine may be attempted by the Observations on the faecal excretion of adenoviruses
demonstration of rising titers of antibody in VN or im- and enteroviruses in conventional and “minimal dis-
munodiffusion tests or by an indirect fluorescent anti- ease” pigs. Vet Rec 79:595–599.
body test (Dea and Elazhary 1984b). Derbyshire, J. B.; Clarke, M. C.; and Collins, A. P. 1975.
Serological and pathogenicity studies with some un-
TREATMENT AND PREVENTION classified porcine adenoviruses. J Comp Pathol
85:437–443.
No specific antiviral treatment is available. Porcine aden- Ducatelle, R.; Coussement, W. ; and Hoorens, J. 1982. Se-
ovirus infections have not been shown to be of sufficient quential pathological study of porcine adenovirus en-
economic importance to justify the development of vac- teritis. Vet Pathol 19:179–189.
cines, although adenovirus vaccines have been used suc- Edington, N.; Kasza, L.; and Christofinis, G. J. 1972.
cessfully in other species. Repopulation of herds with Meningoencephalitis in gnotobiotic pigs inoculated
specific-pathogen-free stock does not appear to be a reli- intranasally and orally with porcine adenovirus 4.
able means of excluding infection with porcine aden- Res Vet Sci 13:289–291.
oviruses (Derbyshire and Collins 1971). Genov, I., and Bodon, L. 1976. Vurkhu tipiziraneto i
razprostranenieto na adenovirus i po svine. Vet Med
REFERENCES Nauki 13:32–88.
Haig, D. A.; Clarke, M. C.; and Pereira, M. S. 1964. Isola-
Benko, M.; Harrach, B.; and D’Halluin, J.-C. 1990. Mole- tion of an adenovirus from a pig. J Comp Pathol
cular cloning and physical mapping of the DNA of 74:81–84.
bovine adenovirus serotype 4; study of the DNA ho- Kasza, L. 1966. Isolation of an adenovirus from the brain
mology among bovine, human and porcine aden- of a pig. Am J Vet Res 27:751–758.
oviruses. J Gen Virol 71:465–469. Kasza, L.; Hodges, R. T.; Betts, A. O.; and Trexler, P. C.
Betts, A. O.; Jennings, A. R.; Lamont, P. H.; and Page, Z. 1969. Pneumonia in gnotobiotic pigs produced by si-
1962. Inoculation of pigs with adenoviruses of man. multaneous inoculation of a swine adenovirus and
Nature 193:45–46. Mycoplasma hyopneumoniae. Vet Rec 84:262–267.
Bibrack, B. 1974. Vorkommen und Verbreitung von klin- Kirkbride, C. A., and McAdaragh, J. P. 1978. Infectious
isch inapparenten Virusinfektionen beim Schwein in agents associated with fetal and early neonatal death
der Bundesrepublik Deutschland. Zentralbl Veter- and abortion in swine. J Am Vet Med Assoc
inärmed (B)20:193–196. 172:480–483.
Brack, M.; Bernhardt, D.; Liess, B.; Bahr, H.; Rohde, G.; Narita, M.; Imada, T.; and Fukosho, A. 1985. Pathologic
and Amtsberg, G. 1969. Untersuchungen über patho- changes caused by transplacental infection with an
gene Eigenschaften von Adenovirustammen des adenovirus-like agent in pigs. Am J Vet Res
Schweines. Zentralbl Veterinärmed (B)16:671–688. 46:1126–1129.
Chandler, R. L. 1965. Demonstration of a porcine aden- Pavlov, N. 1977. Morfologichni promeni v vuvretsite na
ovirus by electron microscopy. Virology 25:143–145. praseta pri adenovirusna infektsiya. Vet Med Nauki
Coussement, W.; Ducatelle, R.; Charlier, G.; and 14:21–25.
Hoorens, J. 1981. Adenovirus enteritis in pigs. Am J Rondhuis, P. R. 1972. Some physicochemical and biolog-
Vet Res 42:1905–1911. ical characteristics of an adenovirus isolated from a
Dea, S., and Elazhary, M. A. S. Y. 1984a. Cultivation of a pig in the Netherlands. Tijdschr Diergeneeskd
porcine adenovirus in porcine thyroid cell cultures. 97:841–851.
Cornell Vet 74:208–217. Sanford, S. E., and Hoover, D. M. 1983. Enteric aden-
———. 1984b. Prevalence of antibodies to porcine ade- ovirus infection in piglets. Can J Comp Med
novirus in swine by indirect fluorescent antibody 47:396–400.
test. Am J Vet Res 45:2109–2112. Shadduck, J. A.; Koestner, A.; and Kasza, L. 1967. The le-
92 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

sions of porcine adenoviral infection in germfree and ovirus. J Comp Pathol 77:45–50.
pathogen-free pigs. Pathol Vet 4:537–552. Smith, I. M.; Betts, A. O.; Watt, R. G.; and Hayward, A.
Shadduck, J. A.; Kasza, L.; and Koestner, A. 1968. Distri- H. S. 1973. Experimental infections with Pasteurella
bution of a porcine adenovirus after inoculation of septica (serogroup A) and an adeno- or enterovirus in
experimental animals. Zentralbl Bakteriol (Orig gnotobiotic piglets. J Comp Pathol 83:1–12.
B)207:152–157. Watt, R. G. 1978. Virological study of two commercial
Sharpe, H. B. A., and Jessett, D. M. 1967. Experimental pig herds with respiratory disease. Res Vet Sci
infection of pigs with two strains of porcine aden- 24:147–153.
African Swine Fever
5 J. M. Sanchez-Vizcaino

African swine fever (ASF) was first described by Mont- carrier pigs are the most important source for virus dis-
gomery in Kenya in 1921. It was subsequently shown to semination thereafter.
be caused by an unusual virus, ASF virus (ASFV), that ASF is currently endemic in Sardinia and in parts
under natural conditions infects only porcine species of Africa, particularly in many sub-Saharan coun-
and whose properties differ markedly from those of any tries. Although ASF has appeared elsewhere in the
other virus of veterinary importance (Murphy et al. world at one time or another, these areas are now ASF-
1995). Depending on the circumstances, the clinical re- free.
sponse to infection with ASFV ranges from peracute to There is no treatment or effective vaccine available
inapparent. The acute form is most dramatic in regard to for ASF, and therefore its control is based on a rapid lab-
signs and lesions and is characterized by high fever, high oratory diagnosis and the enforcement of strict sanitary
mortality, extensive hemorrhages, pulmonary edema, measures.
and extensive necrosis of lymphoid tissue (Mebus 1988;
Gomez-Villamandos et al. 1995). Clinically and patho- ETIOLOGY
logically, ASF can resemble several other diseases of pigs
that sometimes present similar signs and lesions, espe- ASFV is a complex, icosahedral, DNA virus classified as
cially hog cholera and erysipelas. Therefore, laboratory the only member of an unnamed floating genus (Mur-
tests are required to establish a definitive diagnosis phy et al. 1995).
(Sanchez-Vizcaino 1986). ASFV replicates primarily in
mononuclear phagocytic cells (Malmquist and Hay Size and Structure
1960; Minguez et al. 1988). However, it also replicates in Virus particles have an average diameter of 200 nm
endothelial cells (Wilkinson and Wardley 1978), hepato- (Breese and De Boer 1966). The architecture of individ-
cytes (Sierra et al. 1987), renal tubular epithelial cells ual particles (virions) includes several concentric struc-
(Gomez-Villamandos et al. 1995), and neutrophils tures with an external hexagonal membrane (Fig. 5.1)
(Casals et al. 1984; Carrasco et al. 1996). that is acquired during budding through the cell mem-
Immunologically speaking, ASF is characterized by brane (Carrascosa et al. 1984). The ASFV genome is a
lymphopenia due to apoptosis of lymphocytes mainly in double-stranded, linear DNA molecule that, depending
the T area of the lymphoid organs (Carrasco et al. 1996), on the particular strain (Blasco et al. 1989), comprises
but there is no evidence of virus replication in T or B cells between 170 and 190 kilobases (kb). It is further charac-
(Minguez et al. 1988; Gomez-Villamandos et al. 1995). terized by terminal inverted repeats (Sogo et al. 1984), a
Humoral and cell-mediated immunity do not seem to be conserved central region of about 125 kb pairs (kbp),
affected (De Boer 1967). However, the mechanism of and variable ends (Blasco et al. 1989). Recently, the com-
protection is poorly understood. plete DNA sequence of the BA71v strain of ASFV was
Inapparent infections with ASFV are common in two analyzed and found to comprise 170,101 nucleotides and
species of wild boars: warthogs (Phacochoerus aethiopi- 151 open reading frames and to encode five multigene
cus) and bushpigs (Potamochoerus porcus). Both act as families (Yañez et al. 1995).
reservoir hosts in Africa (De Tray 1957; Heuschele and ASFV is a very complex virus. At least 28 structural
Coggins 1965). Soft ticks have been shown to be both proteins have been identified in intracellular virus parti-
reservoirs and vectors of ASFV, especially Ornithodoros cles (Tabares et al. 1980), and more than 100 virus-in-
moubata (Plowright et al. 1970) and O. erraticus (Sanchez duced proteins have been identified in infected porcine
Botija 1963a). macrophages. At least 50 of the latter react with sera
From the epidemiological point of view it is believed from infected or recovered pigs (Alcaraz et al. 1992), and
that the entrance of ASFV into a free area is related main- 40 are incorporated into the viral particle (Carrascosa et
ly to feeding swine uncooked, infected pork products al. 1985). Some of these proteins, such as p73, p54, p30,
from international airports and ports. However, once and p12, are very antigenic. Although their role in in-
ASF is established in domesticated swine, infected and ducing protective immunity is still unresolved, they are

93
94 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

5.1. Electron micrograph of an


ASF virus particle.

very good antigens and are used in ASF serodiagnosis Tomé, and Príncipe. In 1957, ASF was detected for the
(Arias and Sanchez-Vizcaino 1992). first time outside the African continent. It appeared in
Lisbon, Portugal, as a peracute form with a mortality of
Virus Replication and Persistence almost 100% (Manso Ribeiro et al. 1963). In 1960 it reap-
In an infected pig, ASFV replicates primarily in mono- peared near Lisbon, apparently as a new outbreak, and it
cytes and macrophages (Malmquist and Hay 1960; spread through the rest of Portugal and to Spain the
Minguez et al. 1988; Mebus 1988). However, ASFV also same year (Polo Jover and Sanchez Botija 1961). ASF re-
replicates in endothelial cells (Wilkinson and Wardley mained endemic in Portugal and Spain until 1995, when,
1978), hepatocytes (Sierra et al. 1987), renal tubular ep- as a result of an intensive eradication program, both
ithelial cells (Gomez-Villamandos et al. 1995) and neu- countries were declared ASF-free. In 1978, ASF again ap-
trophils (Casals et al. 1984; Carrasco et al. 1996). No in- peared outside Africa, namely, in Malta, Sardinia, Brazil,
fection has been observed in lymphocytes (Minguez et and the Dominican Republic. In 1979 it appeared in
al. 1988; Gomez-Villamandos et al. 1995). In nature, AS- Haiti and 1980 it appeared in Cuba. Today, ASF is pres-
FV also replicates in some soft ticks, principally O. ent only in Africa, mainly in sub-Saharan countries, and
moubata (Plowright et al. 1970) and O. erraticus (Sanchez in Sardinia. Elsewhere it has been successfully eradicat-
Botija 1963a). ASFV has been adapted to grow in a large ed.
number of stable cell lines as well, such as VERO, MS,
and CV (Hess et al. 1965). ASFV is very resistant to inac- Reservoirs and Susceptible Animals
tivation in environmental conditions such as tempera- Several different species of soft ticks have been shown to
ture changes and acid pH. It can be isolated from serum be ASFV reservoirs and vectors, including O. moubata in
or blood kept at room temperature for 18 months. How- Africa (Plowright et al. 1969) and O. erraticus in the Iber-
ever, it is inactivated by heat treatment at 60˚C for 30 ian Peninsula (Sanchez Botija 1963a). Transovarial and
minutes (Plowright and Parker 1967) and by many lipid transstadial transmission of ASFV has been described
solvents and commercial disinfectants. for O. moubata (Plowright et al. 1970). More recently, a
ASFV may persist for several weeks or months in number of other tick species widely distributed in North
frozen or uncooked meat. In meat products such as Par- and South America have been identified as harboring
ma ham, viral infectivity was not demonstrated after 300 and transmitting ASFV (Groocock et al. 1980), and a new
days of processing and curing (McKercher et al. 1987). species of soft ticks, O. Savignyi, which is present in
In typical Spanish dry-cured meat products, such as Ser- Africa, can experimentally transmit ASFV to domesticat-
rano ham and Iberian ham and shoulder, the virus sur- ed pigs (Mellor and Wilkinson 1985).
vives for 140 days in Iberian and Serrano hams and Pigs are the only domesticated animals that are natu-
for 112 days in loin (Mebus et al. 1993). No infectious rally infected by ASFV. Wild boars also are susceptible to
ASFV was found in cooked or canned hams heated at ASFV infection, with clinical signs and mortality rates
70˚C. similar to those observed in naturally infected domesti-
cated pigs in Spain and Portugal (Sanchez Botija 1982)
EPIDEMIOLOGY and in Sardinia (Contini et al. 1981) and in experimen-
tally infected feral pigs in Florida (McVicar et al. 1981).
History and Distribution Wild boars can transmit the disease directly to domesti-
ASF was described for the first time in Kenya by Mont- cated pigs, as well as to one another. In Africa, it has been
gomery in 1921. In this case the virus had spread from in- observed that ASFV induces an inapparent infection in
fected warthogs (Phacochoerus aethiopicus) to domesti- two species of wild boars, warthogs (Phacochoerus
cated pigs (Sus scrofa), causing 100% mortality. Since aethiopicus) and bushpigs (Potamochoerus porcus).
then, ASF has been recognized, and is currently endem-
ic, in many African countries, namely, Angola, Zimbab- ASFV Transmission
we, Sudan, Republic of South Africa, Mozambique, São ASFV is maintained in Africa by a cycle of infection be-
CHAPTER 5 AFRICAN SWINE FEVER Sanchez-Vizcaino 95

tween wild boars and soft ticks. In some of these wild against ASFV have been shown to delay the onset of ASF
boars infection is characterized by low levels of virus in clinical signs, to reduce the levels of viremia, and to pro-
the tissues and low or undetectable levels of viremia tect pigs against the potentially fatal consequence of in-
(Plowright 1981); however, even these low levels of ASFV fection (Schalafer et al. 1984; Onisk et al. 1994). Early ex-
are enough for its transmission to domesticated pigs via periments demonstrated the absence of neutralizing
tick vectors. This transmission cycle makes it very diffi- antibodies against ASFV in sera from naturally or exper-
cult to eradicate ASF in Africa. imentally infected pigs. Yet, recovered pigs produce a
In Sardinia, where ASFV is still present, neither inap- normal level of neutralizing antibodies in response to
parent infection of wild or domesticated pigs nor soft foot-and-mouth disease virus vaccine, an event suggest-
tick populations have been observed. Wild pigs are as ing that humoral responses are not adversely affected by
susceptible as domesticated pigs, and their role in the ASFV (De Boer 1967). Other authors (Ruiz Gonzalvo et
epidemiology of ASF is similar to that of domesticated al. 1986) have demonstrated that different ASFV isolates
pigs. However, in Sardinia, recovered, ASFV carrier, do- are largely neutralized by convalescent porcine sera;
mesticated pigs have been recognized, and their role in however, a persistent 10% fraction of nonneutralized
the epidemiology of ASF is a major consideration in de- virus remained. More recently, Gomez-Puertas et al.
signing a strategy for ASF eradication. Notably, the sero- (1996) reported that ASFV-induced antibodies in serum
logical recognition of carrier pigs was an important facet collected from a convalescent pig effectively neutralized
of the successful ASF eradication program in Spain ASFV before and after it was bound to susceptible cells.
(Arias and Sanchez-Vizcaino 1992). Due to the lack of However, ASFV-specific antibodies have never been
neutralizing antibodies in ASF, the correlation between a demonstrated to entirely fulfill the classic definition of
new outbreak and the possible source of virus strain has virus neutralization. On the other hand, T cytotoxic lym-
been very difficult to establish. Even the use of the phocytes from recovered pigs can destroy infected
hemadsorption inhibition reaction (Malmquist 1963) is macrophages (Martins and Leitao 1994)—suggesting
not conclusive. New studies on molecular epidemiology that cell-mediated immunity may be an important com-
comparing the patterns of DNA from different ASFV iso- ponent of the protective response. However, the relative
lates appear most promising, especially for differentiat- roles of antibodies and cell-mediated immunity in pro-
ing African and European ASFV strains (Blasco et al. tection against ASF are still not well understood.
1989). However, it is epidemiologically well established
that the entrance of ASFV into an ASFV-free country is CLINICAL SIGNS
related to the introduction of uncooked infected pork
through international ports or airports where garbage Clinically, ASF can resemble several other diseases of
containing uncooked pork can be found and used for pig pigs that often have similar signs, especially hog cholera
feeding. Once ASF is established in domesticated pigs, and erysipelas. Therefore, laboratory tests are required
ASFV carrier pigs are the most important source for sub- to establish a definitive diagnosis (Sanchez-Vizcaino
sequent virus dissemination. 1986). Moreover, ASF can present different clinical signs,
depending mostly on virus virulence, infectious dose,
IMMUNOLOGY and route of exposure. The clinical forms of ASF range
from peracute (i.e., sudden death with few, if any, previ-
The immune mechanisms involved in protection against ous clinical signs) to subclinical or inapparent. In Africa
ASF are still poorly understood, and all attempts to de- ASF appears mostly as an acute disease characterized by
velop an effective vaccine have been unsuccessful. The loss of appetite, high temperature (40–41˚C), leukope-
difficulty in inducing an effective immunity may be relat- nia, hemorrhages in internal organs, hemorrhages in the
ed to the great variability observed among different AS- skin (especially in the skin of the ears and flanks), and
FV isolates. It may also be related to the fact that ASFV high mortality (Moulton and Coggins 1968; Mebus et al.
replicates in some cells typically involved in the immune 1983).
response. Although there is no evidence for replication of Outside Africa it is also possible to see an acute ASF
ASFV in either T or B cells (Minguez et al. 1988; Gomez- outbreak; however, subacute or chronic forms of the dis-
Villamandos et al. 1995), it does replicate in monocytes ease are more common. The subacute form is character-
and macrophages. If immune suppression has a role, ized by transitory thrombocytopenia and leukopenia
however, it is not obvious. ASFV is highly antigenic, and and numerous hemorrhagic lesions (Gomez-Villaman-
high titers of antibody are detected by in vitro testing of dos et al. 1997). The chronic form is characterized by res-
sera collected from pigs after exposure to the virus. IgM piratory alteration, abortion, and low mortality (Arias et
and IgG were detected in sera collected at 4 days and at al. 1986).
6–8 days after exposure, respectively (Hortiguela and The incubation periods in natural infection vary from
Sanchez-Vizcaino 1984), and high titers of antibody as short as 4 days to as long as 19 days. In experimental
were detected for a long period of time. Antibodies infections, the incubation period is generally shorter and
96 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

varies from 2 to 5 days depending on the virus dose and thrombocytopenia (Gomez-Villamandos et al. 1996).
the route of inoculation (Mebus et al. 1983). The alveolar edema observed in the last stages of the
acute and subacute forms of ASF (and the main cause of
PATHOGENESIS death) is a consequence of activation of pulmonary in-
travascular macrophages (Sierra et al. 1990; Carrasco et
Generally ASFV is spread in domesticated pigs via oral al. 1996).
and nasal routes of dissemination and exposure (Col-
grove et al. 1969; Plowright et al. 1968). However, pigs al- LESIONS
so can be infected by a number of other routes, including
tick bites (Plowright et al. 1969), cutaneous scarification, A wide variety of lesions have been observed in ASF, de-
and intramuscular, intravenous, subcutaneous, and in- pending on the virus virulence. The acute and subacute
traperitoneal injection (McVicar 1984). forms are characterized by extensive hemorrhages and
Infection usually starts in monocytes and lymphoid tissue destruction. Conversely, lesions may be
macrophages of tonsils and mandibular lymph nodes minimal or absent in the subclinical and chronic forms
and then spreads through the draining lymph nodes and (Mebus et al. 1983; Gomez-Villamandos et al. 1995).
blood to the target organs (lymph nodes, bone marrow,
spleen, lung, liver, and kidney), which are the main sites Gross Lesions
of secondary replication. The viremia in ASF starts be- The main gross lesions are in spleen, lymph nodes, kid-
tween 6 and 8 days after infection and lasts for a long neys, and heart (Sanchez Botija 1982). The spleen may
time thereafter. be darkened, enlarged, infarcted, and friable (Fig. 5.2).
ASFV is associated with red blood cell membranes Sometimes lesions are large infarcts with subcapsular he-
(Quintero et al. 1986) and with platelets (Gomez-Villa- morrhages. Lymph nodes are hemorrhagic, edematous,
mandos et al. 1996) and causes hemadsorption (HA) in and friable (Fig. 5.3). They often look like dark-red
affected pigs (Sierra et al. 1991). The pathogenesis of he- hematomas. Because of congestion and subcapsular he-
morrhages observed in the acute form is believed to be morrhage, cut sections of affected lymph nodes some-
phagocytic activation of endothelial cells aggravated by times have a marmoreal appearance. Kidneys usually
virus replication in the same cells in the final stages of have petechial hemorrhages on the cortical (Fig. 5.4) and
the disease. In the subacute form hemorrhages are due cut surfaces, as well as in the renal pelvis. An intense hy-
mainly to an increase of vascular permeability (Gomez- dropericardium with serohemorrhagic fluid is present in
Villamandos et al. 1995). The pathogenesis of the lym- some cases. Petechial and ecchymotic hemorrhages can
phopenia in the acute form has been related to apoptosis be observed in epicardium and endocardium. Other le-
of lymphocytes mainly on the T area of lymphoid organs sions can also be observed in acute ASF, such as serohe-
(Carrasco et al. 1996), but there is no evidence of virus morrhagic fluid in the abdominal cavity, with edema and
replications in T or B cells (Minguez et al. 1988; Gomez- hemorrhages throughout the alimentary tract. Conges-
Villamandos et al. 1995). tion of the liver and the gall bladder can be observed, as
The subacute form is characterized by a transitory well as petechial hemorrhages in the mucosa of the uri-

5.2. Enlarged and darkened


spleen from acute ASF.
CHAPTER 5 AFRICAN SWINE FEVER Sanchez-Vizcaino 97

5.3. Lymph nodes from a nor-


mal pig (left), a pig with subacute
ASF (center), and a pig with acute
ASF (right).

5.4. Kidney from a pig with


acute ASF showing numerous
petechiae on the cortical surface.

nary bladder. Hydrothorax and petechial hemorrhages macrophages (Carrasco et al. 1997). The lymphoid tissue
of the pleura are frequently found in the thoracic cavity, destruction in the acute form is mainly observed on the
and lungs are usually edematous. Intense congestion is T area of the lymphoid organs, but no evidence of virus
observed in the meninges, chorioid plexus, and en- replications has been observed (Minguez et al. 1988 and
cephalon (Arias et al. 1986). Carrasco et al. 1996). The chronic form of ASF is charac-
For the last fifteen years, the most predominant form terized mainly by alterations in the respiratory tract.
of ASF outside Africa has been the subacute form, which These include fibrinous pleuritis, pleural adhesions,
is similar to the acute form except for milder lesions. The caseous pneumonia, and hyperplasia of the lymphoretic-
subacute form is characterized by large hemorrhages in ular tissues. Fibrinous pericarditis and necrotic skin le-
lymph nodes and kidneys. The spleen is enlarged and he- sions are also common (Moulton and Coggins 1968;
morrhagic. Congestion and edema can be observed in Arias et al. 1986).
lungs and in some cases an interstitial pneumonia has
been found (Arias et al. 1986). DIAGNOSIS

Microscopic Lesions Laboratory studies are essential to establish a definitive


In the acute form of ASF histopathological lesions are diagnosis of ASF due to the great similarity of ASF clini-
present in blood vessels and in lymphoid organs. These cal signs and lesions with those of other hemorrhagic pig
lesions are characterized by hemorrhages, microthrom- diseases (e.g., hog cholera, erysipelas, and septicemic sal-
bosis, and damage of the endothelial cells with accumu- monellosis). As in other virus diseases, the laboratory di-
lations of dead cells in the subendothelium (Gomez-Vil- agnosis of ASF can be based on the demonstration of in-
lamandos et al. 1995). Hemorrhagic splenomegaly, fectious virus, viral antigens, viral DNA, or specific
characteristic of the acute and subacute forms, is a con- antibodies. A wide variety of laboratory tests are avail-
sequence of a loss of splenic architecture caused by the able for detecting either ASFV or homologous antibodies
viral replication and resulting necrosis of splenic fixed (Sanchez-Vizcaino 1986).
98 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

ASFV Detection However, even though HA is the most sensitive test


Several techniques have been adapted for the identifica- for ASFV identification, it is important to point out that
tion of ASFV. Some of them, such as the complement fix- a few field strains of ASFV have been isolated that induce
ation test (Cowan 1961), immunoperoxidase staining cytopathic effects in macrophages but do not induce HA
(Pan et al. 1978), enzyme-linked immunosorbent assay (Sanchez Botija 1982). These strains were identified us-
(ELISA) sandwich (Wardley and Wilkinson 1980), elec- ing DIF sediments of these cell cultures.
tron microscopy, and, more recently, DNA-hybridization The exposure of susceptible pigs to tissues from sus-
methods (Pastor and Escribano 1990), are, for one rea- pected cases of ASF may be used as an additional testing
son or another, not practical for routine diagnosis procedure when the recognition of ASF is especially crit-
(Sanchez-Vizcaino 1986). ical (e.g., the possible introduction of ASFV into an AS-
At present, the most convenient, safe, and frequently FV-free area). Two groups of pigs are necessary: one vac-
used techniques are direct immunofluorescence (DIF) cinated against hog cholera and one not vaccinated and
(Bool et al. 1969) and HA (Malmquist and Hay 1960). susceptible to hog cholera (because ASF and hog cholera
DIF is the test recommended to be used first when ASF is are easily confused on the basis of clinical signs and le-
suspected. It is based on the demonstration of viral anti- sions). After the inoculation of both groups with suspect
gens in impression smears or frozen tissue sections from samples, they should be observed daily for clinical signs,
spleen, lung, lymph node, or kidney, reacted with a con- and blood samples should be collected and tested for AS-
jugated immunoglobulin against ASFV. It is a fast and FV by HA and for homologous antibody.
economical test with high sensitivity for the acute form
of ASF. However, for subacute or chronic forms, DIF has ASFV-DNA Detection
a sensitivity of only 40%. This decrease in sensitivity The detection of a wide range of ASFV isolates by poly-
seems to be related to the formation of antigen-antibody merase chain reaction (PCR) has been made possible by
complexes in tissues of an infected pig, which, in turn, using primers from a highly conserved region of the viral
block the reaction between ASFV antigens and the ASF genome. PCR has been used to identify ASFV isolates
conjugate when such tissues are tested in the laboratory from all of the known virus genotypes, including those of
(Sanchez-Vizcaino 1986). low virulence and those that fail to induce HA. It has
HA, because of its sensitivity and specificity, is useful been particularly useful for identifying viral DNA in tis-
under the widest range of circumstances. It should be sues unsuitable for other diagnostic tests (e.g., tissues in
performed to confirm any new outbreak, and especially which the virus had been inactivated or further degraded
when other tests are negative. In the laboratory the HA by tissue changes such as putrefaction). It is an excellent
test is based on the attachment of erythrocytes to the ex- and rapid technique to be included in ASF diagnostic
ternal (cytoplasmic) membrane of ASFV-infected testing.
porcine macrophages cultured in vitro. Typically the
erythrocytes form a rosette around the infected Antibody Detection
macrophage before the appearance of ASFV-induced cy- The study of antibodies to ASFV is especially important
topathic effects (Fig. 5.5) (Malmquist and Hay 1960). for two main reasons. First, a better understanding of

5.5. Hemadsorption phenome-


non: the attachment of erythro-
cytes to macrophages infected with
ASFV.
CHAPTER 5 AFRICAN SWINE FEVER Sanchez-Vizcaino 99

the humoral immune response may contribute to the de- but some of these pigs become carriers and develop
velopment of an efficacious vaccine. Second, detection of chronic lesions, the likelihood of which increases when
antibodies is a useful tool in the diagnosis of ASF. Spe- large numbers of pigs are vaccinated (Manso Ribeiro et
cific ASFV IgG is detectable in blood from the 6th to 8th al. 1963; Sanchez Botija 1963b). Other studies have
day after inoculation and for a long time, even years, shown that serum from pigs resistant to homologous
thereafter. The early appearance and subsequent persis- and some heterologous strains of ASFV inhibits (in vit-
tence of antibodies is the reason they are so useful in ro) infection of cells with different but related heterolo-
studying subacute and chronic forms of the disease. For gous strains (Ruiz Gonzalvo et al. 1986). However, ASFV-
the same reason they play an important role in testing specific antibodies have never been demonstrated to
strategies that are part of eradication programs (Arias neutralize virus in the classic concept of neutralization.
and Sanchez-Vizcaino 1992). The recent analysis of the complete nucleotide sequence
Several techniques have been adapted to ASF anti- of ASFV (Yañez et al. 1995) offers new opportunities to
body detection. These include complement fixation discover the role of various ASFV genes in protective im-
(Cowan 1961), indirect immunofluorescence (IIF) (Bool munity. However, the successful eradication of ASF from
et al. 1969), immunoelectroosmophoresis (IEOP) (Pan et Portugal and Spain proved that vaccine is not essential
al. 1972), ELISA (Sanchez-Vizcaino et al. 1979, 1982), ra- for an eradication program even where the disease is en-
dioimmunoassay (RIA) (Wardley and Wilkinson 1980), demic.
and immunoblotting assay (IB) (Pastor et al. 1987). IIF,
ELISA, and IB are the most frequently used. PREVENTION
IIF is a fast technique with high sensitivity and speci-
ficity for the detection of ASF antibodies from either sera Because ASF is a costly disease and because there are no
or tissue exudates (Sanchez Botija et al. 1970). It is based effective vaccines for its control, it is especially important
on the detection of ASF antibodies which bind to a that ASF-free areas be maintained free by preventing the
monolayer of cell lines infected with a cell culture adapt- introduction of ASFV. Retrospective epidemiological
ed ASF virus. The antibody-antigen reaction is detected studies have shown that the most frequent source of AS-
by a labeled fluorescein A-protein (Hortiguela and FV has been contaminated garbage from international
Sanchez-Vizcaino 1984). Using both IIF and DIF, it is airports or ports. Therefore, all food leftovers from
possible to detect from 85% to 95% of ASF cases (acute, planes and ships should be incinerated. In infected Euro-
subacute, and chronic) in less than 3 hours (Sanchez-Viz- pean areas such as Sardinia, where the disease is endem-
caino 1986). ic and where mild or inapparent clinical signs have been
At present, ELISA is the most useful method for large- recognized, the most important aspects of prevention
scale ASF serological studies. It is based on the detection are controlling pig movements and implementing exten-
of ASF antibodies bound to the viral proteins by addi- sive serological surveys to detect carrier pigs. In areas of
tion of protein A, which is conjugated with an enzyme Africa where ASF is endemic, the most important aspects
that produces a visible color reaction when it reacts with of prevention are controlling natural reservoirs, namely,
the appropriate substrate. soft ticks (O. moubata) and warthogs, and preventing
The IB test is a highly specific, sensitive, and easy-to- their contact with domesticated pigs. When ASF is sus-
interpret technique that has been used successfully as an pected for any reason, pig movement should be restrict-
alternative method to IIF for confirmation of question- ed and diagnostic tests should be performed immediate-
able ELISA results (Arias and Sanchez-Vizcaino 1992). ly. Moreover, it is important to remember that
Samples that should be collected for ASF laboratory low-virulence ASFV strains do not cause signs or lesions
diagnosis are lymph nodes, kidneys, spleen, lungs, that signal their presence.
blood, and serum. Tissues are used for virus isolation
(HA) and viral antigen detection (DIF), blood is used for REFERENCES
virus isolation (HA), and serum and tissue exudates are
used for antibody detection (IIF, ELISA, IB). Alcaraz, C.; Alvarez, A.; and Escribano, J. 1992. Flow cyto-
metric analysis of African swine fever virus induced
TREATMENT plasma membrane proteins and their humoral immune
response in infected pigs. Virology 189:266–273.
At present, no treatment or effective vaccine against AS- Arias, M., and Sanchez-Vizcaino, J. M. 1992. Manual de di-
FV is available. Since 1963, when the first live-attenuated agnostico serologico de la peste porcina africana. Mono-
vaccine was used in Portugal (Manso Ribeiro et al. 1963), grafias INIA 83:5–44.
many efforts have been made to develop a satisfactory Arias, M. L.; Escribano, J. M.; Rueda, A.; and Sanchez-Viz-
vaccine. Inactivated vaccine does not produce any pro- caino, J. M. 1986. La peste porcina africana. Med Vet
tection. Live-attenuated vaccine protects some pigs 3:333–350.
against challenge with the homologous strain of virus, Blasco, R.; Aguero, M.; Almendral, J. M.; and Viñuela, E.
100 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

1989. Variable and constant region in African swine fever. J Comp Pathol 59:146–151.
fever virus DNA. Virology 168:330–338. Gomez-Villamandos, J.; Bautista, M.; Carrasco, L.; Ca-
Bool, P. H.; Ordas, A.; and Sanchez Botija, C. 1969. The di- ballero, M.; Hervas, J.; Villeda, C.; Wilkinson, P.; and
agnosis of African swine fever by immunofluorescence. Sierra, M. 1997. African swine fever infection on bone
Bull Off Int Epizoot 72:819–839. marrow: Lesions and pathogenesis. Vet Res (in press).
Breese, S. S., and De Boer, C. J. 1966. Electron microscope Groocock, C. M.; Hess, W. R.; and Gladney, W. J. 1980. Ex-
observation of Africa swine fever virus in tissue culture perimental transmission of African swine fever virus by
cells. Virology 28:420–428. Ornithodoros coriaceus, an argasid tick indigenous to the
Carrasco, L.; Chacon, M.; Lara, J.; Martin, J.; Gomez, J.; Her- United States. Am J Vet Res 41:591–594.
vas, J.; Wilkinson, P.; and Sierra, M. 1996. Virus associa- Hess, W. R.; Cox, B. F.; Heuschele, W. P.; and Stone, S. S.
tion with lymphocytes in acute African swine fever. Vet 1965. Propagation and modification of African swine
Res 27:305–312. fever virus in cell cultures. Am J Vet Res 26:141–146.
Carrasco, L.; Bautista, J.; Gome, J.; Martin, J.; Chacon, L.; Heuschele, W. P., and Coggins, L. 1965. Isolation of African
and Sierra, M. 1997. Development of microscopic le- swine fever virus from a giant forest hog. Bull Epiz Dis
sions in splenic cords in pig with acute African swine Afr 13:255–256.
fever. Vet Res (in press). Hortiguela, O., and Sanchez-Vizcaino, J. M. 1984. Valo-
Carrascosa, J. L.; Carazo, J. M.; Carrascosa, A. L.; Garcia, N.; racion epidemiologica de la proteina A marcada con per-
Santisteban, A.; and Viñuela, E. 1984. General morphol- oxidasa en el enzimoinmunoensayo indirecto para la de-
ogy and capsid fine structure of African swine fever teccion de anticuerpos de peste porcina africana. Med
virus particles. Virology 132:160–172. Vet 5:269–274.
Carrascosa, A.; Val, M.; Santaren, J.; and Viñuela, E. 1985. Malmquist, W. A. 1963. Serologic and immunologic studies
Purification and properties of African swine fever virus. with African swine fever virus. Am J Vet Res 24:450–459.
J Virol 54:337–344. Malmquist, W. A., and Hay, D. 1960. Hemadsorption and
Casals, I.; Enjuanes, L.; and Viñuela, E. 1984. Porcine leuko- cytopathic effect produced by African swine fever virus
cyte cellular subsets sensitive to African swine fever in swine bone marrow and buffy coat cultures. Am J Vet
virus in vitro. J Virol 52:37–46. Res 21:104–108.
Colgrove, G.; Haelterman, E. O.; and Coggins, L. 1969. Manso Ribeiro, J.; Azevedo, R.; Teixeira, J.; Braco, M.;
Pathogenesis of African swine fever virus in young pigs. Rodriguez, A.; Oliveira, E.; Noronha, F.; Grave, C.; and
Am J Vet Res 30:1343–1359. Vigario, J. 1963. An atypical strain of swine fever virus in
Contini, A.; Cossu, P.; Rutili, D.; and Firinu, A. 1981. African Portugal. Bull Off Int Epizoot 50:516–534.
swine fever in Sardinia. In African Swine Fever, ed. P. J. Martins, C., and Leitao, A. 1994. Porcine immune respons-
Wilkinson, pp. 1–6. EUR 8466 EN, Proc CEC/FAO Re- es to African swine fever virus infection. Vet Immunol
search Seminar, Sardinia, Sept. Immunopathol 43:99–106.
Cowan, K. M. 1961. Complement fixation test for African McKercher, P. D.; Yedloutschnig, R. J.; Callis, J. J.; Murphy,
swine fever diagnosis. J Immunol 86:465–470. R.; Panina, G.; Civardi, A.; Bugnetti, M.; Foni, E.; Laddo-
De Boer, C. J. 1967. Studies to determine neutralizing anti- mada, A.; Scarano, C.; and Scatozza, F. 1987. Survival of
body in sera from animals recovered from African swine viruses in “Prosciutto di Parma” (Parma ham). Can Inst
fever and laboratory animals inoculated with African Food Sci Technol J 20:267–272.
swine fever virus with adjuvants. Arch Gesamte Virus- McVicar, J. W. 1984. Quantitative aspects of transmission of
forsch 20:164–179. African swine fever virus. Am J Vet Res 45:1535–1541.
De Tray, D. E. 1957. African swine fever in warthogs (Phaco- McVicar, J. W.; Mebus, C. A.; Becker, H. N.; Belden, R. C.;
choerus aethiopicus). J Am Vet Med Assoc 130:537–540. and Gibbs, E. P. J. 1981. Induced African swine fever
Gomez-Puertas, P.; Rodriguez, F.; Oviedo, J.; Ramiro, F.; virus in feral pigs. J Am Vet Med Assoc 179:441–446.
Ruiz, F.; Alonso, C.; and Escribano, J. 1996. Neutralizing Mebus, C. A. 1988. African swine fever. Adv Virus Res
antibodies to different proteins of African swine fever 35:251–269.
virus attachment and internalization. J Virol Mebus, C. A.; McVicar, J. W.; and Dardiri, A. H. 1983. Com-
70(8):5689–5694. parison of the pathology of high and low virulence
Gomez-Villamandos, J.; Hervas, J.; Mendez, A.; Carrasco, L.; African swine fever infections. In African Swine Fever,
Villeda, C.; Wilkinson, P.; and Sierra, M. 1995. Experi- ed. P. J. Wilkinson, pp. 183–194. EUR 8466 EN. Proc
mental African swine fever: Apoptosis of lymphocytes CEC/FAO Research Seminar, Sardinia, Sept. 1981.
and virus replication in other cells. J Gen Virol Mebus, C. A.; House, C.; Ruiz, F.; Pineda, J. M.; Tapiador, J.;
76:2399–2405. Pire, J.; Bergada, J.; Yedlontshning, R.; Sahu, S.; Becerra,
Gomez-Villamandos, J.; Bautista, M.; Hervas, J.; Carrasco, V.; and Sanchez-Vizcaino, J. M. 1993. Survival of foot
L.; Cahcon, F.; Perez, J.; and Sierra, M. 1996. Subcellular and mouth disease, African swine fever and hog cholera
changes in platelets in acute and subacute African swine virus in Spanish serrano cured hams and Iberian cured
CHAPTER 5 AFRICAN SWINE FEVER Sanchez-Vizcaino 101

hams, shoulder and loin. Food Microbiol 10:133–143. Quintero, J.; Wesley, R. D.; Whyard, T. C.; Gregg, D.; and
Mellor, P. S., and Wilkinson, P. 1985. Experimental trans- Mebus, C. A. 1986. In vitro and in vivo association of
mission of African swine fever virus by Ornithodoros sav- African swine fever virus with swine erythrocytes. Am J
ignyi (Audouin). Res Vet Sci 39:353–356. Vet Res 47:1125–1131.
Minguez, I.; Rueda, A.; Dominguez, J.; and Sanchez-Viz- Ruiz Gonzalvo, F.; Carnero, M. E.; and Caballero, C. 1986.
caino, J. M. 1988. Double labeling immunohistological Inhibition of African swine fever infection in the pres-
study of African swine fever virus infected spleen and ence of immune sera in vivo and in vitro. Am J Vet Res
lymph nodes. Vet Pathol 25:193–198. 47:1249–1252.
Montgomery, R. E. 1921. On a form of swine fever ocurring Sanchez Botija, C. 1963a. Reservorios del virus de la peste
in British East Africa (Kenya Colony). J Comp Pathol porcina africana: Investigacion del virus de la PPA en los
34:159–191. artropodos mediante la prueba de la hemoadsorcion.
Moulton, J., and Coggins, L. 1968. Comparison of lesions in Bull Off Int Epizoot 60:895–899.
acute and chronic African swine fever. Cornell Vet ———. 1963b. Modificacion del virus de la peste porcina
58:364–388. africana en cultivos celulares: Contribucion al
Murphy, F.; Fauquet, C.; Bishop, D.; Ghabrial, S.; Harvis, A.; conocimiento de la accion patogena y del poder de pro-
Martinelli, G.; Mayo, M.; and Summer, M. 1995. Virus teccion de las estirpes atenuadas. Bull Off Int Epizoot
Taxonomy. Sixth report of Int Committee on Taxonomy 60:901–919.
of Viruses. Archives Virol Suppl 10. ———. 1982. African swine fever: New developments. Rev
Onisk, D.; Borca, M.; Kutish, G.; Kramer, P.; Irusta, P.; and Sci Technol Off Int Epizoot 1:1065–1094.
Rock, D. L. 1994. Passively transferred African swine Sanchez Botija, C.; Ordas, A.; and Gonzalez, J. 1970. La in-
fever virus antibodies protect swine against lethal infec- munofluorescencia indirecta aplicada a la investigacion
tion. Virology 198:350–354. de anticuerpos de la peste porcina africana: Su valor
Pan, I. C.; De Boer, C. J.; and Hess, W. R. 1972. African swine para el diagnostico. Bull Off Int Epizoot 74:397–417.
fever: Application of immunoelectroosmophoresis for Sanchez-Vizcaino, J. M. 1986. Africa swine fever diagnosis.
the detection of antibody. Can J Comp Med 36:309–316. In African Swine Fever, ed. J. Becker, pp. 63–71. Boston:
Pan, I. C.; Shimizu, M.; and Hess, W. R. 1978. African swine Martinus Nijhoff.
fever: Microplaque assay by an immunoperoxidase Sanchez-Vizcaino, J. M.; Martin, L.; and Ordas, A. 1979.
method. Am J Vet Res 39:491–497. Adaptacion y evaluacion del enzimoinmunoensayo para
Pastor, M. J., and Escribano, J. M. 1990. Evaluation of sensi- la deteccion de anticuepos para la peste porcina
tivity of different antigen and DNA-hybridization meth- africana. Laboratorio 67:311–319.
ods in African swine fever virus detection. J Virol Meth Sanchez-Vizcaino, J. M.; Tabares, E.; Salvador, E.; and Or-
28:67–78. das, A. 1982. Comparative studies of two antigens for
Pastor, M. J.; Laviada, M. D.; Sanchez-Vizcaino, J. M.; and use in the indirect ELISA test for the detection of ASF
Escribano, J. M. 1987. Detection of African swine fever antibodies. In African Swine Fever, ed. P. J. Wilkinson,
virus antibodies by immunoblotting assay. Can J Vet Res pp. 195–325. EUR 8466 EN. Proc CEC/FAO Research
53:105–107. Seminar, Sardinia, Sept. 1981.
Plowright, W. 1981. African swine fever. In Infectious Dis- Schalafer, D.; Mebus, C.; and McVicar, W. 1984. African
eases of Wild Mammals, 2d ed. Ed. J. W. Davis, L. H. swine fever in neonatal pigs: Passive acquired protection
Karstand, and D. O. Trainer, pp. 178–190. Ames: Iowa from colostrum or serum from recovered pigs. Am J Vet
State Univ Press. Res 45:1367–1372.
Plowright, W., and Parker, J. 1967. The stability of African Sierra, M.; Bernabe, A.; Mozos, E.; Mendez, A.; and Jover, A.
swine fever virus with particular reference to heat and 1987. Ultrastructure of the liver in pigs with experimen-
pH inactivation. Arch Gesamte Virusforsch 21:383–402. tal African swine fever. Vet Pathol 24:460–462.
Plowright, W.; Parker, J.; and Staple, R. F. G. 1968. The Sierra, M.; Carrasco, L.; Gomez, J.; Martin, J.; Mendez, A.;
growth of a virulent strain of African swine fever virus and Jover, A. 1990. Pulmonary intravascular
in domestic pigs. J Hyg 66:117–134. macrophages in lung of pig inoculated with African
Plowright, W.; Parker, J.; and Peirce, M. A. 1969. The swine fever virus of different virulence. J Comp Pathol
epizootiology of African swine fever in Africa. Vet Rec 102:323–334.
85:668–674. Sierra, M.; Gomez, J.; Carrasco, L.; Fernandez, A.; Mozos,
Plowright, W.; Perry, C. T.; and Peirce, M. A. 1970. Experi- E.; and Jover, A. 1991. In vivo study of hemadsorption in
mental infection of the Argasid tick, Ornithodoros African swine fever virus infected cells. Vet Pathol
moubata porcinus, with African swine fever virus. Arch 28:178–181.
Gesamte Virusforsch 31:33–50. Sogo, J.; Almendral, J.; Talavera, A.; and Viñuela, E. 1984.
Polo Jover, F., and Sanchez Botija, C. 1961. La peste porcina Terminal and internal inverted repetitions in African
africana en España. Bull Off Int Epizoot 55:107–147. swine fever virus DNA. Virology 133:271–275.
102 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Tabares, E.; Marcotegui, M. A.; Fernandez, M.; and Sanchez Wilkinson, P. J., and Wardley, R. C. 1978. The replication of
Botija, C. 1980. Proteins specified by African swine fever ASFV in pig endothelial cells. Br Vet J 134:280–282.
virus. I. Analysis of viral structural proteins and anti- Yañez, R.; Rodriguez, J.; Nogal, L.; Enriquez, C.; Rodriguez,
genic properties. Arch Virol 66:107–117. J.; and Viñuela, E. 1995. Analysis of the complete nu-
Wardley, R., and Wilkinson, P. J. 1980. Detection of African cleotide sequence of African swine fever virus. Virology
swine fever antigen and antibody by radioimmunoassay. 208:249–278.
Vet Microbiol 5:169–176.
Blue Eye Disease
6 A. H. Stephano

Blue eye (BE) is a disease of swine associated with infec- nomic impact has been considerably reduced. BE has not
tion by a member of the Paramyxoviridae family. It is been reported outside Mexico.
characterized clinically by central nervous system disor- Differences in clinical signs became evident during
ders, corneal opacity, and reproductive failure. the first years of the disease. In 1980 mainly piglets were
In 1980 the disease was reported in central Mexico, affected. Mortality and nervous system disorders in pigs
with numerous outbreaks of encephalitis and corneal older than 30 days were uncommon. In 1983, severe out-
opacity in piglets from which a hemagglutinating virus breaks of encephalitis with high mortality in pigs weigh-
was isolated (Stephano et al. 1981). This virus was iden- ing 15–45 kg were observed on badly managed farms, al-
tified as a serologically distinct (Stephano et al. 1986b) ways with concomitant viral and bacterial diseases
member of the family Paramyxoviridae (Stephano and (Stephano and Gay 1985b, 1986a). These and subse-
Gay 1983, 1984, 1985a) and was subsequently named BE quent observations have indicated that BE is frequently
paramyxovirus (BEP). associated with other infectious diseases. It was not until
Members of the Paramyxoviridae family also have 1983 that reproductive failure in sows and transient in-
been isolated from pigs in other countries. In Japan, fertility in boars were identified (Stephano and Gay
Sasahara et al. (1954) isolated a hemagglutinating virus 1984, 1985a). In 1988 severe problems of orchitis, epi-
from pigs with porcine influenza and encephalitis; this didymitis, and testiclar atrophy in boars became evident
virus, named hemagglutinating virus of Japan or Sendi (Campos and Carbajal 1989; Stephano et al. 1990).
virus, proved to be pathogenic for pigs, producing cen-
tral nervous system, respiratory system, and reproduc- ETIOLOGY
tive system disorders. In Canada, Greig et al. (1971) iso-
lated a paramyxovirus from the brain of a pig during an Recently the family Paramyxoviridae was reorganized and
investigation of pigs with nervous signs. This virus was BEP was classified as a member of the subfamily
not able to produce signs and lesions in experimentally Paramyxovirinae, genus Rubulavirus (mumps virus)
inoculated piglets. In Israel, Lipkind et al. (1986) isolated (Fields et al. 1996). Extensive molecular work has been
a paramyxovirus from nasal swabs of apparently healthy carried out in Sweden (Sundqvist et al. 1990; Berg et al.
pigs before slaughter. In the United States, a paramyxo- 1991; Sundqvist et al. 1992; Berg et al. 1992). BEP repli-
like virus was isolated from finishing pigs with respirato- cates and produces cytopathic effects in a wide variety of
ry system and central nervous system disease. This virus cell cultures and agglutinates erythrocytes of both mam-
was shown to be antigenically related to paramyxovirus malian and avian origin (Stephano and Gay 1985a;
1, 3, and 4b but not to the BEP (Paul et al. 1994). Moreno-Lopez et al. 1986; Stephano et al. 1986b).
The first reported outbreak of BE was observed on a
commercial farm with 2500 sows located in La Piedad, Size and Morphology
Michoacán, Mexico (Stephano et al. 1982). That year Electron microscopic examination of BEP revealed parti-
similar outbreaks were observed on other farms in the cles similar to other paramyxoviruses, measuring from
same area, as well as in the states of Jalisco and Guanaju- 135–148 nm to 257–360 nm (Fig. 6.1). The virion is pleo-
ato. From an economic perspective, BE became especial- morphic but usually more or less spherical; no filamen-
ly important in central Mexico, where it was diagnosed tous forms have been observed. The envelope is covered
in the states of Nuevo León, Hidalgo, Tlaxcala, Estado de with a layer of closely spaced surface projections or
México, Federal District, Querétaro, Tamaulipas, Puebla, spikes. Nucleocapsids from disrupted virus particles are
and Campeche (Stephano et al. 1988b). Serological evi- frequently seen as single entities with a diameter of 20
dence indicated that BEP was present in at least 16 states nm and a length of 1000–1630 nm (Fig. 6.2) or more
(Fuentes et al. 1992). The disease is still recognized occa- (Stephano and Gay 1985a).
sionally in central Mexico in the states of Michoacán,
Jalisco, and Guanajuato as cases of nervous system dis- Physicochemical Properties
orders, corneal opacity, reproductive failure in sows, and The infectivity of BEP is abolished by treatment with
orchitis with epididymitis in boars. However, its eco- ether, chloroform, formalin, and beta propiolactone, but

103
104 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

6.1. Blue eye paramyxovirus particles


showing surface projections by negative-
stain electron microscopy (×108,200).

6.2. Fraction of a nucleocapsid from a dis-


rupted BEP (negative-stain electron mi-
croscopy; ×203,000).
CHAPTER 6 BLUE EYE DISEASE Stephano 105

BEP is resistant to actinomycin D. Formalin treatment monolayer cell cultures of bovine (turbinate, kidney, tes-
abolishes both infectivity and hemagglutinating activity. ticle, skin, palatine, choroid plexus), monkey (GMK),
BEP is inactivated at 56˚C after 4 hours. Intact virions mink (lung), and human (fetal) origin. Syncytia were ob-
have a buoyant density of 1.21 g/mL in sucrose gradients served in BEP-infected monolayer cultures of porcine
(Stephano and Gay 1985a). turbinate cells and bovine turbinate and kidney cells
(Moreno-Lopez et al. 1986). The chick embryo also sup-
Biological Properties and Replication ports BEP replication.
BEP has been grown with cytopathic effect in monolayer Hemagglutination has been tested in erythrocytes
cultures of pig kidney (PK), bovine thyroid, bovine em- from chickens, guinea pigs, mice, rats, rabbits, hamsters,
bryo, equine dermis, swine testicle, cat kidney, and baby horses, pigs, goats, cats, and dogs, as well as in the four
hamster kidney–21 and Vero cells. In monolayer cultures groups (A, B, AB, and O) of human erythrocytes. Spon-
of primary PK cells and cells of an established PK cell taneous elution at 37˚C occurred after 30–60 minutes.
line (PK-15), the cytopathic effects start between 24 and The infected PK-15 cells are also positive to hemadsorp-
48 hours after inoculation and are complete between 5 tion with chicken erythrocytes (Stephano and Gay
and 7 days. They consist of individual rounded cells, cy- 1985a; Stephano et al. 1986b).
toplasmic vacuoles, and syncytium formation (Fig. 6.3);
dead cells detach from the culture vessel surface, leaving Serology
small plaques. In PK-15 cells the virus was seen free in cy- Antiserum prepared against paramyxoviruses 1, 2, 3, 4,
toplasm and within vesicles. Some cells also contained 6, and 7 and parainfluenza viruses 1, 2, 3, 4a, 4b, and 5
viral inclusion bodies (Stephano and Gay 1985a; do not affect BEP infectivity (Stephano et al. 1986b).
Stephano et al. 1986a). Cytopathic effects have also been No morphological, physicochemical, or serological
observed in other types of monolayer cell cultures of differences have been observed among different strains
porcine origin (turbinate, choroid plexus), as well as in (Gay and Stephano 1987).

6.3. Syncytium formation


in pig kidney-15 monolayer culture
72 hours after infection (Giemsa;
×500).
106 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

EPIDEMIOLOGY movements when handled. Other signs include lethargy


with some involuntary movements, dilated pupils, ap-
Pigs are the only animals that are known to be affected parent blindness, and sometimes nystagmus. Some
clinically by BEP in nature. Experimentally BEP affects piglets suffer from conjunctivitis, with swollen eyelids
mice, rats, and chick embryos; rabbits, dogs, cats, and and lacrimation. Often the eyelids are closed and adher-
peccaries do not show clinical signs, but rabbits, cats, ent with exudate. In 1–10% of the affected piglets, either
and peccaries produce antibodies (Stephano and Gay unilateral or bilateral corneal opacity is present. Fre-
1985a; Stephano et al. 1988a; Flores 1991; Arellanes et al. quently, corneal opacity occurs without other signs and
1994; Cuetero et al. 1995). resolves spontaneously. In the first cases observed,
Subclinically infected pigs from affected farms are the piglets usually died within 48 hours of the appearance of
main source of BEP infection. The virus seems to be dis- clinical signs, but in later cases, death occurred after 4–6
seminated by nose-to-nose contact between infected and days.
susceptible pigs. Of the litters farrowed during an outbreak, 20–65%
Transmission through semen has not been estab- are affected. In these litters the piglet morbidity is be-
lished, but BEP can be recovered from testis, epididymis, tween 20% and 50%, and mortality of affected piglets is
prostate, seminal vesicles, and bulbo-urethral glands. between 87% and 90%; deaths occur for 2–9 weeks fol-
The virus may be disseminated by people and vehi- lowing an initial outbreak depending mainly on the sys-
cles, and possibly by birds and wind; other sources of in- tem of management.
fection have not been demonstrated. Most of the sows of affected litters are clinically nor-
The disease is self-limiting in closed herds. Sentinel mal. Some of them show moderate anorexia 1 or 2 days
pigs introduced to the farm 6–12 months after an out- before the appearance of clinical signs in the piglets, and
break remain asymptomatic and do not produce anti- corneal opacity has also been observed in the farrowing
bodies against BEP (Stephano and Gay 1986b; Stephano house during outbreaks.
et al. 1986a). In pregnant sows a reproductive failure which lasts
Naturally infected pigs develop antibodies that usual- 2–11 months (usually 4 months) is observed. During
ly persist throughout their lives. However, the disease outbreaks there is an increase in the number of animals
can recur in susceptible progeny and when susceptible returning to estrus, a reduction in farrowing rate, and an
pigs are added to the herd. Farms with a continuous sys- increase in the weaning-to-service interval and nonpro-
tem of production may have cases periodically. ductive sow days. Also, an increase in stillbirths and
mummified fetuses and, consequently, a reduction in the
CLINICAL SIGNS number of pigs born alive and later the total pigs born
by farrow are observed (Table 6.1). Abortion is not a fea-
BE may start in any area of commercial breeding units, ture, but it is observed in some dams during an acute
but it is usually observed first in the farrowing house, outbreak. Gilts and other adult pigs also occasionally de-
with central nervous system signs and high piglet mor- velop corneal opacity.
tality. At about the same time, the farmer may observe Pigs more than 30 days old show moderate and tran-
corneal opacity in some weaned or fattening pigs sient clinical signs such as anorexia, fever, sneezing, and
(Stephano and Gay 1985a, 1986a; Stephano et al. 1988a). coughing. Nervous system signs are less common and
The mortality rate increases rapidly and then decreases less obvious, but when present they consist of listless-
within a short time. Once the initial outbreak is over, no ness, ataxia, circling, and rarely swaying of the head. As
new clinical cases appear unless susceptible pigs are in- in piglets, unilateral or bilateral corneal opacity and con-
troduced, as has been observed on farms that operate on junctivitis continue to appear on the farm for another
a continuous-flow pattern. month without other signs. Only 1–4% of pigs older than
The clinical signs are variable and depend mainly on 30 days are affected and the mortality is generally low.
the age of the pig. Piglets 2–15 days old are most suscep- Outbreaks with 20% mortality and severe central ner-
tible, and the clinical signs are sudden in onset. Healthy vous system manifestations have been observed in 15–45
piglets may suddenly become prostrate, generally in lat- kg pigs; corneal opacity was present in up to 30% of these
eral recumbency, or show nervous system signs. But the pigs (Stephano and Gay 1985b).
disease usually runs a course that starts with fever, a Boars, like other adult animals, generally do not show
rough hair coat, and an arched back, sometimes accom- clinical signs, but mild anorexia and corneal opacity have
panied by constipation or diarrhea. These signs are fol- been observed. Semen evaluation demonstrated that in
lowed by progressive nervous signs of ataxia, weakness, herds infected by the BEP, 29–73% of the boars show
rigidity (mainly of the hindlegs), muscle tremor, and ab- temporary or permanent infertility, with a decrease in
normal posture, such as a sitting position. Anorexia does concentration, an increase in abnormalities, and a de-
not occur while the piglets can still walk. Some piglets crease in motility and viability of spermatozoa. In some
are hyperexcitable, squealing and showing paddling boars there is azospermia; the ejaculate becomes clear
CHAPTER 6 BLUE EYE DISEASE Stephano 107

Table 6.1. Reproductive parameters affected during BEP infection outbreaks


Duration
Parameter Range (months)
Repeat service (%) Increase 5.8–22.1 2–6
Farrowing rate (%) Decrease 6.0–30.2 1–4
Weaning to first service interval Increase 1.0–2.9 2–8
Bred by 7 days (%) Decrease 10.0–26.1 2–8
Abortions (%) Increase 0–4.7 0–2
Sow deaths (%) Increase 0.1–0.8 0–2
Total born/litter Decrease 0–2.1 1–4
Pigs born alive/litter Decrease 0.8–4.1 4–8
Stillborn pigs (%) Increase 4.5–19.6 2–11
Mummies (%) Increase 6.8–36.2 3–12
Preweaning mortality (%) Increase 32.0–51.8 1–7
Note: Most parameters reach the figures shown above, but some persist with lower numbers. Nine farms were evaluated.

and resembles coconut water. Some boars develop The interstitial pneumonia observed suggests dis-
swollen testicles. The testis and epididymis become semination through the blood. In experimentally inocu-
turgid with marked edema; later some develop a granu- lated piglets, the virus could be isolated from the brain,
lar texture and most atrophy (generally unilateral) or be- lung, tonsil, liver, turbinate, spleen, kidney, mesenteric
come soft and flabby with or without granular epi- lymph node, heart, and blood. Brain, lung, and tonsil are
didymitis. Boars with severe lesions lose libido (Campos the most common sites for isolation (Stephano et al.
and Carbajal 1989; Stephano et al. 1990). 1988a).
The cause of the corneal opacity that is sometimes as-
PATHOGENESIS sociated with BE is unknown. It is not always reproduced
experimentally, but histological lesions such as anterior
It has been presumed that the natural infection with BEP uveitis are commonly observed in the cornea (Stephano
is acquired by inhalation. Experimental intratracheal or and Gay 1986b; Perez et al. 1988). Usually the opacity oc-
intranasal exposures by either instillation or aerosol are curs late in the course of the disease. The histological le-
effective routes of infection and result in clinical signs sions and signs suggest that the opacity is due to an im-
and lesions similar to those observed in natural cases. In munologic reaction similar to that produced by canine
previous experiments 1-day-old piglets developed a ner- adenovirus hepatitis. Recent results indicate that the
vous syndrome at 20–66 hours postinoculation, some virus replicates in the cornea since intracytoplasmic in-
weaned pigs (21–50 days old) developed a nervous syn- clusion body formation in the epithelial cells close to the
drome at 11 days postinoculation, and pregnant sows de- corneo-scleral angle was observed in acutely affected
veloped reproductive failure when inoculated during pigs. The corneal opacity has been observed in pigs oth-
pregnancy. The corneal opacity is also occasionally re- erwise clinically normal and resistant to the infection,
produced in these cases. The disease was also reproduced and it usually disappears after some period.
in susceptible pigs placed in contact with experimentally It has been suggested that BEP reaches the uterus
infected pigs for up to 19 days after experimental infec- through the blood. In pregnant sows this produces re-
tion (Stephano and Gay 1983; Perez et al. 1988; productive failure, with embryonic mortality and return
Stephano et al. 1988b). to estrus when infection is in the first one-third of gesta-
The initial site of BEP replication has not been estab- tion, and stillbirths and mummified fetuses when infec-
lished; however, the nasal mucosa and tonsils have been tion is later in gestation (Stephano and Gay 1984).
suggested based on the fact that infectious virus has been Experimental inoculation of young hybrid boars
recovered from nasal and tonsillar swabs. Moreover, vi- showed that nasal instillation of BEP resulted in inflam-
ral antigens are easily detected by immunofluorescence mation and edema of the testis and epididymis by 15
in these tissues when they are collected from either natu- days after inoculation. By 30 days after inoculation there
rally or experimentally infected pigs. The virus also has was necrosis of the seminiferous tubules and rupture of
been observed in the axon of neurons. From the initial the epithelial wall of the epididymis with leakage of sper-
site of replication BEP spreads early in the infection to matozoa out from the lumen leading to abscess forma-
the brain and lung, and histological lesions and central tion. Boars sacrificed 80 days after infection showed fi-
nervous system manifestations occur early in the disease. brosis and granuloma formation in the epididymis as
The brain is the best tissue for isolation and immunoflu- well as testicular atrophy (Ramirez et al. 1995).
orescence. BEP was recovered from testis, epididymis, prostate,
108 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

seminal vesicles, and bulbo-urethral glands in mature


Mexican hairless boars from 10 to 45 days after inocula-
tion (Vizuet et al. 1996).
The BEP infection has commonly been associated
with pneumonia, especially that due to Actinobacillus
pleuropneumoniae, but experimental inoculation of BEP
prior to infection with Pasteurella multocida types A and
D did not result in colonization of the bacteria in the
lung (Garcia et al. 1988).

LESIONS

Gross Lesions
There are no specific gross changes. In piglets, a mild
pneumonia is frequently observed at the ventral tips of
the cranial lung lobes; also mild gastric distension with
milk, distension of the urinary bladder with urine, and a
slight accumulation of fluid with fibrin in the peritoneal
cavity may be observed. Brain congestion and an in-
6.5. Corneal opacity in a 7-day-old piglet.
crease in the cerebrospinal fluid occur. Conjunctivitis,
chemosis (Fig. 6.4), and varied degrees of corneal opaci-
ty (Fig. 6.5), usually unilateral, are observed. Vesicle for-
mation, ulcers, and queratocono (keratoconus) have been epididymis, or testis (Campos and Carbajal 1989;
observed in the cornea, as well as exudate in the anterior Stephano et al. 1990; Ramirez et al. 1995; Vizuet et al.
chamber. Pericardial and kidney hemorrhages are occa- 1996).
sionally observed (Stephano and Gay 1985a, 1986b).
Boars develop swollen testicles and epididymis, with Microscopic Lesions
increase in diameter and weight due to edema. These The main histological changes are located in the brain
changes are frequently unilateral. Orchitis, epididymitis, and spinal cord. These reflect a nonsuppurative en-
and, later, atrophy of the testicle with or without granu- cephalomyelitis affecting mainly the gray matter of the
lomatous formation in the epididymis occur. Hemor- thalamus, midbrain, and cerebral cortex and include a
rhages are occasionally observed in the tunica albuginia, multifocal and diffuse gliosis; perivascular cuffing with
lymphocytes (Fig. 6.6), plasma cells, and reticular cells;
neuronal necrosis; neuronophagia; meningitis; and
choroiditis (Ramirez and Stephano 1982). Intracytoplas-
mic inclusion bodies are found in neurons. There are
variations in the severity and extent of these lesions
(Stephano and Gay 1986b; Perez et al. 1988; Stephano et
al. 1988a).
The lungs have scattered localized areas of interstitial
pneumonia characterized by thickened septa with
mononuclear cell infiltration.
Changes in the eye are mainly corneal opacity, char-
acterized by corneal edema, and anterior uveitis. Neu-
trophils, macrophages, or mononuclear cells infiltrate
the iridocorneal endothelium, corneo-scleral angle, and
cornea. The external sheet of the cornea often has cyto-
plasmic vesicles (Fig. 6.7) (Stephano and Gay 1986b;
Perez et al. 1988; Stephano et al. 1988a).
Many affected pigs have a mild tonsillitis with
desquamated epithelium and inflammatory cells in the
crypts.
In boars the affected testes show degeneration and
6.4. Thickening of the cornea due to edema and necrosis of the germinal epithelium. The interstitial tis-
inflammatory cell infiltration. sue shows Leydig cell hyperplasia, mononuclear cell in-
CHAPTER 6 BLUE EYE DISEASE Stephano 109

6.6. Perivascular cuffing and diffuse gliosis in the cerebellum (H&E; ×225).

6.7. Mononuclear cell


infiltration and edema of the
lamina propria of the cornea,
with cytoplasmic vesicles
in the external epithelium
(H&E; ×225).
110 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

filtration, vascular wall hialinization, and fibrosis. The out effective results. Antimicrobial therapy is commonly
epididymis shows vesicle formation, loss of cilia of the used to treat and prevent secondary infections. Medica-
epithelial cells, rupture of the epithelial wall, presence of tion is commonly used for controlling associated respira-
spermatozoa in the intertubular space, and severe infil- tory problems. Good management, such as maintaining
tration of inflammatory mononuclear cells with a healthy environment and providing proper housing
macrophage phagocytosis of fragmented sperm. Fibrosis and nutrition, reduces the detrimental effect of the dis-
and spermatic granuloma are organized. ease in the herd.

DIAGNOSIS PREVENTION AND CONTROL

Clinical signs such as encephalitis, corneal opacity, and Health control programs are the most reliable method of
reproductive failure in the sow and orchitis and epi- preventing the introduction of BEP to a farm. Swine pop-
didymitis in the boar are consistent with a diagnosis of ulations must be established or replaced from a healthy
BE. Additional diagnostic evidence is provided by histo- pig herd. Perimeter fencing; separate load-out areas,
logical lesions such as nonsuppurative encephalitis, ante- changing rooms, and showers; control of personnel, vis-
rior uveitis, keratitis, orchitis, and epididymitis. The itors, and vehicles; control of birds, rats, and mice; waste
presence of intracytoplasmic inclusion bodies in neu- removal and dead pig disposal; and a quarantine—all
rons and corneal epithelium confirms the diagnosis. provide insurance against infection. Serological analysis
Serological tests such as hemagglutination inhibition should be performed in the replacement animals.
(HI), virus neutralization, microtitration serum-virus Elimination of BEP from infected herds has been ac-
neutralization, and enzyme-linked immunosorbent as- complished by management practices such as closing the
say (ELISA) have been used diagnostically to identify an- herd, cleaning and disinfecting, all-in/all-out, elimina-
tibody-positive pigs. HI is the most used test, but false- tion of clinically affected animals (pigs with nervous
positive titers of 1:16 have been detected when chicken signs or infertile boars), and dead pig disposal. These
erythrocytes are used or when the BEP antigen is grown procedures followed by serological testing, herd perfor-
in chicken embryos; therefore, bovine erythrocytes are mance analyses, and introduction of sentinel BEP
recommended. Direct immunofluorescence has been seronegative pigs confirm the elimination of BEP
performed with tissue sections and monolayers, using a (Stephano et al. 1986b).
conjugate prepared with rabbit or pig serum (Stephano To reduce the economic impact of BEP, different
and Gay 1985a; Stephano et al. 1988a; Ramirez et al. measures have to be taken. Infertile boars, with or with-
1996). Paired serum samples, 15 days apart, are recom- out orchitis, must be eliminated. Artificial insemination
mended for the diagnosis of the disease. should be performed if necessary. Sows and gilts that are
BEP is easily isolated by adding a homogenated or presumed to be pregnant should be carefully observed
triturated suspension of brain or tonsil from infected for signs of estrus and, if possible, examined by ultra-
pigs to the medium of monolayer cultures of PK-15 cells sound to confirm pregnancy. It is unknown whether pur-
or primary porcine kidney cells. The virus-induced cyto- poseful, and thus quicker, dissemination of the virus in
pathic effect is characterized by syncytium formation. an infected herd has a tendency to limit the total eco-
A differential diagnosis must include other causes of nomic impact of the disease.
encephalitis and reproductive disease, especially porcine Various experimental killed-virus vaccines have been
reproductive and respiratory syndrome virus and Au- produced in cell monolayer cultures and chicken em-
jeszky’s disease virus. Only BEP produces corneal opaci- bryos and suspended in oil or OH2Al3 adjuvant
ty (in up to 30% of infected pigs) as well as orchitis and (Stephano et al. 1992; Martinez et al. 1996).
epididymitis in boars (Stephano and Gay 1985b;
Stephano 1986; Stephano et al. 1988a; Campos and Car- REFERENCES
bajal 1989; Stephano et al. 1990).
Arellanes, A. E.; Fuentes, R. M. C.; Carreon, N. R.; and
TREATMENT Ramirez, M. H. 1994. Inoculacion experimental del
paramixovirus del ojo azul en el gato domestico (Felix
As with most viral diseases of swine, there are no specif- catus). Vet Mex 25:239–241.
ic treatments for BEP. Once the clinical signs are evident, Berg, M.; Sundqvist, A.; Moreno-Lopez, J.; and Linne, T.
nothing can be done to modify the course of the disease. 1991. Identification of the porcine paramyxovirus LP-
Pigs with corneal opacity frequently spontaneously re- MV matrix protein gene: Comparative sequence analysis
cover whereas pigs with central nervous disturbances with other paramyxoviruses. J Gen Virol 72:1045–1050.
generally die. Berg, M.; Hjertner, B.; Moreno-Lopez, J.; and Linne, T.
Serum from sows who previously had affected litters 1992. The P gene of porcine paramyxovirus LPMV en-
has been administered orally to piglets, apparently with- codes three possible polypeptides P, V and C: The P pro-
CHAPTER 6 BLUE EYE DISEASE Stephano 111

tein mRNA is edited. J Gen Virol 73:1195–1200. testis. In Mem 20th Reun Acad Invest Biol Repr, pp.
Campos, H. R., and Carbajal, S. M. 1989. Trastornos repro- 211–214. Ixtapa, Mex.
ductivos de los sementales de una granja porcina de ciclo Ramirez, M. H.; Carreon, N. R.; Mercado, G. C.; and Ro-
completo ante un brote de ojo azul. In Mem 24th Congr driguez, T. J. 1996. Hemoaglutinacion e inhibicion de la
Asoc Mex Vet Esp Cerdos, pp. 62–64. Morelia, Mex. hemoaglutinacion del paramixovirus porcino a traves de
Cuetero, R. S.; Ramirez, M. H.; Careon, N. R.; and Cam- la modificacion de algunas variables que participan en la
puzano, G. J. 1995. Inoculacion experimental del prueba. Vet Mex 27:257–259.
paramixovirus de ojo azul en rats de laboratorio (Cepa Ramirez, T. C. A., and Stephano, A. H. 1982. Histological
Wistar) via intramuscular. Vet Mex 26:231–235. central nervous system lesions produced by an haemag-
Fields, B. N.; Knipe, D. M.; and Howley, P. M. 1996. Fields’ glutinating virus in naturally infected piglets. Proc Int
Virology, 3d ed., pp. 33–34. Philadelphia: Lippincott- Congr Pig Vet Soc 7:154.
Raven. Sasahara, J.; Hayashi, S.; Kumagai, T.; Yamamoto, Y.; Hira-
Flores, J. J. 1991. Inoculacion experimental del sawa, K.; Munekata, K.; Okaniwa, A.; and Kato, K. 1954.
paramixovirus del ojo azul (POA) en el pecari de collar On a swine virus disease newly discovered in Japan. 1.
(Dicotiles tajacu). Tesis FMVZ. UNAM. Isolation of the virus. 2. Some properties of the virus.
Fuentes, R. M.; Carreon, N. R.; Ramirez, M. H.; Trujillo, M. Virus 4:131–139.
E.; and Fraire, I. B. 1992. Estudio piloto de la frecuencia Stephano, A. H. 1986. Diagnostico diferencial entre Au-
de anticuerpos contra el paramixovirus del ojo azul en jeszky y syndrome del ojo azul. Sint Porc 5(12):41–48.
cerdos de la Republica Mexicana. Vet Mex 23:37–39. Stephano, A. H., and Gay, G. M. 1983. El syndrome del ojo
Garcia, G. J.; Camacho, M. J.; Mendoza, E. S.; Ciprian, C. A.; azul. Estudio experimental. In Mem Reun Inv Pecu Mex-
Gonzalez, G. S.; Diaz, C.; and Stephano, A. H. 1988. In- ico, pp. 523–528. Mexico, D.F.
feccion experimental con el virus de ojo azul y Pasteurel- ———. 1984. Experimental studies of a new viral syn-
la multocida en cerdos convencionales. In Mem 23d Con- drome in pigs called “blue eye” characterized by en-
gr Asoc Mex Vet Esp Cerdos, pp. 88–89. Leon, Mex. cephalitis and corneal opacity. Proc Int Congr Pig Vet
Gay, G. M., and Stephano, A. H. 1987. Stain analysis of a Soc 8:71.
new paramyxovirus isolated from 12 outbreaks of en- ———. 1985a. El syndrome del ojo azul en cerdos I. Sint
cephalitis and corneal opacity in pigs (blue eye syn- Porc 4(5):42–49.
drome). In Proc 23d World Vet Congr, p. 161. Montreal. ———. 1985b. El syndrome del ojo azul en granjas engor-
Greig, A. S.; Johnson, C. M.; and Bouillant, A. M. P. 1971. dadoras. In Mem 19th Congr Asoc Mex Vet Esp Cerdos,
Encephalomyelitis of swine caused by a haemagglutinat- pp. 71–74. Merida, Mex.
ing virus. VI. Morpholoy of the virus. Res Vet Sci ———. 1986a. El syndrome del ojo azul: Una nueva enfer-
12:305–307. medad en cerdos asociada a un paramyxovirus. Vet Mex
Lipkind, M.; Shoham, D.; and Shihmanter, E. 1986. Isola- 17:120–122.
tion of a paramyxovirus from pigs in Israel and its anti- ———. 1986b. Encefalitis, falla reproductiva y opacidad de
genic relationship with avian paramyxoviruses. J Gen Vi- la cornea, ojo azul. Sint Porc 5(12):26–39.
rol 67:427–439. Stephano, A. H.; Gay, G. M.; Ramirez, T. C.; and Maqueda,
Martinez, L. A.; Correa, G. P.; Coba, A. M.; and Fajardo, M. A. J. J. 1981. Estudio de un brote de encefalitis en le-
R. 1996. Antigenicidad e inocuidad de una vacuna inac- chones por un virus hemoaglutinante. In Mem 17th
tivada contra la enfermedad del ojo azul (EOA) de los Congr Asoc Mex Vet Esp Cerdos, p. 43. Puerto Vallarta,
cerdos al ser aplicada en lechones. In Mem 31st Congr Mex.
Asoc Mex Vet Esp Cerdos, p. 56. Veracruz, Mex. ———. 1982. An outbreak of encephalitis in piglets pro-
Moreno-Lopez, J.; Correa-Giron, P.; Martinez, A.; and Eric- duced by an hemagglutinating virus. Proc Int Congr Pig
sson, A. 1986. Characterization of a paramyxovirus iso- Vet Soc 7:153.
lated from the brain of a piglet in Mexico. Arch Virol Stephano, A. H.; Doporto, J. M.; and Gay, M. 1986a. Estudio
91:221–231. epidemiologico en dos granjas afectadas por el ojo azul.
Paul, P. S.; Janke, B. H.; Battrell, M. A.; Lyoo, Y. S.; Halbur, Proc Int Congr Pig Vet Soc 9:456.
P. G.; Landgraf, J.; and Huinker, C. 1994. Isolation of a Stephano, A. H.; Gay, M.; and Kresse, J. 1986b. Properties of
paramyxo-like virus from pigs with interstitial pneumo- a paramyxovirus associated to a new syndrome (blue
nia and encephalitis. Proc Int Congr Pig Vet Soc 13:72. eye) characterized by encephalitis, reproductive failure
Perez, P. F.; Stephano, A. H.; and Gay, G. M. 1988. Estudio and corneal opacity. Proc Int Congr Pig Vet Soc 9:455.
histologico en lechones inoculados experimentalmente Stephano, A. H.; Fuentes, R. M.; Hernandez, J. P.; Herrado-
con el paramyxovirus de ojo azul. In Mem 23d Congr ra, L. M.; and Carreon, R. 1988a. Encefalitis y opacidad
Asoc Mex Vet Esp Cerdos, pp. 81–83. Leon, Mex. de la cornea en cerdos destetados, inoculados experi-
Ramirez, M. H.; Reyes, L. J.; Kennedy, S.; and Hernandez, J. mentalmente con paramyxovirus de ojo azul. In Mem
P. 1995. Studies on the pathogenesis of the pig paramyx- 23d Congr Asoc Mex Vet Esp Cerdos, pp. 90–92. Leon,
ovirus of the blue eye disease on the epididymis and Mexico.
112 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Stephano, A. H.; Gay, G. M.; and Ramirez, T. C. 1988b. En- and Moreno-Lopez, J. 1990. The structural proteins of a
cephalomyelitis, reproductive failure and corneal opaci- porcine paramyxovirus (LPMV). J Gen Virol
ty in pigs, associated with a new paramyxovirus infec- 71:609–613.
tion (blue eye). Vet Rec 122:6–10. Sundqvist, A.; Berg, M.; Moreno-Lopez, J.; and Linne, T.
Stephano, A. H.; Hernandez, D.; Perez, C.; Gonzalez, C. T.; 1992. The haemagglutinin-neuraminidase glycoprotein
Ramirez, M. H.; and Cervantes, A. 1990. Boar infertility of the porcine paramyxovirus LPMV: Comparison with
and testicle atrophy associated with blue eye paramyx- other paramyxoviruses revealed the closest relationship
ovirus infection. Proc Int Congr Pig Vet Soc 11:211. to Simian virus 5 and mumps virus. Arch Virol
Stephano, A. H.; Olvera, M. J.; Garcia, V. P.; Ramirez, M. H.; 122:331–340.
and Cordoba, D. J. 1992. Eficacia de una vacuna inacti- Vizuet, A. O.; Martinez, G. R.; Carreon, N. R.; Lopez, M. J.
vada para la prevencion de la infeccion por el paramyx- R.; Pradal-Roa, P.; Mendoza, G. R.; and Hernandez, M.
ovirus de ojo azul. In Mem 27th Congr Asoc Mex Vet Esp L. 1996. Experimental inoculation in Mexican Hairless
Cerdos, pp. 24–28. Acapulco, Gro. boars with the paramyxovirus of the blue eye disease.
Sundqvist, A.; Berg, M.; Hernandez-Jauregui, P.; Linne, T.; Proc Int Congr Pig Vet Soc 14:127.
Bovine Viral Diarrhea and
7 Border Disease
P. Vannier and E. Albina

One of the best known and most widely studied viral changes in cell culture, while others were noncytopathic
pathogens of the pig is hog cholera (HC) virus (HCV), a (Gillespie et al. 1960). The relationship between the cy-
pestivirus of the family Flaviviridae. There are, however, topathic effect and the pathogenesis of the disease has
other pestiviruses to which pigs may be exposed, includ- been investigated. It is now well established that mucos-
ing bovine viral diarrhea (BVD) virus (BVDV) and bor- al disease occurs only in immunotolerant calves persis-
der disease (BD) virus (BDV). tently infected with a noncytopathic strain of BVDV and
The discovery by Darbyshire (1961) of an antigenic subsequently superinfected with a cytopathic strain of
relationship between the pestiviruses stimulated further BVDV with the similar antigenic properties (Brownlie et
investigations into natural and experimental BVDV and al. 1984, 1987). Infection with the cytopathic strain may
BDV infections of swine. The first report of natural in- be of endogenous origin (i.e., a cytopathic mutant of the
fection of swine with BVDV came from Australia in noncytopathic strain) or it may be the result of exoge-
1964, but BVDV was not isolated from a naturally infect- nous exposure to a strain that, by chance, is antigenical-
ed pig until 1973 (Fernelius et al. 1973). The presence of ly similar. The third member of the Pestivirus genus is the
BVD-BD antibodies in pig sera interferes with HC eradi- causative agent of BD in sheep. This disease, character-
cation programs. Therefore, it is essential to identify the ized by congenital disorders in lambs, was first recog-
origin of any pestivirus antibodies encountered in such nized in 1959 in Great Britain near the border between
programs. In addition, the teratogenic property of pes- England and Wales (Hughes et al. 1959), but the im-
tiviruses has been well established (Terpstra and munological relationship of BDV with BVDV was dis-
Wensvoort 1988; Vannier et al. 1988; Wensvoort and covered later (Hamilton and Timoney 1972).
Terpstra 1988), and the infection of pregnant sows by The terms BVDV and BDV are used to indicate that
BVDVs and BDVs induces a pathology partially resem- the virus was isolated from either cattle or sheep, al-
bling that of congenital HC. though these two viruses cannot be differentiated mor-
phologically or structurally (Laude 1979). There is also
ETIOLOGY evidence that BVDV can infect sheep and BDV can infect
cattle (Barlow et al. 1980; Terlecki et al. 1980). In pigs,
BVDVs and BDVs have been classified together with the pestivirus isolates are usually HCV. However, BVDV and
HCVs in the Pestivirus genus, Flaviviridae family (Wen- BDV can be isolated from naturally infected pigs (Car-
gler et al. 1995). Morphological and structural character- brey et al. 1976; Terpstra and Wensvoort 1988). More-
istics of the three pestiviruses are similar (Laude 1979; over, it has been demonstrated through cross-neutraliza-
Collett et al. 1989). Their genome is single-stranded RNA tion tests and tests using monoclonal antibodies
of positive polarity. Virions are enveloped and range in (Wensvoort et al. 1989b; Leforban 1990) that, in the
diameter from 25 to 120 nm. This wide range in size is past, BVDV may have been isolated from pigs but misla-
the result of either the entire particle or only the core be- beled as HCV on the basis of tests using polyclonal anti-
ing measured (Hsien-Jue Chu and Yuan Chung Zee bodies only.
1984). Their density ranges from 1.09 to 1.16 g/cm3 and Up to now, pestiviruses have been named according
is dependent on the type of cell host used for their prop- to the species in which they cause disease. As previously
agation (Horzinek and Van Berlo 1987; Laude 1979). indicated, cross-species transmission within the Artio-
BVD was first described in 1946 as an infectious and dactyla has been reported for BVDV as well as BDV. The
contagious bovine disease (Olafson et al. 1946), and molecular analysis and the use of a panel of 76 mono-
“mucosal disease” was reported in 1953 (Ramsey and clonal antibodies revealed a fourth group within the
Chivers 1953). Further studies showed that the viruses genus Pestivirus which consists of ovine and bovine iso-
responsible for these two conditions were similar. They lates (Becher et al. 1995; Paton et al. 1995). The authors
have been referred to by most authors as BVDV proposed that the taxonomy of pestiviruses be reconsid-
(Gillespie et al. 1961). Some isolates induced cytopathic ered and suggested the following: pestivirus type 1 (clas-

113
114 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

sical BVDV strains), pestivirus type 2 (HCV strains), pes- piglets show persistent infection and immunotolerance
tivirus type 3 (true BDV strains), and pestivirus type 4 (Vannier et al. 1988). When the sow is experimentally in-
(BVDV-like strains of bovine and ovine origins). fected, the litter may comprise a mixture of virus-posi-
More recently, Harasawa (1996) constructed a phylo- tive and antibody-positive pigs. This suggests some vari-
genetic tree of pestiviruses by analyzing their 5′-untrans- ability in the time at which individual fetuses became
lated region. His results indicated that the genetic rela- infected (Edwards et al. 1995). The course of the infec-
tionship between BDV and HCV is much closer than that tion is quite similar to that described after a BVDV infec-
between genotypes of various BVDVs. Therefore, Hara- tion in pregnant cows (Baker 1987). Such congenitally
sawa proposed five groups of pestiviruses. Genetic dis- BDV-infected piglets appear to excrete large amounts of
criminations between pestiviruses actually rely on se- virus, since other young animals kept in contact have a
quence comparison of short fragments of the pestivirus rapid seroconversion with high antibody titers. Con-
genome. It would not be surprising that in the future, ad- versely, when the pigs are infected after birth, spread of
ditional subgroups will be identified through sequencing infection to in-contact pigs under the same experimental
and analysis of other parts of the genome. Studies on conditions does not occur, which suggests a low or nil ex-
pestiviruses isolated from wild ruminants or wild boars cretion of virus (Vannier et al. 1988).
could also give interesting new pestivirus genotypes. BVDVs and BDVs can be isolated or detected by im-
munofluorescence from blood, tonsils, spleen, kidneys,
EPIDEMIOLOGY ileum, and lymph nodes. Isolation of virus from ileum
indicates that fecal excretion of the virus is a possibility.
From HC-free countries like Australia, Ireland, Great
Britain, and Denmark, prevalences of BVD antibodies CLINICAL SIGNS
within the pig population have been described as varying
from 1.6% to 43.5% depending on the age of the pigs and Under Natural Conditions
possibly to some extent on the degree of their contact Natural infection of pigs with BVDV usually occurs with-
with cattle (Holm Jensen 1985). In countries where HC is out clinical signs. However, in some cases, natural infec-
present, the situation with regard to BVD antibodies tion of pig herds with pestiviruses other than HC is asso-
seems to be about the same. In the Netherlands, even ciated with breeding problems like poor conception,
though up to 15% of slaughtered sows had antibodies small litters, and a few abortions. Hyperthermia and
against BVDV, only 13 naturally occurring infections clonic spasms have also been described (Carbrey et al.
have been encountered over a 10-year period (Terpstra 1976; Stewart et al. 1977). More recently, in the Nether-
and Wensvoort 1988). Cattle are usually regarded as the lands and France, signs resembling those of congenital
source of BVDV infection in pigs. One source of infec- HC infections were described, namely, an increased
tion is the feeding of whey or milk to sows, a common death rate in piglets of up to 2–5 weeks of age which
practice in units with dairy farming (Terpstra and were born to sows vaccinated 4 months earlier against
Wensvoort 1988). In some cases, pigs had contact with HC or Aujeszky’s disease with batches of vaccines acci-
cattle recently vaccinated with BVDV (Stewart et al. dentally contaminated with a ruminant pestivirus (Terp-
1971). In other cases the pigs and cattle were kept in sep- stra and Wensvoort 1988; Vannier et al. 1988). The clini-
arate lots and buildings, but personnel and equipment cal signs in the piglets included anemia, rough hair coat,
moved freely between the different farm units (Carbrey growth retardation, wasting, and congenital tremors.
et al. 1976). Conjunctivitis, diarrhea, polyarthritis, petechiae in the
Nevertheless, pigs can be infected without any con- skin, and blue eartips were also observed (Terpstra and
tact with cattle and without being fed with bovine milk Wensvoort 1988).
or offal (Terpstra and Wensvoort 1988). The use of con- Natural infection of sows with BDV often resulted in
taminated modified-live-virus vaccines (HC or Aujeszky’s repeat-breeding and many litters containing dead and
disease) or contaminated biological products can be the mummified fetuses. Clinical signs of eyelid edema and
source of infection in pigs (Vannier et al. 1988; locomotor disorders, sometimes associated with diar-
Wensvoort and Terpstra 1988). In these cases, ovine or rhea and arthritis, occurred in a high proportion of
bovine contaminants are involved in the disorders. piglets from these sows. The mortality rate up to 2 days
In all probability, persistent infection of congenitally of age varied between 30% and 70% (Vannier et al. 1988).
infected pigs is responsible for the dissemination of
BVDV or BDV within a herd and virus transmission to Under Experimental Conditions
other susceptible pregnant sows (Terpstra and Several trials of BVDV and BDV inoculation of pigs,
Wensvoort 1988; Vannier et al. 1988). For example, it has mainly pregnant sows, have been done using oral, in-
been shown that persistent BDV infection of litters oc- tranasal, intramuscular, or intrauterine routes with in-
curs when sows are infected during early pregnancy; consistent results (Fernelius et al. 1973; Wrathall et al.
then the fetuses are transplacentally infected and most 1978; Stewart et al. 1980; Mengeling 1988; Leforban et
CHAPTER 7 BOVINE VIRAL DIARRHEA AND BORDER DISEASE Vannier, Albina 115

al. 1990b). The result is mainly dependent on the strain fetuses or piglets are observed when the sows are infect-
used and on the stage of pregnancy. The inoculation of ed at 25–41 days of gestation (Mengeling 1988; Leforban
pregnant sows with the NADL strain of BVDV (Gutekun- et al. 1990b).
st and Malquist 1963) between the 28th and 54th day of Under experimental conditions, most of the congen-
gestation did not result in subsequent transplacental in- itally infected piglets showed persistent infection and im-
fection of the fetuses (Stewart et al. 1980), but eyelid ede- munotolerance. After the disappearance of the maternal
ma was seen in a few piglets (Leforban et al. 1990b). The passively acquired antibodies, no active immunity was
inoculation of 9–18 kg piglets with the Singer strain of detected in the serum of a majority of piglets. The virus
BVDV (Coria and McClurkin 1978) did not produce dis- could be isolated from slaughtered piglets and was ex-
ease, but the virus was recovered from blood and tissues creted by some of them, as evidenced by the contamina-
of inoculated pigs, and antibodies were detected in their tion of young pigs placed in contact with them (Vannier
sera after 3 weeks. When later challenged with a virulent et al. 1988). In some experimental infections of pregnant
HCV strain, these inoculated piglets developed a severe sows with BDV, for unknown reasons, the onset of the
disease, but 6 of 7 survived (Stewart et al. 1971). The clinical signs in the piglets was delayed until 13–14 days
same strain used to inoculate fetuses between 41 and 65 after birth. It can be assumed that colostral antibodies
days by transuterine injection caused either death or ingested by the piglets blocked the multiplication of the
small fetuses (Mengeling 1988). In another study, groups pestiviruses or delayed disease in transplacentally infect-
of weaner pigs were intranasally inoculated with BVDV ed piglets (Vannier et al. 1988; Leforban et al. 1990b).
strain OSLOSS/2482. Four weeks later, the animals were However, the delayed postnatal clinical expression of the
challenged with decreasing doses of HCV. After HCV infection in piglets when sows are infected during early
challenge, fever occurred in only one animal, while none pregnancy remains unexplained.
of the others reacted clinically. However, most animals The pathogenicity of BVDVs and BDVs seems vari-
developed viremia (Dahle et al. 1993). able depending on strains used and on experimental con-
Field BDV strains inoculated into pregnant sows be- ditions. BDV seems to be more consistently pathogenic
tween the 30th and 34th day of gestation infected the for fetuses, whereas variable results are obtained with
corresponding fetuses and caused disease characterized BVDV strains. The Singer strain, adapted to replicate in
by low body weights and shorter lengths of newborn porcine cells, and strain 87/6 can infect and cause death
piglets (Wrathall et al. 1978). In another experiment of porcine fetuses, whereas the NADL strain does not in-
(Leforban et al. 1990b) perinatal death rate was in- duce a real disease in the piglets (Mengeling 1988; Lefor-
creased; and eyelid edema, increased rectal temperature, ban et al. 1990b; Edwards et al. 1995).
and anemia were observed in survivors during their sec-
ond week of life. Subsequently, growth retardation asso- LESIONS
ciated with respiratory signs and diarrhea developed in
some pigs which eventually died within 2 months. Pigs Pigs infected after birth with BVDVs or BDVs have very
without respiratory and enteritic signs survived and had mild or no lesions. Stewart et al. (1971) saw hyperemia of
normal growth despite a marked snout deformation with the small intestine of a pig euthanized and necropsied 11
prognathism observed in one of them. Virus was isolat- days after it had been placed in contact with NADL-
ed from blood and organs of all dead piglets but not strain-infected calves. Carbrey et al. (1976) identified a
from those that survived. When 40-day-old new specific- transient leucopenia during the first week following ex-
pathogen-free pigs were placed in contact with these perimental infection of pigs with a pig isolate of BVDV.
transplacentally infected piglets, they did not show any The prenatal infection of fetuses by diaplacental
disease, but they developed a high level of antibody to transmission of the viruses from the infected sow is fol-
BDV which protected them completely against a chal- lowed by consistent pathological disorders in fetuses or
lenge with a virulent strain of HCV. piglets. In the 13 naturally occurring BVD outbreaks in
the Netherlands, chronic gastroenteritis and septicemia
PATHOGENESIS with hemorrhages in lymph nodes, epicardium, and kid-
neys were the most consistent lesions reported. The in-
The ability of BVDV and BDV to establish intrauterine flammation of the digestive tract was frequently charac-
infection in swine has been well demonstrated by several terized by catarrh, hypertrophy, or ulceration of the
authors (Wrathall et al. 1978; Stewart et al. 1980; Van- mucosa. Necrotic tonsillitis, icterus, polyserositis, pol-
nier et al. 1988). These two pestiviruses are embryotoxic, yarthritis, and atrophy of the thymus were also noted
but they have little or no pathogenicity for pigs infected (Terpstra and Wensvoort 1988).
after birth. The intensity of the signs is associated with When a pig isolate of BVDV was given to gilts be-
the stage of gestation: clinical signs are more serious if tween 42 and 46 days of pregnancy, significant micro-
the sows are infected during the first third of pregnancy. scopic lesions were observed in the leptomeninges and
Indeed, the most spectacular clinical signs or lesions in the choroid plexus of the fetus as collections of lympho-
116 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

cytes and histiocytes and cellular accumulation in the ments. The threshold of sensitivity was 100 TCID50
vascular adventitia and perivascular spaces (Stewart et (median tissue culture infectious doses) for HCV and 1
al. 1980). TCID50 for BVDV. The RT-PCR provides a rapid and sen-
After experimental inoculation of 34-day-pregnant sitive diagnostic tool for the detection and differentia-
sows with BDV, postmortem dissection of newborn tion of HCV from ruminant pestiviruses (Wageck Canal
piglets revealed cerebellar hypoplasia in 9 out of 19 live- et al. 1996).
born piglets, associated with a small meningocoele in 1 Since pestiviruses share common antigenic struc-
of the 9 (Wrathall et al. 1978). The French-isolated Avey- tures (or patterns), the serological tests for the detection
ron strain of BDV (Chappuis et al. 1984) inoculated into of antibodies to HCV also detect antibodies to ruminant
30-day-pregnant sows was responsible for lesions in lym- pestiviruses. The practical importance of this is that the
phoid tissues in some piglets: marked hemorrhages in presence of ruminant pestivirus antibodies in pig sera
lymph nodes and other lymphoid tissues were found in very often causes a false-positive result in serological sur-
stillborn fetuses or in piglets that died shortly after birth. veys for HC (Holm Jensen 1985).
The histological examination of lymph nodes, spleen, These cross-reactivities are causing difficulties in
and tonsil revealed marked subacute inflammatory le- eradication and epidemiosurveillance programs for HC.
sions characterized by accumulations of lymphocytes, However, the differentiation between antibodies to HCV
plasmocytes, and eosinophilic polymorphonuclear and antibodies to ruminant pestivirus can be done by the
leukocytes, numerous secondary follicles, increased pop- neutralization test (Holm Jensen 1981) or the ELISA test
ulations of reticulocytes, and lymphoid hypoplasia with (Leforban et al. 1990a) by comparing the antibody titers
pyknosis and karyorrhexis. Thymus, liver, and nervous to the two viruses (i.e., HCV and ruminant pestivirus) or
tissues were normal (Leforban et al. 1990b). through a monoclonal-antibody-based HC ELISA that is
able to specifically detect antibodies to HCV while ignor-
DIAGNOSIS ing antibodies to other pestiviruses (Wensvoort et al.
1988).
Isolation of the virus may be achieved from tissues sub-
mitted for HC diagnosis (i.e., tonsil, lymph node, spleen, PREVENTION
and heparinized blood). In HC-free countries BVD and
BD must be considered as differential diagnoses of HC To prevent the infection of pigs by BVDVs or BDVs, it is
and all suspect cases of HC should be tested for BVDVs necessary to prevent direct or indirect contact with cattle
and BDVs. or sheep. Also, because natural infection with BVDV of-
The diagnosis can be readily achieved using immuno- ten occurs when pigs are fed cow’s milk or bovine offal,
fluorescence or immunoenzymatic tests on cryostat sec- such additions to pig feed should be avoided.
tions of tissues. Isolation of the virus in pestivirus-free The biological risk is also important when live-virus
cell culture can also be used. Cells can be of either vaccines are used. Indeed, the cells used for multiplica-
porcine or ruminant origin. If BVDV or BDV is isolated tion of master seed virus to prepare vaccine batches can
from pigs, it is reported to grow better with a higher titer be contamined by BVDVs or BDVs. HC and Aujeszky’s
in cells from ruminant origin than in porcine cells disease vaccines were contaminated by a pestivirus
(Wensvoort et al. 1989a). However, the definitive identi- (probably BDV) because secondary lamb kidney cells
fication of an isolate can only be done by using a panel of were used to multiply the vaccinal strains (Wensvoort
monoclonal antibodies able to link specifically to rumi- and Terpstra 1988; Vannier et al. 1988).
nant pestiviruses or to HCV. Three types of monoclonal Thirty-five of 158 tested bovine kidney or testis pri-
antibodies are used for this purpose: those that detect all mary cells were found to be contaminated by BVDV
strains of pestivirus, including HCV; those that detect (Wellemans and Van Opdenbosch 1987). For this reason,
HCV only; and those that detect ruminant pestivirus the use of primary or secondary cells to prepare modi-
strains only (Peters et al. 1986; Bolin et al. 1988; Edwards fied-live-virus vaccines has to be prohibited. Moreover,
et al. 1988; Hess et al. 1988; Wensvoort et al. 1989b; Pa- both bovine and nonbovine cell lines might be infected,
ton et al. 1995). and all cell lines have to be controlled very carefully in re-
Molecular genetic probes for the detection of pes- gard to pestiviruses (Wellemans and Van Opdenbosch
tiviruses were generated from genomic RNA of HCV af- 1987; Potts et al. 1989). The main source of contamina-
ter synthesis of cDNA and cloning. A panel of probes tion for cells is the bovine serum that is added to the nu-
was selected according to their place on the viral trient medium. The high prevalence of BVDV in the
genome—as determined by sequencing and comparison world and the existence of persistently infected calves
with the BVDV sequence. This panel seems to be able to and bovine fetuses increase the risk of contamination of
differentiate between pestiviruses (Cruciere et al. 1991; bovine serum. Rossi et al. (1980) reported that up to 62%
Schelp et al. 1991). A reverse transcriptase-polymerase of examined batches of nonirradiated bovine fetal sera
chain reaction (RT-PCR) allowed differentiation between might be found positive for BVDV. Therefore, the sys-
pestiviruses by the expected size of the amplified frag- tematic test and treatment of bovine serum and of bio-
CHAPTER 7 BOVINE VIRAL DIARRHEA AND BORDER DISEASE Vannier, Albina 117

logical products used for the preparation of vaccines are tions in experimental pigs and sheep. Br Vet J
firmly recommended. 151:181–188.
Fernelius, A. L.; Amtower, W. C.; Lambert, G.; McClurkin,
REFERENCES A. W.; and Matthews, P. J. 1973. Bovine viral diarrhoea
virus in swine: Characteristics of virus recovered from
Baker, J. C. 1987. Bovine viral diarrhoea virus: A review. J naturally and experimentally infected swine. Can J
Am Vet Med Assoc 190:1449–1458. Comp Med 37:13–20.
Barlow, R. M.; Rennie, J. C.; and Gardiner, A. C. 1980. Infec- Gillespie, J. H.; Baker, J. A.; and McEntee, K. 1960. A cyto-
tion of pregnant sheep with the NADL strain of bovine pathic strain of bovine diarrhoea virus. Cornell Vet
virus diarrhoea virus and their subsequent challenge 50:73–79.
with Border Disease II B pool. J Comp Path 90:67–72. Gillespie, J. H.; Coggins, L.; Thompson, J.; and Baker, J. A.
Becher, P.; König, M.; Paton, D. J.; and Thiel, H. J. 1995. Fur- 1961. Comparison by neutralisation tests of strains of
ther characterization of border disease virus isolates: Ev- virus isolated from virus diarrhoea and mucosal disease.
idence for the presence of more than three species with- Cornell Vet 51:155–159.
in the genus Pestivirus. Virology 209:200–206. Gutekunst, D. E., and Malquist, W. A. 1963. Separation of a
Bolin, S.; Moennig, V.; Kelso Gourley, N. E.; and Ridpath, J. soluble antigen and infectious particles of bovine virus
1988. Monoclonal antibodies with neutralizing activity diarrhoea and their relationship to hog cholera. Can J
segregate isolates of bovine viral diarrhoea virus into Comp Med Vet Sci 27:121–123.
groups. Arch Virol 99:117–123. Hamilton, A., and Timoney, P. J. 1972. BVD virus and bor-
Brownlie, J.; Clarke, M. C.; and Howard, C. J. 1984. Experi- der disease. Vet Rec 91:468.
mental production of fatal mucosal disease in cattle. Vet Harasawa, R. 1996. Phylogenetic analysis of pestivirus
Rec 114:535–536. based on the 5′-untranslated region. Acta virologica
———. 1987. Clinical and experimental mucosal disease 40:49–54.
defining a hypothesis for pathogenesis. In Pestivirus In- Hess, R. G.; Coulibaly C. O. Z.; Greiser-Wilke, I.; Moennig,
fections of Ruminants, pp. 147–156. CEC Publication, V.; and Liess, B. 1988. Identification of hog cholera virus
Eur. 10 238. Luxembourg. isolates by use of monoclonal antibodies to pestiviruses.
Carbrey, E. A.; Stewart, W. C.; Kresse, J. I.; and Snyder, M. L. Vet Microbiol 16:315–321.
1976. Natural infection of pigs with bovine viral diar- Holm Jensen, M. 1981. Detection of antibodies against hog
rhoea virus and its differential diagnosis from hog cholera virus and bovine viral diarrhoea virus in porcine
cholera. J Am Vet Med Assoc 169:1217–1219. serum: A comparative examination using CF, PLA and
Chappuis, G.; Brun, A.; Kato, F.; Dufour, R.; and Durant, M. MLVA assays. Acta Vet Scand 22:85–98.
1984. Isolement et caractérisation d’un pestivirus dans ———. 1985. Screening for neutralizing antibodies against
un foyer d’enterocolite leucopenie chez des moutons de hog cholera and/or bovine viral diarrhoea virus in Dan-
l’Aveyron. Epidemiol Santé Anim 6:117–118. ish pigs. Acta Vet Scand 26:72–80.
Collett, M.; Moennig, V.; and Horzinek, M. C. 1989. Recent Horzinek, M. C., and Van Berlo, M. F. 1987. The pestivirus-
advances in pestivirus research. J Gen Virol 70:253–266. es: Where do they belong? Ann Rech Vet 18:115–119.
Coria, M. F., and McClurkin, A.-W. 1978. Duration of active Hsien-Jue Chu and Yuan Chung Zee. 1984. Morphology of
and colostrum-derived passive antibodies to bovine vi- bovine viral diarrhoea virus. Am J Vet Res 45:845–850.
ral diarrhoea virus in calves. Can J Comp Med Vet Sci Hughes, L. E.; Kershaw, G. F.; and Shaw, I. G. 1959. “B” or
42:239–243. border disease: An undescribed disease of sheep. Vet
Cruciere, C.; Bakkali, L.; Gonzague, M.; and Plateau, E. Rec 71:313–317.
1991. cDNA probes for the detection of pestiviruses. Laude, H. 1979. Nonarbo—Togoviridae: Comparative hy-
Arch Virol Suppl 3:191–197. drodynamic properties of the pestivirus genus. Archs of
Dahle, J.; Shagemann, G.; Moennig, V.; and Liess, B. 1993. Virol 62:347–352.
Clinical, virological and serological findings after in- Leforban, Y. 1990. Profils épitopiques comparés de 18
tranasal inoculation of pigs with bovine diarrhoea virus souches de peste porcine classique: Comparaison des
and subsequent intranasal challenge with hog cholera souches isolées de formes chroniques et des autres
virus. J Vet Med B 40:46–54. souches. Rec Med Vet 166(5):455–461.
Darbyshire, J. M. 1961. A serological relationship between Leforban, Y.; Edwards, S.; Ibata, G.; and Vannier, P. 1990a.
swine fever and mucosal disease of cattle. Vet Res A blocking ELISA to differentiate hog cholera virus anti-
72:331. bodies in pig sera from those due to other pestiviruses.
Edwards, S.; Sand, J. J.; and Harkness, J. W. 1988. The ap- Ann Rech Vet 21:119–129.
plication of monoclonal antibody panels to characterize Leforban, Y.; Vannier, P.; and Cariolet, R. 1990b. Patho-
pestivirus isolates from ruminants in Great Britain. genicity of border disease and bovine viral diarrhoea
Archs Virol 102:197–206. viruses for pig: Experimental study on the vertical and
Edwards, S.; Roehe, P. M.; and Ibata, G. 1995. Comparative horizontal transmission of the viruses. Proc Int Congr
studies of border disease and closely related virus infec- Pig Vet Soc 11:204.
118 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Mengeling, W. L. 1988. The possible role of bovine viral di- terson, D. S. P.; and Sweasey, D. 1980. Pathogenicity for
arrhoea virus in maternal reproductive failure of swine. sheep foetus of bovine virus diarrhoea—mucosal dis-
Proc Int Congr Pig Vet Soc 10:228. ease virus of bovine origin. Br Vet J 136:602–611.
Olafson, P.; MacCallum, A. D.; and Fox, F. H. 1946. An ap- Terpstra, C., and Wensvoort, G. 1988. Natural infections of
parently new transmissible disease of cattle. Cornell Vet pigs with bovine viral diarrhoea virus associated with
36:205–213. signs resembling swine fever. Res Vet Sci 45:137–142.
Paton, D. J.; Sands, J. J.; Lowings, J. P.; Smith, J. E.; Ibata, G.; Vannier, P.; Leforban, Y.; Carnero, R.; and Cariolet, R. 1988.
and Edwards, S. 1995. A proposed division of the pes- Contamination of a live virus vaccine against pseudora-
tivirus genus using monoclonal antibodies, supported bies (Aujeszky’s disease) by an ovine pestivirus pathogen
by cross neutralization assays and genetic sequencing. for the pig. Ann Rech Vet 19:283–290.
Vet Res 26:92–109. Wageck Canal, C.; Hotzel, I.; Lopes de Almeida, L.; Roehe, P.
Peters, W.; Greiser-Wilke, I.; Moennig, V.; and Liess, B. M.; and Masuda, A. 1996. Differentiation of classical
1986. Preliminary serological characterisation of bovine swine fever virus from ruminant pestiviruses by reverse
viral diarrhoea virus strains using monoclonal antibod- transcription and polymerase chain reaction (RT-PCR).
ies. Vet Microbiol 12:195–200. Vet Microbiol 48:373–379.
Potts, B. J.; Sawyers, M.; Shekarchi, I. C.; Wismer, T.; and Wellemans, G., and Van Opdenbosch, E. 1987. Presence of
Huddleston, D. 1989. Peroxidase-labeled primary anti- bovine viral diarrhoea (BVD) virus in several cell lines.
body method for detection of pestivirus contamination Ann Rech Vet 18:99–102.
in cell cultures. J Virol Methods 26:119–124. Wengler, G.; Bradley, D. W.; Collett, M. S.; Heinz, F. X.;
Ramsey, F. K., and Chivers, W. H. 1953. Mucosal disease of Schlesinger, R. W.; and Strauss, J. H. 1995. Family Fla-
cattle. North Am Vet 34:629–633. viviridae. Arch Virol Suppl 10:415–427.
Rossi, C. R.; Bridgman, C. R.; and Kiesel, G. K. 1980. Viral Wensvoort, G., and Terpstra, C. 1988. Bovine viral diar-
contamination of bovine fetal lung cultures and bovine rhoea virus infections in piglets born to sows vaccinated
fetal serum. Am J Vet Res 41:1680–1681. against swine fever with contaminated vaccine. Res Vet
Schelp, C.; Dahle, J.; Krietsch, T.; Kaaden, O. R.; and Liess, Sci 45:143–148.
B. 1991. Differentiation of pestiviruses by a hog cholera Wensvoort, G.; Bloemraad, M.; and Terpstra, C. 1988. An
virus-specific genetic probe. Arch Virol Suppl enzyme immunoassay employing monoclonal antibod-
3:209–215. ies and detecting specifically antibodies to classical
Stewart, W. C.; Carbrey, E. A.; Jenney, E. W.; Brown, C. L.; swine fever virus. Vet Microbiol 17:129–140.
and Kresse, J. I. 1971. Bovine viral diarrhoea infection in Wensvoort, G.; Terpstra, C.; and De Kluijver, E. P. 1989a.
pigs. J Am Vet Med Assoc 159:1556–1563. Characterization of porcine and some ruminant pes-
Stewart, W. C.; Carbrey, E. A.; Kresse, J. I.; and Snyder, M. L. tivirus by cross neutralization. Vet Microbiol
1977. Experimental and naturally occurring bovine virus 20:291–306.
diarrhoea infections of pigs. In Proc Agric Res Seminar Wensvoort, G.; Terpstra, C.; De Kluijver, E. P.; Kragten, C.;
on Hog Cholera/Classical Swine Fever and African and Warnaar, J. C. 1989b. Antigenic differentiation of
Swine Fever, p. 262. E.E.C. EUR 5904 EN. pestivirus strains with monoclonal antibodies against
Stewart, W. C.; Miller, C. D.; Kresse, J. I.; and Snyder, M. L. hog cholera virus. Vet Microbiol 21:9–20.
1980. Bovine viral diarrhoea infection in pregnant Wrathall, A. E.; Bailey, J.; Done, J. T.; Shaw, I. G.; Winkler, C.
swine. Am J Vet Res 41:459–462. E.; Gibbons, D. F.; Patterson, D. S. P.; and Sweasey, D.
Terlecki, S.; Richardson, C.; Done, J. T.; Harkness, J. W.; 1978. Effects of experimental border disease infection in
Sand, J. J.; Shaw, I. G.; Winkler, C. E.; Duffel, S. J.; Pat- the pregnant sow. Zbl Vet Med B 25:62–69.
8 Porcine Circovirus
P. D. Lukert and G. M. Allan

In 1982 a novel virus, subsequently named porcine cir- ETIOLOGY


covirus (PCV), was identified by Tischer et al. (1982) as a
contaminant of a porcine kidney cell line (PK-15, ATCC- PCV is a member of the Circoviridae family. Its mean di-
CCLL33). It was found to contain a single-stranded, cir- ameter is 17 nm (Fig. 8.1) and its buoyant density in CsCl
cular, DNA genome and to be smaller than other DNA is 1.33–1.34. It resists inactivation when exposed to an
viruses. Largely on the basis of these properties it has re- acidic environment (pH 3), chloroform, or high temper-
cently been placed in a new family of DNA viruses called atures (56˚C and 70˚C) (Allan et al. 1994b). Its entire sin-
Circoviridae by the International Committee on Taxono- gle-stranded, circular, DNA genome comprising 1759
my of Viruses (Lukert et al. 1995). Serological surveys in bases has been cloned and sequenced (Buhk et al. 1985;
Germany (Tischer et al. 1982), Canada (Dulac and Af- Meehan et al. 1997).
shar 1989), New Zealand (Horner 1991), Great Britain PCV replicates in some porcine cell lines and is de-
(Edwards and Sands 1994), Northern Ireland (Allan et al. pendent upon cellular proteins expressed during the S-
1994b), and the United States (Hines and Lukert 1995) phase of the cell cycle (Tischer et al. 1982, 1987). Tisch-
indicated widespread infection with PCV in adult pigs in er et al. found that viral replication was enhanced
all of the countries. It is likely that the virus is ubiquitous following treatment of cell cultures with d-glucosamine,
throughout the world. resulting in an increase of up to 30% in the number of
Hines and Lukert (1994) reported PCV as a cause of cells containing PCV antigens. The virus does not cause
congenital tremors in newborn pigs, and more recently, detectable cytopathic effects (CPE) in cell culture, and vi-
Canadian and Northern Ireland workers (Clark 1997) as- ral-infected cells are detected using immunostaining
sociated PCV with a new disease called postweaning mul- techniques (Fig. 8.2). Persistent infections of cell cul-
tisystemic wasting syndrome (PMWS) in weanling pigs. tures with PCV, notably PK-15 cell lines, have been
The infectious nature of congenital tremors has been recorded. Allan et al. (1994a) have demonstrated replica-
known for many years, but PMWS appears to be an tion of PCV in monocyte/macrophage cultures derived
emerging disease in swine. If the association of PCV to from porcine bone marrow, peripheral blood, lung wash-
these clinical diseases is confirmed, this virus would rep- ings, and lymph nodes (Fig. 8.3). Mononuclear cell cul-
resent a very significant pathogen of swine. tures derived from the peripheral blood of a bovine were

8.1. Electronmicrograph of
negatively stained 17 nm virions of
PCV from a PK-15 cell culture. Bar
represents 100 nm.

119
120 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

8.2. PCV-infected cells in PK-15 cell


cultures detected using a monoclonal
antibody to PCV in an IIF assay.

8.3. Porcine macrophage cultures,


inoculated with PCV and immuno-
stained 48 hours after infection with
a monoclonal antibody to PCV using
an IIF test. Note PCV replication in
some cells.

also susceptible to infection with PCV, but not similar tative replication-associated protein of plant circoviruses
cultures from sheep or humans. McNeilly et al. (1996) and geminiviruses. A similar ORF region has been iden-
studied the effect of PCV infection on porcine alveolar tified in the genome of PBFDV. Another animal cir-
macrophage immune function in vitro. They found no ef- covirus, CAV, has no significant nucleic acid or protein
fect on the expression of Fc and complement receptors similarities (Meehan et al. 1997).
or phagocytosis. They did observe an up-regulation of
MHC class I antigen at 4 days after infection and a re- EPIDEMIOLOGY
duction in the number of cells expressing MHC class II
antigen at 8 days after infection. A significant reduction As previously stated, serological surveys in several coun-
in macrophage-mediated, mitogen-induced lymphocyte tries indicated that PCV infections are widespread and
proliferation was also observed following PCV infection. the seropositive incidence of PCV in most herds ranges
Such results could indicate that PCV infections may in- from 20% to 80%. It is quite likely that the virus is present
terfere with normal immune function. Two other cir- worldwide. Allan et al. (1994b) performed a longitudinal
coviruses, chicken anemia virus (CAV) and psittacine survey of PCV antibody profiles in sera from two litters
beak-and-feather disease virus (PBFDV), have been asso- of piglets in a PCV-infected breeder-finisher herd in
ciated with immune dysfunction (Adair et al. 1991; La- Northern Ireland. They found that maternally derived
timer et al. 1992). The genome of PCV has either six antibody to PCV disappeared at 8–9 weeks after birth
(Bukh et al. 1988) or seven (Meehan et al. 1997) overlap- and serum antibody reappeared at 13–15 weeks, indicat-
ping open reading frames (ORFs) and an absence of in- ing exposure of the animals to PCV challenge around
tergenic regions. The largest predicted ORF encodes a 36 11–13 weeks of age. This corresponded to the movement
kDa protein which exhibits high similarity with the pu- of pigs to the fattening unit within the farm.
CHAPTER 8 PORCINE CIRCOVIRUS Lukert 121

Tischer et al. (1982) and Allan et al. (1994b) were un- common signs are diarrhea, coughing, and central ner-
able to detect PCV antibodies in sera from cattle, sheep, vous system disturbances. Morbidity is usually low, with
chickens, turkeys, goats, mice, rabbits, or humans using mortality high in clinical cases. The morbidity and mor-
indirect immunofluorescence microscopy (IIF). In a later tality rates in postweaning pigs have been reported to be
study by Tischer et al. (1995) using enzyme-linked im- as high as 50% in some swine populations (Harding
munosorbent assay (ELISA) and IIF, a low level of PCV 1997).
antibody was reported in 30% of human sera, 12–69% of
mouse sera, and 35% of cattle sera. They suggest that this PATHOGENESIS
reactivity reflects infection of other species with a cir-
covirus related to PCV. These findings have yet to be con- Tischer et al. (1986) reported the first experimental
firmed. pathogenesis study with PCV. They infected 6 nine-
month-old seronegative pigs with virus purified from PK-
CLINICAL SIGNS 15 infected cells. They recovered virus from nasal swabs
from 3–6 days postinfection and from feces 13–15 days
The discussion of clinical signs will relate to two diseases postinfection. A noninfected contact pig became infect-
that have recently been associated with PCV infections: ed, and virus was recovered from the same anatomical lo-
infectious congenital tremors and PMWS. Earlier studies cations, indicating lateral transmission of the virus. No
with experimental infections of seronegative swine of clinical or histologic changes were observed. Experimen-
several ages with PCV failed to induce clinical disease tal infections of 4 one-day-old pigs also failed to produce
(Tischer et al. 1986; and Allan et al. 1995). Tischer et al. signs of disease. Antibody to PCV first appeared 1 week
(1986) were able to reisolate virus from infected pigs, after exposure and increased up to 5 weeks after infec-
and Allan et al. (1995) demonstrated the organs and tis- tion. The IIF antibody titers ranged from 1:40 to 1:160 at
sues where the virus replicated using immunostaining. the peak of the response.
Both of these studies used PCV that originated in the PK- Allan et al. (1995) infected colostrum-deprived, one-
15 (ATCC-CCLL33) cell line, and the possibility exists day-old pigs and the infection was followed by a combi-
that this is an attenuated strain of PCV since it may have nation of virus isolation and immunofluorescent (IF)
resided in this cell line for a large number of cell and staining of cryostat sections of organs and tissues. The
virus generations. virus was found to replicate in several lymphoid tissues
(thymus, spleen, and the mesenteric, bronchial, and
Infectious Congenital Tremors retropharyngeal lymph nodes), nasal mucosa, lung, and
The clinical signs of congenital tremors are highly vari- small intestine, and it was associated with
able and the tremors vary from mild to severe. The num- macrophage/monocytes, histocytes, and thymic
ber of pigs affected within a litter may also vary consid- macrophages (Figs. 8.4 and 8.5). The virus was not de-
erably. The severe tremors can cause death during the tected in kidney, brain, or liver.
first week of life because of the inability to nurse; pigs Allan et al. (1995) also reported a survey of a large
die of starvation. Pigs that live for a week usually survive, number of sera and tissues from fetuses involved in out-
and most will recover by 3 weeks. The tremors are bilat- breaks of reproductive disorders in Northern Ireland.
eral, affecting the skeletal muscle, and the tremors sub- No antibody to PCV was detected in any sera from fetus-
side when the pigs are recumbent or asleep. Tremors may es; however, two isolations of PCV were made from still-
be initiated or accentuated by external stimuli such as born pigs. They concluded that the absence of antibody
sudden noises or chilling. Some producers report that to PCV in fetal sera suggests that transplacental infection
some pigs never fully recover and continue to shake would be either extremely rare or occurs prior to fetal im-
throughout the growing and fattening period. The affect- munocompetence.
ed litters usually are from young breeding stock recently Hines (1994) isolated PCV from primary kidney cell
introduced to a farm, which would indicate exposure of cultures derived from a pig with congenital tremors. The
seronegative breeding stock to the virus at a critical stage isolate was used to infect four seronegative sows in the
of gestation. third trimester of gestation. They were infected with
5000 TCID50 (median tissue culture infectious doses) of
PMWS PCV by intranasal/oral and subcutaneous routes. The
PMWS primarily affects pigs between 6 and 8 weeks of majority of pigs that were delivered from these sows ex-
age and rarely affects suckling pigs. It has been diag- hibited tremors which persisted for 2–3 weeks. These
nosed in porcine reproductive and respiratory syndrome tremors were milder than those of the pig from which
(PRRS) positive and negative herds but the diagnosis of the virus was isolated. The virus was reisolated in cell cul-
PMWS is complicated in PRRS-positive herds by sec- tures from the kidney, small intestine, cecum, and colon
ondary infections. The clinical signs include weight loss, of five pigs from the experiment with congenital
emaciation, tachypnea, dyspnea, and jaundice. Less tremors. The virus was visualized by immunostaining of
122 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

8.4. IIF staining of cryostat section,


using monoclonal antibodies of spleen
from a pig inoculated with PCV and
sacrificed 4 days after inoculation.
Note foci of PCV antigen in spleen
cells.

8.5. IIP staining of tissue section of


thymus from a pig inoculated with PCV
and sacrificed 8 days after infection.
Note immunostaining of PCV antigen
throughout the section.

all of the same tissues plus the mesenteric lymph nodes Congenital Tremors
and liver. The only reported lesion associated with severe cases of
Gustafson and Kanitz (1974) had previously de- congenital tremors is retardation of myelin deposition in
scribed transmission of a virus causing congenital the spinal cord. Lamar (1971) reported lack of myelina-
tremors. The electron micrograph of the virus that they tion on nerves in the spinal cord from 1 to 13 days of age
isolated (Gustafson 1981) was similar to PCV, and like in affected pigs. There are no gross lesions in pigs with
PCV it did not cause a CPE in cell culture. They reported congenital tremors. The PCV antigens have not been
that the virus could induce tremors when sows were ex- demonstrated in the cells of the spinal cord nor has the
posed at several different stages of gestation. virus been isolated from these tissues.
Detailed studies on the pathogenesis of PMWS await
a definitive identification of its cause and a better under- PMWS
standing of what is needed to consistently produce the The gross and histopathologic lesions of PMWS have
associated signs and lesions. been described by Clark (1997) and Daft et al. (1996).
Carcasses of pigs with PMWS are in poor condition and
LESIONS display varying degrees of muscle wasting. The skin
shows moderate pallor and is icteric in about 20% of the
All of the pathogenesis studies with PCV have failed to cases. All lymph nodes are enlarged three to four times
report either macroscopic or microscopic changes in or- and are homogeneously white on cut surfaces.
gans or tissues from experimentally infected swine. The Lungs are diffusely noncollapsed and are heavy and
lesions normally found with congenital tremors and with firm or rubbery. Lung surfaces are mottled with gray-tan
PMWS will be discussed separately. lobules interspersed with normal yellow to pink lobules.
CHAPTER 8 PORCINE CIRCOVIRUS Lukert 123

In severe cases alveolar hemorrhages are indicated by PREVENTION


large patches of dark red or brown lobules. Gray-red at-
electatic or consolidated areas are common in cranial There are no vaccines available and none have been ex-
and middle lung lobes. perimentally tried. When PCV was shown to cause
The liver is grossly normal in half the cases, and the tremors, many in the swine industry believed that the
rest show varying degrees of mottling with mild to mod- condition was not economically severe enough to war-
erate liver atrophy. The interlobular connective tissue is rant the development of a vaccine. The emergence of
prominent, and in severe cases, all lobules are smaller PMWS may change this attitude in the future, especially
and more prominent than normal. Spleens are usually in those areas where the syndrome is apparent. Artificial
enlarged and meaty and noncongested on cut surfaces. exposure of new breeding stock to the environment of a
In half the cases the kidneys are spotted with white new farm might prevent the occurrence of congenital
foci visible on the subcapsular surfaces. All kidneys with tremors. However, until we understand the epidemiolo-
visible lesions are enlarged and pale and may be up to gy and pathogenesis of both congenital tremors and
five times normal size due to edema. There may be mot- PMWS, it is difficult to make recommendations for con-
tled areas in the cecum, and spiral colon mucosa may be trol of PCV infections.
hyperemic and petechiated.
Histopathologic changes are found in the lungs, kid- REFERENCES
neys, liver, pancreas, and all lymphoid tissues. The com-
mon finding in all of the tissues is lymphocytic-histiocyt- Adair, B. M.; McNeilly, F.; McConnell, C. D. G.; Todd, D.;
ic infiltrations. Lungs exhibit partial to complete Nelson, R. T.; and McNulty, M. S. 1991. Effects of
sloughing of epithelium with fibroplasia. Lymphoid or- chicken anemia agent on lymphokine production
gans and tissues show loss of B-cell follicles and expan- and lymphocyte transformation. Avian Dis
sion of T-cell areas. Kidneys may show cortical and 35:783–792.
medullary atrophy and edema of the intertubular con- Allan, G. M.; McNeilly, F.; Foster, C.; and Adair, B. M.
nective tissues. Glomeruli are not involved. The livers 1994a. Infection of leucocyte cell cultures from dif-
show mild to severe hepatocyte necrosis, and existing he- ferent species with porcine circovirus. Vet Microbiol
patocytes are swollen. 41:267–279.
Electron microscopy and immunoperoxidase stain- Allan, G. M.; Phenix, K. V.; Todd, D.; and Adair, B. M.
ing indicate that PCV is involved in lesions found in all 1994b. Some biological and physico-chemical prop-
organs. erties of porcine circovirus. J Vet Med 41:17–26.
Allan, G. M.; McNeilly, F.; Cassidy, J. P.; Reilly, G. A. C.;
DIAGNOSIS Adair, B. M.; Ellis, W. A.; and McNulty, M. S. 1995.
Pathogenesis of porcine circovirus: Experimental in-
The diagnosis of PCV infection can be made by isolation fections of colostrum-deprived piglets and examina-
of the virus in primary pig kidney cell cultures or in per- tion of pig foetal material. Vet Microbiol 44:49–64.
manent cell lines of pig kidney. The presence of virus is Buhk, H. J.; Tischer, I.; and Koch, M. 1985. Cloning and
detected using immunostaining. Both IIF and indirect sequencing of the porcine circovirus (PCV) genome.
immunoperoxidase staining have been used. Detection Zentralblatt für Bakteriologie, Mikrobiologie, und
of a rising titer of PCV antibodies would also indicate a Hygiene Ser A, Meeting Abstr 260:465.
recent infection. Congenital tremors is recognized by the Buhk, H. J.; Blab, I.; and Tischer, I. 1988. Replication of
clinical signs and ruling out either genetic or chemical negative strand DNA of the single-stranded porcine
causes of the tremors. As the association of PCV with circovirus genome. In Abstr Joint Meeting of Sektion
congenital tremors and PMWS becomes more substanti- Virologie and Virus Group of Soc Gen Microbiol, p.
ated, the use of immunofluorescence and in situ hy- 54.
bridization to detect virus antigen or nucleic acid in tis- Clark, E. G. 1997. Post-weaning multisystemic wasting
sue and organs will be available for diagnosis. syndrome. In Proc Am Assoc Swine Pract, Quebec
City, Canada, pp. 499–501.
TREATMENT Daft, B.; Nordhausen, R. W.; Latimer, K. S.; and Niagro,
F. D. 1996. Interstitial pneumonia and lym-
There are no known treatments for PCV infections. Pigs phadenopathy associated with circoviral infection in
severely affected with congenital tremors may have a a six-week-old pig. In Proc Amer Assoc Vet Lab Diag,
greater survival rate if help in nursing is provided by the Little Rock, Ark, p. 32.
caretaker. Dulac, G., and Afshar, A. 1989. Porcine circovirus anti-
gens in PK15 cell line (ATCC CCLL 33) and evidence
of antibodies to circovirus in Canadian pigs. Can J Vet
Res 53:431–433.
124 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Edwards, S., and Sands, J. J. 1994. Evidence of circovirus Lukert, P. D.; de Boer, G. F.; Dale, J. L.; Keese, P.; McNul-
infection in British pigs. Vet Rec 134:180–181. ty, M. S.; Randles, J. W.; and Tischer, I. 1995. The Cir-
Gustafson, D. P. 1981. Congenital tremors. In Diseases of coviridae. In Virus Taxonomy: Sixth Report of the In-
Swine, 5th ed. Ed. A. D. Leman, R. D. Glock, W. L. ternational Committee on Taxonomy of Viruses. Ed.
Mengeling, R. H. C. Penny, E. Scholl, and B. Straw. F. A. Murphy, C. M. Fauquet, D. H. L. Bishop, S. A.
Ames: Iowa State Univ Press, pp. 335–338. Ghabrial, A. W. Jarvis, G. P. Martelli, M. A. Mayo,
Gustafson, D. P., and Kanitz, C. L. 1974. Experimental and M. D. Summers. Vienna and New York: Springer-
transmission of congenital tremors in swine. Proc US Verlag, pp. 166–168.
Anim Health Assoc 78:338–345. McNeilly, F.; Allan, G. M.; Foster, J. C.; Adair, B. M.; and
Harding, J. C. 1997. Post-weaning multisystemic wasting McNulty, M. S. 1996. Effect of porcine circovirus on
syndrome (PMWS): Preliminary epidemiology and porcine alveolar macrophage function. Vet Immunol
clinical presentation. In Proc Amer Assoc Swine Immunopathol 49:295–306.
Pract, Quebec City, Canada, p. 503. Meehan, B. M.; Creelan, J. L.; McNulty, M. S.; and Todd,
Hines, R. K. 1994. Porcine Circovirus Causes Congenital D. 1997. Sequence of porcine circovirus DNA: Affini-
Tremors Type A-II Proved by Fulfilling Koch’s Postu- ties with plant circoviruses. J Gen Virol 78:221–
lates. Ph.D. diss., Univ Georgia. 227.
Hines, R. K., and Lukert, P. D. 1994. Porcine circovirus as Tischer, I.; Gelderblom, H.; Vettermann, W.; and Koch,
a cause of congenital tremors in newborn pigs. In M. 1982. A very small porcine virus with circular sin-
Proc Amer Assoc Swine Pract, Chicago, Ill, p. 344. gle stranded DNA. Nature 295:64–66.
———. 1995. Porcine circovirus: A serological survey of Tischer, I.; Mields, W.; Wolf, D.; Vaght, M.; and Griem,
swine in the United States. Swine Health and Prod W. 1986. Studies on the epidemiology and patho-
3:71–73. genicity of porcine circovirus. Arch Virol
Horner, G. 1991. Pig circovirus antibodies present in 91:271–276.
New Zealand pigs. Surveillance Wellington 18:23. Tischer, I.; Peters, D.; Rasch, R.; and Pociuli, S. 1987.
Lamar, C. H. 1971. Characterization of Peculiar Cellular Replication of porcine circovirus: Induction by glu-
Structures in the Spinal Cords of Pigs Affected with cosamine and cell cycle dependence. Arch Virol
Myoclonia Congenita. M.S. thesis, Purdue Univ. 96:37–57.
Latimer, K. S.; Steffens, W. L.; Rakich, P. M.; Niagro, F. Tischer, I.; Bode, L.; Apodaca, J.; Timm, H.; Peters, D.;
D.; Kircher, I. M.; and Lukert, P. D. 1992. Cryp- Rasch, R.; Pociuli, S.; and Gerike, E. 1995. Presence of
tosporidiosis in four cockatoos with psittacine beak antibodies reacting with porcine circovirus in sera of
and feather disease. J Am Vet Med Assoc 5:707–710. humans, mice, and cattle. Arch Virol 140:1427–1435.
Cytomegalovirus
9 N. Edington

Large basophilic intranuclear inclusions in cytomegalic separated by a translucent halo from the envelope; the
cells were first described in the mucous glands of porcine envelope of extracellular and cytoplasmic virions has ex-
turbinate mucosa by Done (1955). Their occurrence in ternal projections and a clear unit-membrane structure
pigs with rhinitis led to the designation “inclusion body (Valiček et al. 1973). As with other herpesviruses, parti-
rhinitis.” Experimental transmission aligned with ultra- cles without cores or with translucent cores, as well as cy-
structural examination (Duncan et al. 1965; Valiček et al. toplasmic or extracellular nucleocapsids without en-
1970) indicated that herpeslike virions were the causal velopes, are frequently seen in vivo and in vitro.
agent and that infection frequently also involved the
lachrymal and salivary glands, as well as renal tubular Physicochemical Properties
epithelium. Subsequently, the characteristic histologic PCMV is sensitive to chloroform and to ether (Booth et
lesions have been described in the turbinates of pigs al. 1967), but other physicochemical properties have not
from countries throughout the world. The nature and been investigated.
distribution of the lesions suggested that the agent be-
longed to that group of herpesviruses affecting humans Cultivation
and animals described as “salivary gland viruses” (Smith Propagation of PCMV in vitro has been problematic. L’E-
1959) and then as cytomegaloviruses (Weller et al. 1960; cuyer and Corner (1966) passaged an isolate five times in
Plummer 1973) and finally as Betaherpesvirinae primary pig lung cells, while Watt et al. (1973), investi-
(Matthews 1982). Inasmuch as they form a subgroup of gating the susceptibility of a wide range of tissues, found
the herpesviruses, they are slow growing and produce cy- that only lung macrophages from 3- to 5-week-old pigs
tomegaly with distinctive intranuclear inclusions; they were highly sensitive for both primary isolation and the
tend to be species-specific; they usually induce a clinical- serial propagation of virus. Cytomegaly and the forma-
ly silent infection in the adult but often a fatal, general- tion of intranuclear and, occasionally, small intracyto-
ized infection in the young animal; and they have the plasmic inclusions (Fig. 9.1) were seen 3–14 days
ability to cross the placenta and infect the fetus. Since the postinoculation (DPI), with the time of their appearance
virus of “inclusion body rhinitis” shows all these fea- dependent on the titer of PCMV used as inoculum. Prog-
tures, it is justifiable to call it porcine cytomegalovirus eny virus reaches a maximum titer of up to 5.5 median
(PCMV). PCMV also shares with the other herpesviruses cell culture infectious doses of virus/ml at 11–14 DPI.
the capacity to induce latent infection and to be excreted However, the limitations of lung macrophages are that
in the presence of circulating antibody. In susceptible they do not replicate, and therefore, primary cultures
herds the virus may cause fetal and piglet deaths, runt- must be screened for contaminant viruses, including
ing, rhinitis, and pneumonia, as well as poor liveweight PCMV and porcine reproductive and respiratory syn-
gain. However, in herds under good management the drome (PRRS) virus (PRRSV). Also, the absence of an
virus may be endemic without any apparent economic identifiable cytopathic effect (CPE) in unstained prepara-
loss. tions necessitates the reading of the macrophages either
with Giemsa or labeled antibody (Watt et al. 1973;
ETIOLOGY Plowright et al. 1976) (Fig. 9.2). Bouillant et al. (1975) re-
ported that a cell line derived from porcine fallopian
Size and Structure tube will support viral replication, while Kawamura and
The morphology of PCMV is typical of a herpes virion Matsuzaki (1996) reported that 12-O-tetrade-
with an electron-dense core 45–70 nm in diameter, a sur- canoylphorbol 13-acetate accelerated viral replication in
rounding icosahedral capsid of 80–100 nm, and an out- this system.
er single or, more rarely, double membrane 120–150 nm
across (Duncan et al. 1965; Valiček et al. 1970). The core Replication
has a usually elongated, variable outline that is oval, rec- Systematic studies of in vitro replication have not been
tangular, or dumbbell in shape. The nuclear nucleocap- reported. Infected cells are about six times as large as
sids appear to acquire an electron-dense coat, which is normal cells, with swelling of the mitochondria, endo-

125
9.1. Cultured pig lung macrophages
11 days after infection with PCMV. The
basophilic intranuclear inclusions stand
out clearly in the enlarged cells (May-
Grünwald-Giemsa; ×720). (Courtesy
R. G. Watt.)

plasmic reticulum, and Golgi apparatus (Duncan et al. (Plowright et al. 1976) (Fig. 9.2). IIF titers up to 1:64 to
1965). Much smaller acidophilic inclusions are seen, 1:128 were frequent in commercial pig sera, the test be-
sometimes in the nucleus but more often in the cyto- ing at least eightfold more sensitive than virus-neutraliz-
plasm. Ultrathin sections show that the large intranu- ing assays (VN). There was no evidence of serologic vari-
clear inclusions correlate with the formation of nucleo- ations among the limited number of U.K. isolates. Assaf
capsids, often in crystalloid arrays. The capsids acquire et al. (1982) and Tajima et al. (1993) describe ELISAs that
an electron-dense coat in the nucleus, and the envelope is are more sensitive than VN and have been adapted to
taken from the inner nuclear membrane, the virions distinguish IgG and IgM responses (Tajima et al. 1994).
coming to lie free or within membranous sacs in the cy-
toplasm. In the late stages of replication, as the cell is dis- Host Range
integrating, crystalloid arrays of viruses may also be seen PCMV appears to be host-specific both in vivo and in vit-
in the cytoplasm (Valiček et al. 1973). ro. The virus has failed to replicate in rabbits, mice, ham-
sters, chick embryos, and cattle.
Serology
An indirect immunofluorescence (IIF) test on infected EPIDEMIOLOGY
lung macrophages fixed in acetone has been described
The virus has worldwide distribution. Serologic evidence
in the United Kingdom and Japan indicates that over 90%
of herds have been exposed to infection, with more than
98% of pigs being seropositive by ELISA or IIF. The pos-
sibility of transplacental infection demands that hystero-
tomy-derived litters be carefully monitored.
The virus may be recovered from nasal and ocular se-
cretions, urine, and cervical fluids of pregnant sows ex-
posed to PCMV for the first time; isolation of virus from
the testis and epididymis (Booth et al. 1967; Shirai et al.
1985) indicates that boars should be carefully examined.
Dissemination of PCMV and infection by PCMV proba-
bly most commonly occurs via the nasal route, with the
9.2. Lung macrophage cultures showing fluorescence environment also being contaminated by urine.
after IIF, using specific PCMV antiserum. The nuclear In a longitudinal survey of winter and summer co-
staining (N) is most intense at the membrane. Cytoplas- horts of pigs, the majority shed virus nasally between 3
mic and discrete paranuclear fluorescence (P) can also be and 8 weeks of age, correlating with the mixing of stock
seen (×480). in the early postweaning period. Antibody levels de-
CHAPTER 9 CYTOMEGALOVIRUS Edington 127

creased during the period of virus excretion but rose produce a mild rhinitis in young pigs. In short-term trials
again at 8–11 weeks, continuing up to slaughter at 23 no synergistic effect was observed between PCMV and
weeks (Plowright et al. 1976). The pattern suggested that Bordetella bronchiseptica (Edington et al. 1976b), but pro-
maternal antibody was replaced by active immunity, longed exposures warrant investigation, as these results
which has been confirmed by Tajima et al. (1994). contradict field observations (Cameron-Stephen 1961;
In a smaller number of pigs, virus excretion was pre- Corner et al. 1964).
dominant at 3 weeks and had terminated at 5 weeks, sug-
gesting that early postnatal or congenital infection had PATHOGENESIS
occurred. This has been associated with mummification
and stillbirths, neonatal deaths, and runt pigs with rhini- Pregnant sows inoculated with virus were lethargic and
tis and/or pneumonia in field situations (Cameron- anorexic 14–21 DPI, coinciding with the period of
Stephen 1961; Rac 1961; Corner et al. 1964) and has been viremia, and this was immediately followed by the recov-
confirmed experimentally (L’Ecuyer et al. 1972; Edington ery of virus from nasal swabs. PCMV was isolated from
et al. 1977). The reactivation of PCMV excretion after cervical fluids later, at 30–35 DPI, and radiographic as-
the administration of corticosteroids (Edington et al. sessment of growth arrest indicated that the majority of
1976c; Narita et al. 1985) points to the possible reactiva- fetal deaths occurred in this period. This suggests that
tion of replication when new breeding stock is intro- the virus takes a further 14–20 days to replicate in the fe-
duced or when routine is otherwise disturbed. tus and that cervical shedding is of fetal, rather than ma-
The recovery of virus from the lung macrophages of ternal, origin. Neither virus nor inclusions were detected
quiescent pigs indicates that the macrophage is a reser- in the cervix or endometrium at this stage. In a small
voir of infection; this finding has serious implications in number of fetuses, the virus was detected as late as
relation to the restricted in vitro tropism of European 60–80 DPI, but it was not clear whether this represented
isolates of PRRSV (see Chapter 18). PCMV may also in utero spread by contact infection, delayed replication
modify the outcome of concurrent infections, as does in these fetuses, or virus crossing the placenta on more
murine cytomegalovirus, which modifies the host defen- than one occasion. The small number of congenitally in-
sive mechanism, particularly inhibiting T-cell function fected animals that were examined excreted virus persis-
(Booss and Wheelock 1977; Kelsey et al. 1977). This ef- tently and died suddenly with the histologic lesions of
fect has been more elegantly demonstrated for human generalized infection within 7 days of birth (Edington et
cytomegalovirus (Bonifacino 1996). Nowhere has the al. 1977). Late-term fetuses and congenitally infected
“opportunistic” component of cytomegaloviruses been neonates were all seronegative.
more devastating than in humans infected with HIV. Superinfection of sows with low levels of circulating
antibody has been associated with transplacental infec-
CLINICAL SIGNS tion (Edington et al. 1988a). Infection of the embryo
shortly after implantation certainly can occur and may
The susceptible pregnant sow is frequently listless and result in embryonic deaths. The virus was most com-
anorexic while viremic but shows no pyrexia or any oth- monly found in the meninges, Kupffer cells, and peri-
er clinical abnormality throughout pregnancy. Piglets toneal macrophages of these early fetuses (Edington et
may be born dead or die soon after birth without clinical al. 1988b).
symptoms. Others are stunted, are pale due to anemia, In experimental intranasal infections of young pigs
and show a variable edema which is often most notice- the occasional isolation of virus from nasal and conjunc-
able around the jaw and tarsal joints. Field outbreaks tival swabs prior to viremia suggests that the primary site
have included symptoms of shivering, sneezing, and res- of replication is in the nasal mucous glands or the lachry-
piratory distress. Up to 25% of the litter may be lost, with mal or harderian glands. Viremia was detected 14–16
the remainder showing poor liveweight gains. The possi- DPI in 3-week-old pigs inoculated intranasally, and over
bility of surviving pigs being persistent excreters must an extended period (5–19 DPI) in neonatal pigs (Eding-
also be considered. ton et al. 1976c). Infectivity in blood was leukocyte asso-
Uncomplicated infection with PCMV is usually a clin- ciated. Subsequently, the virus may be recovered from
ically silent event in pigs older than 3 weeks but can be nasal secretions for up to 32 days, pharyngeal and con-
fatal for the fetus or newborn pig. Such a statement is junctival shedding being of shorter duration, while the
qualified by the observations that some adult animals ex- persistence of viruria has been difficult to determine.
posed to PCMV for the first time may develop the lesions The site of tertiary viral replication varies with age.
of a generalized infection and be anorexic and lethargic At an age of 3 weeks or more, when most animals ex-
without pyrexia during the period of viremia (i.e., 14–21 perience infection, the virus disseminates to epithelial
DPI), while some piglets less than 3 weeks old develop sites—particularly the glands of the nasal mucosa, hard-
only the lesions of disseminated epithelial infection and erian and lachrymal glands, kidney tubules, and more
survive. rarely the epididymis and mucous glands of the esopha-
PCMV does not induce atrophic rhinitis but it will gus—to hepatocytes, and to duodenal epithelium.
128 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

In the fetal or neonatal pig there is a predilection for


reticuloendothelial (RE) cells, particularly capillary en-
dothelium and the sinusoids of lymphoid tissues, thus
giving rise to generalized lesions (Edington et al. 1988b).
While there is a tendency for replication to predominate
in either RE or epithelial cells, they are not mutually ex-
clusive, some individuals developing inclusions in both
groups of cells. Infectivity persists at least in lung
macrophages, since the virus is recoverable from cul-
tured lung washings from pigs that were not excreting
virus. Viral shedding has been restimulated in quiescent
animals following administration of corticosteroids (Ed-
ington et al. 1976c; Narita et al. 1985). Experimentally,
IIF antibody is detected 3 weeks after exposure to PCMV,
rising to a peak at 6 weeks and persisting until slaughter.
However, in rearing units in which PCMV is endemic, the
virus appears to be released in the presence of circulating
maternal antibody (Plowright et al. 1976).
Although inclusion bodies have occasionally been
seen in the epididymis, attempts to isolate virus from se-
men have been unsuccessful. Nor has it been possible to
establish infection by preputial inoculation. Neverthe-
less, boars can excrete virus in urine and nasal secretions.

LESIONS

It is useful to distinguish between disseminated infection


of epithelial tissues in the older pig and generalized in-
fection of RE tissues in the fetus or neonate (Edington et
al. 1976a).
9.3. The basophilic intranuclear inclusion, translucent
halo, and defined nuclear membrane are prominent in
Epithelial Lesions the enlarged superficial mucous gland epithelium of an
No macroscopic changes are seen. Histologically, the animal 18 days after experimental intranasal inoculation
characteristic basophilic intranuclear inclusion bodies (H&E; ×480).
and cytomegaly are seen in the nasal mucous glands (Fig.
9.3), acinar and duct epithelium of harderian and lachry-
mal glands, and renal tubular epithelium. In these tis- reparative lesions in the kidney are those of an intersti-
sues the number of affected cells may be extensive. Iso- tial nephritis (Kelly 1967). Sparsely distributed focal
lated inclusions are more rarely seen in the mucous gliosis is seen in the central nervous system (CNS), with
glands of the esophagus, the epithelial lining of the duc- inclusions occasionally seen in the glial cells.
tus epididymis, and the seminiferous epithelium, as well
as the epithelial lining of the duodenum and jejunum. In Generalized Lesions
these minor sites the desquamation of infected cells The salient macroscopic lesions in young pigs are wide-
leaves no trace of infection; but in the major sites lym- spread petechiae and edema. The edema most common-
phocytes, plasma cells, and macrophages accumulate ly involves the thoracic cavity and subcutaneous tissues.
around the affected epithelial tissue and invade the acini In the thorax, pericardial and pleural effusions are seen,
as the cells are shed. Where complete acini are lost, there while the pulmonary edema occurs throughout the
may be some attempted replacement by a more squa- lungs, the interlobular septae being most distended and
mous type of epithelium from the duct; but frequently in the ventral tips of the lobes appearing purple and con-
the nasal mucosa the remnants of the acinus have the ap- solidated (Fig. 9.5). The subcutaneous edema was most
pearance of focal lymphoid hyperplasia (Fig. 9.4). In nat- marked around the throat and tarsal joints. The lymph
ural cases the simultaneous infection with B. bronchisep- nodes were all enlarged and edematous, with petechiae.
tica or possibly Pasteurella spp. will produce changes in Although petechiae were also distinct at the other sites of
the ciliated epithelium lining the nasal mucosa and in the edema, they were most extensive in the kidneys, particu-
lesion of atrophic rhinitis, but these ostoid changes are larly subcapsular, so that the appearance varied from
not seen in pigs that are infected with PCMV only. The speckling to completely purple or black. In the small in-
CHAPTER 9 CYTOMEGALOVIRUS Edington 129

9.5. This pig was inoculated with PCMV when 1 day


old. It died at 16 days of age with widespread petechiae
and subcutaneous edema. The interlobular septae of the
lungs are distended with transudate, and the tips of the
apical, cardiac, and diaphragmatic lobes are also consoli-
dated.

9.4. The lamina propria is heavily infiltrated with


lymphocytes and plasma cells 24 days after inoculation ing renal tissue and in glomerular capillary endothelium
of PCMV. Many of the acini of the mucous glands still (Fig. 9.6). Hemorrhage and gliosis occur throughout the
show cytomegaly and prominent inclusion bodies (H&E; CNS, with a predilection for the choroid plexus, cerebel-
×120). lum, and olfactory lobes (Stephano-Hornedo and Eding-
ton 1987).
In field outbreaks these lesions may be complicated
by concurrent infections—mucopurulent rhinitis, pneu-
testine, hemorrhages were seen, but rarely, varying from monia, and enteritis being reported (Cameron-Stephen
complete involvement to focal areas less than 1 cm long. 1961; Corner et al. 1964).
Fetal infection did not result in pathognomonic
macroscopic lesions. The pattern was one that is typical- DIAGNOSIS
ly seen in cases of reproductive failure characterized by
stillbirth, mummified fetuses, embryonic death, and in- The presence of PCMV in a herd of pigs is most easily
fertility (SMEDI). The mummified fetuses were random- confirmed by detecting antibody by IIF or ELISA on ran-
ly distributed and sometimes of variable age. In the dom samples of sera from fattening pigs; this is also the
acute fatal syndrome most inclusions are seen in capil- most practical way of monitoring hysterotomy-derived
lary endothelial and sinusoidal cells and thus occur in all herds.
lymphoid and parenchymatous tissues. The fact that In sows showing SMEDI-like reproductive disorders,
these cells are smaller than the epithelial cells of the dis- differentiation must be made from parvovirus and possi-
seminated disease means that the cytomegaly and inclu- bly from Aujeszky’s disease. The virus may be isolated
sion bodies are not so prominent and may be overlooked from neonates or fetuses by sampling nasal mucosa,
(Fig. 9.6). The damaged endothelium is associated with lung, and kidney and by making direct cultures of lung
the local edema and/or hemorrhage and with macrophages if possible. If the carcass is kept at 4˚C, vi-
macrophages and erythrocytes in the distended extracel- ral antigen can be detected by immunostaining on frozen
lular space. Mononuclear cells with inclusions are found sections of these tissues at least 24 hours after infectivity
in blood vessels and also in the spleen; infected has been lost. Alternatively, the pathognomonic inclu-
macrophages are prevalent in alveolar tissues. Replica- sions and cytomegaly may be detected in histologic sec-
tion in hepatocytes results in focal necrosis. In the kidney tions.
the inclusions are most common in areas of differentiat- In determining the presence of PCMV as a compo-
130 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

9.6. Section of kidney from a


piglet with viral inclusions (arrows)
in capillary endothelium in both the
glomerulus and interstitial tissue.
Infected cells show enlargement but
only reach the size of normal tubu-
lar epithelium (H&E; ×240).

nent of infectious rhinitis in a herd, virus isolation must tates that offspring must be carefully monitored serolog-
be made from nasal swabs or confirmed by immuno- ically for at least 70 days.
staining from nasal scrapings.
The relatively recent emergence of PRRS as a wide- REFERENCES
spread and economically important disease of swine
leads to the speculation that PCMV may be recognized Assaf, R.; Bouillant, A. M. P.; and Di Franco, E. 1982. En-
more often in the future. Diagnostic testing for PRRSV is zyme-linked immunosorbent assay (ELISA) for the de-
now common, and porcine lung macrophages, which are tection of antibodies to porcine cytomegalovirus. Can J
often used for primary isolation of PRRSV, may fortu- Comp Med 46:183–185.
itously reveal the presence of PCMV in some cases. Bonifacino, J. S. 1996. Reversal of fortune for nascent pro-
Detecting PCMV will likely become an even more im- teins. Nature 344:405–406.
portant consideration in the future because of the ex- Booss, J., and Wheelock, E. E. 1977. Role of viraemia in the
panding field of xenotransplantation and the potential suppression of T cell function during murine cy-
for transfer of porcine tissues and organs to people. tomegalovirus infection. Infect Immunol 17:378–381.
Booth, J. C.; Goodwin, R. E. W.; and Whittlestone, P. 1967.
TREATMENT Inclusion-body rhinitis of pigs: Attempts to grow the
causal agent in tissue cultures. Res Vet Sci 8:338–345.
No specific treatment can be given, although medication Bouillant, A. M. P.; Dulac, G.; Willis, N.; Girard, A.; Greig,
against concurrent bacterial infection is used in out- A. S.; and Boulanchier P. 1975. Viral susceptibility of a
breaks of infectious rhinitis. Where rhinitis and/or cell line derived from the pig oviduct. Can J Comp Med
SMEDI have been associated with the detection of 39:450–456.
PCMV, the outbreaks have usually been self-limiting Cameron-Stephen, I. D. 1961. Inclusion-body rhinitis of
(Cameron-Stephen 1961; Corner et al. 1964). swine. Aust Vet J 37:87–91.
Corner, A. H.; Mitchell, D.; Julian, R. J.; and Meads, E. B.
PREVENTION 1964. A generalised disease in piglets associated with the
presence of cytomegalic inclusions. J Comp Pathol
In populations where PCMV is endemic, it does not ap- 74:192–199.
pear to be a problem under good management systems. Done, J. T. 1955. An “inclusion body” rhinitis of pigs. Vet
However, the introduction of new stock must always be Rec 67:525–527.
regarded as hazardous since it may stimulate latent in- Duncan, J. R.; Ramsey, E. K.; and Switzer, W. P. 1965. Elec-
fection in the presence of circulating antibody or give tron microscopy of cytomegalic inclusion disease of
rise to the problems of primary infection in susceptible swine (inclusion body rhinitis). Am J Vet Res
herds. Virus-free herds can be established by hysteroto- 26:939–946.
my, but the ability of PCMV to cross the placenta dic- Edington, N.; Plowright, W.; and Watt, R. G. 1976a. Gener-
CHAPTER 9 CYTOMEGALOVIRUS Edington 131

alised cytomegalic inclusion disease: Distribution of cy- prednisolone induced recrudescence of herpesvirus in-
tomegalic cells and virus. J Comp Pathol 86:191–202. fection. Am J Vet Res 46:1506–1510.
Edington, N.; Smith, I. M.; Plowright, W.; and Watt R. G. Plowright, W.; Edington, N.; and Watt, R. G. 1976. The be-
1976b. Relationship of porcine cytomegalovirus and B. haviour of porcine cytomegalovirus in commercial pig
bronchiseptica to atrophic rhinitis in gnotobiotic piglets. herds. J Hyg (Camb) 75:125–135.
Vet Rec 98:42–45. Plummer, G. 1973. Cytomegaloviruses of man and animals.
Edington, N.; Watt, R. G.; and Plowright, W. 1976c. Cy- Prog Med Virol 15:92–125.
tomegalovirus excretion in gnotobiotic pigs. J Hyg Rac, R. 1961. Infectious rhinitis in pigs: Laboratory aspects.
(Camb) 77:283–290. Aust Vet J 37:91–93.
Edington, N.; Watt, R. G.; Plowright, W.; Wrathall, A. E.; Shirai, J.; Narita, M.; and Iijima, Y. 1985. A cytomegalovirus
and Done, J. T. 1977. Experimental transmission of isolation from swine testicle culture. Jpn J Vet Sci
porcine cytomegalovirus. J Hyg (Camb) 78:243–251. 47:697–703.
Edington, N.; Broad, S. C.; Wrathall, A. E.; and Done, J. T. Smith, M. G. 1959. The salivary gland viruses of man and
1988a. Superinfection with PCMV initiating transpla- animals. Prog Med Virol 2:171–202.
cental infection. Vet Microbiol 16:189–193. Stephano-Hornedo, A., and Edington, N. 1987. Encefalitis
Edington, N.; Wrathall, A. E.; and Done, J. T. 1988b. Porcine experimental por citomegalovirus porcino en cerdos
cytomegalovirus in early gestation. Vet Microbiol gnotobioticos: Estudio histopatologica. Vet Mex
17:117–128. 18:189–202.
Kawamura, H., and Matsuzaki, S. 1996. Influence of 12-O- Tajima, T.; Hironao, T.; Kajikawa, T.; and Kawamura, H.
tetradecanoylphorbol 13-acetate on replication of 1993. Application of ELISA for the seroepizootiological
porcine cytomegalovirus in the 19PFT cell line. J Vet survey of antibodies against porcine cytomegalovirus. J
Med Sci 58:263–265. Vet Med Sci 55:421–424.
Kelly, D. E. 1967. Pathology of extranasal lesions in experi- Tajima, T.; Hironao, T.; Kajikawa, T.; Suzuki, Y.; and Kawa-
mental inclusion body rhinitis of pigs. Res Vet Sci muru, H. 1994. Detection of antibodies against porcine
8:472–478. cytomegalovirus from whole blood collected on the
Kelsey, D. K.; Olsen, G. A.; Overall, J. C.; and Glasgow, L. A. blood sampling paper. J Vet Med Sci 56:189–190.
1977. Alteration of host defense mechanisms by murine Valiček, L.; Smid, B.; Pleva, V.; and Meusik, J. 1970. Porcine
cytomegalovirus. Infect Immunol 18:754–760. cytomegalic inclusion disease virus: Electron microscop-
L’Ecuyer, C., and Corner, A. H. 1966. Propagation of porcine ic study of the nasal mucosa. Arch Ges Virus 32:19–30.
cytomegalic inclusion disease virus in cell cultures: Pre- Valiček, L.; Smid, B.; and Meusik, J. 1973. Electron mi-
liminary report. Can J Comp Med 30:321–326. croscopy of porcine cytomegalovirus. Arch Ges Virus
L’Ecuyer, C.; Corner, A. H.; and Randall, G. C. B. 1972. 41:344–353.
Porcine cytomegalic inclusion disease: Transplacental Watt, R. G.; Plowright, W.; Sabo, A.; and Edington, N. 1973.
transmission. Proc Int Congr Pig Vet Soc 2:99. A sensitive cell culture system for the virus of porcine in-
Matthews, R. E. E. 1982. Classification and Nomenclature of clusion body rhinitis (cytomegalic inclusion disease).
Viruses (1982). Basel, London, New York: S. Karger, pp. Res Vet Sci 14:119–121.
49–50. Weller, T. H., Hanshaw, J. B.; and Scott, D. E. 1960. Serolog-
Narita, M.; Shimizu, M.; Kawanuru, H.; Haritari, M.; and ic differentiation of viruses responsible for cytomegalic
Moriwaki, M. 1985. Pathologic changes in pigs with inclusion disease. Virology 12:130–132.
Eastern Equine Encephalomyelitis
10 F. Elvinger and C. A. Baldwin

Eastern equine encephalomyelitis (EEE) is endemic in the ed from WEEV in 1933 (TenBroeck and Merrill 1933).
eastern United States. Naturally occurring EEE in swine Nucleic acid analysis has demonstrated that EEEV has
was first reported in 1972 (Pursell et al. 1972) when 160 evolved independently into North and South American
of 200 pigs in one herd died in an outbreak in the state of antigenic varieties (Casals 1964; Weaver et al. 1991), each
Georgia. Exposure of domestic and feral swine to the with distinct virulence characteristics (Walder et al.
agent had been described earlier in serologic surveys in 1980). Isolates appear to be genetically stable over a dis-
the states of Georgia, Massachusetts, and Wisconsin crete time period within a geographic region of trans-
(Karstad and Hanson 1958, 1959; Feemster et al. 1958). mission (Roehrig et al. 1990).
The disease is diagnosed sporadically in porcine case ma-
terial at veterinary diagnostic laboratories but is likely EPIDEMIOLOGY
underrecognized in the field.
EEE first was recognized in horses on the eastern EEEV circulates endemically among many bird species
coast of the United States (Giltner and Shahan 1933; considered to be reservoirs and amplifiers of virus. Virus
TenBroeck and Merrill 1933). Its etiologic agent, EEE is transmitted from bird to bird mainly by the
virus (EEEV), soon was reported to be a human pathogen ornithophilic mosquito vector Culiseta melanuris. Other
as well (Fothergill et al. 1938). A median annual number mosquito species from genera such as Aedes and
of three human cases has been reported in the United Anopheles also transmit the EEEV. Mosquito species with
States since 1955 (Anonymous 1990). In addition, EEE opportunistic feeding habits on both birds and mam-
has been diagnosed and reported in cattle, dogs, goats, mals are responsible for the epidemic virus transmission
swine, and a variety of wild mammalian species (Pursell to mammals. The appearance of EEE in mammals is cor-
et al. 1972, 1976, 1983; Bigler et al. 1976; McGee et al. related to climatic conditions that influence population
1992). Wild and domesticated birds are susceptible to dynamics of vectors (Letson et al. 1993; Francy and Wag-
disease caused by EEEV (Scott and Weaver 1989). Clini- ner 1992). In Florida, virus circulates among birds and
cal signs range from inapparent to severe with high death infects mammals year-round, but farther north, virus ac-
losses. Histologic lesions in birds include inflammation tivity is limited to summer months.
and hemorrhage or necrosis of visceral organs, which Exposure of swine to EEEV was first noted in the late
differs from the central nervous system disease seen in 1950s in serosurveys in Georgia, Massachusetts, and
mammals (Dein et al. 1986; Tully et al. 1992; Guy et al. Wisconsin (Karstad and Hanson 1958, 1959; Feemster et
1994). al. 1958), and clinical outbreaks have been reported from
The virus is transmitted by mosquito vectors in two Georgia and Florida (Pursell et al. 1972; Elvinger et al.
distinct cycles. The endemic cycle, which involves or- 1994, 1996b). Swine tested on 9 (20%) of 45 farms in
nithophilic mosquito species, maintains the virus in southern Georgia had antibodies to EEEV, and 60 (16%)
birds considered to be reservoirs and amplifiers of the of 376 feral swine on a Georgia barrier island had serum-
virus. The epidemic cycle, which involves a variety of virus neutralization test titers that ranged from 4 to 128
mosquito species feeding on various avian and mam- (Elvinger et al. 1996b). In general, mammals have been
malian hosts, leads to the occasional transmission of considered to be dead-end hosts due to low virus titers
virus to mammalian hosts (Scott and Weaver 1989). that are insufficient to infect vectors. However, nursing
pigs that were experimentally infected developed high-
ETIOLOGY titer viremia that lasted up to 168 hours. Virus could be
recovered from oropharyngeal and rectal swabs up to 96
The virus of EEE belongs to the genus Alphavirus in the hours after virus administration, and virus could be iso-
family Togaviridae. Presently there are 26 confirmed lated from tonsils of pigs up to 20 days after experimen-
members in the genus Alphavirus, including western tal infection. Contact control pigs, in addition, have se-
equine encephalomyelitis (WEE) virus (WEEV) and roconverted in several experimental infection studies
Venezuelan equine encephalomyelitis virus (Hahn et al. (Karstad and Hanson 1958; Baldwin et al. 1997a). Thus,
1988). EEEV was described and serologically differentiat- it is conceivable that infected nursing pigs are a source of

133
134 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

virus both for vectors and, by direct transmission, for LESIONS


mammals in close proximity.
No gross lesions indicative of EEE have been observed in
CLINICAL SIGNS naturally or experimentally infected pigs (Karstad and
Hanson 1959; Pursell et al. 1972; Elvinger et al. 1994,
EEE predominantly affects nursing pigs. Clinical signs 1996a; Baldwin et al. 1997a). Microscopically, necrotiz-
include depression, anorexia, ataxia, prostration, lateral ing hepatitis appears within 12 hours of experimental in-
recumbency, convulsions, and ultimately death. Howev- fection. Lesions increase in size over the next 24 hours
er, clinical disease is not commonly observed following and resolve partially by 48 hours and completely by 72
experimental or natural exposure to EEEV. Most nursing hours after infection (Baldwin et al. 1997a). Central ner-
pigs exhibit only a transient temperature increase, even vous system lesions may not be present in pigs that die in
when high doses of EEEV are experimentally given by the acute phase of disease. Earliest lesions appear in
oral, intradermal, or intravenous routes (Elvinger et al. brain approximately 48 hours after infection. The ap-
1996a; Baldwin et al. 1997a). Predisposing factors, in- pearance of these lesions is concurrent with the appear-
cluding adverse environmental conditions or concurrent ance of mild lesions in heart and resolution of lesions in
disease, may contribute to the high mortality observed in liver. Encephalitis is characterized by inflammatory cell
natural outbreaks of EEE in swine (Pursell et al. 1972; infiltration, disseminated perivascular cuffing, neuronal
Elvinger et al. 1994). Experimental infection of older an- necrosis and neuronophagia, nodules of glial prolifera-
imals did not lead to clinical disease (Karstad and Han- tion, and malacia (Pursell et al. 1972; Elvinger et al. 1994,
son 1959), and EEE has not been described in adult 1996a; Baldwin et al. 1997a). Viral inclusions have not
swine. The oldest pig recorded with clinical EEE was a 2- been observed. Initially, neutrophils predominate in in-
month-old female (Pursell et al. 1983). filtrates and perivascular cuffs, whereas lymphocytes are
more common at later stages. In addition, histiocytes,
PATHOGENESIS adventitial cells, eosinophils, and cellular debris are ob-
served. Hyaline or granular thrombi are found in blood
Natural infection of pigs likely occurs through transmis- vessels. Lesions occur primarily in gray matter of brain
sion of virus by hematophagous vectors; experimental and spinal cord, but white matter may also be affected.
infection has been achieved by intracranial, intradermal, Patches of inflammatory cells are noted in the meninges.
intravenous, and oral routes of virus administration Multifocal myocardial necrosis has been reported in nat-
(Karstad and Hanson 1959; Pursell et al. 1972; Baldwin urally infected pigs and reproduced in experimental in-
et al. 1997a). Rectal temperature increases within 24 fection (Elvinger et al. 1994, 1996a; Baldwin et al.
hours of virus administration and is elevated for less 1997a). Swine have not developed hemorrhagic intesti-
than 12 hours in all experimentally infected pigs; howev- nal lesions such as those described in whooping cranes
er, clinical signs of central nervous system disease are ob- and emus (Dein et al. 1986; Tully et al. 1992). Pigs that
served in only a few pigs between 18 and 72 hours after survived experimental infection with no apparent clini-
virus administration (Baldwin et al. 1997a). Circulating cal signs occasionally had lesions of mild encephalitis
virus was isolated 6 hours after experimental infection, with lymphocytic perivascular cuffs, focal areas of glio-
regardless of route of infection, and was present in sis, and foci of myocardial necrosis that were partially
blood for up to 168 hours. Neutralizing antibodies are mineralized and surrounded by macrophages (Baldwin
detectable approximately 120 hours after experimental et al. 1997a).
infection. The virus also was recovered from oropharyn-
geal and rectal swabs 6–96 hours after infection. After DIAGNOSIS
the acute viremic phase, EEEV was isolated only from
tonsils and central nervous tissues. The virus was isolat- Death losses in nursing pigs following clinical signs of
ed or demonstrated by oligonucleotide probe in tonsils central nervous system disease in EEE endemic areas are
up to 20 days after experimental infection. Consequent- indicators of virus activity, particularly when climatic
ly, tonsillar tissues of infected pigs could be a significant conditions are conducive to the development of vector
source of virus dispersion, especially since contact pigs populations. Histopathologic lesions may first appear in
become infected (Karstad and Hanson 1958; Baldwin et liver and myocardium, but lesions in brain consistent
al. 1997a). Oligonucleotide probing revealed EEEV in with EEE may not have developed yet in pigs that died
transient liver lesions that appeared early during the during the early viremic phase. A definitive diagnosis can
viremic phase of the infection, prior to the appearance of be supported by virus isolation or detection of virus
lesions in myocardium and the central nervous system RNA or antigen. Serologic evidence of exposure in sur-
(Baldwin et al. 1997a, b). The presence of the virus in liv- viving pigs is a good indicator of EEEV activity, although
er lesions may indicate virus tropism for hepatic tissue antibody titers may be due to earlier exposure of sows
and viral replication in the liver. and transmission of antibodies through colostrum.
CHAPTER 10 EASTERN EQUINE ENCEPHALOMYELITIS Elvinger, Baldwin 135

A wide spectrum of tests to isolate virus, demon- developed. Although two of six experimentally infected
strate and identify EEEV RNA or antigen, or detect anti- pigs from vaccinated sows were viremic, virus titers were
bodies to EEEV is used to diagnose natural and experi- lower and duration of viremia was shorter than in pigs
mental infections with EEEV. The virus has been isolated from nonvaccinated sows. Pigs from vaccinated sows are
in cell cultures using the Vero (continuous African green not likely to be a source of virus for infection of mosqui-
monkey) cell line, BHK-21 (baby hamster kidney) cells, to vectors. Naturally infected sows should transfer pro-
or other vertebrate cell lines susceptible to EEEV (Fergu- tective antibodies against EEEV to their litters. Dams
son et al. 1979; Monath et al. 1981; Scott et al. 1988; transferred maternal antibodies 5 months after vaccina-
Elvinger et al. 1996a). Virus antigen has been demon- tion even though antibody levels in serum from dams
strated in tissues by immunohistochemistry and en- were below detection level by virus-serum neutralization
zyme-linked immunosorbent assay (Monath et al. 1981; test. Maternally derived antibodies were measured in
Scott et al. 1988; Patterson et al. 1996), and EEEV RNA pigs for up to 11 weeks after colostrum intake and should
was detected with DNA in situ hybridization or after am- be protective until natural susceptibility to clinical dis-
plification with polymerase chain reaction (Vodkin et al. ease is outgrown.
1993; Armstrong et al. 1995; Gregory et al. 1996). Meth- The second approach to prevention of EEE is control
ods used to detect antibodies to EEEV include virus- of exposure to EEE vectors. Incident cases of EEE in any
serum neutralization test, enzyme-linked immunosor- mammalian or avian species can serve as sentinel cases
bent assay, complement fixation test, and signaling the need to initiate application of insecticides
hemagglutination inhibition test (Ferguson et al. 1979; by local and state governments. Some communities in
Tesh and McCammon 1979; Sahu et al. 1994). endemic areas have established surveillance programs
Fresh and formalin-fixed specimens of brain, spinal for testing sera of sentinel birds for the appearance of
cord, liver, heart, and tonsil should be submitted for antibodies to EEE and other arboviral diseases and/or
histopathologic examination and virus isolation when for trapping mosquitoes to determine the presence of
EEE is suspected in a swine herd. Virus isolation can be EEEV or other viruses. Pheasants are more sensitive for
attempted on oropharyngeal swabs from herd mates, detection of EEEV activity than chickens; however, chick-
since EEEV can persist in tonsillar tissue. Precautions ens are more commonly used as sentinel birds (Morris et
should be taken to avoid accidental human infection al. 1994; Day and Stark 1996). Detection of virus activity
when collecting the specimens. Serum from affected and in an area prompts the release of warnings to the public
nonaffected pigs should be submitted for serologic ex- through news media and implementation or expansion
amination, although disease and death likely will pre- of vector control measures, including a general, area-
cede the onset of a detectable immune response. wide application of pesticides like malathion or fenthion
(Carter et al. 1981). One southern Georgia county initiat-
TREATMENT AND PREVENTION ed chemical mosquito control following the diagnosis of
EEE in swine (Elvinger et al. 1994). Insecticidal interven-
Treatment of swine clinically affected with EEE has not tions are costly and are designed to prevent human cas-
been attempted. No treatment of affected horses has es. Costs for treatment of a human case have been esti-
been described, and therapeutic efforts in human cases mated to range from $20,000, when there are no residual
are directed toward management of symptoms (Craven sequelae, up to a lifetime cost of $3 million for patients
1991). that suffer permanent sequelae (Villari et al. 1995). Aeri-
Two routes for prevention of EEE can be taken: (1) al application of insecticides benefits swine producers in
vaccination of animals at risk and (2) vector control. endemic areas directly by preventing outbreaks and eco-
In endemic areas, most horses are routinely vaccinat- nomic losses and indirectly by preventing amplification
ed at least once, but usually twice, per year to prevent of EEEV in pigs that would put the producer and farm la-
EEEV infection (Wilson et al. 1986; Gibbs et al. 1988). bor at risk of contracting infection.
Human experimental vaccines are available for research
and for laboratory personnel who may become exposed REFERENCES
to the virus. Vaccination of sows is an option for protect-
ing swine producers from catastrophic losses due to Anonymous. 1990. Arboviral infections of the central ner-
EEEV infection in pigs. Sows vaccinated experimentally vous system—United States, 1989. Morb Mort Weekly
with commercial equine vaccines have developed mater- Rep 39:407–417.
nal antibodies that were transmitted to pigs via Armstrong, P.; Borovsky, D.; Shope, R. E.; Morris, C. D.;
colostrum (Elvinger et al. 1996a). Maternally derived an- Mitchell, C. J.; Karabatsos, N.; Komar, N.; and Spielman,
tibodies are protective for experimentally infected, early- A. 1995. Sensitive and specific colorimetric dot assay to
weaned pigs. Pigs from vaccinated sows did not develop detect eastern equine encephalomyelitis viral RNA in
the fever, clinical signs, or histopathologic lesions related mosquitoes (Diptera: Culicidae) after polymerase chain
to EEEV infection that the pigs from nonvaccinated sows reaction amplification. J Med Entomol 32:42–52.
136 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Baldwin, C. A.; Liggett, A. D.; Elvinger, F.; and Tang, K. N. weather variables to develop a predictive model for
1997a. Experimental inoculation of pigs with a porcine equine cases. In Animal Health Insight.
isolate of eastern equine encephalomyelitis virus. Un- USDA:APHIS:VS Animal Health Information. Spring
published. 1992, pp. 1–8.
———. 1997b. Demonstration of eastern equine en- Gibbs, E. P. J.; Wilson, J. H.; and All, B. P., III. 1988. Studies
cephalomyelitis virus by DNA in situ hybridization in on passive immunity and the vaccination of foals
liver lesions of experimentally infected pigs. Unpub- against eastern equine encephalitis in Florida. In Equine
lished. Infectious Diseases V. Ed. D. G. Powell. Proc 5th Int
Bigler, W. J.; Lassing, E. B.; Buff, E. E.; Prather, E. C.; Beck, E. Conf, Lexington, Ky, pp. 201–205.
C.; and Hoff, G. L. 1976. Endemic eastern equine en- Giltner, L. T., and Shahan, M. S. 1933. The 1933 outbreak of
cephalomyelitis in Florida: A twenty-year analysis. infectious equine encephalomyelitis in the eastern
1955–1974. Am J Trop Med Hyg 25:884–890. states. No Am Vet 14:25–27.
Carter, J. H.; Lee, K. R.; and Chancy, O. J., Sr. 1981. The op- Gregory, C. R.; Latimer, K. S.; Niagro, F. D.; Campagnoli, R.
erational response to the 1980 outbreak of eastern P; Steffens, W. L.; and Ritchie, B. W. 1996. Detection of
equine encephalomyelitis in southeastern Georgia. Mos- eastern equine encephalomyelitis virus RNA in forma-
quito News 41:785–789. lin-fixed, paraffin-embedded tissues using DNA in situ
Casals, J. 1964. Antigenic variants of eastern equine en- hybridization. J Vet Diagn Invest 8:151–155.
cephalitis virus. J Exp Med 119:547–565. Guy, J. S.; Barnes, H. J.; and Smith, L. G. 1994. Experimen-
Craven, R. B. 1991. Togaviruses. In Textbook of Human Vi- tal infection of young broiler chickens with eastern
rology, 2d ed. Ed. R. B. Belshe. St. Louis: Mosby-Year equine encephalitis virus and highlands J virus. Avian
Bk., pp. 663–674. Dis 38:572–582.
Day, J. F., and Stark, L. M. 1996. Eastern equine encephalitis Hahn, C. S.; Lustig, S.; Strauss, E. G.; and Strauss, J. H. 1988.
transmission to emus (Dromaius novaehollandiae) in Vo- Western equine encephalitis virus is a recombinant
lusia County, Florida: 1992 through 1994. J Am Mosqui- virus. Proc Nat Acad Sci 85:5997–6001.
to Contr Assoc 12:429–436. Karstad, L. H., and Hanson, R. P. 1958. Infections in wildlife
Dein, F. J.; Carpenter, J. W.; Clark, G. G.; Montali, R. J.; with the viruses of vesicular stomatitis and eastern
Crabbs, C. L.; Tsai, T. F.; and Docherty, D. E. 1986. Mor- equine encephalomyelitis. In Trans 23d North Am
tality of captive whooping cranes caused by eastern Wildlife Conf, pp. 175–185.
equine encephalitis virus. J Am Vet Med Assoc ———. 1959. Natural and experimental infections in swine
189:1006–1010. with the virus of eastern equine encephalitis. J Infect Dis
Elvinger, F.; Liggett, A. D.; Tang, K. N.; Harrison, L. R.; Cole, 105:293–296.
J. R., Jr.; Baldwin, C. A.; and Nessmith, W. B. 1994. East- Letson, G. W.; Bailey, R. E.; Pearson, J.; and Tsai, T. F. 1993.
ern equine encephalomyelitis virus infection in swine. J Eastern equine encephalitis (EEE): A description of the
Am Vet Med Assoc 205:1014–1016. 1989 outbreak, recent epidemiologic trends, and the as-
Elvinger, F.; Baldwin, C. A.; Liggett, A. D.; Tang, K. N.; and sociation of rainfall with EEE occurrence. Am J Trop
Dove, C. R. 1996a. Protection of pigs by vaccination of Med Hyg 49:677–685.
pregnant sows against eastern equine encephalomyelitis McGee, E. D.; Littleton, C. H.; Mapp, J. B.; and Brown, R. J.
virus. Vet Microbiol 51:229–239. 1992. Eastern equine encephalomyelitis in an adult cow.
Elvinger, F.; Baldwin, C. A.; Liggett, A. D.; Tang, K. N.; and Vet Pathol 29:361–363.
Stallknecht, D. E. 1996b. Prevalence of exposure to east- Monath, T. P.; McLean, R. G.; Cropp, C. B.; Parham, G. L.;
ern equine encephalomyelitis virus in domestic and fer- Lazuick, J. S.; and Calisher, C. H. 1981. Diagnosis of east-
al swine in Georgia. J Vet Diagn Invest 8:481–484. ern equine encephalomyelitis by immunofluorescent
Feemster, R. F.; Wheeler, R. E.; Daniels, J. B.; Rose, H. D.; staining of brain tissue. Am J Vet Res 42:1418–1421.
Schaeffer, M.; Kissling, R. E.; Hayes, R. O.; Alexander, E. Morris, C. D.; Baker, W. G.; Stark, L.; Burgess, J.; and Lewis,
R.; and Murray, W. A. 1958. Field and laboratory studies A. L. 1994. Comparison of chickens and pheasants as
on equine encephalitis. New England J Med sentinels for eastern equine encephalitis and St. Louis
259:107–113. encephalitis viruses in Florida. J Am Mosquito Contr As-
Ferguson, J. A.; Reeves, W. C.; and Hardy, J. L. 1979. Studies soc 10:545–548.
on immunity to alphaviruses in foals. Am J Vet Res Patterson, J. S.; Maes, R. K.; Mullaney, T. P.; and Benson, C.
40:5–10. L. 1996. Immunohistochemical diagnosis of eastern
Fothergill, L. D.; Dingle, J. H.; and Connerley, M. L. 1938. equine encephalomyelitis. J Vet Diagn Invest 8:156–160.
Human encephalitis caused by virus of eastern variety of Pursell, A. R.; Peckham, J. C.; Cole, J. C., Jr.; Stewart, W. C.;
equine encephalomyelitis. New Engl J Med and Mitchell, F. E. 1972. Naturally occurring and artifi-
219:411–422. cially induced eastern encephalomyelitis in pigs. J Am
Francy, D. B., and Wagner, B. A. 1992. Equine en- Vet Med Assoc 161:1143–1147.
cephalomyelitis in the United States, 1980–1990: Use of Pursell, A. R.; Mitchell, F. E.; and Seibold, H. R. 1976. Natu-
CHAPTER 10 EASTERN EQUINE ENCEPHALOMYELITIS Elvinger, Baldwin 137

rally occurring and experimentally induced eastern en- Tesh, M. J., and McCammon, J. R. 1979. Detection of ar-
cephalomyelitis in calves. J Am Vet Med Assoc bovirus antibodies in avian sera by the complement fix-
169:1101–1103. ation-inhibition test. Am J Vet Res 40:299–301.
Pursell, A. R.; Hall, R. F.; Sangster, L. T.; Waller, E.; and Be- Tully, T. N.; Shane, S. M.; Poston, R. P.; England, J. J.; Vice,
dell, D. M. 1983. Eastern encephalomyelitis: The Geor- C. C.; Cho, D. Y.; and Panigrahy, B. 1992. Eastern equine
gia experience 1982–1983. In 26th Annu Proc Amer As- encephalitis in a flock of emus (Dromaius novaehol-
soc Vet Lab Diagn, pp. 383–392. landiae). Avian Dis 36:808–812.
Roehrig, J. T.; Hunt, A. R.; Chang, G. J.; Sheik, C. B.; Bolin, Villari, P.; Spielman, A.; Komar, N.; McDowell, M.; and
R. A.; Tsai, T. F.; and Trent, D. W. 1990. Identification of Timperi, R. J. 1995. The economic burden imposed by a
monoclonal antibodies capable of differentiating anti- residual case of eastern encephalitis. Am J Trop Med
genic varieties of eastern equine encephalitis viruses. Hyg 52:8–13.
Am J Trop Med Hyg 42:394–398. Vodkin, M. H.; McLaughlin, G. L.; Day, J. F.; Shope, R. E.;
Sahu, S. P.; Alstad, A. D.; Pedersen, D. D.; and Pearson, J. E. and Novak, R. J. 1993. A rapid diagnostic assay for east-
1994. Diagnosis of eastern equine encephalomyelitis ern equine encephalomyelitis viral RNA. Am J Trop Med
virus infection in horses by immunoglobulin M and G Hyg 49:772–776.
capture enzyme-linked immunosorbent assay. J Vet Di- Walder, R.; Jahrling, P. B.; and Eddy, G. A. 1980. Differenti-
agn Invest 6:34–38. ation markers of eastern equine encephalitis (EEE)
Scott, T. W., and Weaver, S. C. 1989. Eastern equine en- viruses and virulence. Zbl Bact Microbiol Hyg Suppl
cephalomyelitis virus: Epidemiology and evolution of 9:237–250.
mosquito transmission. Adv Virus Res 37:277–328. Weaver, S. C.; Scott, T. W.; and Rico-Hesse, R. 1991. Molec-
Scott, T. W.; Olson, J. G.; All, B. P., III; and Gibbs, E. P. J. ular evolution of eastern equine encephalomyelitis virus
1988. Detection of eastern equine encephalomyelitis in North America. Virol 182:774–784.
virus antigen in equine brain tissue by enzyme-linked Wilson, J. H.; Rubin, H. L.; Lane, T. J.; and Gibbs, E. P. J.
immunosorbent assay. Am J Vet Res 49:1716–1718. 1986. A survey of eastern equine encephalomyelitis in
TenBroeck, C., and Merrill, M. H. 1933. A serological differ- Florida horses: Prevalence, economic impact, and man-
ence between eastern and western equine en- agement practices, 1982–1983. Prev Vet Med 4:261–
cephalomyelitis virus. In 31st Proc Soc Exp Biol Med, pp. 271.
217–220.
Encephalomyocarditis Virus
11 H. S. Joo

Encephalomyocarditis virus (EMCV) has been recog- Many properties of EMCV are shared by other picor-
nized as a swine pathogen since it was first isolated dur- naviruses. It is ether-resistant and stable over a wide
ing an outbreak of acute disease in Panama (Murnane et range of pH. It is inactivated after 30 minutes at 60˚C,
al. 1960). Subsequently, outbreaks have been reported in but some strains have shown a marked thermal stability.
many different countries. Major epizootics with high EMCV replicates well in cell cultures originating from
mortality in baby pigs have been reported in Florida several animal species, including rodents, swine, and hu-
from 1960 to 1966 (Gainer 1967), New South Wales, mans. The virus also replicates in mice and chicken em-
Australia, in 1970 and 1984 (Acland and Littlejohns bryos and is pathogenic to many laboratory animal
1975; Seaman et al. 1986), and Cuba (Ramos et al. 1983). hosts.
Clinical disease has also been reported in New Zealand Acute fatal disease is most often produced in mice
(Sutherland et al. 1977), South Africa (Williams 1981), and hamsters following inoculation by various routes.
Brazil (Roehe et al. 1985), Italy (Sidori et al. 1988), Neurologic disease due to encephalitis is observed, but
Switzerland (Hani et al. 1992), Greece (Paschaleri-Pa- myocarditis may also be seen at necropsy. Pathogenicity
padopoulou et al. 1992), and Canada (Dea et al. 1991), in rats, guinea pigs, rabbits, and monkeys appears to
while detection of EMCV antibodies without clinical dis- vary depending on the age of the animals and the virus
ease has been reported in the United Kingdom (Sangar et strains used.
al. 1977) and Japan (Kudo et al. 1995). The virus has hemagglutinating ability with guinea
Besides baby pig mortality, EMCV has been implicat- pig, rat, horse, and sheep erythrocytes, and most EMCV
ed as a cause of reproductive failure. Reproductive prob- strains require KCl-borate (0.12M KCl; 0.05M H3BO3)
lems associated with EMCV infections have been report- buffered solution for an optimal hemagglutination reac-
ed in Cuba (Gomez et al. 1982), Australia (Littlejohns tion. Some differences in hemagglutinating activity be-
1984; Links et al. 1986), Canada (Dea et al. 1991), South tween EMCV strains have been reported (Sangar et al.
Korea (Park et al. 1990), Belgium (Koenen et al. 1992), 1977; Kim et al. 1991b). The EMCV virions contain a sin-
and the United States (Joo et al. 1988; Kim et al. 1989a; gle strand of RNA of molecular weight 2.6 × 106 daltons,
Christianson et al. 1992). which makes up 31% of the virion mass and is enclosed
in a protein capsid shell. The viral proteins show four
ETIOLOGY nonidentical polypeptide bands in SDS-polyacrylamide
gel electrophoresis. Other molecular characteristics, in-
The EMCV group is classified as genus Cardiovirus of the cluding the nucleotide sequence of the RNA, are rela-
family Picornaviridae. The first EMCV isolation was tively well understood.
made from a chimpanzee with myocarditis in Florida
(Helwig and Schmidt 1945), and subsequently, the virus EPIDEMIOLOGY
was isolated from a variety of animal species over a wide
geographic area (Tesh and Wallace 1977). During the The EMCV group is generally regarded as a rodent virus,
1940s several virus strains, including Columbia-SK, although the virus naturally infects a wide range of ver-
Mengo, and M.M., were isolated. These viruses were tebrate species. The host range includes chimpanzees,
found to be antigenically similar and are considered to monkeys, elephants, lions, squirrels, mongooses,
be in the same group as EMCV. EMCV has been reported racoons, and swine. Pigs are the domestic animals most
to be antigenically related to the cricket paralysis virus of susceptible to clinical disease by EMCV infection.
insects (Tinsley et al. 1984). Rats and mice are believed to be the principal reser-
voir of the virus (Tesh and Wallace 1977). EMCV neu-
tralizing antibodies have been demonstrated in sera
from wild rats, Rattus spp., trapped in several areas of
the United States and Canada. Many rodents are suscep-
The author deeply acknowledges Dr. H. M. Acland and Dr. I. R. Little-
johns, the authors of the EMCV chapter in the 6th edition of Diseases of tible to experimental infection, showing high levels of
Swine. The present chapter is based on their chapter in the 6th edition. the virus in their tissues, and infected rodents excrete the

139
140 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

virus in their feces and urine. EMCV has been isolated shown temperatures up to 41˚C and death between days
from dried feces and from intestines of rats and mice 2 and 11, usually 3–5 days, postinoculation or occasional
captured on farms where swine disease had previously recovery with chronic myocarditis. The severity of the
occurred (Gainer 1967; Acland and Littlejohns 1975). Al- disease in pigs appears to vary depending on the virus
though the virus has been isolated from mosquitoes strain and the age of the pigs at the time of infection.
caught in Africa, Brazil, and the United States, and from Mortality approaching 100% is usually confined to pigs
ticks in India (Tesh and Wallace 1977), there is no evi- of preweaning age. Infections in pigs from postweaning
dence that natural EMCV infection in swine is vector- age to adulthood are usually subclinical, although some
borne. mortality may be observed even in adult pigs.
The most important sources for swine infection ap- In breeding females, clinical signs vary from no obvi-
pear to be feed or water contaminated with the virus by ous illness to severe reproductive problems. The first
rats, other rodents, or infected carcasses. Feed contami- clinical signs of anorexia and fever may be observed in
nated with infected carcasses may contain high doses of infected sows. Such sows may show various forms of re-
the virus, since a large amount of the virus is found in the productive failure, including abortions and increased
carcasses. An episode of lion deaths at a zoo was found numbers of mummified and stillborn fetuses (Dea et al.
to be due to feeding carcasses of African elephants that 1991). Increased neonatal deaths and preweaning mor-
had died of EMCV infection (Simpson et al. 1977). tality will also be observed.
The mode of virus transmission is not clear, but ro-
dent-to-pig transmission is probably common. Several PATHOGENESIS
outbreaks in Australia were found to be closely associat-
ed with rat and mouse plagues (Acland and Littlejohns The natural infection of swine occurs most likely by the
1975; Seaman et al. 1986). Pig-to-pig transmission has oral route. Following experimental oral infection in
been questioned because sentinel pigs failed to become piglets, viremia was demonstrated as early as 2 days
infected after contact with experimentally infected and postinoculation and persisted for 2–4 days (Craighead et
sick pigs (Littlejohns and Acland 1975; Horner and al. 1963). The virus was present in the feces for as long as
Hunter 1979). However, the role of the infected pigs in 9 days following oral administration. The persistence of
natural transmission, either directly or indirectly, cannot the virus beyond the viremic period suggests some viral
be excluded, since infected pigs have been shown to ex- replication in the intestine. Large amounts of the virus
crete the virus at least for a short period. were found in the spleen and mesenteric lymph nodes,
The virulence of EMCV isolates in pigs may vary de- indicating that lymphatic tissue was a site of virus repli-
pending on the virus strains maintained in reservoir cation. The highest virus titers were recovered from heart
hosts within geographical areas. For example, Australian muscle in both experimental and natural infections, and
strains were shown to be more virulent in experimental myocardial lesions were predominant at necropsy. Liver,
infection of pigs than New Zealand strains (Littlejohns pancreas, and kidney also contained virus, usually at a
and Acland 1975; Horner and Hunter 1979), and certain greater concentration than in blood. Animals that sur-
isolates in Florida were found to cause only myocarditis, vived the acute disease produced EMCV antibodies. Af-
without death (Gainer et al. 1968). ter antibody formation, the virus was no longer recover-
To date there is no clear evidence to support the role able. The course of infection in swine appears to be
of EMCV as a pathogen in livestock other than swine. Ev- influenced by virus strain, viral dose, history, and level of
idence for human infection with EMCV has been demon- viral passage and susceptibility of the individual animal.
strated by antibody detection in human populations The pathogenesis of transplacental infection with
(Tesh 1978), but there are no reports that the virus caus- EMCV in pregnant sows is not well understood. Follow-
es human heart disease. However, EMCV has been uti- ing intramuscular infection of pregnant sows with EM-
lized in different research models for human diseases, in- CV, a transplacental infection with fetal deaths was ob-
cluding the investigation of the pathogenesis of served in one of the three sows infected in late gestation,
myocarditis, vasculitis, and viral-induced diabetes melli- while fetal infection in sows during early pregnancy was
tus. not conclusive (Love and Grewal 1986). Infected and
dead fetuses showed myocardial lesions varying from
CLINICAL SIGNS multiple small foci to large diffuse patches. Some diffi-
culty in producing experimental reproductive disease in
EMCV infection in young pigs is characterized most pregnant sows was also observed with a U.S. isolate.
commonly by acute disease with sudden deaths due to However, transplacental infection was successful when
myocardial failure. Other clinical signs, such as anorexia, the virus was passaged in young pigs rather than cell cul-
listlessness, trembling, staggering, paralysis, or dyspnea, ture before inoculation (Christianson et al. 1992). Fetal
are also observed. Experimentally infected swine (Craig- deaths following infection in sows appear to occur as ear-
head et al. 1963; Littlejohns and Acland 1975) have ly as 2 weeks postinfection. At this time it is not known
CHAPTER 11 ENCEPHALOMYOCARDITIS VIRUS Joo 141

whether all EMCV strains can cause both the typical my-
ocarditis observed in young pigs and reproductive fail-
ure.
Pathogenic variability in swine fetuses by different
EMCV isolates has been reported (Kim et al. 1989b). Lit-
tle pathogenicity was observed following infection of
swine fetuses in utero with laboratory-passaged strains.
Pathogenic effects of field isolates were obvious in fetus-
es of both mid- and late gestational ages. It has been sug-
gested that virulent strains may have been attenuated
and lost pathogenicity by prolonged laboratory pas-
sages.
Among the laboratory animals, clinical manifesta-
tions and the pathogenesis of EMCV infection were vari-
able. Experimental infection has resulted in fatal my-
ocarditis in adult guinea pigs and in some strains of
white rats. Owl monkeys (night monkeys) and mar-
mosets were reported to be highly susceptible to infec-
tion. The virus has seldom been pathogenic to rabbits
and rhesus monkeys, causing only inapparent infections
despite high viremic levels. It is interesting to note that
the pathogenicity of EMCV can be modified by laborato- 11.1. Heart of pig with EMCV infection showing
ry manipulations, and EMCV variants that differ in or- multiple white foci in myocardium.
gan tropism and in pathogenicity have been developed.
Certain strains cause predominantly fatal en-
cephalomyelitis (encephalotropism) or widespread my- tion of mononuclear cells (Fig. 11.3), vascular conges-
ocardial damage (cardiotropism) or even specific de- tion, edema, and degeneration of the myocardial fibers
struction of pancreatic beta cells (pancreotropism) with necrosis. Mineralization of necrotic heart muscle is
(Craighead 1966; Cerutis et al. 1989). common but not always present. Congestion with
meningitis, perivascular infiltration with mononuclear
LESIONS cells, and some neuronal degeneration may be observed
in the brain (Murnane et al. 1960; Gainer et al. 1968;
Pigs dying in the acute phase of cardiac failure may show Acland and Littlejohns 1975). Nonsuppurative en-
only epicardial hemorrhage or no gross lesions. At cephalitis and myocarditis have also been observed in
necropsy of experimentally infected young pigs, hy- swine fetuses with natural EMCV infection (Kim et al.
dropericardium, hydrothorax, pulmonary edema, and 1989a).
ascites are frequently observed, and gross myocardial le-
sions are prominent. The heart is usually enlarged, soft, DIAGNOSIS
and pale, with visible yellowish or white necrotic foci
(2–15 mm in diameter) or large ill-defined pale areas The clinical history of reproductive failure, along with
(Fig. 11.1). The lesions are most commonly observed on high preweaning mortality, is a useful tip in the diagno-
the epicardium of the right ventricle and may extend to sis of EMCV infection. EMCV-induced reproductive
varying depths within the myocardium. The virus is problems should be differentiated from those of infec-
present in heart muscle in most cases, even when my- tion with other pathogens. EMCV causes reproductive
ocardial lesions are minimal or occasionally absent problems in sows of all parities, along with high neona-
(Gainer et al. 1968; Littlejohns and Acland 1975). tal mortality, while porcine parvovirus infection is mani-
Infected fetuses become mummified in various sizes fested in an increase in mummified fetuses mainly in gilt
depending on the stage of infection and may be hemor- litters, without neonatal mortality. Other infections,
rhagic, edematous, or apparently normal (Fig. 11.2). The such as porcine reproductive and respiratory syndrome
myocardial lesions may be seen in some infected fetuses virus, pseudorabies virus, and Leptospira spp., should al-
but are difficult to observe under field conditions. The so be considered.
gross abnormalities of the fetuses are not easy to distin- Dyspnea manifested as rapid abdominal breathing
guish from those caused by other viral infections, unless due to heart failure may be observed among EMCV-in-
the myocardial lesions are observed. fected newborn pigs. Gross lesions of white necrotic ar-
Histopathologically, the most significant finding in eas in the heart muscle are characteristic of EMCV infec-
young pigs is myocarditis with focal or diffuse accumula- tion, although such lesions resemble those of vitamin E
142 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

11.2. Normal-appearing,
edematous, hemorrhagic, and
mummified swine fetuses (left to
right) from a sow infected intra-
muscularly with EMCV at 50
days of gestation and examined
28 days after infection.

11.3. Focal infiltration of


mononuclear cells in myocardium
of a stillborn swine fetus with
natural EMCV infection.

and selenium deficiency and heart infarcts by septic em- show a rapid and complete cytolysis, and viral identifica-
bolism. tion can then be made by staining the infected monolay-
A definitive diagnosis should be based on virus isola- ers with EMCV fluorescent antibody conjugate or by the
tion and identification. Heart is the best tissue for virus inhibition of the cytopathic effects by specific immune
isolation from swine fetuses and young pigs. Tissue sam- serum. Virus isolation is usually successful from pigs
ples can be frozen without any detectable loss of virus in- during the acute phase but is difficult from pigs in the
fectivity. The virus can be isolated by inoculating sam- stage after development of circulating antibody.
ples into mice, chicken embryos, or cell cultures. Infected Histopathologic lesions consistent with EMCV infec-
mice or chicken embryos will die 3–6 days after inocula- tion may play an important role in making a diagnosis.
tion. For virus isolation in cell culture, baby hamster kid- As described previously, myocarditis of varying stages
ney (BHK-21), HeLa, or Vero cell line cultures are com- with infiltration of mononuclear cells, along with non-
monly used. For primary cell culture, fetal mouse suppurative encephalitis, is indicative of EMCV infec-
fibroblasts are suitable. Infected cell monolayers will tion. Serologic methods may also help in the diagnosis of
CHAPTER 11 ENCEPHALOMYOCARDITIS VIRUS Joo 143

EMCV infection. Detection of antibody specific to EM- post-weaning pigs in Quebec. Arch Virol 117:121–128.
CV from stillborn or large mummified fetuses is particu- Gainer, J. H. 1967. Encephalomyocarditis virus infection in
larly significant for fetal infection (Joo et al. 1988; Kim et Florida, 1960–1966. J Am Vet Med Assoc 151:421–
al. 1991a, b) because there is no transmission of mater- 425.
nal immunoglobulins across the placenta in swine. Inter- Gainer, J. H.; Sauderfur, J. R.; and Bigler, W. J. 1968. High
pretation of sow serology is often confusing, since posi- mortality in a Florida swine herd infected with the en-
tive antibody titers have not always been associated with cephalomyocarditis virus: An accompanying epizootio-
clinical disease. Both serum neutralization and hemag- logic survey. Cornell Vet 58:31–47.
glutination inhibition tests (Joo et al. 1988) can be used Gomez, L.; Lorenzo, M.; Ramos, J. R.; Luya, M. J.; Mayo, D.;
to detect EMCV antibody in serum samples. Although and Giral, T. 1982. Isolation of the encephalomyocardi-
the specificity of each test is not well defined, antibody tis virus in a sow and her fetus. Rev Cub Cienc Vet
titers of ≥1:16 appear to be significant. An enzyme- 13:21–24.
linked immunosorbent assay can also be used to detect Hani, H.; Zimmermann, W.; Bestetti, G. E.; and Muller, H.
EMCV antibody (Shanley 1980). K. 1992. A disease outbreak associated with severe my-
ocarditis resembling encephalomyocarditis virus infec-
TREATMENT AND PREVENTION tion in two Swiss farms: Clinical and pathological char-
acterization of the disease and aetiological studies. Proc
There is no treatment, but mortality may be minimized Int Congr Pig Vet Soc 12:105.
by avoiding stress or excitement of the pigs at risk. It is Helwig, F. C., and Schmidt, E. D. H. 1945. A filter-passing
important to control rodents on pig farms or minimize agent producing interstitial myocarditis in anthropoid
their contact with pigs either directly or indirectly via apes and small animals. Sciences 102:31–33.
contamination of feed or water. Although pig-to-pig Horner, G. W., and Hunter, R. 1979. Experimental infection
transmission is not clear, introduction of pigs from pre- in pigs with encephalomyocarditis virus. NZ Vet J
viously infected farms should be avoided. Basic rules of 27:202–203.
sanitation and hygiene should be applied. Animals dying Joo, H. S.; Kim, H. S.; and Leman, A. D. 1988. Detection of
of the disease should be promptly and sanitarily dis- antibody to encephalomyocarditis virus in mummified
posed. The virus can be inactivated in water containing or stillborn pigs. Arch Virol 100:131–134.
0.5 ppm residual chlorine. For disinfectants, iodine- Kim, H. S.; Joo, H. S.; and Bergeland, M. E. 1989a. Serolog-
based preparations or mercuric chloride can be used. An ic, virologic and histopathologic observations of en-
inactivated vaccine for EMCV infection in swine is com- cephalomyocarditis virus infection in mummified and
mercially available in the United States. The vaccine ap- stillborn pigs. J Vet Diag Invest 1:101–104.
pears to be effective, since high humoral immunity is de- Kim, H. S.; Christianson, W. T.; and Joo, H. S. 1989b. Path-
tected in vaccinated pigs. ogenic properties of encephalomyocarditis virus isolates
in swine fetuses. Arch Virol 109:51–57.
REFERENCES ———. 1991a. Characterization of encephalomyocarditis
virus isolated from aborted swine fetuses. Am J Vet Res
Acland, H. M., and Littlejohns, I. R. 1975. Encephalomy- 52:1649–1652.
ocarditis virus infection of pigs. I. An outbreak in New Kim, H. S.; Joo, H. S.; Christianson, W. T.; and Morrison,
South Wales. Aust Vet J 51:409–415. R. B. 1991b. Evaluation of serologic methods for the
Cerutis, D. R.; Bruner, R. H.; Thomas, D. C.; and Giron, D. J. detection of antibodies to encephalomyocarditis virus
1989. Tropism and histopathology of the D, B, K, and in swine fetal thoracic fluids. J Vet Diagn Invest
MM variants of encephalomyocarditis virus. J Med Vi- 3:283–286.
rol 29:63–69. Koenen, F.; Declercq, K.; and Strobbe, R. 1992. Isolation of
Christianson, W. T.; Kim, H. S.; Yoon, I. J.; and Joo, H. S. encephalomyocarditis virus in the offspring of swine
1992. Transplacental infection of midgestation sows with reproductive failure in Belgium. Proc Int Congr Pig
with encephalomyocarditis virus. Am J Vet Res Vet Soc 12:104.
53:44–47. Kudo, H.; Yoshizawa, S.; Hiroike, T.; and Hirose, O. 1995. A
Craighead, J. E. 1966. Pathogenicity of M and E variants of retrospective serological survey of the encephalomy-
the encephalomyocarditis (EMC) virus. I. Myocar- ocarditis virus among pigs in Chiba Prefecture, Japan. J
diotropic and neurotropic properties. Am J Pathol Vet Med Sci 57:793–795.
48:333–343. Links, I. J.; Whittington, R. J.; Kennedy, D. J.; Grewal, A.;
Craighead, J. E.; Peralta, P. H.; Murnane, T. G.; and and Sharrock, A. J. 1986. An association between en-
Shelokov, A. 1963. Oral infection of swine with the en- cephalomyocarditis virus infection and reproductive
cephalomyocarditis virus. J Infec Dis 112:205–212. failure in pigs. Aust Vet J 63:150–152.
Dea, S. A.; Bilodeau, R.; and Martineau, G. P. 1991. Isolation Littlejohns, I. R. 1984. Encephalomyocarditis virus from
of encephalomyocarditis virus among stillborn and stillborn pigs. Aust Vet J 61:93.
144 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Littlejohns, I. R., and Acland, H. M. 1975. Encephalomy- Seaman, J. T.; Boulton, J. G.; and Carrigan, M. J. 1986. En-
ocarditis virus infection in pigs. II. Experimental dis- cephalomyocarditis virus disease of pigs associated with
eases. Aust Vet J 51:416–422. a plague of rodents. Aust Vet J 63:292–294.
Love, R. J., and Grewal, A. S. 1986. Reproductive failure in Shanley, J. D. 1980. Enzyme-linked immunosorbent assay
pigs caused by encephalomyocarditis virus. Aust Vet J for immunoglobulin G antibody to encephalomyocardi-
63:128–129. tis virus. J Clin Microbiol 12:663–666.
Murnane, T. G.; Craighead, J. E.; Mondragon, H.; and Sidori, L.; Barigazzi, G.; Foni, E.; Marcato, P. S.; and Barbi-
Shelokov, A. 1960. Fetal disease of swine due to en- eri, G. 1988. Encephalomyocarditis due to cardiovirus in
cephalomyocarditis virus. Science 131:498–499. Po Valley swine: Preliminary observations. Proc Italian
Park, N. Y.; Chung, C. Y.; Ri, C. Y.; Kee, H. Y.; Bae, S. Y.; Lee, Soc Swine Pathol, pp. 249–260.
B. J.; Jung, B. T.; and Kim, D. S. 1990. Encephalomy- Simpson, C. F.; Lewis, A. L.; and Gaskin, J. M. 1977. En-
ocarditis virus infection in pigs associated with repro- cephalomyocarditis virus infection of captive elephants.
ductive failure. Korean J Vet Sci 30:441–446. J Am Vet Med Assoc 171:902–905.
Paschaleri-Papadopoulou, E.; Anastasiadis, G.; Skoufos, J.; Sutherland, R. J.; Horner, G. W.; Hunter, R.; and Fyfe, B. H.
Kyriakis, S. C.; and Papadopoulos, O. 1992. En- 1977. An outbreak of viral encephalomyocarditis in
cephalomyocarditis in Greece: A follow up. Proc Int pigs. NZ Vet J 25:225.
Congr Pig Vet Soc 12:106. Tesh, R. B. 1978. The prevalence of encephalomyocarditis
Ramos, J. R.; Gomez, L.; Mayo, M.; and Sanchez, G. 1983. virus neutralizing antibodies among various human
Infection due to encephalomyocarditis virus in swine populations. Am J Trop Med Hyg 27:144–149.
and other species over the 1975–1981 period. Rev Cub Tesh, R. B.; and Wallace, G. D. 1977. Observations on the
Cienc Vet 14:71–77. natural history of encephalomyocarditis virus. Am J
Roehe, P. M.; Rodrigues, N. C.; and Jose de Oliveira, S. 1985. Trop Med Hyg 27:133–143.
Encephalomyocarditis virus (EMC) in swine in the state Tinsley, T. W.; MacCallum, F. O.; Robertson, J. S. and
of Rio Grande do Sul, Brazil. Rev Microbiol (São Paulo) Brown, F. 1984. Relationship of encephalomyocarditis
16:117–120. virus to cricket paralysis virus of insects. Intervirol
Sangar, D. V.; Rowlands, D. J.; and Brown, F. 1977. En- 21:181–186.
cephalomyocarditis antibodies in sera from apparently Williams, M. C. 1981. Encephalomyocarditis virus infec-
normal pigs. Vet Rec 100:240–241. tion. J S Africa Vet Assoc 52:76.
Enterovirus
12 J. B. Derbyshire

Porcine enteroviruses are ubiquitous, and no conven- is surrounded by a cubic capsid. Lipoprotein is lacking,
tional herd of pigs has been shown to be free of infec- and the viruses are stable when treated with lipid sol-
tion. While the majority of infections are asymptomatic, vents. Porcine enteroviruses are also relatively stable to
porcine enteroviruses have been associated with a variety heat and pH values between 2 and 9. Hemagglutination
of clinical conditions, including polioencephalomyelitis, has not been demonstrated for porcine enteroviruses.
female reproductive disorders, enteric disease, and pneu- They are relatively resistant to many disinfectants; of 10
monia. They have also been isolated from the male geni- commonly used disinfectants tested by Derbyshire and
tal tract (Phillips et al. 1972), although the insemination Arkell (1971) against Talfan virus, only sodium
of gilts with contaminated semen did not influence their hypochlorite or 70% ethanol completely inactivated it.
fertility (De Meurichy and Pensaert 1977). A possible as- Porcine enteroviruses are also highly resistant to the en-
sociation between porcine enteroviruses and cutaneous vironment, with Teschen disease virus surviving for
lesions resembling swine vesicular disease has been pro- more than 168 days at 15˚C (Ottis 1976). They survive
posed (Knowles 1988). for long periods in liquid manure, in which they are in-
The first evidence of porcine enterovirus infection to activated more rapidly if the manure is aerated (Lund
be reported was the occurrence of Teschen disease, a po- and Nissen 1983); they are also inactivated in liquid ma-
lioencephalomyelitis with high mortality, in Czechoslo- nure by ionizing radiation (Simon et al. 1983), and by
vakia over 60 years ago. This severe disease has contin- anaerobic digestion (Derbyshire et al. 1986).
ued to occur sporadically, mainly in Central Europe but
also in Africa; milder forms of polioencephalomyelitis Laboratory Cultivation
(Talfan disease, benign enzootic paresis), caused by sero- Porcine enteroviruses are readily cultivated in the labora-
logically related but less virulent strains of porcine en- tory in cell cultures of porcine origin. They are most fre-
terovirus, have been reported in the last 35 years in West- quently grown in primary or secondary pig kidney (PK)
ern Europe, North America, and Australia. In France, the cell cultures or in established kidney cell lines such as
encephalomyelitis is intermediate in severity between IBRS-2, but they may also be cultivated in other cells of
Teschen and Talfan diseases (Métianu 1986). The associ- porcine origin such as the SST cell line. Some strains can
ation of porcine enteroviruses with reproductive, en- be grown in HeLa, monkey kidney, or baby hamster kid-
teric, and respiratory disorders in swine is less well es- ney (BHK) cell lines. Different strains produce one of
tablished than their role in polioencephalomyelitis. three kinds of cytopathic effect (CPE) in PK cultures, and
Strains that have not been shown to be pathogenic have this characteristic, together with the cytopathogenicity
been referred to as enteric cytopathogenic swine orphan of the viruses for BHK-21, HeLa, and Vero cells, enables
(ECSO) or enteric cytopathogenic porcine orphan strains to be placed in one of four groups (Knowles et al.
(ECPO) viruses. The only known natural host for porcine 1979).
enteroviruses is the pig, although experimentally infect-
ed pregnant guinea pigs developed placental lesions Serologic Classification
(Lieu 1976). The serologic classification of porcine enteroviruses is
based upon the virus-neutralization (VN) test; numerous
ETIOLOGY attempts to achieve such a classification have been
recorded in the literature. The early work in this area was
Physicochemical Characteristics by Dunne et al. (1971) and extended by Knowles et al.
Porcine enteroviruses resemble the enteroviruses of oth- (1979); Table 12.1 is based mainly upon these authors’
er species in their basic properties and are classified findings. A complement-fixation test, suitable for the
within the family Picornaviridae. The virions are spheri- rapid screening and typing of porcine enteroviruses, has
cal, 25–31 nm in diameter, and nonenveloped, with a also been described (Knowles and Buckley 1980). Subse-
buoyant density in cesium chloride of 1.34. They contain quent findings (Knowles 1983) suggest that additional
a core of single-stranded ribonucleic acid (RNA), which serogroups may exist. Some limited cross-reactivity

145
146 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

among the existing serogroups is evident, and Hazlett CLINICAL SIGNS


and Derbyshire (1978) showed that gastrointestinal anti-
bodies were more broadly specific than serum antibod- Although porcine enterovirus infection is most frequent-
ies. ly asymptomatic, various clinical syndromes have been
associated with certain serogroups, as indicated in Table
EPIDEMIOLOGY 12.2 and outlined below.

The virulent serogroup 1 strains associated with classical Polioencephalomyelitis


Teschen disease appear to be restricted to those areas in The most severe form of polioencephalomyelitis is that
which the disease occurs, and they have not been isolat- produced by the highly virulent serogroup 1 strains that
ed in North America. Less virulent serogroup 1 viruses cause Teschen disease. This is a disease of high morbidi-
and representatives of the other serogroups appear to be ty and high mortality, affecting all ages of swine and as-
ubiquitous (Odend’hal 1983). Transmission of porcine sociated with major economic losses. The early signs of
enterovirus infection is most frequently by the fecal-oral Teschen disease include fever, anorexia, and listlessness,
route, and indirect transmission by fomites is extremely rapidly followed by locomotor ataxia. In severe cases
likely to occur since the viruses are relatively resistant. there may be nystagmus, convulsions, opisthotonus, and
Endemic infection with several serogroups of porcine en- coma. Paralysis ensues, and the animal may assume a
teroviruses can always be demonstrated in conventional dog-sitting posture or remain in lateral recumbency.
herds and is probably maintained in groups of weaned Stimulation by sound or touch may elicit uncoordinated
piglets. Singh and Bohl (1972) demonstrated waves of limb movements or opisthotonus. Death is common
infection with six different serotypes over a period of 26 within 3 or 4 days of the onset of clinical signs. Since the
months in a long-term study of enterovirus infection in a appetite returns after the acute phase, some animals may
single herd. Infection is normally acquired by piglets be kept alive by careful nursing, but these cases show
shortly after weaning when maternally derived antibod- muscle wasting and residual paralysis. The less virulent
ies are withdrawn and pigs from several litters are mixed type 1 strains (Talfan disease, benign enzootic paresis)
together, and it persists for at least several weeks. Adults and the other serogroups associated with polioen-
rarely excrete virus but have high antibody levels. Pigs of cephalomyelitis produce a milder disease with relatively
any age are, however, fully susceptible to infection with a low morbidity and mortality. Mainly young piglets are
porcine enterovirus belonging to a serogroup to which affected, and the disease rarely progresses to complete
they have not previously been exposed. paralysis.

Table 12.1. Serologic classification of porcine en- Reproductive Disorders


teroviruses The term SMEDI was introduced initially (Dunne et al.
Sero- 1965) to designate a group of viruses, subsequently
group Reference shown to be porcine enteroviruses, that had been isolat-
No. CPE Strain Other Strains ed in association with stillbirth (S), mummified fetuses
1 I Teschen Talfan, PS34 (SMEDIC), PS35, (M), embryonic death (ED), and infertility (I). Subse-
F65, J1, WR1, T1, ECPO-3, E1, quent studies by the same group of workers and by oth-
PE6 ers (De Meurichy et al. 1976) indicated that the syn-
2 I T80 T52A, O3b, F17, F59, E4, J2, T3 drome could be reproduced experimentally. However, it
3 I O2b PS2–PS13, PS14 (SMEDI B),
PS15–PS20, F34, PE1, PE3, is now well established that parvovirus infection may al-
PE10 so lead to embryonic death and fetal mummification,
4 I PS36 PS38, F78, DE8 and this virus may be more frequently associated with
5 I F26 F12, J3 these disorders of early and midgestation. Other find-
6 I PS37 (SMEDI E) T4, WR4, F7, ECPO-2, J5
7 I F43 WR2
8 II PS27 (SMEDI A) PS23, PS25, PS26, PS28–PS30,
PS32 (SMEDI D), V13, A1, Table 12.2. Natural or experimental clinical syn-
WR3, WR5, WR6, ECPO-1, dromes associated with porcine enterovirus infection
ECPO-5, CHICO, PE4, PE5, Syndrome Serogroups
PE7
9 III UKG410/73 UKG139/3, UKG194/73, Polioencephalomyelitis 1, 2, 3, 5
UKG298/73(c), UKG380/73, Reproductive disorders 1, 3, 6, 8
Pd2 Diarrhea 1,2,3,5,8
10 III LP54 GF50 Pneumonia 1,2,3,8
11 I UKG173/74 MV1-76 Pericarditis and myocarditis 2,3
CHAPTER 12 ENTEROVIRUS Derbyshire 147

ings (Cropper et al. 1976) substantiate a role for en- of the lamina propria is involved. Epithelial cell destruc-
teroviruses as well as parvoviruses in these disorders, tion is not a feature of enterovirus infections. Viremia
and experimental (Bielanski and Raeside 1977) and field follows regularly in infections with the virulent
(Kirkbride and McAdaragh 1978) data confirm an asso- serogroup 1 viruses but occurs less regularly with the less
ciation between enterovirus infection and abortion in virulent strains, leading to infection of the central ner-
swine. These reproductive disorders are not usually ac- vous system (CNS) (Holman et al. 1966). It may be as-
companied by clinical signs in the sow or gilt. sumed that the pregnant uterus is also infected by
viremic spread of the virus, since embryonic or fetal in-
Diarrhea fections were demonstrated in gilts following nasal or
The role of porcine enteroviruses as enteric pathogens is oral inoculation of enterovirus (Huang et al. 1980). In-
uncertain. They have frequently been isolated from the tranasal inoculation of the virus may lead experimental-
feces of piglets with diarrhea; but since they can be read- ly to lung infection (Meyer et al. 1966), but the signifi-
ily isolated from normal piglets, particularly postwean- cance of the natural inhalation of viral aerosols is not
ing, and since diarrhea can be caused by a variety of oth- known. It has also been clearly demonstrated that when
er viral and bacterial agents, their presence may be piglets are inoculated parenterally with swine en-
coincidental. However, diarrhea has been produced ex- teroviruses, the virus rapidly infects the intestine. Ex-
perimentally by enteroviruses in piglets believed to be traintestinal infections are relatively transient, whereas
free of other pathogens. The diarrhea is mild and rela- the virus persists in the large intestine for several weeks.
tively transient, and it seems clear that porcine en-
teroviruses are considerably less important enteric LESIONS
pathogens than rotaviruses or coronaviruses. When
piglets were infected with porcine enteroviruses together No specific changes have been associated with intestinal
with rotaviruses, the disease was less severe than in enterovirus infections. They do not appear to cause vil-
piglets infected only with the rotavirus (Janke et al. lous atrophy, which is characteristic of primary intesti-
1988). nal pathogens such as coronaviruses and rotaviruses.
Other than muscle atrophy in chronic cases, no gross le-
Pneumonia, Pericarditis, and Myocarditis sions are found in polioencephalomyelitis. The histolog-
The role of enteroviruses as respiratory pathogens is also ic lesions associated with the latter are widely distributed
uncertain. It is probable that alone they rarely cause clin- in the CNS but are especially numerous in the ventral
ical signs of respiratory disease, although Pospisil et al. columns of the spinal cord, the cerebellar cortex, and the
(1971) noted increased respiration, coughing, snorting, brain stem. The changes are more marked and extensive
reduced appetite, and depression in piglets exposed to in Teschen disease than in milder encephalomyelitides
an aerosol of porcine enterovirus. While pathogenic such as Talfan disease. The neurons show progressive,
studies indicate some degree of tropism of these viruses diffuse chromatolysis (Koestner et al. 1966), and there
for the lung, the pneumonia produced is usually subclin- are focal areas of gliosis and perivascular lymphocytic
ical. Two serotypes of porcine enterovirus have been cuffing. Some meningeal infiltration with lymphocytes,
shown experimentally to be capable of producing peri- particularly over the cerebellum, may also occur.
carditis, and in one experiment myocardial involvement The SMEDI syndrome is remarkable for the lack of
occurred (Long and Koestner 1969). These findings specific lesions in stillborn or neonatal piglets, although
might lead to a suspicion of enterovirus infection in the mild focal gliosis and perivascular cuffing in the brain
case of sudden death in piglets, although encephalomy- stem have been found occasionally. Placental changes are
ocarditis virus might be a more likely candidate. restricted to nonspecific degeneration.
Pneumonic lesions have been produced by several in-
PATHOGENESIS vestigators. Smith et al. (1973) described areas of grayish
red consolidation in the ventral anterior lobes of lungs
Natural infection occurs by ingestion of the virus, and it infected with a serogroup 2 strain. There were exudates
is well established (Long 1985) that initial replication oc- in the alveoli and bronchi, slight perivascular and peri-
curs in the tonsil and intestinal tract. The large intestine bronchiolar cuffing, and some hyperplasia of the bron-
and ileum are infected more frequently than the upper chiolar epithelium.
small intestine, and the former tissues contain higher A serogroup 3 strain consistently produced serofibri-
titers of virus. It has not been clearly established which nous pericarditis experimentally, and the more severely
cells in the intestine support viral replication, but by affected piglets showed focal myocardial necrosis (Long
analogy with experiments on poliovirus (Kanamitsu et and Koestner 1969).
al. 1967) it is probable that the reticuloendothelial tissue
148 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

DIAGNOSIS ed Teschen disease virus by cell culture passage and


showed that live or formalin-inactivated vaccines pre-
The occurrence of clinical signs associated with polioen- pared from this virus induced similar levels of protection
cephalomyelitis is suggestive of viral infection, but the in piglets. Success has been claimed for a Teschen disease
differentiation of enteroviral infection from other neu- eradication program involving ring vaccination and
rotropic viruses requires isolation of the virus from the slaughter (Schaupp 1968). Restrictions on the import of
CNS or the demonstration of viral antigen by specific im- swine and pork products from areas in which Teschen
munofluorescence. Virus isolation from the CNS re- disease is endemic seem to be effective in limiting the
quires the collection of tissues from a piglet showing ear- spread of virulent serogroup 1 viruses. If such strains
ly nervous signs; animals that have been paralyzed for were introduced into North America, they would be con-
several days may no longer contain infectious virus in the trolled by a policy of quarantine and slaughter.
CNS (Lynch et al. 1984). The virus may be isolated in PK Vaccination has not been practiced against the milder
cell cultures from suspensions of spinal cord, brain stem, forms of polioencephalomyelitis or against the other
or cerebellum; it may subsequently be identified on the clinical manifestations of enterovirus infection in swine.
basis of its physicochemical characteristics, by immuno- Of the latter, only the SMEDI syndrome is of sufficient
fluorescence (Watanabe 1971), or immunoperoxidase economic importance to justify specific control measures
staining (Sulochana and Derbyshire 1978). Serologic in the field, but the multiplicity of serogroups that may
identification of the isolate is desirable. The isolation of be involved complicates the development of an effective
an enterovirus from the gastrointestinal tract of a piglet vaccine. The best current approach to the prevention of
with nervous signs does not establish the etiology of the reproductive disorders associated with enteroviruses
disease, since the enteric infection may be coincidental. would appear to be the application of management prac-
An enzyme-linked immunosorbent assay, suitable for tices that ensure that gilts are exposed to infection with
mass screening, has been described for the detection of endemic enteroviruses at least 1 month before breeding.
antibodies against Teschen disease virus (Hubschle et al. This can be achieved naturally if the animals remain in a
1983). In the SMEDI syndrome, mummified fetuses car- single building from birth to breeding, with thorough
ried to term rarely contain live virus but may contain vi- mixing of piglets from different litters at weaning; but if
ral antigen detectable by immunofluorescence. En- breeding stock is segregated at an early age, they should
terovirus isolation in PK cell culture may be attempted be contaminated with fecal material from recently
from tissues of aborted or stillborn fetuses. Lung tissue weaned piglets. This can be readily accomplished by
appears to be the most reliable source for the isolation of adding fresh feces to the feed of gilts or by dosing gilts
porcine enteroviruses from fetuses (Huang et al. 1980). with capsules of feces, which should be a pooled sample
VN tests on the body fluids of such fetuses can be carried collected from weaned piglets in several pens to ensure
out against the SMEDI-associated serogroups. Serology exposure to as wide a range as possible of the virus pres-
on the sow is not of value. In the investigation of pneu- ent in the herd. The operation of a closed-herd system re-
monia or diarrhea, virus isolation from the respiratory or duces the risk of introducing extraneous viruses, but it is
intestinal tract may be attempted, but the virologic find- not possible to eliminate this risk, since the relatively re-
ings should be cautiously interpreted, especially in rela- sistant enteroviruses can be transmitted by a variety of
tion to diarrhea, since enteric infections with en- fomites. If the introduction of fresh stock is essential for
teroviruses are common in healthy piglets. breeding purposes, before the gilts or sows are bred they
should be exposed (by fecal contamination as described
TREATMENT above) to any virus that may be present or introduced.
Exclusion of porcine enteroviruses by repopulation
As in most viral infections, control measures for porcine of herds with specific-pathogen-free (SPF) stock seems to
enteroviruses depend on prevention rather than treat- be difficult or impossible to achieve over a prolonged pe-
ment. Potential antiviral chemotherapeutics for porcine riod, since enteroviruses have been isolated from a com-
enteroviruses have received little attention. Piglets with mercial SPF herd (Derbyshire et al. 1966) and the acci-
mild polioencephalomyelitis may recover if nursing care dental introduction of Talfan virus into SPF gilts
is provided during the period of transient paresis. maintained under strict isolation has been described
(Parker et al. 1981).
PREVENTION
REFERENCES
Vaccination has been practiced in the field only for the
control of Teschen disease. The earlier Teschen disease Bielanski, A., and Raeside, J. I. 1977. Plasma concentration
vaccines, containing inactivated virus of pig tissue ori- of steroid hormones in sows infected experimentally
gin, have been superseded by attenuated or inactivated with Leptospira pomona or porcine enterovirus T1 in late
cell culture vaccines. Mayr and Correns (1959) attenuat- gestation. Res Vet Sci 22:28–34.
CHAPTER 12 ENTEROVIRUS Derbyshire 149

Cropper, M.; Dunne, H. W.; Leman, A. D.; Starkey, A. L.; Knowles, N. J. 1983. Isolation and identification of porcine
and Hoefling, D. C. 1976. Prevalence of antibodies to enteroviruses in Great Britain, 1979 to 1980. Br Vet J
porcine enteroviruses and porcine parvovirus in body 139:19–22.
fluids of fetal pigs from small vs. large litters. J Am Vet ———. 1988. The association of group III porcine en-
Med Assoc 168:233–235. teroviruses with epithelial tissues. Vet Rec 122:441–442.
De Meurichy, W., and Pensaert, M. 1977. Effect of an en- Knowles, N. J., and Buckley, L. S. 1980. Differentiation of
terovirus in gilts inseminated with a semen-virus mix- porcine enterovirus serotypes by complement fixation.
ture. Zentralbl Veterinärmed (B) 24:97–103. Res Vet Sci 29:113–115.
De Meurichy, W.; Pensaert, M.; and Bonte, P. 1976. Het Knowles, N. J.; Buckley, L. S.; and Pereira, H. G. 1979. Clas-
SMEDI-syndrome bij het varken: Rol van de en- sification of porcine enteroviruses by antigenic analysis
terovirussen en het parvovirus. Vlaam Diergeneeskd Ti- and cytopathic effects in tissue culture: Description of 3
jdschr 45:241–261. new serotypes. Arch Virol 62:201–208.
Derbyshire, J. B., and Arkell, S. 1971. The activity of some Koestner, A.; Kasza, L.; and Holman, J. E. 1966. Electron mi-
chemical disinfectants against Talfan virus and porcine croscopic evaluation of the pathogenesis of porcine po-
adenovirus type 2. Br Vet J 127:137–142. lioencephalomyelitis. Am J Pathol 49:325–337.
Derbyshire, J. B.; Clarke, M. C.; and Collins, A. P. 1966. Ob- Lieu, C. I. 1976. The experimental infection of pregnant
servations on the faecal excretion of adenoviruses and guinea pigs with porcine enterovirus: “SMEDI A” virus.
enteroviruses in conventional and minimal disease pigs. Taiwan J Vet Med Anim Husb 28:1–14.
Vet Rec 79:595–599. Long, J. F. 1985. Pathogenesis of porcine polioen-
Derbyshire, J. B.; Monteith, H. D.; and Shannon, E. E. 1986. cephalomyelitis. In Comparative Pathobiology of Viral
Virological studies on an anaerobic digestion system for Diseases, vol. 1. Ed. R. A. Olsen, S. Krakowka, and J. R.
liquid manure. Agri Wastes 18:309–312. Blakeslee. Boca Raton, Fla.: CRC Press, pp. 179–197.
Dunne, H. W.; Gobble, J. L.; Hokanson, J. F.; Kradel, D. C.; Long, J. F., and Koestner, A. 1969. Pericarditis and my-
and Bubash, G. R. 1965. Porcine reproductive failure as- ocarditis in germ-free and colostrum-deprived pigs ex-
sociated with a newly identified “SMEDI” group of pi- perimentally infected with a porcine polioen-
cornaviruses. Am J Vet Res 26:1284–1297. cephalomyelitis virus. J Infect Dis 120:245–249.
Dunne, H. W.; Wang, J. T.; and Ammerman, E. H. 1971. Lund, E., and Nissen, B. 1983. The survival of enteroviruses
Classification of North American porcine enteroviruses: in aerated and unaerated cattle and pig slurry. Agri
A comparison with European and Japanese strains. In- Wastes 7:221–223.
fect Immun 4:619–631. Lynch, J. A.; Binnington, B. D.; and Hoover, D. M. 1984.
Hazlett, D. T. G., and Derbyshire, J. B. 1978. Broad speci- Virus isolation studies in an outbreak of porcine en-
ficity of gastrointestinal antibodies following vaccina- cephalomyelitis. Can J Comp Med 48:233–235.
tion of piglets with a porcine enterovirus. J Comp Pathol Mayr, A., and Correns, H. 1959. Experimentelle Unter-
88:467–471. suchungen über Lebend- und Totimpfstoffe aus einem
Holman, J. E.; Koestner, A.; and Kasza, L. 1966. Histopatho- modifizierten Gewebekulturstamm des Teschenvirus
genesis of porcine polioencephalomyelitis in the germ- (poliomyelitis suum). Zent ralbl Veterinärmed
free pig. Pathol Vet 3:633–651. 6:416–428.
Huang, J.; Gentry, R. F.; and Zarkower, A. 1980. Experimen- Métianu, T. 1986. La maladie de Teschen-Talfan en France.
tal infection of pregnant sows with porcine enterovirus- Bull Acad Vet Fr 59:291–302.
es. Am J Vet Res 41:469–473. Meyer, R. C.; Woods, G. T.; and Simon, J. 1966. Pneumoni-
Hubschle, O. J. B.; Rajanarison, I.; Koko, E.; Rakotondra- tis in an enterovirus infection in swine. J Comp Pathol
mary, E.; and Rasiofomanana, P. 1983. ELISA zur pru- 76:397–405.
fung von schweinseren auf antikorper gegen Teschen- Odend’hal, S. 1983. The Geographical Distribution of Ani-
virus. DTW 90:86–88. mal Virus Diseases. New York: Academic Press, p. 415.
Janke, B. H.; Morehouse, L. G.; and Solorzano, R. F. 1988. Ottis, K. 1976. Vergleichende Untersuchungen über die
Single and mixed infections of rotaviruses and en- Tenazitat von Viren in Trink- und Oberflächenwasser.
teroviruses: Clinical signs and microscopic lesions. Can J Inaug. diss., Munich.
Vet Res 52:364–369. Parker, B. N. J.; Wrathall, A. E.; and Cartwright, S. F. 1981.
Kanamitsu, M.; Kasamaki, A.; Ogawa, M.; Kasahara, S.; and Accidental introduction of porcine parvovirus and Tal-
Inamura, M. 1967. Immunofluorescent study on the fan virus into a group of minimal disease gilts and their
pathogenesis of oral infection of poliovirus in monkey. effects on reproduction. Br Vet J 137:262–267.
Jpn J Med Sci Biol 20:175–194. Phillips, R. M.; Foley, C. W.; and Lukert, P. D. 1972. Isola-
Kirkbride, C. A., and McAdaragh, J. P. 1978. Infectious tion and characterization of viruses from semen and the
agents associated with fetal and early neonatal death reproductive tract of male swine. J Am Vet Med Assoc
and abortion in swine. J Am Vet Med Assoc 161:1306–1316.
172:480–483. Pospisil, Z.; Gois, M.; Veznikova, D.; and Cerny, M. 1971.
150 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

The pathogenesis of experimental infection of gnotobi- Smith, I. M.; Betts, A. O.; Watt, R. G.; and Hayward, A. H. S.
otic piglets with enterovirus strain Kr 69TK. Acta Vet 1973. Experimental infections with Pasteurella septica
Brno 40(Suppl 2):43–46. (serogroup A) and an adeno- or enterovirus in gnotobi-
Schaupp, W. 1968. Eradication of contagious paralysis otic piglets. J Comp Pathol 83:1–12.
(Teschen disease) in Austria. Wien Tierärztl Wochen- Sulochana, S., and Derbyshire, J. B. 1978. Use of indirect
schr 55:346–356. immunoperoxidase test for detection of porcine en-
Simon, J.; Mocsari, E.; Digleria, M.; and Felkai, V. 1983. Ef- teroviral antigens in infected PK15 cell cultures. Kerala J
fect of radiation on certain animal viruses in liquid Vet Sci 9:111–119.
swine manure. Int J Appl Radioisot 34:793–795. Watanabe, H. 1971. Fluorescent antibody technique in cul-
Singh, K. V., and Bohl, E. H. 1972. The pattern of enterovi- tured cells infected with porcine enteroviruses. Jpn J Vet
ral infection in a herd of swine. Can J Comp Med Res 19:1–6.
36:243–248.
Hemagglutinating
13 Encephalomyelitis Virus
M. B. Pensaert

In Canada in 1962, Greig et al. isolated a previously un- chloride (Mengeling and Coria 1972) and 1.18 g/cm3 in
recognized viral pathogen of swine from the brain of potassium tartrate (Greig and Bouillant 1972).
suckling pigs with encephalomyelitis. The virus respon-
sible for this disease was called hemagglutinating en- Physicochemical Properties
cephalomyelitis virus (HEV) because of its hemaggluti- The virus is stable between pH 4 and 10 and moderately
nating properties; it was later classified as a coronavirus sensitive to heat (Chappuis et al. 1975). All viral infectiv-
(Greig et al. 1971; Phillip et al. 1971). ity is lost after 30 minutes at 56°C, but the infectivity titer
In 1969, an antigenically similar, if not identical, is reduced by only 0.8 log10 after 7 days at 4°C. HEV was
virus was isolated in England from suckling pigs showing shown by Greig and Girard (1969) to be sensitive to lipid
anorexia, depression, and vomiting, but without signs solvents, including sodium desoxycholate. Lipid solvent
clearly associated with encephalomyelitis (Cartwright et agents are thus suitable for disinfection. Ultraviolet irra-
al. 1969). Animals that did not die remained stunted in diation also results in a significant reduction of viral in-
growth; the condition was therefore called vomiting and fectivity (Pensaert and Callebaut 1974).
wasting disease (VWD). Mengeling and Cutlip (1976)
were later able to experimentally reproduce both of the Biologic Properties
major forms of the disease (i.e., the clinically apparent The natural host of HEV is the pig. The virus also has
encephalomyelitis and VWD) using the same field iso- been adapted experimentally to replicate in mice (Kaye
lates. Although epizootiologic studies have revealed that et al. 1977; Yagami et al. 1986) and Wistar rats (Hirano
infection of swine with HEV is prevalent, naturally oc- et al. 1993). In mice, the virus is neurotropic, and mouse
curring disease is uncommon. Neonatal pigs are usually susceptibility was found to be influenced by age and in-
protected by passively acquired colostral antibody, and oculation routes (Yagami et al. 1993). Four-week-old
they subsequently develop an age-related resistance to Wistar rats died of encephalitis after inoculation of HEV
the potential clinical effects of the virus. Therefore, stud- via different routes (Hirano et al. 1993).
ies on interactions between the virus and swine have HEV was first isolated in primary cultures of pig kid-
been scarce in recent years. ney (PK) cells by Greig et al. (1962), who described a cy-
topathic effect (CPE) characterized by the appearance of
ETIOLOGY syncytia. Many of these syncytia degenerated soon after
their formation, detached from the cell sheet, and float-
Virus Structure ed in the medium as semiopaque gelatinous masses. The
HEV has an electron microscopic appearance similar to incorporation of a noncytotoxic amount of diethylami-
other coronaviruses. Negatively stained particles have a noethyl-dextran may enhance the CPE (Sato et al. 1983).
spherical shape, with an overall diameter of 120 nm Using the immunofluorescence (IF) test, HEV was al-
(Greig et al. 1971). Club-shaped surface projections so shown to propagate in several other porcine cell cul-
arranged as a “solar corona” protrude from the envelope. tures, such as adult thyroid gland, embryonic lung, testi-
Lamontagne et al. (1981) showed that the viral particle cle cell line, PK-15 cell line (Pirtle 1974), IBRS2 cell line
contains two concentric membranes (an external enve- (Chappuis et al. 1975), SK cell line (Lucas and Napthine
lope and an inner membranous bag) encircling a central 1971), SK-K cell line (Hirano et al. 1990), and swine em-
core. bryo kidney cell line KSEK6 (Kadoi et al. 1994). Non-
Studies on the chemical composition revealed that porcine cell cultures were shown to have little suscepti-
the virus contains five polypeptides, four of which are bility for growth of HEV.
glycosylated, with molecular weights from 31,000 to Virus particles can be seen by electron microscopy in
180,000 daltons (Pocock and Garwes 1977; Callebaut cytoplasmic vesicles of infected cells. Assembly occurs
and Pensaert 1980). The viral nucleic acid is considered by budding through membranes of the endoplasmic
to be of the RNA type since the growth of HEV is not af- reticulum (Ducatelle et al. 1981).
fected by inhibitors of DNA metabolism (Greig and Gi- HEV was demonstrated to possess a virion-associated
rard 1969). The buoyant density is 1.21 g/cm3 in cesium hemagglutinin. The virus spontaneously agglutinates

151
152 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

erythrocytes of mice, rats, chickens, and several other with respiratory illness in Japan in 1984 (Hirahara et al.
animals (Girard et al. 1964). Elution of HEV from red 1987). The first isolation of this virus in Taiwan was ob-
blood cells has not been observed. The S protein of HEV tained from 30- to 50-day-old pigs with central nervous
is shown to be a hemagglutinin requiring N-acetyl-9-O- disease (Chang et al. 1993).
acetylneuraminic acid as a receptor determinant on the Under experimental conditions, disease has been
surface of erythrocytes (Schultze and Herrler 1993). Us- produced in most instances when nonimmune pigs have
ing the same kinds of erythrocytes, a hemadsorption test been exposed oronasally to HEV sometime during the
can be used to demonstrate viral growth in inoculated first few weeks of life (Alexander 1962; Appel et al.
cell cultures. 1965). Clinical signs may vary, however, and in a study in
which several field isolates of HEV were compared as to
Antigenic and Genomic Characteristics virulence, the severity of such signs was related to a dif-
Although HEV is known to be the cause of different clin- ference in host susceptibility (even among littermates) as
ical syndromes, only one serotype of the virus is known well as to the apparent virulence of each isolate (Men-
to exist. The susceptibility of the infected pigs and strain geling and Cutlip 1976). In contrast, older pigs and
differences in virulence are probably responsible for the neonatal pigs that had received antibody in colostrum
fact that usually only one of the syndromes is seen in a were usually clinically unaffected when exposed to HEV
particular outbreak. An antigenic relationship exists be- under otherwise similar conditions (Appel et al. 1965).
tween HEV and the bovine coronavirus (BCV), as shown These observations are believed to explain why naturally
by seroneutralization (SN), hemagglutination inhibition occurring disease is relatively uncommon even though
(HI) (Sato et al. 1980), IF, and immunoelectron mi- HEV is ubiquitous among swine. In herds where HEV in-
croscopy (Pensaert et al. 1981). HEV is also related to hu- fection is enzootic, most pigs receive protective antibody
man respiratory coronavirus OC43 (Kaye et al. 1977) and in colostrum, and circulating maternal antibodies persist
mouse hepatitis virus (Pedersen et al. 1978). Nucleotide for about 4–18 (mean 10.5) weeks (Paul and Mengeling
sequence analysis of the region covering the S2 probe re- 1984). By the time such antibody wanes, the pigs have al-
vealed 92.6% nucleotide sequence homology to BCV and ready developed an age-related resistance to the disease.
91.9% to HCV-OC43 (Vieler et al. 1995). Moderate cross- Additional support for this idea is provided by a serolog-
reactivity was observed between HEV and turkey enteric ic study on two Belgian breeding farms, which showed
coronavirus (Dea and Tijssen 1989). that passively acquired colostral immunity was replaced
by active immunity as a consequence of subclinical in-
EPIDEMIOLOGY fection of pigs between 8 and 16 weeks of age (Pensaert
et al. 1980).
Pigs are the only species known to be naturally suscepti-
ble to HEV infection. Most of the infections in this CLINICAL SIGNS
species are subclinical, and the economic importance of
the disease is low. The spread of HEV in the pig popula- Upon infection with HEV, two clinical syndromes are
tion has been studied in several countries. The HI and SN possible: an acute, clinically apparent encephalomyelitis
tests proved to be almost equally sensitive in the demon- and VWD. Both syndromes are mainly confined to pigs
stration of specific antibodies to HEV in swine sera less than 3 weeks of age, although older swine may occa-
(Mengeling 1975). Serologic surveys revealed that infec- sionally vomit and have a brief period of inappetence,
tion of swine with HEV is very common and probably listlessness, and nervous signs. Encephalomyelitis
worldwide. In fattening pigs, 31% of the sera were posi- caused by HEV was indeed diagnosed in 30- to 50-day-
tive in Canada (Girard et al. 1964), 46% in Northern Ire- old pigs in Taiwan (Chang et al. 1993). Prior to the Tai-
land (McFerran et al. 1971), 49% in England (Cartwright wanese report, a typical encephalomyelitic form had
and Lucas 1970), 52% in Japan (Hirai et al. 1974), 75% in been described only in Canada (Alexander et al. 1959)
Germany (Hess and Bachmann 1978), and 0–89% in the and the United States (Werdin et al. 1976). Both syn-
United States, depending on the region surveyed (Men- dromes show many signs in common, and between the
geling 1975). The percentage of sows with antibodies at acute encephalomyelitis and the chronic VWD, all de-
slaughter varied from 43% in Northern Ireland to 98% in grees of severity may occur.
the United States. A high number of seropositive ani- At the start of a VWD outbreak, sneezing or cough-
mals was also found in Denmark (Sorensen 1975), ing may occur. The primary sign seen after an incubation
France (Vannier et al. 1981), Australia (Forman et al. period of 4–7 days is repeated retching and vomiting.
1979), Belgium (Pensaert et al. 1980), and Austria (Möstl Pigs under 4 weeks of age start suckling but soon stop,
1990). Conversely, Neuvonen et al. (1982) found 40 withdraw from the sow, and vomit the milk they have
Finnish elite breeding pig herds to be free of seropositive taken in. They huddle together, look pale and listless,
animals. and often have an arched back. Body temperature can be
HEV was isolated from the respiratory tracts of pigs elevated at the beginning of the disease but returns to
CHAPTER 13 HEMAGGLUTINATING ENCEPHALOMYELITIS VIRUS Pensaert 153

the normal range within 1–2 days. Affected pigs are often PATHOGENESIS
observed to grind their teeth. They dip their mouths into
water bowls but drink little, if at all, indicating a possible Several observations indicate that HEV is able to repli-
pharyngeal paralysis. The persistent vomiting and de- cate in the upper respiratory tract with or without pro-
creased food intake result in constipation and a rapid de- ducing clinical signs. Mengeling et al. (1972), Pensaert et
cline of condition. The youngest pigs become severely al. (1980), and Hirahara et al. (1989) recorded the isola-
dehydrated after a few days, exhibit dyspnea and tion of HEV from the nasal cavity, trachea, and lungs of
cyanosis, fall into a coma, and die. Older pigs lose their diseased and healthy pigs. Respiratory signs such as
appetite and rapidly become emaciated. They continue sneezing and nasal secretion were produced after in-
to vomit, although less frequently than in the early stage tranasal or oral inoculation of conventional pigs at the
of the disease. Some pigs develop a large distension of age of 50 days but not after subcutaneous inoculation
the cranial abdomen. This “wasting” state may persist for and not in intranasally inoculated 70-day-old animals
several weeks until the pigs die of starvation. Mortality (Hirahara et al. 1989). Hirahara et al. (1989) obtained
approaches 100% within the same litter, and survivors only sneezing and nasal secretion in 10-day-old,
remain permanently stunted. colostrum-deprived piglets upon intranasal inoculation;
An outbreak of the encephalomyelitic form may start intracerebral inoculation resulted in nervous signs in on-
as a VWD outbreak. Some pigs begin to vomit 4–7 days ly two of seven animals. The virus is excreted for 8–10
after birth. The vomiting is continued intermittently for days in oronasal secretions (Pensaert and Callebaut
1–2 days but is rarely severe and does not result in dehy- 1974; Hirahara et al. 1989). The mode of transmission
dration. In other outbreaks, the first sign is acute depres- occurs through nasal secretions. Most infections under
sion and a tendency to huddle. Pigs may become sick as field circumstances have a subclinical course.
soon as 2 days after birth. Occasionally, sneezing, cough- Typical clinical disease was reproduced by oronasal
ing, or upper respiratory embarassment is observed. The inoculation of colostrum-deprived piglets. In a series of
pigs lose weight rapidly and their hair loses its luster and studies on the pathogenesis of the disease, Andries and
becomes rough. Pensaert (1980c) inoculated newborn colostrum-de-
After 1–3 days, symptoms of a severe en- prived pigs oronasally with an HEV strain from pigs
cephalomyelitis arise. Younger pigs are most severely af- showing the VWD syndrome. Anorexia and vomiting
fected and exhibit various combinations of nervous were seen after an incubation period of 4 days. Pigs were
signs. Generalized muscle tremors and hyperesthesia are killed at different times after inoculation; results of the
common findings. Pigs that are able to stand usually examination by the immunofluorescent antibody tech-
have a jerky gait, and they tend to walk backward, often nique revealed that the epithelial cells of nasal mucosa,
ending in a dog-sitting position. They soon become very tonsils, lungs, and small intestine served as sites of pri-
weak, are unable to rise, and paddle their limbs. Their mary viral replication.
noses and feet become cyanotic. Blindness, opistho- After local replication near the sites of entry, the
tonus, and nystagmus can also occur. Finally, the pigs be- virus spread via the peripheral nervous system to the
come prostrate, lying on their sides, and have dyspnea. central nervous system (CNS). At least three pathways
In most cases, coma precedes death. Mortality in appeared to be involved. A first pathway led from the
younger pigs is usually 100%. nasal mucosa and tonsils to the trigeminal ganglion and
Older pigs usually suffer a mild transient illness in the trigeminal sensory nucleus in the brain stem. A sec-
which posterior paralysis may be the most common sign. ond pathway occurred along the vagal nerves via the va-
The paresis in a few cases is accompanied by blindness. gal sensory ganglion to the vagal sensory nucleus in the
The outbreak described in Taiwan (Chang et al. 1993) brain stem. A third pathway led from the intestinal
in 30- to 50-day-old pigs was characterized by fever, con- plexuses to the spinal cord, also after replication in local
stipation, hyperesthesia, muscular tremor, progressive sensory ganglia. Earlier studies had already shown that
anterior paresis, posterior paresis, prostration, recum- viremia is probably of little or no importance in the
bency, and paddling movements with a morbidity rate of pathogenesis (Andries and Pensaert 1980b).
4% and a mortality rate of 100%. The pigs died within In the CNS, the infection started in well-defined nu-
4–5 days after the onset of clinical signs. clei of the medulla oblongata but progressed later into
The interval for onset of the disease in the first litter the entire brain stem, the spinal cord, and sometimes al-
to cessation of the disease or its failure to appear in a lit- so the cerebrum and cerebellum. Fluorescence in the
ter is usually 2–3 weeks (Werdin et al. 1976). Disappear- brain was always restricted to the perikaryon and
ance of the disease coincides with the time it takes sows processes of neurons (Fig. 13.1). Vomiting was induced
to develop immunity and to pass this protection on to by viral replication in the vagal sensory ganglion (gan-
their offspring. It has been shown that pigs exposed to glion distale vagi) or by impulses to the vomiting center
HEV develop seroneutralizing and hemagglutination-in- produced by infected neurons at different sites (Andries
hibiting antibodies (Pensaert and Callebaut 1974). 1982).
154 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

To elucidate the pathogenesis of the wasting, radio- A nonsuppurative encephalomyelitis was reported in
logic studies were performed on chronically infected 70–100% of pigs with nervous signs and in 20–60% of
pigs, vagotomized pigs, and controls (Andries 1982). pigs showing the VWD syndrome. The lesions are char-
The stomach of control pigs was always empty within 10 acterized by perivascular cuffing, gliosis, and neuronal
hours, whereas barium was retained in the stomach for degeneration (Richards and Savan 1960; Alexander
2–7 days in pigs with HEV. In pigs with a bilateral ab- 1962; Hoorens et al. 1977; Narita et al. 1989b; Chang et
dominal vagotomy, the stomach emptying was less dis- al. 1993). They are most pronounced in the gray matter
turbed. This indicated not only that the delayed empty- of the pons Varolii, the medulla oblongata, and the dor-
ing in pigs with HEV was due to earlier viral replication sal horns of the upper spinal cord. It was suggested that
in the vagal ganglion and vagal nuclei in the brain but encephalitic lesions are a specific immune response to
that the virus-induced lesions in the intramural plexuses HEV following its replication in the CNS (Narita et al.
of the stomach were probably also responsible for the 1989b). Neuritis of peripheral sensory ganglia, particu-
gastric stasis. The disturbance of the stomach emptying larly the trigeminal ganglia, also occurs.
was considered to play an important role in the patho- Microscopic changes in the stomach wall and the
genesis of the wasting. lungs were found only in pigs showing the VWD syn-
drome. Degeneration of the ganglia of the stomach wall
LESIONS and perivascular cuffing were present in 15–85% of dis-
eased animals. The lesions were most pronounced in the
The only significant gross lesions reported in natural pyloric gland area (Steinicke and Nielsen 1959; Schlen-
HEV infections are cachexia and a distension of the ab- stedt et al. 1969; Hoorens et al. 1977).
domen, which develops in some chronically affected pigs
(Schlenstedt et al. 1969; Hoorens et al. 1977). The stom- DIAGNOSIS
achs of such pigs are dilated and filled with gas.
Microscopic lesions are found in the tonsils, the ner- To effect diagnosis by virus isolation, the tonsils, brain,
vous system, respiratory system, and stomach of acutely and lungs are dissected aseptically from young diseased
diseased pigs. The lesions tend to disappear in animals piglets slaughtered as soon as possible after the first
surviving acute stages of the disease. Tonsillar changes signs of infection. It is very difficult to isolate the virus
are characterized by epithelial degeneration and lym- from pigs that have been sick for more than 2 days.
phatic cell infiltration in the crypts (Narita et al. 1989a). Crude suspensions of selected tissues are inoculated into
Degeneration of the epithelial cells in the turbinates, primary PK cells or secondary pig thyroid cells. The pres-
bronchi, and alveoli and interstitial peribronchiolar ence of HEV is shown by the developing of syncytia,
pneumonia with infiltration of neutrophils and hemadsorption, and hemagglutination (Andries and
macrophages were observed in 20% of naturally infected Pensaert 1980a). A single blind passage with cells and
pigs (Hoorens et al. 1977) and to a much larger extent in culture fluid is recommended, since clinical specimens
experimentally infected pigs (Cutlip and Mengeling from pigs infected with HEV often contain very small
1972; Hirahara et al. 1989). amounts of infectious virus.

13.1. Viral multiplication in the


brain stem of a pig inoculated with
HEV. Fluorescence is seen in the
axon and the perikaryon of a neuron
(×500).
CHAPTER 13 HEMAGGLUTINATING ENCEPHALOMYELITIS VIRUS Pensaert 155

Antibodies to the virus can be detected by the SN, Andries, K. 1982. Pathogenese en epizoötiologie van “vom-
plaque reduction, or HI test (Mengeling 1975; Sato et al. iting and wasting disease,” een virale infektie bij het
1983). Since subclinical infections with the virus are very varken. Ph.D. diss., Med Fac Diergeneeskd Rijksuniv,
common, antibody titers must be evaluated very careful- Ghent, 24:164.
ly. Moreover, a significant rise in antibody titer can be Andries, K., and Pensaert, M. 1980a. Propagation of
obtained only if acute sera are taken as soon as possible hemagglutinating encephalomyelitis virus in porcine
after the appearance of clinical signs. Pigs that become ill cell cultures. Zentralbl Veterinärmed (B) 27:280–290.
after an incubation period of 6–7 days may indeed have ———. 1980b. Virus isolation and immunofluorescence in
already built up a high antibody titer at that moment, different organs of pigs infected with hemagglutinating
making an interpretation of paired sera very difficult. encephalomyelitis virus. Am J Vet Res 41:215–218.
Differential diagnosis must be made between HEV in- ———. 1980c. Immunofluorescence studies on the patho-
fection, Teschen-Talfan disease, and Aujeszky’s disease. genesis of hemagglutinating encephalomyelitis virus af-
Clinical signs of encephalomyelitis associated with the ter oronasal inoculation. Am J Vet Res 41:1372–1378.
latter two diseases are usually more severe than those as- Appel, M.; Greig, A. S.; and Corner, A. H. 1965. En-
sociated with HEV infection and may appear among old- cephalomyelitis of swine caused by a haemagglutinating
er pigs as well as piglets. Respiratory signs in older pigs virus. IV. Transmission studies. Res Vet Sci 6:482–489.
and abortions in sows are typical for Aujeszky’s disease. Callebaut, P. E., and Pensaert, M. B. 1980. Characterization
The viruses can be grown in PK and pig thyroid cells; in and isolation of structural polypeptides in haemaggluti-
PK cells they are distinguishable by their CPE. They can nating encephalomyelitis virus. J Gen Virol 48:193–204.
be further differentiated by virus identification tests and Cartwright, S. F., and Lucas, M. 1970. Vomiting and wasting
the production of hemagglutinin by HEV. disease in piglets. Vet Rec 86:278–280.
Cartwright, S. F.; Lucas, M.; Cavill, P. J.; Gush, A. F.; and
PREVENTION Blandford, T. B. 1969. Vomiting and wasting disease of
piglets. Vet Rec 84:175–176.
On most breeding farms, HEV infection persists enzoot- Chang, G. N.; Chang, T. C.; Lin, S. C.; Tsai, S. S.; and Chern,
ically and is maintained through a subclinical infection R. S. 1993. Isolation and identification of hemaggluti-
of the respiratory pathways. Sows usually will have come nating encephalomyelitis virus from pigs in Taiwan. J
into contact with the virus before the time of first far- Chinese Soc Vet Sci 19:147–158.
rowing. They will protect their offspring against clinical Chappuis, G.; Tektoff, J.; and Leturdu, Y. 1975. Isolement en
signs by colostral antibodies. When infection occurs in France et identification du virus de la maladie du vom-
such pigs, it will remain subclinical. Only when sows are issement et du deperissement des porcelets (coronalike
not immune at the time of farrowing (on newly populat- virus). Rec Med Vet 151:557–566.
ed farms or on small farms in which the virus is not Cutlip, R. C.; and Mengeling, W. L. 1972. Lesions induced
maintained through lack of sufficient numbers of litters) by hemagglutinating encephalomyelitis virus strain 67N
will an infection of pigs within the first weeks after birth in pigs. Am J Vet Res 33:2003–2009.
result in clinical signs. Maintaining the virus on farms to Dea, S., and Tijssen, P. 1989. Detection of turkey enteric
obtain immune sows at the time of their first farrowing coronavirus by enzyme-linked immunosorbent assay
creates a favorable situation in preventing break- and differentiation from other coronaviruses. Am J Vet
throughs of disease in piglets. Res 50:226–231.
Once clinical signs are evident, the disease will run its Ducatelle, R.; Coussement, W.; and Hoorens, J. 1981. Mor-
course; spontaneous recoveries are rare. Litters born 2–3 phogenesis of hemagglutinating encephalomyelitis in
weeks after the onset of disease are usually protected by vivo and in vitro. Vlaams Diergeneeskd Tijdschr
maternal immunity. Before that time, piglets born from 50:326–336.
nonimmune sows can be protected by specific hyperim- Forman, A. J.; Hale, C. J.; Jones, R. T.; and Conaughton, I. D.
mune serum injected at birth. However, the time lapse 1979. Haemagglutinating encephalomyelitis virus infec-
between diagnosis and cessation of the disease is usually tion of pigs. Aust Vet J 55:503–504.
too short to gain much profit from this procedure. Girard, A.; Greig, A. S.; and Mitchell, D. 1964. En-
cephalomyelitis of swine caused by a haemagglutinating
REFERENCES virus. III. Serological studies. Res Vet Sci 5:294–302.
Greig, A. S., and Bouillant, A. M. P. 1972. Studies on the
Alexander, T. J. L. 1962. Viral encephalomyelitis of swine in hemagglutination phenomenon of hemagglutinating
Ontario: Experimental and natural transmission. Am J encephalomyelitis virus (HEV) of pigs. Can J Comp Med
Vet Res 32:756–762. 36:366–370.
Alexander, T. J. L.; Richards, W. R. C.; and Roe, C. K. 1959. Greig, A. S., and Girard, A. 1969. Encephalomyelitis of
An encephalomyelitis of suckling pigs in Ontario. Can J swine caused by a haemagglutinating virus. V. Response
Comp Med 23:316–319. to metabolic inhibitors and other chemical compounds.
156 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Res Vet Sci 10:509. glutinating encephalomyelitis virus in serums from


Greig, A. S.; Mitchell, D.; Corner, A. H.; Bannister, G. L.; swine in the United States. Am J Vet Res 36:821–823.
Meads, E. B.; and Julian, R. J. 1962. A hemagglutinating Mengeling, W. L., and Coria, M. F. 1972. Buoyant density of
virus producing encephalomyelitis in baby pigs. Can J hemagglutinating encephalomyelitis of swine: Compar-
Comp Med 26:49–56. ison with avian bronchitis virus. Am J Vet Res
Greig, A. S.; Johnson, C. M.; and Bouillant, A. M. P. 1971. 33:1359–1363.
Encephalomyelitis of swine caused by a haemagglutinat- Mengeling, W. L., and Cutlip, R. C. 1976. Pathogenicity of
ing virus. VI. Morphology of the virus. Res Vet Sci field isolants of hemagglutinating encephalomyelitis
12:305–307. virus for neonatal pigs. J Am Vet Med Assoc
Hess, R. G., and Bachmann, P. A. 1978. Erbrechen und 168:236–239.
Kummern der Ferkeln: Vorkommen und Verbreitung in Mengeling, W. L.; Boothe, A. D.; and Ritchie, A. E. 1972.
Suddeutschland. Tierärtzl Umschau 33:571–574. Characteristics of a coronavirus (strain 67N) of pigs.
Hirahara, T.; Yasuhara, H.; Kodama, K.; Nakai, M.; and Am J Vet Res 33:297–308.
Sasaki, N. 1987. Isolation of hemagglutinating en- Möstl, K. 1990. Erhebungen über porcine Coronaviren in
cephalomyelitis virus from respiratory tract of pigs in Österreich. III. Das hemagglutinating Encephalomyelitis
Japan. Jpn J Vet Sci 49:85–93. Virus (HEV) der Schweine. Wien Tierärztl Monatsschr
Hirahara, T.; Yamanaka, M.; Yasuhara, H.; Matsui, O.; Ko- 77(4):117–120.
dama, K.; Nakai, M.; Sasaki, N.; and Matumoto, M. Narita, M.; Kawamura, H., Tsuboi, T.; Haritani, M.; and
1989. Experimental infection of pigs with porcine Kobayashi, M. 1989a. Immunopathological and ultra-
hemagglutinating encephalomyelitis virus. Jpn J Vet Sci structural studies on the tonsil of gnotobiotic pigs in-
51:827–830. fected with strain 67N of haemagglutinating en-
Hirai, K.; Chang, C.; and Shimakura, S. 1974. A serological cephalomyelitis virus. J Comp Pathol 100:305–312.
survey on haemagglutinating encephalomyelitis virus Narita, M.; Kawamura, H.; Haritani, M.; and Kobayashi, M.
infection in pigs in Japan. Jpn J Vet Sci 36:375–380. 1989b. Demonstration of viral antigen and im-
Hirano, N.; Ono, K.; Takasawa, H.; Murakami, T.; and Haga, munoglobulin (IgG and IgM) in brain tissue of pigs ex-
S. 1990. Replication and plaque formation of swine perimentally infected with haemagglutinating en-
haemagglutinating encephalomyelitis virus (67N) in cephalomyelitis virus. J Comp Pathol 100:119–128.
swine cell line, SK-K culture. J Virol Methods 27:91–100. Neuvonen, E.; Ek-Kommonen, C.; Veijalainen, P.; and Schul-
Hirano, N.; Haga, S.; and Fujiwara, K. 1993. The route of man, A. 1982. Absence of hemagglutinating en-
transmission of hemagglutinating encephalomyelitis cephalomyelitis virus in Finnish elite breeding pig herds.
virus (HEV) 67N strain in 4-week-old rats. Adv Exp Med Nord Vet Med 34:334–335.
Biol 342:333–338. Paul, P. S., and Mengeling, W. L. 1984. Persistence of pas-
Hoorens, J.; Thoonen, H.; Gheyle, M.; and Van Buyten, A. sively acquired antibodies to haemagglutinating en-
1977. Braakziekte bij biggen. Vlaams Diergeneeskd Tijd- cephalomyelitis virus in swine. Am J Vet Res
schr 46:209–224. 45:932–934.
Kadoi, K.; Sakai, T.; Yukawa, M.; Kamata, H.; and Iwabuchi, Pedersen, N. C.; Ward, J.; and Mengeling, W. L. 1978. Anti-
M. 1994. The propagation of a porcine hemagglutinat- genic relationship of the feline infectious peritonitis
ing encephalomyelitis virus in swine kidney cell cultures. virus to coronaviruses of other species. Arch Virol
Microbiologica 17:341–344. 58:45–53.
Kaye, H. S.; Yarbrouch, W. B.; Reed, C. J.; and Harrison, A. Pensaert, M. B.; and Callebaut, P. E. 1974. Characteristics of
K. 1977. Antigenic relationship between human coron- a coronavirus causing vomiting and wasting in pigs.
avirus strain OC43 and hemagglutinating en- Arch Gesamte Virusforsch 44:35–50.
cephalomyelitis virus strain 67N of swine: Antibody re- Pensaert, M. B.; Andries, K.; and Callebaut, P. 1980. A
sponses in human and animal sera. J Infect Dis seroepizootiologic study of vomiting and wasting dis-
135:201–209. ease virus in pigs. Vet Q 2:142–148.
Lamontagne, L.; Marois, P.; Marsolais, G.; diFranco, E.; and Pensaert, M. B.; DeBouck, P.; and Reynolds, D. J. 1981. An
Assaf, R. 1981. Inner structure of some coronaviruses. immunoelectron microscopic and immunofluorescent
Can J Comp Med 45:177–181. study on the antigenic relationship between the coron-
Lucas, M. H., and Napthine, P. 1971. Fluorescent antibody avirus-like agent CV777 and several coronaviruses. Arch
technique in the study of three porcine viruses. J Comp Virol 68:45–52.
Pathol 81:111–117. Phillip, J. I. H.; Cartwright, S. F.; and Scott, A. C. 1971. The
McFerran, J. B.; Clarke, J. K.; Connor, T. J.; and Knox, E. R. size and morphology of T.G.E. and vomiting and wast-
1971. Serological evidence of the presence of haemag- ing disease viruses of pigs. Vet Rec 88:311–312.
glutinating encephalomyelitis virus in Northern Ireland. Pirtle, E. C. 1974. Titration of two porcine respiratory virus-
Vet Rec 88:339. es in mammalian cell cultures by direct fluorescent anti-
Mengeling, W. L. 1975. Incidence of antibody for hemag- body staining. Am J Vet Res 35:249–250.
CHAPTER 13 HEMAGGLUTINATING ENCEPHALOMYELITIS VIRUS Pensaert 157

Pocock, D. H., and Garwes, F. J. 1977. The polypeptides of mark. Nord Vet Med 27:208–212.
haemagglutinating encephalomyelitis virus and isolated Steinicke, O., and Nielsen, A. 1959. Histological changes of
subviral particles. J Gen Virol 37:487–499. the mesenteric plexus of the stomach in “baby pig dis-
Richards, W. P. C., and Savan, M. 1960. Viral en- ease.” Nord Vet Med 11:399–429.
cephalomyelitis of pigs: A preliminary report on the Vannier, P.; Chappuis, G.; Labadie, J. L.; Renault, L.; and
transmissibility and pathology of a disease observed in Josse, J. 1981. A serological survey of the virus of vomit-
Ontario. Cornell Vet 50:132–155. ing and wasting disease in piglets. Ann Zootech 30:379.
Sato, K.; Inaba, Y.; and Matumoto, M. 1980. Serological re- Vieler, E.; Schlapp, T.; Anders, C.; and Herbst, W. 1995. Ge-
lation between calf diarrhea coronavirus and hemagglu- nomic relationship of porcine hemagglutinating en-
tinating encephalomyelitis virus. Arch Virol cephalomyelitis virus to bovine coronavirus and human
66:157–159. coronavirus OC43 as studied by the use of bovine coro-
Sato, K.; Inaba, Y.; Miura, Y.; Tokuhisa, S.; and Matumoto, navirus S gene-specific probes. Arch Virol
M. 1983. Inducement of cytopathic changes and plaque 140:1215–1223.
formation by porcine haemagglutinating en- Werdin, R. E.; Sorensen, D. K.; and Stewart, W. C. 1976.
cephalomyelitis virus. Vet Microbiol 8:521–530. Porcine encephalomyelitis caused by haemagglutinating
Schlenstedt, V. D.; Barnikol, H.; and Plonait, H. 1969. Er- encephalomyelitis virus. J Am Vet Med Assoc
brechen und Kummern bei Saugferkeln. DTW 168:240–246.
76:694–695. Yagami, K.; Hirai, K.; and Hirano, N. 1986. Pathogenesis of
Schultze, B., and Herrler, G. 1993. Recognition of N-acetyl- haemagglutinating encephalomyelitis virus (HEV) in
9-O-acetylneuraminic acid by bovine coronavirus and mice experimentally infected by different routes. J Comp
hemagglutinating encephalomyelitis virus. Adv Exp Pathol 96:645–657.
Med Biol 342:299–304. Yagami, K.; Izumi, Y.; Kajiwara, N.; Sugiyama, F.; and
Sorensen, K. J. 1975. Haemagglutinerende en- Sugiyama, Y. 1993. Neurotropism of mouse-adapted
cephalomyelitis virus (HEV) infektioner hos grise: Serol- haemagglutinating encephalomyelitis virus. J Comp
ogisk indikation for infektionens tilstedevaerelse i Den- Pathol 109:21–27.
Classical Swine Fever (Hog Cholera)
14 J. T. Van Oirschot

To avoid confusion with the acronym HCV for hepatitis ETIOLOGY


C virus of humans, the term “classical swine fever virus”
instead of “hog cholera virus” is increasingly used. Con- CSFV belongs to the genus Pestivirus. Bovine viral diar-
sequently, this designation will be adopted in this chap- rhea virus (BVDV) and border disease virus (BDV),
ter. which can infect cattle, sheep, and swine, are the two oth-
Classical swine fever (CSF) (hog cholera) is a highly er members of this genus. In 1991, the genus Pestivirus
contagious viral disease of swine. The infection can run was allocated to the family Flaviviridae.
an acute, subacute, chronic, atypical, or inapparent CSFV measures 40–50 nm in diameter, with a nucle-
course. Acute CSF is caused by virulent virus and gener- ocapsid of about 29 nm. It is an enveloped RNA virus,
ally results in high morbidity and mortality, whereas in- the membrane surrounding an isometric core. Fringelike
fections with low-virulence virus can go unnoticed. projections of 6–8 nm have been demonstrated on the
The question of whether CSF originated in the Unit- surface of the virion. The buoyant density, depending on
ed States or elsewhere will probably remain conjectural. the gradient material and on the cells used to propagate
According to Hanson (1957), the first description of a the virus, has been reported to be between 1.12 and 1.17
cholera-like disease of swine was from Tennessee, in g/mL. Sedimentation coefficient values of 140–180 S
about 1810. Later outbreaks were reported from Ohio in have been found (Horzinek 1981).
the early 1830s. CSF may possibly have occurred in The virion has a single-stranded RNA of positive po-
France in 1822 and in Germany in 1833, but other re- larity and is about 12.3 kb long. The genome, which has
ports suggest that the disease first appeared outside the been completely sequenced, contains one large open
United States in England in 1862 and subsequently reading frame, coding for one large polyprotein of ap-
spread to the European continent (Fuchs 1968). CSF was proximately 3900 amino acids (Meyers et al. 1989;
reported from South America in 1899 and from South Moormann et al. 1990) that is cleaved co- and posttrans-
Africa in 1900. lationally by host-cell- and virus-encoded proteases to
Classical swine fever virus (CSFV) still has a world- yield mature viral proteins. The open reading frame is
wide distribution, but gradually more countries succeed flanked by a 5′ noncoding region of almost 400 nu-
in eradicating the virus. Countries free of the disease cleotides and a 3′ noncoding region of approximately
are Australia, Belgium, Canada, France, Great Britain, 200 nucleotides. The order of the gene products along
Iceland, Ireland, New Zealand, Portugal, the Scandina- the open reading frame is as follows:
vian countries, Spain, Switzerland, and the United
States. NH 2 -(N pro -C-E rns -E1-E2-p7-NS2.3-NS4A-NS4B -
A state-federal CSF eradication program was begun NS5A-NS5B)-COOH
in the United States in 1962. The last U.S. outbreak oc-
curred in 1976. The total cost of the effort to eradicate The (putative) function of individual proteins has
CSF amounted to about $140 million. been described (Meyers and Thiel 1996). The nucleocap-
The European Community (EC) supports a program sid protein precedes the Erns protein (formerly called
to eliminate CSFV from its member countries. It is based gp44/48); the latter is located at the surface of the virion
on the killing of infected herds and is supported by oth- as a homodimer but is also secreted from cells. The Erns
er veterinary legislative and zoo sanitation measures. In protein has ribonuclease activity, which is unique among
spite of these combined efforts, outbreaks of CSF are viruses (Schneider et al. 1993; Hulst et al. 1994). The
still reported from the EC countries, causing severe eco- function of this enzymatic activity is as yet unclear. The
nomic losses; for example, in the Netherlands the direct E1 protein (gp33) is present in the viral envelope as an
cost of the control program amounted to $93 million E1-E2 heterodimer, and the E2 (gp55), which is the most
in the period 1983–85. Severe epizootics plagued Bel- immunogenic protein of CSFV, is present as a homo-
gium and Germany in 1994, and the Netherlands in dimer and as a heterodimer with E1. The p7 protein is
1997. probably not incorporated in the virion, and the remain-

159
160 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

ing C-terminal part of the open reading frame is coding growth. Virus replication is restricted to the cytoplasm of
solely for nonstructural proteins. the cell and does not result in a cytopathic effect. The
The analysis of nucleotide sequences of the 5′ non- first-progeny virus is released from the cells at 5–6 hours
coding region, the N-terminal part of E2, and a region of postinfection. Under single growth cycle conditions,
NS5B yielded indications to divide CSFV strains into two there is an exponential increase in virus titer until 15
major groups (Hofmann et al. 1994; Lowings et al. 1996). hours postinfection, after which virus production con-
A marked antigenic variation also exists among strains of tinues at a high level for several days. In cell cultures, CS-
CSFV (Wensvoort 1989; Edwards and Sands 1990) and FV spreads by means of the medium, by cytoplasmic
probably resides on the nonconserved antigenic unit on bridges to neighboring cells, and from mother to daugh-
the N-terminal half of E2 (Van Rijn et al. 1994) and on ter cells. CSFV can persist in cell culture. The virus seems
E1 as well (Weiland et al. 1992). Even within some strains to mature at intracytoplasmic membranes, which is in
of CSFV, antigenic heterogeneity is present (Wensvoort accordance with the inability to detect CSFV antigens at
et al. 1989). The use of monoclonal antibodies (MABs) the surface of the infected cell (Van Oirschot 1980).
to study the diversity of CSFV strains supports the ge- Thermal and pH stability may vary per strain of CS-
netic division of CSFV into two major groups (Lowings FV. The higher the temperature, the quicker CSFV is in-
et al. 1996). No relation was observed between antigenic activated. Inactivation is partly dependent on the medi-
variants and virulence (Wensvoort et al. 1989). On the um containing the virus. Thus cell culture fluid
other hand, field isolates that were neutralized more infectivity is lost after 10 minutes at 60˚C, whereas in de-
readily by BVDV antibody than by CSFV antibody were fibrinated blood the virus is not inactivated after 30 min-
of reduced virulence (Kamijyo et al. 1977). utes at 68˚C. The virus is quite stable at pH 5–10. Above
Although CSFV differs from BVDV and BDV on the and below these pH values, infectivity is relatively rapid-
genetic and antigenic level, it also shows many similari- ly destroyed. The inactivation rate below pH 5 is depen-
ties with these other pestiviruses. The differences mostly dent on the temperature: at pH 4 a half-life of 260 hours
occur on the E2 protein, because most of the MABs that was found at 4˚C and of 11 hours at 21˚C (Depner et al.
differentiate CSFV from BVDV and BDV are directed 1992). Lipid solvents such as ether, chloroform, and de-
against E2 (Wensvoort 1989; Edwards et al. 1991; Paton oxycholate quickly inactivate the virus. For disinfection,
1995). 2% sodium hydroxide is still considered most suitable. In
The close antigenic relationship between CSFV and pens the virus appears to be inactivated in a few days; in
other pestiviruses is evidenced by cross-reactions in im- liquid manure of pigs, CSFV (initial titer of 105.5 TCID50
munodiffusion, immunofluorescence, and, to a lesser de- [median tissue culture infectious doses] per mL) may
gree, neutralization tests. Certain strains of CSFV also in- survive for 2 weeks at 20˚C and more than 6 weeks at 4˚C
duce neutralizing antibody to BVDV, and pigs can be (Haas et al. 1995). In pork and pork products it can re-
partially immunized against CSF with BVDV. The com- main infective for months, which is of great epizootio-
mon antigens among pestiviruses reside largely on the logic importance.
nonstructural protein NS2.3. There is an overall homol-
ogy of about 70% on the amino acid level between CSFV EPIDEMIOLOGY
and BVDV strains (Paton 1995).
Field strains of CSFV vary widely in virulence. Strains The pig is the only natural host and is the significant
of high virulence induce acute disease and high mortali- source of spread of CSFV. Direct contact between infect-
ty, whereas moderately virulent strains generally give rise ed and susceptible pigs is the principal means of viral
to subacute or chronic infections. Postnatal infection transmission. Infected pigs may shed the virus before the
with low-virulence CSFV results in mild disease or sub- onset of disease and continue to do so during the entire
clinical infection. However, such low-virulence strains disease period. CSFV is notably excreted with oronasal
can produce mortality in porcine fetuses and newborn and lacrimal secretions, urine, and feces. Pigs that recov-
piglets. The outcome of CSFV infections with strains of er from CSF generally shed virus until specific antibodies
moderate virulence is partially determined by host fac- have developed. Thus pigs infected with virulent CSFV
tors such as breed, age, immune competence, and nutri- may shed large amounts of virus for 10–20 days, where-
tional condition, whereas in infections with highly viru- as postnatal infections with low-virulence strains are
lent or avirulent CSFV, host factors seem to play a minor characterized by short virus excretion periods. Conse-
role. It has been reported that the virulence of CSFV may quently, virulent CSFV will usually spread faster in a
be an unstable property, since enhancement of virulence herd and induce higher morbidity than low-virulence
has been observed after one or more passages in pigs strains. Chronically infected pigs shed the virus continu-
(Dunne 1975). ously or intermittently till death.
Although CSFV can replicate in nonporcine cells, When a pregnant sow is exposed to CSFV strains of
porcine kidney cells are used most frequently for virus low virulence, the infection initially will often go unno-
CHAPTER 14 CLASSICAL SWINE FEVER (HOG CHOLERA) Van Oirschot 161

ticed, but the virus can be transmitted to the fetuses in persistent and inapparent infections that are clinically
utero. Such a congenital infection usually results in still- hard to recognize and therefore may be diagnosed only
birth and/or birth of weak pigs, which die shortly after after the virus has spread to other herds.
birth. Because CSFV persists in fetuses, large quantities
of virus may then be disseminated at farrowing. Howev- CLINICAL SIGNS
er, if congenitally infected piglets are born healthy and
remain so for months, they act as a hardly recognizable When virulent CSFV first appears in a herd, only a few
and continuous source of viral spread (Van Oirschot and pigs will show clinical signs of disease. Initially, the pigs
Terpstra 1977). Thus CSFV infection may smolder in a may only appear drowsy or less active; if they are dis-
herd for months before the eventual diagnosis is made. turbed and made to stand, some will have arched backs
These persistent congenital infections are therefore of and others may appear chilled. Still others may stand
utmost importance in the epidemiology of CSF, particu- with drooping heads and straight tails. At this time, a re-
larly in regions where viruses of reduced virulence pre- duced appetite is noticed. Later, it progresses to a
dominate. marked anorexia exemplified by the pig nosing around
The introduction of newly purchased, healthy-looking in the feed for a short while before returning to its resting
infected pigs into a herd is the most common cause of CSF place.
outbreaks. The infection may originate from breeding At the first sign of inactivity, fever can be recorded.
farms, or pigs may be exposed to the virus at places where Within 6 days of exposure to CSFV, the temperature of
many pigs are assembled or during transport in contami- an affected animal may become higher than 42˚C, al-
nated vehicles. An important source of infection is virus- though values between 41˚C and 42˚C are more common
containing garbage that has not been properly sterilized. during the course of the disease. Concurrent with the
This mode of virus spread was responsible for 22% of the temperature rise is a corresponding drop in the leuko-
outbreaks of CSF in the United States in 1973 (Dunne cyte count. Total leukocyte counts of 9000 to as low as
1975). The virus can be carried over long distances in pork 3000/mm3 of blood may be found.
and (frozen) pork products, which may lead to the in- Early in the course of the disease, the eyes show a
troduction of CSF into countries free of the disease. marked discharge associated with conjunctivitis, which
Mechanical vectors contribute to the spread of CSFV may progress until the eyelids are completely adhered.
between premises. In this respect, trucks, farmers, vet- Constipation commonly develops during the period of
erinarians, and other personnel with their equipment initial high temperature, followed by a severe, watery,
constitute a great risk, but virus may also be mechanical- yellowish gray diarrhea. Sick pigs become chilled and
ly transmitted by pets, birds, and arthropods (Stewart will huddle or pile on each other seeking warmth. It is
1981). Airborne transmission has been demonstrated ex- not uncommon for pigs to vomit a yellowish fluid con-
perimentally (Hughes and Gustafson 1960; Terpstra taining much bile. Convulsions may occur in a few swine,
1987) and thus may be a (probably infrequent) mode of which usually die within hours or at most a few days af-
spread of CSFV between herds at close proximity. ter the convulsions begin. Hyperemia of the skin may oc-
CSFV can circulate in wild pig populations, in some cur concurrently with the initial temperature rise.
countries posing a threat to domestic pig husbandry. As the disease progresses, more pigs become affected,
Several countries, including those of the EC, have and those that were sick first become gaunt and tucked
eradication programs in force, based on rapid diagnosis up in the flank and have a characteristic weaving, stag-
and killing of infected herds, supplemented by other gering gait that appears to be directly related to a weak-
control measures. Despite these efforts, CSF has still not ness in the hindquarters. This is usually followed by a
been eliminated in many countries. This may be ac- posterior paresis. A purplish discoloration extending
counted for by the high density of pigs in certain areas, over the abdomen, snout, ears, and medial sides of the
the movement over long distances of pigs and pork and legs may occur near the terminal stage of the disease
pork products, and the frequent inability to trace the (Stewart 1981). Most pigs that suffer from acute CSF die
source of outbreaks. During the 1982–85 epizootic in between 10 and 20 days postinfection. In subacute CSF,
the Netherlands, the source of infection was recorded as pigs show less severe signs of disease and succumb with-
“unknown” in 54% of breeding and mixed herds and in in 30 days (Dunne 1975).
29% of fattening herds (Terpstra 1987). In Europe, the Persistence of CSFV in the host can result in different
origin of 13 out of 32 outbreaks (42%) in 1988 was un- clinical syndromes. Mengeling and Packer (1969) de-
known (Picard 1989); in Germany 50 of the 117 cases scribed three phases of chronic CSF based on the clinical
(43%) in 1994 had an unknown origin (Pittler et al. signs observed. In the first, or acute, phase of illness,
1995). The emergence of CSFV strains of reduced viru- anorexia, depression, elevated temperature, and
lence is also a major factor in explaining hitherto unsuc- leukopenia were present. After several weeks, the ap-
cessful eradication programs. Such strains give rise to petite and general appearance of the pigs improved
162 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

markedly, and their temperatures decreased to normal infected piglets. Among piglets infected in utero, skin he-
or slightly above normal values. Leukopenia usually per- morrhages are common and neonatal mortality is high.
sisted. This general clinical improvement characterized However, piglets can recover from an in utero–acquired
the second phase of illness. In the third phase, pigs again CSFV infection.
became anorectic and depressed, with temperatures of-
ten elevated until shortly before death. Runt pigs can de- PATHOGENESIS
velop during the course of chronic CSF. Such pigs are se-
verely retarded in growth, have skin lesions, and Under natural conditions, the mode of entry of CSFV in
frequently stand with arched backs. Pigs with chronic the pig is the oronasal route. Occasionally, the virus gains
CSF may survive for more than 100 days. access to the host through conjunctival or genital mu-
Van Oirschot and Terpstra (1977) reported a “late-on- cous membranes or skin abrasions. The tonsil is the pri-
set” disease as a sequel of congenital CSFV infection. The mary site of virus replication after oral and parenteral in-
syndrome was characterized by an initial, relatively long fection.
period during which pigs remained free of disease. Not The virus initially infects epithelial cells of the tonsil-
until a few months after primary exposure did the pigs lar crypts and subsequently spreads to the surrounding
develop mild anorexia and depression, conjunctivitis, lymphoreticular tissue. From the tonsil, CSFV is trans-
dermatitis, diarrhea, and locomotive disturbances lead- ferred via lymphatic vessels to the lymph nodes draining
ing to posterior paresis. Body temperatures were normal. the tonsillar region. The virus replicates in the regional
Most pigs survived for more than 6 months, but all even- lymph nodes, then reaches the peripheral blood, and
tually died (Table 14.1). from that time grows to high titers in spleen, bone mar-
Persistent, virtually inapparent infections can also row, visceral lymph nodes, and lymphoid structures lin-
develop after postnatal inoculation with CSFV strains of ing the small intestine. As a result of the growth in lym-
reduced virulence (Carbrey et al. 1977). Leukopenia is a phoid tissue and in circulating leukocytes and
consistent manifestation of persistent CSF, but in the mononuclear cells, the level of viremia is high. The virus
terminal stages of disease, leukocytosis may develop. probably does not invade parenchymatous organs until
Low-virulence strains of CSFV generally induce mild dis- late in the viremic phase. Generally, CSFV titers in lym-
ease or subclinical infections, which can be accompanied phoid tissues are higher than in parenchymatous organs.
by leukopenia. The spread of virulent virus throughout the pig is usual-
A congenital CSFV infection can result in abortion, ly completed in 5–6 days (Ressang 1973).
fetal mummification, malformations, stillbirth, and the The multiple hemorrhages seen in acute CSF are
birth of weak pigs with tremors or of healthy-looking yet caused by degeneration of endothelial cells in conjunc-

Table 14.1. General features of acute, chronic, and late-onset CSF


Feature Acute Chronic Late-Onset
Virulence High Moderate Low
of virus
Time of Postnatal Postnatal Prenatal
infection
Course of Short incubation period, severe Short incubation period, three Late onset of disease, gradually
illness depression, high fever, anorexia, phases of illness: (1) depression, aggravating depression and
conjunctivitis, constipation, fever, anorexia; (2) clinical anorexia, normal to slightly
diarrhea, convulsions, incoor- improvement; (3) terminal, elevated body temperatures,
dination, hemorrhages of skin exacerbation of disease conjunctivitis, dermatitis,
locomotion disturbances
Viremia High level Temporary reduction or disappearance Persistent high level
Leukopenia Develops quickly Develops quickly, followed by Develops late during infection
leukocytosis
Immune Absent Present Absent
response to
CSF
Death 10–20 days 1–3 months 2–11 months
Gross lesions Multiple hemorrhages (especially Ulcers of cecum and colon, Lymph node swelling, thymic
in lymph nodes and kidney), infarction of spleen, rib atrophy
infarction of spleen lesions
Microscopic Degeneration of endothelial cells, Degeneration of endothelial cells, Degeneration of endothelial
proliferation of reticular cells, severe lymphocyte depletion, cells, severe lymphocyte
encephalitis histiocytic hyperplasia, depletion, histocytic hyper-
glomerulonephritis plasia
CHAPTER 14 CLASSICAL SWINE FEVER (HOG CHOLERA) Van Oirschot 163

tion with severe thrombocytopenia and disturbance in fi- developmental age of the fetus and the virulence of the
brogen synthesis. Virtually all pigs die from acute CSF. virus strain largely determine the outcome of a congeni-
The mechanism actually responsible for death is not tal infection. Generally, the risk for fetal damage is high-
clear, but severe disturbance of the circulatory system er the earlier infection occurs during pregnancy.
appears to be the most likely cause (Fuchs 1968). Pigs that have recovered from CSF possess antibodies
During acute CSF, the pig’s immune reactivity to CSFV, but neutralizing antibodies can also be pro-
changes. A depressed secondary antibody response to duced in subacute fatal cases. Whereas pigs with chronic
lysozyme (Charley et al. 1980), highly abnormal respons- CSF are able to elicit a specific antibody response, pigs
es of peripheral blood and organ lymphocytes to T- and with congenital persistent CSF appear to be immunotol-
B-cell mitogens (Van Oirschot et al. 1981, 1983), and B- erant to the virus. Although porcine fetuses gain im-
lymphocyte deficiency (Susa et al. 1992) have been re- munocompetence around midgestation, only some of
ported. them may produce antibodies to CSFV in the last phase
Persistent CSFV infections are generally caused by of fetal development. Occasionally, after postnatal infec-
strains of reduced virulence. Arbitrarily, two forms of tion with certain strains of CSFV, neutralizing antibodies
persistence may be distinguished: chronic and late-onset are only transiently detectable or completely absent.
CSF (Table 14.1). This may be the consequence of a poor immunogenicity
The first phase of chronic CSF resembles acute CSF, of the virus. Such strains are generally of reduced viru-
but the virus spreads more slowly and virus titers in lence.
serum and organs tend to be lower. In the period of clin- In pigs that do not mount a normal antibody re-
ical improvement, virus titers in serum are low or absent sponse after a primary contact with CSFV, two phenom-
and viral antigen is usually limited to epithelial cells of ena have repeatedly been observed. Reexposure of such
tonsil, salivary gland, ileum, and kidney. A specific anti- pigs to CSFV can result in a kind of hyperreactivity char-
body response and/or decrease in the number of cells acterized by a shorter incubation period and a more se-
producing virus probably account for the temporary dis- vere illness than in primarily infected pigs. In contrast
appearance of CSFV from the serum. The simultaneous with this sensitization phenomenon, an enhanced resis-
circulation of viral antigens and antibody may result in tance to virulent CSFV has been described. Pigs persis-
deposition of antigen-antibody complexes in the kid- tently infected with CSFV of reduced virulence have a
neys, which eventually can lead to glomerulonephritis. markedly prolonged survival when inoculated with viru-
During the exacerbation of acute disease, virus is again lent CSFV. It is conceivable that these phenomena play a
distributed throughout the body. Its spread may be pro- role under field conditions.
moted by the immune exhaustion that appears to devel- The role of cell-mediated immunity in CSF has not
op in pigs with chronic CSF (Cheville and Mengeling yet been elucidated. Corthier (1978) and Remond et al.
1969). (1981) detected a transient blastogenic response to CSFV
Late-onset CSF initially runs an inapparent course. It antigens in subclinically affected or vaccinated pigs. A
may not be until several months after primary contact proliferative response of T lymphocytes against CSFV
with the virus that pigs develop signs of disease. These has been induced by infective virus, which appeared not
infections are the sequel of exposure to CSFV strains of to be directed to the immunodominant E2 protein. In ad-
low virulence during fetal life (Van Oirschot and Terpstra dition, a considerable nonspecific lymphoproliferation
1977). The pigs have a lifelong high level of viremia, has been observed (Kimman et al. 1993). An epitope rec-
which can be transiently decreased after the ingestion of ognized by CSFV-specific cytotoxic T lymphocytes has
colostral antibody. CSFV antigen is widespread through- been identified near the cleavage site between the pro-
out epithelial, lymphoidal, and reticuloendothelial tis- teins NS2.3 and NS4A (Pauly et al. 1995).
sues. Pigs with congenital persistent CSF do not mount a The precise pathogenic mechanisms of CSFV infec-
neutralizing antibody response to the virus. However, tions are not well understood. Some hypotheses are de-
the antibody response to antigens unrelated to CSFV is scribed in extensive reviews by Fuchs (1968) and Mahnel
generally normal, indicating a specific immune tolerance and Mayr (1974).
to the virus. The response of peripheral blood lympho-
cytes to mitogens is only slightly depressed in these per- LESIONS
sistently infected pigs (Van Oirschot 1979).
The pathogenesis of postnatal infections with low- In peracute cases of CSF, pathologic lesions are often
virulence CSFV is not well known. The growth of virus absent. In acute to subacute CSF, the pathologic picture
seems to be mainly restricted to tonsil and regional is that of a septicemic disease characterized by multiple
lymph nodes, but the virus can be disseminated via the hemorrhages of various sizes (caused by hydropic degen-
bloodstream, as evidenced by the frequently occurring eration and necrosis of endothelial cells lining the vascu-
transplacental transmission in pregnant sows. CSFV ap- lar system) in conjunction with defects in the blood
pears to grow across the placental barrier at one or more coagulation mechanism. In addition, catarrhal, fibri-
sites, and it subsequently spreads from fetus to fetus. The nous, and hemorrhagic inflammatory reactions are
164 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

present in digestive, respiratory, and urogenital tracts. heart, intestinal mucosa, serous and mucous mem-
Pathologic changes are most frequently observed in branes, and skin. The skin may also become cyanotic.
lymph nodes and kidney. The lymph nodes become Infarction is the result of a disrupted blood flow into
swollen, edematous, and then hemorrhagic. They com- a certain area resulting from the occlusion of blood cap-
monly have peripheral or more diffuse hemorrhages, giv- illaries by thrombi. The thrombi in turn may be caused
ing the nodes a marbled or red to near black appearance by the hydropic degeneration of endothelial cells cou-
respectively (Fig. 14.1). Virtually all lymph nodes may be pled with the tendency of erythrocytes to clump in a
affected. Microscopically, depletion of lymphocytes and manner termed “sludging.” Infarction of the spleen (Fig.
reticular hyperplasia are seen. 14.3) is considered to be almost pathognomonic for CSF.
Hemorrhages of the kidney may vary in size from Infarctions occur as dark blebs of various sizes, raised
hardly visible petechiae to ecchymotic hemorrhages. slightly above the surrounding surfaces. They may ap-
They frequently occur on the surface of the cortex (Fig. pear singly or as a series of lesions coalescing to form a
14.2) and are less common in the medullary pyramids continuous border of infarcts along the edge of the
and hilus. Petechial to ecchymotic hemorrhages can also spleen. Infarction of the gallbladder and tonsil may oc-
be observed in urinary bladder, larynx and epiglottis, cur. In the latter organ it causes necrosis, which upon

14.1. Peripheral hemorrhage


of the mandibular lymph node.
(Courtesy W. C. Stewart.)

14.2. Kidney showing numerous


petechial hemorrhages. (Courtesy
W. C. Stewart.)
CHAPTER 14 CLASSICAL SWINE FEVER (HOG CHOLERA) Van Oirschot 165

14.3. Infarction of the spleen.


(Courtesy L. D. Miller.)

bacterial invasion leads to suppurative tonsillitis. and focal necrosis can also be observed in the brain.
Pigs with acute to subacute CSF can show infarctions In persistent CSF, hemorrhages and infarctions are
and hemorrhages in the lung. Presumably as a conse- less pronounced or are completely absent. Although de-
quence of secondary bacterial infections, catarrhal to generation of endothelial cells is a consistent finding in
fibrinous bronchopneumonia and pleuritis may develop. persistently infected pigs, it does not result in severe cir-
The heart is usually flabby and shows some myocardial culatory disturbances. The most outstanding lesions in
congestion. In an animal dying of CSF, the stomach is pigs with persistent CSF are atrophy of the thymus and
usually empty except for a yellow, bilious fluid and a severe depletion of lymphocytes and germinal follicles in
small amount of feed. The fundus is often markedly con- peripheral lymphoid organs. Histiocytic hyperplasia
gested and hemorrhagic. The mucosal surface may con- with phagocytosis of lymphocytic debris is also fre-
tain petechiae and mild to severe erosions. A mild to quently seen. The plasmacytosis and glomerulonephritis
moderate catarrhal or necrotic enteritis is found in the that occur in chronic CSF were not present in pigs with
intestines. Mesenteric blood vessels are usually marked- late-onset CSF. Persistent CSFV infections induce adren-
ly engorged. Occasionally, subserous ecchymotic and al cortical hyperplasia characterized by an increased
suffusive hemorrhages occur in the small and large in- width of the zona fasciculata and atrophy of the zona
testines (Stewart 1981). glomerulosa and zona reticulata (Cheville and Men-
Most CSFV-infected swine show an encephalitis at geling 1969; Van der Molen and Van Oirschot 1981).
necropsy, of which the principal lesion is perivascular Necrosis and ulcerations, sometimes in the form of but-
cuffing. Proliferation of endothelial cells, microgliosis, ton ulcers (Fig. 14.4), in the cecum and colon and rib le-

14.4. Button ulcers in the cecum


and colon. (Courtesy L. D. Miller.)
166 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

sions are common in persistent CSF. Chronic rib lesions with swine. In acute CSF there is a rapid spread of the
are caused by a sudden calcification of large numbers of disease among pigs of all ages and high mortality at 1–2
mature cartilage cells and appear as a marked transverse weeks after the onset of clinical signs. Leukopenia is a
line of semisolid bone structure across the rib proximal consistent finding in pigs with CSF. At necropsy, lesions
from the costochondral junction (Dunne 1975) (Table of diagnostic significance are hemorrhages in lymph
14.1). nodes, kidney, and other organs and infarction of the
Congenital CSFV infections can result in mummifica- spleen.
tion, stillbirth, and malformations (Fig. 14.5). General- Acute CSF may be confused on clinical and patholog-
ized subcutaneous edema, hydrops ascites, and hy- ic grounds with African swine fever, porcine reproduc-
drothorax are the most pronounced lesions in stillborn tive and respiratory syndrome, septicemic salmonellosis,
pigs. Malformations consist of deformities of head and pasteurellosis, streptococcosis, erysipelas, or
limbs, hypoplasia of cerebellum and lungs, and hy- Haemophilus suis infections; therefore, it is recommend-
pomyelogenesis. In utero–infected piglets that die short- ed that clinical diagnosis be confirmed in the laboratory.
ly after birth often show petechial hemorrhages of skin In many countries that have eradication programs that
and internal organs. include destruction of positive herds, any tentative field
diagnosis has to be confirmed by laboratory investiga-
DIAGNOSIS tions.
Unlike in acute CSF, it is virtually impossible to make
Outbreaks of typical acute CSF can be diagnosed in the a clinical diagnosis in cases of subacute, chronic, or late-
field with reasonable certainty on the basis of an accu- onset CSF because of the wide variability of clinical signs
rate anamnesis and a thorough clinical and pathologic and pathologic lesions. Clinical signs are milder than in
investigation. An anamnesis of a CSF outbreak often in- acute CSF; they can occur intermittently, or the infection
cludes one or more of the following: recent purchase of may go unnoticed for months. In addition, persistent in-
pigs, cases of CSF on adjacent or nearby farms, garbage fections may involve only a few pigs in a herd. In such
feeding, or a recent visit by persons having close contact cases, counting of blood leukocytes may prove useful. Fi-
nally, infected swine may not have signs indicative of
CSF because of concurrent infections with other infec-
tious agents. Persistent CSFV infections are difficult to
recognize, so the diagnosis is often delayed. Consequent-
ly, such an infected herd may act as a source for further
dissemination of the virus. Therefore, in countries where
CSF is present, samples should be submitted to the labo-
ratory when the clinical signs, combined with the anam-
nesis, raise even the slightest suspicion of CSF. CSFV in-
fections by strains of reduced virulence seem to increase
in importance, particularly in areas where CSF has been
endemic for a long period and in final stages of eradica-
tion programs. In the United States 55% of field isolates
in the period 1965–75 were characterized as being of re-
duced virulence (Carbrey et al. 1977).
Laboratory diagnosis of CSF is based on detection of
viral antigen, isolation of virus, or demonstration of vi-
ral antibody.
The direct fluorescent antibody (FA) test on frozen
tissue sections is the method of choice for detecting viral
antigen. Samples should be collected from a number of
dead or sick pigs and submitted fresh (preferably on ice)
and without the addition of any preservatives to the lab-
oratory. Tissues to submit for laboratory diagnosis are
tonsil, spleen, kidney, and distal part of the ileum. The
tonsil, which is the first tissue to become positive after
exposure to CSFV, is by far the most important organ for
14.5. Mummified fetuses and stillborn pigs of a sow detecting viral antigen (Fig. 14.6). The ileum is frequent-
exposed to low-virulence CSFV at day 43 of gestation. ly found to be positive in more prolonged cases of CSF.
(Courtesy W. C. Stewart.) An absence of viral antigen in several tissues does not ex-
CHAPTER 14 CLASSICAL SWINE FEVER (HOG CHOLERA) Van Oirschot 167

14.6. CSF viral antigen in epithelial cells of tonsillar crypts detected by FA test. (Courtesy C. Terpstra.)

clude CSF as the cause of the outbreak; where suspicion Molecular biology has provided genetic assays that
remains, more pigs should be examined. The FA test on rapidly differentiate between CSFV and other pestivirus-
frozen tissue sections is a simple, rapid (can be complet- es. These are based on reverse transcription followed by
ed in 2 hours), and reliable technique and has become polymerase chain reaction (RT-PCR) and direct DNA se-
the official diagnostic test in many CSF eradication pro- quencing of the amplified cDNA from the 5′ noncoding
grams. region (Hofmann et al. 1994), on discriminatory RT-PCR
CSFV shares common antigens with BVDV and BDV. of sequences of E2 (Katz et al. 1993) or of a nonstruc-
Therefore, the direct FA test fails to discriminate be- tural protein (Wirz et al. 1993), or on RT-PCR followed
tween CSFV and BVDV/BDV antigens. Because pigs can by restriction enzyme mapping of the 5′ noncoding re-
become infected with BVDV or BDV, the FA test may gion (Vilcek et al. 1994).
yield false-positive results. It is crucial to distinguish CS- In pigs vaccinated with the attenuated Chinese (C)
FV from BVDV/BDV, because a BVDV or BDV infection strain of CSF, viral antigen can be demonstrated by the
erroneously diagnosed as CSF leads, in most countries, direct FA test until about 2 weeks postvaccination (Terp-
to killing of the herd and other control measures and stra 1978). Thus in areas where vaccination is practiced,
may result in loss of the CSF-free status. To differentiate it may sometimes be necessary to differentiate between
between CSFV and BVDV/BVD it no longer is necessary field virus and vaccine virus. For this purpose, a pair of
to perform time-consuming cross-neutralization tests conjugated MABs can be used, one of which reacts with
with sera of in-contact pigs or pigs inoculated with the a conserved epitope of CSFV and the other with most
isolate. A rapid differentiation can be made by staining CSFV strains but not with vaccine strains. A positive re-
frozen tissue sections or infected cell cultures with conju- action by the first MAB and a negative by the second in-
gates of MABs directed against conserved epitopes of dicate that vaccine virus may be present (Fig. 14.7) and
CSFV. When the selected MABs react with the isolate, it warrant further investigation. Lapinized vaccine strains
is CSFV, and when antigen is not detected, BVDV/BVD is can be distinguished from field strains by their ability to
involved (Fig. 14.7) (Wensvoort et al. 1986, 1989). Such induce fever and antibodies to CSFV after intravenous in-
nonreactive isolates need to be confirmed as BVDV or oculation in rabbits.
BDV. CSFV may be isolated by inoculation of a porcine kid-
14.7. Frozen sections of tonsils of pigs infected with the virulent Brescia strain (1, 2, 3) or the
attenuated vaccine strain C (4, 5, 6) of CSVF or a strain of BVDV (7, 8, 9). Sections were reacted
with peroxidase conjugates prepared from a polyclonal anti-CSFV serum (1, 4, 7) or from MABs that
recognize different epitopes of CSFV (MAB 1 [2, 5, 6], MAB 2 [3, 6, 9]). (Courtesy G. Wensvoort.)
CHAPTER 14 CLASSICAL SWINE FEVER (HOG CHOLERA) Van Oirschot 169

ney (PK-15) cell line with a 2–10% mixed homogenate of called upon and zoo sanitation measures may be insti-
tonsil and spleen of a suspected pig. After 24–72 hours, tuted to help eradicate the virus. All pigs in infected
the cultures are examined for viral antigen by the FA test herds are destroyed, the source of infection and possible
(Fig. 14.7). The virus isolation procedure is more sensi- contacts are traced, “standstill” restrictions become op-
tive than the FA on frozen tissue sections. erative, and infected farms are intensively disinfected.
A promising method to support a rapid diagnosis of Liquid manure can best be disinfected by chemical treat-
CSF in large numbers of live pigs is antigen detection by ment. Countries with sporadic outbreaks of the disease
enzyme-linked immunosorbent assay (ELISA) in blood usually apply similar control and eradication proce-
and tissue samples. However, this method is not as sensi- dures.
tive as virus isolation, in that virus isolation can detect In countries where CSF is enzootic, vaccination is of-
infection some days earlier. In addition, the tests are not ten practiced, and in some countries vaccination is sup-
yet specific for CSFV (Shannon et al. 1993; Depner et al. plementary to the killing of infected herds. Vaccination
1995). usually ceases when no more outbreaks occur or when a
Antibody detection can be a useful diagnostic tool for stage is reached where destruction of infected herds
suspected farms where the usual virus detection proce- alone may eliminate the residual virus.
dures have failed and in the last phase of an eradication In the Netherlands, a strict vaccination regime pur-
program to detect subclinically infected herds. Serologic sued for 1 year and supported by the usual veterinary po-
tests are imperative if a country desires to become inter- lice and zoo sanitation measures succeeded in eradicat-
nationally recognized as free from CSF. ing CSF from three enzootic areas. The program
Several tests are available for the detection of anti- consisted of mass vaccination of all pigs over 2 weeks of
bodies to CSFV. The virus neutralization test in its vari- age and supplementary vaccination of previously unvac-
ous modifications (Holm Jensen 1981; Terpstra et al. cinated 6- to 8-week-old pigs and newly introduced stock
1984) is often used. However, it may measure antibodies at monthly intervals. Vaccination was compulsory, and
to BVDV because low neutralizing antibody titers to CS- all vaccinated pigs were identified by ear tagging. The
FV can be induced by infections with BVDV (Liess et al. number of outbreaks in the vaccinated areas declined in
1977); therefore, such sera need to be tested in a BVDV- 2 weeks, and the areas were free of CSF from the fifth
neutralization test. Higher antibody titers to BVDV than month after the start of the program (Terpstra and
to CSFV indicate infection with BVDV, but a concurrent Robijns 1977). In an emergency vaccination campaign,
infection with CSFV may not be completely excluded. In supplementary vaccination was conducted in piglets
such cases, more pigs from the herd should be tested. born from vaccinated sows at the age of 7–9 weeks, and
ELISAs, which are rapid and simple to perform, have breeding gilts born from vaccinated sows were revacci-
been developed for large-scale serologic investigations. nated when 6–7 months old to boost herd immunity
An ELISA that differentiates between antibodies to CSFV (Terpstra and Wensvoort 1987). The European Union
and BVDV employs two MABs directed against different has adopted a nonvaccination policy for CSF, which im-
epitopes on glycoprotein E2. Because neither MAB rec- plies that emergency vaccinations are also prohibited.
ognizes BVDV, the chance that BVDV antibodies are de- Vaccines originally attenuated by serial passages in
tected is virtually negligible (Wensvoort et al. 1988). The rabbits (C strain) or attenuated by passages in cell cul-
E2 antigen is produced by recombinant baculovirus in tures (Japanese GPE− strain, French Thiverval strain) are
insect cells. This ELISA is used for seroepizootiologic sur- used as an aid in the control of CSF. The C strain has an
veys in various countries (Wensvoort et al. 1988). insertion of 13 nucleotides in the 3′ noncoding region
Because antibodies to field strains of CSFV cannot compared with that of virulent CSFV strains (Moor-
yet be distinguished from antibodies to vaccine virus, mann et al. 1996). The GPE− strain differs in 225 nu-
vaccination limits the use of serologic tests for diagnostic cleotides from its parental strain (Ishikawa et al. 1995).
purposes. These vaccines are highly efficacious and safe (Aynaud
1988). The simultaneous vaccination method and use of
PREVENTION inactivated vaccines have been abandoned in most coun-
tries. In addition, the early attenuated CSFV vaccines are
To prevent the reintroduction of CSFV, countries free of no longer used because they induced a variety of fetal ab-
CSF ban the import of live pigs, pork, and insufficiently normalities after vaccination of pregnant sows.
heated pork products from countries where CSF is pres- The C strain is the most extensively used vaccine. The
ent. In addition, swill is generally destroyed or effective- growth of the C strain appeared to be mainly restricted
ly sterilized before being fed to pigs. Cleansing and dis- to lymphoid tissues, especially the tonsil, although C vi-
infecting of mechanical vectors, such as trucks, are too ral antigen has occasionally been detected in kidneys
often neglected as preventive measures against reintro- (Terpstra 1978). The C strain can pass the placental bar-
duction of CSFV into an area. Should a case of CSF arise rier of pregnant sows but does not seem to produce any
in countries free of CSF, veterinary “police” may be abnormality in infected fetuses (Tesmer et al. 1973). It is
170 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

innocuous for pregnant sows and newborn piglets. The C of virus replication in the tonsils of pigs previously vac-
strain can be transmitted from vaccinated pigs to non- cinated with a Chinese strain vaccine and challenged
vaccinated contact pigs (Van Bekkum 1977). However, oronasally with a virulent strain of classical swine fever
the chance of reversion to virulence appears to be mini- virus. Vet Microbiol 14:105–113.
mal, because it retained its attenuated characteristics af- Carbrey, E. A.; Stewart, W. C.; Kresse, J. L.; and Snijder, M.
ter 20–30 serial passages in pigs (Aynaud 1988) and be- L. 1977. Inapparent hog cholera infection following the
haved similarly in pigs treated with corticosteroids as in inoculation of field isolates. In CEC Seminar on Hog
nontreated pigs (Florent et al. 1969; Kamijyo et al. 1976). Cholera/Classical Swine Fever and African Swine Fever,
The attenuated vaccines usually confer immunity Hannover, EUR 5904, pp. 214–230.
within 1 week after inoculation, and the immunity per- Charley, B.; Corthier, G.; Houdayer, M.; and Rouze, P. 1980.
sists for at least 2–3 years, and probably lifelong. Vacci- Modifications des reactions immunitaires au cours de la
nation not only protects against disease but also greatly peste porcine classique. Ann Rech Vét 11:27–33.
reduces the replication of virulent CSFV upon challenge Cheville, N. F., and Mengeling, W. L. 1969. The pathogene-
(Biront et al. 1987) and prevents transmission of chal- sis of chronic hog cholera (swine fever): Histologic, im-
lenge virus from pigs with moderate neutralizing anti- munofluorescent, and electron microscopic studies. Lab
body titers to in-contact pigs (Terpstra and Wensvoort Invest 20:261–274.
1988). Vaccinated sows transmit antibodies to their off- Corthier, G. 1978. Cellular and humoral immune response
spring via colostrum. Because the intestine of the new- in pigs given vaccinal and chronic hog cholera viruses.
born piglet is permeable for immunoglobulins during Am J Vet Res 39:1841–1844.
the first 36–48 hours of life, antibodies to CSFV enter di- Depner, K.; Bauer, T.; and Liess, B. 1992. Thermal and pH
rectly into the circulation. These maternally derived anti- stability of pestiviruses. Rev Sci Tech OIE (Office Inter-
bodies have a half-life of approximately 14 days. Piglets national des Epizooties) 11:885–893.
born to vaccinated sows are protected for 5–8 weeks Depner, K.; Paton, D. J.; Cruciere, C.; De Mia, G. M.; Müller,
against mortality from CSF but not against replication A.; Koenen, F.; Stark, R.; and Liess, B. 1995. Evaluation
and shedding of virulent virus (Terpstra 1977). of the enzyme-linked immunosorbent assay for the
Maternally derived antibodies inhibit the develop- rapid screening and detection of classical swine fever
ment of active immunity after vaccination. Although a virus antigens in the blood of pigs. Rev Sci Tech OIE (Of-
high percentage of piglets born to vaccinated sows can fice International des Epizooties) 14:677–689.
effectively be vaccinated around 6 weeks of age, revacci- Dunne, H. W. 1975. Hog cholera. In Diseases of Swine, 4th
nation of such pigs around 5 months will probably in- ed. Ed. H. W. Dunne and A. D. Leman. Ames: Iowa State
crease herd immunity. The suppression by maternally Univ Press.
derived antibodies can be reduced by substantially in- Edwards, S., and Sands, J. J. 1990. Antigenic comparisons of
creasing the amount of vaccine virus per dose (Lin et al. hog cholera virus isolates from Europe, America and
1982). Aerosol immunization of pigs has yielded varying Asia using monoclonal antibodies. Dtsch Tierärztl
results with regard to efficacy. Wochenschr 97:79–81.
The development of marker vaccines that allow dif- Edwards, S.; Moennig, V.; and Wensvoort, G. 1991. The de-
ferentiation between infected and vaccinated pigs is in velopment of an international reference panel of mono-
progress. Such a marker vaccine can be composed of the clonal antibodies for the differentiation of hog cholera
E2 protein that is excreted from insect cells infected with virus from other pestiviruses. Vet Microbiol 29:101–108.
recombinant E2-baculovirus and that is subsequently Florent, A.; Thomas, J.; and Leunen, J. 1969. Contrôle des
formulated in an adjuvant (Hulst et al. 1993). In this vaccins vivants contre la peste porcine: Intérêt de l’im-
case, serologic differentiation can be based on detection munodépression pour la mise en évidence de la viru-
of antibody against Erns. A CSFV marker vaccine may al- lence résiduelle. Bull l’Office Int Epizooties 72:665–669.
so be based on a genetically engineered live deletion mu- Fuchs, F. 1968. Schweinepest. In Handbuch der Virusinfek-
tant virus. The engineering of full-length cDNA of the tionen bei Tieren, vol. 3. Ed. H. Rohrer. Jena: Gustav Fis-
genomes of the C strain (Moormann et al. 1996) and of cher.
a virulent strain (Meyers et al. 1996) from which infec- Haas, R.; Ahl, R.; Böhm, R.; and Strauch, D. 1995. Inactiva-
tious RNA can be transcribed provides the basis for the tion of viruses in liquid manure. Rev Sci Tech OIE (Of-
development of live marker vaccines against CSF. fice International des Epizooties) 14:435–445.
Hanson, R. P. 1957. Origin of hog cholera. J Am Vet Med As-
REFERENCES soc 131:211–218.
Hofmann, M. A.; Brechtbühl, K.; and Stäuber, N. 1994.
Aynaud, J. M. 1988. Principles of vaccination. In Classical Rapid characterization of new pestivirus strains by di-
Swine Fever and Related Viral Infections. Ed. B. Liess. rect sequencing of PCR-amplified cDNA from the 5′
Boston: Martinus Nijhoff, pp. 165–180. noncoding region. Arch Virol 139:217–229.
Biront, P.; Leunen, J. I.; and Vandeputte, J. 1987. Inhibition Holm Jensen, M. 1981. Detection of antibodies against hog
CHAPTER 14 CLASSICAL SWINE FEVER (HOG CHOLERA) Van Oirschot 171

cholera virus and bovine viral diarrhea virus in porcine Mengeling, W. L., and Packer, R. A. 1969. Pathogenesis of
serum: A comparative examination using CF, PLA and chronic hog cholera: Host response. Am J Vet Res
NPLA assays. Acta Vet Scand 22:85–98. 30:409–417.
Horzinek, M. C. 1981. Non-Arthropod-Borne Togaviruses. Meyers, G., and Thiel, H.-J. 1996. Molecular characteriza-
New York: Academic Press. tion of pestiviruses. Adv Virus Res 47:53–118.
Hughes, R. W., and Gustafson, D. P. 1960. Some factors that Meyers, G.; Rumenapf, T.; and Thiel, H.-J. 1989. Molecular
may influence hog cholera transmission. Am J Vet Res cloning and nucleotide sequence of the genome of hog
21:464–471. cholera virus. Virology 171:555–567.
Hulst, M. M.; Westra, D. F.; Wensvoort, G.; and Moormann, Meyers, G.; Thiel, H.-J.; and Rümenapf, T. 1996. Classical
R. J. M. 1993. Glycoprotein E1 of hog cholera virus ex- swine fever virus: Recovery of infectious viruses from
pressed in insect cells protects swine from hog cholera. J cDNA constructs and generation of recombinant cy-
Virol 67:5435–5442. topathogenic defective interfering particles. J Virol
Hulst, M. M.; Himes, G.; Newbigin, E.; and Moormann, R. 70:1588–1595.
J. M. 1994. Glycoprotein E2 of classical swine fever Moormann, R. J. M.; Warmerdam, P. A. M.; Van der Meer,
virus: Expression in insect cells and identification as a ri- B.; Schaaper, W. W. M.; Wensvoort, G.; and Hulst, M.
bonuclease. Virology 200:558–565. M. 1990. Molecular cloning and nucleotide sequence of
Ishikawa, K.; Nagai, H.; Katayama, K.; Tsutsui, M.; Tan- hog cholera virus strain Brescia and mapping of the ge-
abayashi, K.; Takeuchi, K.; Hishiyama, M.; Saitoh, A.; nomic region encoding envelope protein E1. Virology
Takagi, M.; Gotoh, K.; Maramatsu, M.; and Yamada, A. 177:184–198.
1995. Comparison of the entire nucleotide and deduced Moormann, R. J. M.; Van Gennip, H. G. P.; Miedema, G. K.
amino acid sequences of the attenuated hog cholera vac- W.; Hulst, M. J.; and Van Rijn, P. A. 1996. Infectious
cine strain GPE− and the wild-type parental strain ALD. RNA transcribed from an engineered full-length cDNA
Arch Virol 140:1385–1391. template of the genome of a pestivirus. J Virol
Kamijyo, Y.; Onkuma, S.; Shimizu, M.; and Shimizu, Y. 70:763–770.
1976. Effect of dexamethasone on the multiplication of Paton, D. J. 1995. Pestivirus diversity. J Comp Pathol
attenuated strains of hog cholera virus in pigs. Vet Mi- 112:215–236.
crobiol 1:475–477. Pauly, T.; Elbers, K.; König, M.; Lengsfeld, T.; Saalmüller, A.;
———. 1977. Differences in pathogenicity and antigenicity and Thiel, H.-J. 1995. Classical swine fever virus: Specif-
among hog cholera virus strains. Nat Inst Anim Health ic cytotoxic T lymphocytes and identification of a T cell
Q (Tokyo) 17:133–140. epitope. J Gen Virol 76:3039–3049.
Katz, J. B.; Ridpath, J. F.; and Bolin, S. R. 1993. Presumptive Picard, M. 1989. Les pestes porcines en France et en Europe
diagnostic differentiation of hog cholera virus from en 1988. Epidémiol Santé Anim 16:27–32.
bovine viral diarrhea and border disease viruses by us- Pittler, H.; Fielder, J.; Jentsch, D.; Hasselbach, P.; and
ing a cDNA nested-amplification approach. J Clin Mi- Kramer, M. 1995. Der Schweinepest-Seuchenzug
crobiol 31:565–568. 1993/1994, Probleme und Konsequenzen. Tierärztl Um-
Kimman, T. G.; Bianchi, A. T. J.; Wensvoort, G.; De Bruin, T. schau 50:522–530.
G. M.; and Meliefste, C. 1993. Cellular immune re- Remond, M.; Plateau, E.; and Cruciere, C. 1981. In vitro
sponse to hog cholera virus (HCV): T cells of immune study of the cellular response of pigs vaccinated against
pigs proliferate in vitro upon stimulation with live HCV, classical swine fever. Zentralblatt für Veterinärmed (B)
but the E1 envelope glycoprotein is not a major T-cell 28:743–748.
antigen. J Virol 67:2922–2927. Ressang, A. A. 1973. Studies on the pathogenesis of hog
Liess, B.; Frey, H. R.; and Prager, D. 1977. Antibody response cholera. I. Demonstration of hog cholera virus subse-
of pigs following experimental infections with strains of quent to oral exposure. Zentralblatt für Veterinärmed
hog cholera and bovine viral diarrhea virus. In CEC Sem- (B) 20:256–271.
inar on Hog Cholera/Classical Swine Fever and African Schneider, R.; Unger, G.; Stark, R.; Schneider-Scherzer, E.;
Swine Fever, Hannover, EUR 5904, pp. 200–213. and Thiel, H.-J. 1993. Identification of a structural gly-
Lin, T. T. C.; Lai, S. S.; Chen, C. S.; and Lee, R. C. T. 1982. Im- coprotein of an RNA virus as a ribonuclease. Science
mune response to different doses of LPC-China virus in 261:1169–1171.
pigs with different levels of colostral hog cholera anti- Shannon, A. D.; Morrissy, C.; Mackintosh, S. G.; and West-
body. Proc Int Congr Pig Vet Soc 7:132. bury, H. A. 1993. Detection of hog cholera virus anti-
Lowings, P.; Ibata, G.; Needham, J.; and Paton, D. 1996. gens in experimentally infected pigs using an antigen-
Classical swine fever virus diversity and evolution. J Gen capture ELISA. Vet Microbiol 34:233–248.
Virol 77:1311–1321. Stewart, W. C. 1981. Hog cholera. In Diseases of Swine, 5th
Mahnel, H., and Mayr, A. 1974. Schweinepest. In Infektion- ed. Ed. A. D. Leman, R. D. Glock, W. L. Mengeling, R. H.
skrankheiten und ihre Erreger, vol. 16. Jena: Gustav Fis- C. Penny, E. Scholl, and B. Straw. Ames: Iowa State Univ
cher. Press.
172 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Susa, M.; König, M.; Saalmüller, A.; Reddehase, M. J.; and immune system. Thesis, State Univ Utrecht.
Thiel, H.-J. 1992. Pathogenesis of classical swine fever; Van Oirschot, J. T., and Terpstra, C. 1977. A congenital per-
B-lymphocyte deficiency caused by hog cholera virus. J sistent swine fever infection. I. Clinical and virological
Virol 66:1171–1175. observations. Vet Microbiol 2:121–132.
Terpstra, C. 1977. The immunity against challenge with Van Oirschot, J. T.; De Jong, D.; and Huffels, A. D. N. H. J.
swine fever virus of piglets from sows vaccinated with C- 1981. Effect of infections with swine fever on immune
strain virus. Tijdschr voor Diergeneeskunde functions. I. Response of lymphocytes from blood and
102:1293–1298. lymphoid organs from infected and normal pigs to anti-
———. 1978. Detection of C-strain virus in pigs following immunoglobulin serum and protein A. Vet Microbiol
vaccination against swine fever. Tijdschr voor Dierge- 6:41–57.
neeskunde 103:678–683. ———. 1983. Effect of infections with swine fever virus on
———. 1987. Epizootiology of swine fever. Vet Quart (Sup- immune functions. II. Lymphocyte response to mito-
pl)9:50–60. gens and enumeration of lymphocyte subpopulations.
Terpstra, C., and Robijns, K. G. 1977. Experience with re- Vet Microbiol 8:81–95.
gional vaccination against swine fever in enzootic areas Van Rijn, P. A.; Miedema, G. K. W.; Wensvoort, G.; Van
for limited periods using C-strain virus. Tijdschr voor Gennip, H. G. P.; and Moormann, R. J. M. 1994. Anti-
Diergeneeskunde 102:106–112. genic structure of envelope glycoprotein E1 of hog
Terpstra, C., and Wensvoort, G. 1987. Influence of the vac- cholera virus. J Virol 68:3934–3942.
cination regime on the herd immune response for swine Vilcek, S.; Herring, A. J.; Herring, J. A.; Nettleton, P. F.; Low-
fever. Vet Microbiol 13:143–151. ings, J. P.; and Paton, D. J. 1994. Pestiviruses isolated
———. 1988. The protective value of vaccine-induced neu- from pigs, cattle and sheep can be allocated into at least
tralising antibody titres in swine fever. Vet Microbiol three genogroups using polymerase chain reaction and
16:123–128. restriction endonuclease analysis. Arch Virol
Terpstra, C.; Bloemraad, M.; and Gielkens, A. L. J. 1984. The 136:309–323.
neutralizing peroxidase-linked assay for detection of an- Weiland, E.; Ahl, R.; Stark, R.; Weiland, F.; and Thiel, H.-J.
tibody against swine fever virus. Vet Microbiol 1992. A second envelope glycoprotein mediates neutral-
9:113–120. ization of a pestivirus, hog cholera virus. J Virol
Tesmer, S.; Urbaneck, D.; Kaden, V.; Wittmann, D.; and 66:3677–3682.
Hahnefeld, H. 1973. Zur Wirkung von Schweinepest- Wensvoort, G. 1989. Epitopes on structural proteins of hog
Lebendvirusvakzine aus dem Impfvirusstam “C” bei tra- cholera (swine fever) virus. Thesis, State Univ Utrecht.
genden Sauen und deren Nachzucht. Monatsh für Vet- Wensvoort, G.; Terpstra, C.; Boonstra, J.; Bloemraad, M.;
erinärmed 28:251–254. and Van Zaane, D. 1986. Production of monoclonal an-
Van Bekkum, J. G. 1977. Experience in the Netherlands with tibodies against swine fever virus and their use in labo-
the lapinised, so-called Chinese (C) strain of vaccine. In ratory diagnosis. Vet Microbiol 12:101–108.
CEC Seminar on Hog Cholera/Classical Swine Fever and Wensvoort, G.; Bloemraad, M.; and Terpstra, C. 1988. An
African Swine Fever, Hannover, EUR 5904, pp. 379–391. enzyme immunoassay employing monoclonal antibod-
Van der Molen, E. J., and Van Oirschot, J. T. 1981. Congeni- ies and detecting specifically antibodies to classical
tal persistent swine fever (hog cholera). I. Pathomorpho- swine fever virus. Vet Microbiol 17:129–140.
logical lesions in lymphoid tissues, kidney and adrenal. Wensvoort, G.; Terpstra, C.; De Kluijver, E. P.; Kragten, C.;
Zentralblatt für Veterinärmed (B) 28:89–101. and Warnaar, J. C. 1989. Antigenic differentiation of
Van Oirschot, J. T. 1979. Experimental production of con- pestivirus strains with monoclonal antibodies against
genital persistent swine fever infections. II. Effect on hog cholera virus. Vet Microbiol 21:9–20.
functions of the immune system. Vet Microbiol Wirz, B.; Tratschin, J.-D.; Müller, H. K.; and Mitchell, D. B.
4:133–147. 1993. Detection of hog cholera virus and differentiation
———. 1980. Persistent and inapparent infections with from other pestiviruses by polymerase chain reaction. J
swine fever virus of low virulence: Their effects on the Clinical Microbiol 31:1148–1154.
Japanese B Encephalitis
15 H. S. Joo and R. M. Chu

Japanese B encephalitis (JE) is a mosquito-borne viral


disease in animals and humans. Most domestic animals
are vulnerable to the virus, including horses, cattle,
sheep, goats, and pigs. Other animals, such as rabbits,
rats, pigeons, dogs, ducks, chickens, wild birds, and rep-
tiles, are also susceptible. Experimental infection has
been reported in mice and in a few species of lizards.
The pig is considered the most important natural am-
plifying animal for JE virus (JEV). The disease is impor-
tant in pregnant sows because infection causes stillbirth
and other reproductive disturbances; affected boars may
have an acute inflammatory reaction in the testes. Hors-
es infected by the virus develop lesions in the central ner-
vous system (CNS), and similar lesions have been found
in donkeys and monkeys. In other animals, infection is
usually subclinical.
JE is of primary significance in humans, epidemics
having been recorded in Japan (1935), Korea (1949), and
India and Nepal (1978). The disease is one of the most
common mosquito-borne diseases of the human CNS in 15.1. Geographic distribution of JE outbreaks in Asia.
Japan, China, and other western Pacific countries. Isola-
tion and identification of the virus were first described
by Fujita (1933) and Taniguchi et al. (1936). Episodes of There are three structural and several nonstructural pro-
infection in humans occur annually during mosquito teins. The structural proteins are envelope glycoprotein
(Culex tritaeniorhynchus) season. In most people disease E (54 kDa), nonglycosylated envelope protein M (8 kDa),
is subclinical or mild, but fatal encephalitis develops in and capsid protein C (14 kDa). Envelope protein E carries
some children, and abortion is reported in pregnant epitopes that elicit neutralization antibody. At least eight
women (Chaturvedi et al. 1980). epitopes are found on protein E, and one of the critical N
The geographic distribution of the disease is restrict- sites on the protein shows JEV specificity (Kimura-Kuro-
ed to eastern Asia (Fig. 15.1); infection has been recog- da and Yasui 1986). The oligonucleotide fingerprinting
nized in Japan, eastern Soviet Union, Korea, China, Tai- technique (Hori et al. 1986) reveals that mutations of
wan, the Philippines, Indonesia, Singapore, Malaysia, JEV are present in strains that were isolated years apart
Hong Kong, Vietnam, Laos, Bangladesh, Nepal, Thai- in Japan and Thailand.
land, Burma, Sri Lanka, India, and the Pacific islands. Use of monoclonal antibodies in hemagglutination
inhibition, neutralization, enzyme-linked immunosor-
ETIOLOGY bent assay (ELISA), and competitive binding assay has
shown that the closest relative of the JEV is the Murray
Physicochemical Properties Valley encephalitis virus; the next closest relative is the
JE is caused by a filterable virus, of the genus Flavivirus, West Nile virus; the St. Louis encephalitis virus is the
the only genus in the family Flaviviridae, formerly least closely related (Kimura-Kuroda and Yasui 1986).
grouped in the family Togaviridae. The virus is spherical, These relationships are confirmed by comparing the ho-
about 40 nm in diameter, and enveloped, and it contains mology of the viruses’ nucleotides and amino acids
single-stranded ribonucleic acid (RNA) with icosahedral (McAda et al. 1987).
capsids. The complete nucleotide sequence of the JEV The JEV is unstable in the environment and easily in-
genome RNA contains 10,976 nucleotides correspond- activated by disinfectants. The virus is sensitive to ether,
ing to 3432 amino acid residues (Sumiyoshi et al. 1987). chloroform, and sodium desoxycholate and also to pro-

173
174 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

teolytic or lipolytic enzymes. It is readily inactivated at of Southeast Asia, as vector activity is modified by dif-
56˚C after 30 minutes and has an optimal pH stability of fering climatic conditions.
8.5. Other animals and different mosquito species could
alter infection cycles and are factors involved in the
Replication transmission of JE. The disease is a growing threat in In-
The JEV replicates well and produces a cytopathic effect dia, where pigs are much less numerous compared with
in a wide range of host cell culture systems, such as pri- other endemic areas. In southern Thailand, 70% of the
mary cells and cell lines from mammals, including Vero pigs are infected with JEV, and the JEV vector mosquito is
cells, baby hamster kidney (BHK)-21 cells, and L-M prevalent; however, the encephalitis in humans is rare
mouse fibroblast cells. Cells originating from mosquitoes (Burke et al. 1985).
are also frequently used. Examples include C6/36 cell In temperate zones, JEV survives the winter, but in-
lines from larval cells of Aedes albopictus and the A. ae- fection of pigs in winter is not an important problem
gypti cell line from embryonic tissue. Leukocyte cultures (Takashima et al. 1988). Chickens and wild birds, espe-
from various animals are good media for growing the cially herons and egrets, are able to maintain positive
virus (Kadarnath et al. 1987). Hormones such as insulin, sera year-round (Hammon et al. 1958; Bhattacharya et al.
adrenocorticotropic hormone (ACTH), hydrocortisone, 1986). The JEV may also be carried by cold-blooded ver-
and concanavalin A have been shown to enhance JEV tebrates throughout the winter; experimentally, the virus
replication in cell culture systems (Kelkar 1985; Kokorev survives in two species of lizard in hibernation (Doi et al.
and Kolotvinova 1986). 1983). The virus can also be isolated naturally from
Like the other flaviviruses, JEV proteins are encoded snakes, and bats are possible reservoirs. Multiplication
by a single open reading frame that continues uninter- of JEV has been demonstrated in a variety of mosqui-
rupted throughout the region that is sequenced (McAda toes, including the species in Culex, Aedes, and Anopheles.
et al. 1987). A short pulse of actinomycin D has no effect The role of ticks has been described in animals with over-
on early replication of the virus, but continuous expo- winter infection. Transovarial vertical infection is ob-
sure partially inhibits replication. Mitomycin, a DNA- served in some mosquitoes, such as Aedes, which may be
synthesis inhibitor, does not exert any influence on repli- another mechanism for the infection.
cation of the virus. In addition, nuclear involvement
apparently does not occur (Leary and Blair 1983). CLINICAL SIGNS

EPIDEMIOLOGY Although young susceptible piglets may occasionally


show clinical signs, clinical illness is not a feature of JEV
The epidemiologic features of JE have been described in infection in adults or pregnant pigs. However, various de-
detail by Konno et al. (1966) and Kono and Kim (1969). grees of abnormality in fetuses may be evident when in-
In nature, infection with JEV is maintained cyclically, in- fected pregnant pigs come to term. Litters commonly
volving vector mosquitoes (Self et al. 1973), birds (arde- contain varying numbers of stillborn and mummified fe-
ids), and mammals. tuses (Fig. 15.2), weak piglets with nervous system signs,
JE is of public health concern. A correlation between and apparently normal piglets. Experimental induction
infection in pigs and infection in humans is apparent, of reproductive failure by infection with JEV causes sim-
with evidence indicating that swine play an important ilarly damaged litters, an additional observation being
role in the buildup of the virus within a population. In the presence of subcutaneous edema and hydrocephalus
enzootic zones, pigs are invariably found in high concen- in some stillborn piglets (Shimizu et al. 1954). Experi-
tration and are favored feeding sources for mosquitoes. mental infection of susceptible pregnant sows causes no
Consistent development of viremia in susceptible pigs clinical signs in dams but results in infection of fetuses in
ensures a continued supply of infected mosquitoes. In utero and subsequent abnormal farrowings: varying
Japan and Korea, the mosquito season starts in late June, numbers of mummified fetuses of different sizes and
varying somewhat according to latitude, and the pig- stillborn and weak piglets with subcutaneous edema and
mosquito cycle becomes evident shortly afterward. At hydrocephalus (Shimizu et al. 1954). Abortion has not
that time, the pig population contains a high proportion been a feature of intrauterine infection in experimental
of young, susceptible breeding stock in which passive studies.
immunity to JEV has waned during the winter (Konno et Infertility in boars in summer appears to be associat-
al. 1966). This buildup of susceptible stock is not as ed with JEV infection. Hashimura et al. (1976) isolated
prevalent in tropical areas of Southeast Asia, where the JEV from the testicles of boars with orchitis, and Ogasa
pig-mosquito cycle may continue throughout the year, al- et al. (1977) demonstrated that JEV infection of suscepti-
lowing most young stock to develop an early active im- ble boars resulted in invasion of the sexual organs and
munity. Epidemiologic patterns may differ among areas disturbance of spermatogenesis. Such boars developed
CHAPTER 15 JAPANESE B ENCEPHALITIS Joo, Chu 175

15.2. Mummified and stillborn


fetuses from a gilt following a
natural infection with JEV.

edematous, congested testicles, hardening of the epi- in mice, the virus replicates first in the peritoneal
didymis, and reduced libido, and virus was excreted in macrophages and later, on day 3, in the splenic
the semen, which had significantly depressed total and macrophages of the prefollicular region. Viral produc-
motile sperm counts, with numerous abnormalities of tive infection is observed in macrophages and T cells.
spermatozoa. In most boars, damage was temporary, Macrophages are also claimed to be involved in the de-
with subsequent complete recovery, but occasionally, velopment of latent infection.
boars with severe infection became permanently infer- Experimentally, JEV infection induces the generation
tile. of suppressor T cells through the production of a soluble
suppressor factor and suppresses the humoral- and cell-
PATHOGENESIS mediated immune responses of the infected animal. This
is manifested by depressed activities of plaque-forming
Pigs become infected with JEV via the bites of mosqui- cells and delayed hypersensitivities. These suppressions
toes carrying the virus. Infected pigs develop viremia, involve at least two generations of suppressor T cells. In-
which persists for approximately 12 hours to a few days. fection with JEV initially stimulates the production of
The multiplication model of the JEV in pigs has not been first-generation phenotypic Ly 1–2 suppressor cells,
thoroughly studied; however, some data are available for which release the suppressor factor, which has a molecu-
humans, monkeys, and mice. lar weight of 12,000 Da. This factor is nondialyzable and
After the initial viremia, the virus disseminates to sensitive to hydrocortisone. Suppressor factor adheres to
vascular tissues such as the liver, spleen, and muscle, the peritoneal or splenic macrophages and further stim-
where further replication augments the viremia. The ulates the generation of another subpopulation of sup-
virus enters the CNS via cerebral spinal fluid; by en- pressor T cells, which then exert their suppressor activi-
dothelial cell, macrophage, or lymphocyte infection; or ties on plaque-forming cells and delay hypersensitivities
by a hematogenous route. In humans and mice, JEV in- (Mathur et al. 1986; Rawat et al. 1986).
fects and destroys neurons selectively, mostly in the areas Transplacental infection of JEV has been reported in
of the brain stem, thalamus, basal ganglion, and the pigs and mice. In pregnant pigs, fetuses may become in-
lower layer of the cortex (Johnson 1987). fected during the viremic period. Experimentally, after
In mosquitoes, JEV infection is noncytopathic; the intravenous infection of pregnant pigs with JEV, the
virus is multiplied in phagocytic or hemolymph cells of virus is recoverable from fetuses as early as 7 days postin-
the fat body in 2 days (Johnson 1987). After infection, fection. In some animals, the virus fails to cross the pla-
the viral antigens spread widely in many organs, but se- centa. Shimizu et al. (1954) have suggested that success-
lective infection in the CNS (Leake and Johnson 1987) is ful transplacental infection may depend on the time of
always present 4 days postinfection; 1 or 2 days later the gestation at which the dam was infected or on the strain
virus can be found in salivary glands. of virus. When infection of pregnant dams takes place in
Macrophages play an important role in the patho- the mid-third of gestation, transplacental infection and
genesis of the disease. After intraperitoneal inoculation pathogenic effects are more obvious (Shimizu et al.
176 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

1954). Field observations show that fetal death and pigs. Neuronal degeneration is prominent in the gray
mummification are associated with JEV infection of matter and Purkinje layers. Most cells in perivascular
dams between 40 and 60 days of gestation; fetuses from cuffs in humans are T-helper/inducer cells; however,
gilts infected after 85 days of gestation were little affect- one-fourth of the cells are suppressor/cytotoxic T cells.
ed (Sugimori et al. 1974). The data are similar for mice, in which T-suppressor cells
Fetal deaths are assumed to be associated with un- proliferate during JEV infection. Involvement of visceral
controlled multiplication of the virus and subsequent organs, including lung, liver, kidney, and spleen, is docu-
destruction of vital stem cells in fetuses that have not mented in humans and experimentally in monkeys and
reached the stage of immunocompetence. The stage of horses. Pathologic changes are seen mostly in the reticu-
gestation at which fetuses become immunocompetent loendothelial system, such as focal hyperplasia of germi-
for JEV has not yet been determined. The average time of nal centers in the spleen; interstitial pneumonia is also
immunocompetence for other viruses, such as porcine seen (Rawat et al. 1986; Johnson 1987; R. M. Chu and M.
parvovirus, is 70 days of gestation (Joo et al. 1976), and Y. Liao, unpublished data—1990).
the findings of Sugimori et al. (1974) suggest that there Pathologic changes in the testes associated with JEV
are no pathogenic effects of JEV on the fetus when the infection are not well described. In naturally affected
virus crosses the placenta after this stage of gestation. pigs, a large amount of mucous fluid is observed in the
Thus, the pathogenesis of JEV for the porcine conceptus cavity tunica vaginalis, as well as fibrous thickening
seems comparable to that described for porcine par- along the edge of the epididymis and the visceral layer of
vovirus (Joo et al. 1976); that is, for pathogenic effects to the tunica vaginalis. Microscopically, such testicles show
occur, the virus must reach the conceptus before the edema and inflammatory changes, with cellular infiltra-
stage of immunocompetence. tions in the interstitial tissue of the epididymis and tuni-
In mice (Fujisaki et al. 1982), transplacental infection ca vaginalis. Cell infiltration and hemorrhage are also ev-
was demonstrated when pregnant females were exposed ident in the interstitial tissue of the testes. Degenerative
to the virus any time after the third day of gestation— changes are often seen in the seminiferous epithelium
with the incidence of infection highest when exposure (Hashimura et al. 1976; Ogasa et al. 1977).
was between 7 and 10 days of gestation. It was conclud-
ed that transplacental infection of JEV depends on the DIAGNOSIS
degree of development between placenta and fetal tis-
sues, not on the intensity of viremia. The definitive diagnosis of JEV infection is based upon
isolation of the virus from fetuses and infected pigs. A
LESIONS differential diagnosis must include porcine reproductive
and respiratory syndrome, parvovirus infection, Au-
Major pathologic lesions have not been found in pigs jeszky’s disease, toxoplasmosis, and hog cholera. Other
postnatally infected with JEV. However, various abnor- infections associated with reproductive failure such as
malities have been observed in litters from dams infected cytomegalovirus, leptospirosis, and enterovirus infec-
with the virus during pregnancy. Gross pathologic le- tions are also considered. Lack of clinical signs in sows
sions noted in stillborn or weak neonatal piglets include and piglets with JEV infection is useful in excluding many
hydrocephalus, subcutaneous edema, hydrothorax, as- diseases. Seasonal distribution also indicates JEV infec-
cites, petechial hemorrhages on serous membranes, con- tion.
gestion of lymph nodes, necrotic foci in liver and spleen, Several serologic tests are available that detect anti-
and congested meninges or spinal cord (Burns 1950). It body titers of JEV infection in pig serum, such as the
was further observed that the CNS appeared hypoplastic hemagglutination inhibition test, ELISA, antigen biotin-
in areas, the cerebral cortex particularly being extremely labeled ELISA, single radial hemolysin, and serum neu-
thin in some hydrocephalic piglets (Shimizu et al. 1954). tralization technique. Epitope-blocking immunoassay
Cerebellar hypoplasia and spinal hypomyelinogenesis (Burke et al. 1987) and detection of cerebral spinal fluid
have also been described (Morimoto 1969). IgM, using ELISA diagnostic kits, are used in humans.
Histopathologically, significant lesions in affected However, in an area where vaccination is routine, the di-
piglets or stillborn pigs are restricted to the CNS; few da- agnostic value of the serologic tests is challenged, and
ta on changes in other tissues have been recorded. Most paired serum samples should be considered. Presence of
CNS lesions, mainly involving the cortex, basal ganglion, the antibody in fetuses is also of diagnostic value.
brain stem, and spinal cord, occur in pigs up to 6 months Although isolation of the virus from suitable speci-
of age. Diffuse nonsuppurative encephalitis and mens is essential to make a definitive diagnosis, detect-
spondylitis are also reported. The highly selective dam- ing viral antigens in infected tissues such as brains, pla-
age to neurons found in humans is not well described in centa, and mummified fetuses is also diagnostically
pigs; however, scattered neuronal degeneration and important. Methods such as avidin-biotin staining and
necrosis are found in the cerebrum and cerebellum in fluorescent antibody staining have been used to detect JE
CHAPTER 15 JAPANESE B ENCEPHALITIS Joo, Chu 177

viral antigen in formalin-fixed tissues following treat- vishnui and appearance of JE antibody in sentinel chicks
ment of such tissues with trypsin or another proteolytic and wild birds in relation to Japanese encephalitis cases.
enzyme (Kurata et al. 1983; Iwasaki et al. 1986). Trop Geogr Med 38(l):46–50.
Isolation of the virus can be performed by intracere- Burke, D. S.; Tingpalapong, M.; Ward, G. S.; Andre, R.; and
bral inoculation of brain extracts into suckling mice be- Leake, L. J. 1985. Intense transmission of Japanese en-
tween 1 and 5 days of age. Signs of CNS disturbance or cephalitis virus to pigs in a region free of epidemic en-
death follow between 4 and 14 days postinoculation, and cephalitis. SE Asian J Trop Med Public Health
virus in mouse brain tissue can readily be identified by in 16(2):199–206.
vivo neutralization tests in suckling mice or in cell cul- Burke, D. S.; Nisaiak, A.; and Gentry, M. K. 1987. Detection
ture. The commonly used cell cultures are derived from of flavivirus antibodies in human serum by epitope-
hamster, pig kidney, and mosquito, in all of which the blocking immunoassay. J Med Virol 23(2):165–173.
virus causes a cytopathic effect; the mosquito cell line Burns, K. F. 1950. Congenital Japanese B encephalitis infec-
from A. albopictus clone C6/36 is the most efficient. It tion of swine. Proc Soc Exp Biol Med 75:621–625.
should be recognized that tissue suspected of being in- Chaturvedi, U. C.; Mathur, A.; Chandra, A.; Das, S. K.; Tan-
fected with JEV must be handled with care, because the don, H. O.; and Singh, U. K. 1980. Transplacental infec-
virus is not only heat labile but also pathogenic to hu- tion with Japanese encephalitis virus. J Infect Dis
mans. 141:712–714.
Doi, R.; Oya, A.; Shirasaka, A.; Yabe, S.; and Sasa, M. 1983.
PREVENTION Studies on Japanese encephalitis virus infection of rep-
tiles. II. Role of lizards on hibernation of Japanese en-
The treatment of JE in humans with human recombinant cephalitis virus. Jpn J Exp Med 53(2):125–134.
interferon-αA gives clinically satisfactory results. This Fujisaki, Y.; Sugimori, T.; Morimoto, T.; Miura, Y.; Kawaka-
treatment of JE in swine is not available. Breaking the in- mi, Y.; and Nakano, K. 1975. Immunization of pigs with
fectious cycle in this arthropod-borne disease is usually a the attenuated S strain of Japanese encephalitis virus.
good step toward management; however, JEV multiplies Natl Inst Anim Health Q (Tokyo) 15:55–60.
in a variety of species of mosquitoes, and the infectious Fujisaki, Y.; Miuya, Y.; Sugimori, T.; Murakami, Y.; Ino, T.;
patterns vary according to local ecology. It is impractical and Miura, K. 1982. Experimental studies on vertical in-
to control the insect; therefore, immunizing the breeders fection of mice with Japanese encephalitis virus. III. Ef-
with JEV vaccine is widely applied as an applicable con- fect of gestation days at the time of inoculation on pla-
trol and preventive measure. cental and fetal infection. Natl Inst Anim Health Q
A variety of mostly live-attenuated vaccines have 22(3):95–101.
been developed for the prevention of JE in pigs and are Fujita, T. 1933. Studies on the causative agent for epidemic
being used successfully in the field (Hsu et al. 1972; Fu- encephalitis. Jpn J Exp Med 17:1441–1501.
jisaki et al. 1975; Kwon et al. 1976). Inactivated vaccines Hammon, W. M.; Sather, G. E.; and McClure, H. E. 1958.
have proved to be less efficient and have become less Serological survey of Japanese B encephalitis virus infec-
popular. It is recommended that attenuated vaccines be tion in birds in Japan. Am J Hyg 67:118–134.
given to young gilts and boars twice at an interval of 2–3 Hashimura, K.; Uemiyada, S.; Komemura, S.; Fukumoto, S.;
weeks before the start of the mosquito season. As an ex- Okuda, G.; Miura, K.; and Hayashi, S. 1976. Isolation of
tra precaution, it is advisable that during the mosquito Japanese encephalitis virus from orchitis in pigs. Sum-
season, young gilts and boars selected for breeding be mary 81st Meet Jpn Soc Vet Sci, p. 136.
vaccinated before mating. The vaccine may be used si- Hori, H.; Igarashi, A.; Yoshida, I.; and Takagi, M. 1986.
multaneously with other viral vaccines such as hog Oligonucleotide fingerprint analysis on Japanese en-
cholera vaccine. cephalitis virus strains after passage histories. Acta Virol
Attempts have also been made to develop a genetical- 30(5):428–431.
ly engineered vaccine, but no commercial product is Hsu, S. T.; Chang, L. C.; Lin, S. Y.; Chuang, T. Y.; Ma, C. H.;
available. Some studies have demonstrated that mono- Inoue, Y. K.; and Okuno, T. 1972. The effect of vaccina-
clonal antibodies to epitopes on the E envelope of JEV tion with a live attenuated strain of Japanese encephali-
prevent mice from being infected by JEV (Kimura-Kuro- tis virus on stillbirths in swine in Taiwan. Bull WHO
da and Yasui 1988). However, application of this to pigs 46:465–471.
needs further investigation. Iwasaki, Y.; Zhao, J. X.; Yamamota, T.; and Kouuo, H. 1986.
Immunohistochemical demonstration of viral antigens
REFERENCES in Japanese encephalitis. Acta Neuropathol 70(l):79–81.
Johnson, R. T. 1987. The pathogenesis of acute viral en-
Bhattacharya, S.; Chakraborty, S. K.; Chakraborty, S.; cephalitis and postinfectious encephalomyelitis. J Infect
Ghosh, K. K.; Palit, A.; Mukherjee, K. K.; Chakraborty, Dis 155(3):359–364.
M. S.; Tandon, N.; and Hati, A. K. 1986. Density of Culex Joo, H. S.; Donaldson-Wood, C. R.; and Johnson, R. H. 1976.
178 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Observations on the pathogenesis of porcine parvovirus 67(2):171–179.


infection. Arch Virol 51:123–129. McAda, P. C.; Mason, P. W.; Schimaljohn, C. S.; Dalrymple,
Kadarnath, N.; Dayaraj, C.; Goverdhan, M. K.; and Ghosh, J. M.; Mason, T. L.; and Fournier, M. J. 1987. Partial nu-
S. N. 1987. Replication of Japanese encephalitis virus in cleotide sequence of the Japanese encephalitis virus
monkey, pig and chick leucocyte cultures. Trans R Soc genome. Virology 158(2):348–360.
Trop Med Hyg 81(5):829–832. Morimoto, T. 1969. Epizootic swine stillbirth caused by
Kelkar, S. D. 1985. Enhancement of Japanese encephalitis Japanese encephalitis virus. Proceedings of a sympo-
virus growth in vitro by concanavalin A. Indian J Med sium on factors producing embryonic and fetal abnor-
Res 81:437–440. malities, death, and abortion in swine. US ARS
Kimura-Kuroda, J., and Yasui, K. 1986. Antigenic compari- 91–73:137–153.
son of envelope protein E between Japanese encephalitis Ogasa, A.; Yokoki, Y.; Fujisaki, Y.; and Habu, A. 1977. Re-
virus and some other flaviviruses using monoclonal an- productive disorders in boars infected experimentally
tibodies. J Gen Virol 67:2663–2672. with Japanese encephalitis virus. Jpn J Anim Reprod
———. 1988. Protection of mice against Japanese en- 23:171–175.
cephalitis virus by passive administration with mono- Rawat, S.; Mathur, A.; and Chaturvedi, U. C. 1986. Charac-
clonal antibodies. J Immunol 141(10):3606–3611. terization of Japanese encephalitis virus-specific sup-
Kokorev, V. S., and Kolotvinova, E. G. 1986. Stimulation of pressor T cells and their product in suppression of the
arbovirus reproduction in cell cultures by hormones. Vo- humoral immune response in mice. Ann Inst Pasteur
pr Virusol 31(5):623–629. Immunol 1370(3):391–401.
Konno, J.; Endo, K.; Agatsuma, H.; and Ishida, N. 1966. Self, L. S.; Shin, H. K.; Kim, K. H.; Lee, K. W.; Chow, C. Y.;
Cyclic outbreaks of Japanese encephalitis among pigs and Hong, H. K. 1973. Ecologic studies on Culex tritae-
and humans. Am J Epidemiol 84:292–300. niorhynchus as a vector of Japanese encephalitis. Bull
Kono, R., and Kim, K. H. 1969. Comparative epidemiologi- WHO 49:41–47.
cal features of Japanese encephalitis in the Republic of Shimizu, T.; Kawakami, Y.; Fukuhara, S.; and Matsumoto,
Korea, China (Taiwan) and Japan. Bull WHO M. 1954. Experimental stillbirth in pregnant swine in-
40:263–277. fected with Japanese encephalitis virus. Jpn J Exp Med
Kurata, T.; Hondo, R.; Sato, S.; Oda, A.; Aoyama, Y.; and 24:363–375.
McCormick, J. B. 1983. Detection of viral antigens in Sugimori, T.; Morimoto, T.; Fujisaki, Y.; Sugawara, S.;
formalin-fixed specimens by enzyme treatment. Ann NY Tomishima, S.; and Ogata, M. 1974. A status quo survey
Acad Sci 420:192–207. on stillbirth and abortion in swine. III. Relationship be-
Kwon, H. J.; Kang, B. J.; Lim, Y. M.; Lee, C. K.; Kwon, Y. B.; tween the day of pregnancy at the time of outbreak of
Hur, W.; and Jeon, Y. S. 1976. Studies on Japanese en- Japanese encephalitis and the occurrence of stillbirth
cephalitis live virus vaccine. III. Pathogenicity of tissue and abortion. J Jpn Vet Med Assoc 27:282–285.
culture attenuated strain of virus (Anyang strain). Res Sumiyoshi, H.; Mori, C.; Fuke, I.; Morita, K.; Kuhara, S.;
Rep Off Rural Dev (Korea) 18:21–28. Kondou, J.; Kikuchi, Y.; Nagamatu, H.; and Igarashi, A.
Leake, C. J., and Johnson, R. T. 1987. The pathogenesis of 1987. Complete nucleotide sequence of the Japanese en-
Japanese encephalitis virus in Culex tritaeniorhynchus cephalitis virus genome RNA. Virology 16(2):497–510.
mosquitos. Trans R Soc Trop Med Hyg 81(4):681–685. Takashima, I.; Watanabe, T.; Ouchi, N.; and Hashimoto, N.
Leary, K. R., and Blair, C. D. 1983. Japanese encephalitis 1988. Ecological studies of Japanese encephalitis virus in
virus replication: Studies on host cell nuclear involve- Hokkaido: Interepidemic outbreaks of swine abortion
ment. Exp Mol Pathol 38(2):264–270. and evidence for the virus to overwinter locally. Am J
Mathur, A.; Rawat, S.; Chaturvedi, U. C.; and Misra, V. S. Trop Med Hyg 38(2):420–427.
1986. Macrophage transmission of suppressor signal for Taniguchi, T.; Hosokawa, M.; and Kuga, S. 1936. A virus iso-
suppression of delayed hypersensitivity and humoral re- lated in 1935 epidemic of summer encephalitis of Japan.
sponse in JEV-infected mice. Br J Exp Pathol Jpn J Exp Med 14:185–196.
Porcine Epidemic Diarrhea
16 M. B. Pensaert

In 1971, previously unrecognized acute outbreaks of di- that of other coronaviruses. Assembly of the virus oc-
arrhea were observed in feeder pigs and fattening swine curs by budding through intracytoplasmic membranes
in England (Oldham 1972). The clinical appearance was (Ducatelle et al. 1981b; Sueyoshi et al. 1995).
similar to that of an infection with transmissible gas- Physicochemical and biological characterization
troenteritis (TGE) virus except for the important differ- studies have confirmed the classification of PEDV as a
ence that suckling pigs (under 4–5 weeks of age) did not member of the Coronaviridae family. The virus is ether
become sick. TGE virus (TGEV) and other known en- and chloroform sensitive. Its density in sucrose is 1.18
teropathogenic infectious agents were ruled out, but a vi- g/mL. Concentrated and purified virus containing in-
ral etiology of unknown identity was suspected. The dis- testinal perfusate from infected piglets failed to aggluti-
ease spread to other European countries and the name nate erythrocytes of 12 different animal species (Calle-
“epidemic viral diarrhea” (EVD) was adopted. baut and DeBouck 1981; Witte et al. 1981). Cell
In 1976, TGE-like outbreaks of acute diarrhea were culture–adapted PEDV loses its infectivity when heated
observed in swine of all ages, including suckling pigs to ≥60˚C for 30 minutes but is moderately stable at 50˚C.
(Wood 1977), but again TGEV and other known en- The virus is stable between pH 5.0 and 9.0 at 4˚C and be-
teropathogenic agents were ruled out as the cause. The tween pH 6.5 and 7.5 at 37˚C. Viral infectivity is not im-
name “EVD type 2” was used to differentiate them from paired by ultrasonication or by multiple freezing and
the outbreaks described in the early 1970s (designated thawing. Replication is not inhibited by 5-iodo-
type 1). The difference between types 1 and 2 was that in 2′-deoxyuridine, indicating that the viral nucleic acid is
type 2 outbreaks baby piglets were involved. RNA (Hofmann and Wyler 1989).
In 1978, a coronavirus-like agent was found to be as- The pattern of the structural proteins of PEDV is
sociated with the type 2 outbreaks (Chasey and similar to that of other coronaviruses. The virus possess-
Cartwright 1978; Pensaert and DeBouck 1978). Experi- es a glycosylated peplomer protein with a molecular
mental inoculations with an isolate designated CV777 re- weight of 85,000–135,000 daltons, a glycosylated enve-
vealed its enteropathogenic character both for piglets lope protein of 20,000–32,000 daltons, and an unglyco-
(DeBouck and Pensaert 1980) and for fattening swine. It sylated RNA-binding nucleocapsid protein of 58,000 dal-
appeared that this coronavirus could be involved in out- tons (Egberink et al. 1988).
breaks of type 1 as well as of type 2, and the name Using direct immunofluorescence (IF) and immuno-
“porcine epidemic diarrhea” (PED) was proposed (Pen- electron microscopy, PEDV was found to be antigenical-
saert et al. 1982) and is still used. An explanation for the ly distinct from the two known porcine coronaviruses
clinical difference between type 1 and type 2 has not (TGEV and hemagglutinating encephalomyelitis virus)
been provided until now. and canine coronavirus, neonatal calf diarrhea virus, in-
fectious bronchitis virus, and feline infectious peritonitis
ETIOLOGY virus (Pensaert et al. 1981; Witte et al. 1981). However,
examination with the aid of more sensitive techniques
On a morphologic basis, PED virus (PEDV) particles such as immunoblotting and immunoprecipitation
show characteristics of the family Coronaviridae (Chasey showed that PEDV has antigenic determinants in
and Cartwright 1978; Pensaert and DeBouck 1978). The common with feline infectious peritonitis virus. These
particles detected in fecal material are pleomorphic, with determinants are located on the nucleocapsid protein
a tendency to a spherical shape. Their mean diameter, (Yaling et al. 1988). Identification of the membrane gly-
projections included, is 130 nm, with a range of 95–190 coprotein supports classification with group I coron-
nm. Many particles have an electron-opaque central aviruses (Utiger et al. 1995). On the basis of genomic
area. The club-shaped projections measure 18–23 nm analysis, PEDV holds an intermediate position between
and are radially spaced from the core. An internal struc- human coronavirus 229E and TGEV (Bridgen et al.
ture cannot be recognized. The morphogenesis of PEDV 1993). All these data definitely confirm that PEDV is a
in intestinal epithelial cells was found to be identical to coronavirus.

179
180 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Cultivation of the virus was originally accomplished 97% homology with that of the CV777 strain isolated in
by orally inoculating piglets and subsequently collecting Belgium (Park et al. 1995).
the small intestine and its contents during the early
stages of diarrhea. A virus stock containing 105 pig infec- EPIDEMIOLOGY
tious doses per milliliter was easily obtained (DeBouck
and Pensaert 1980). Serologic examinations to determine the presence of
The adaptation of PEDV to grow in an artificial host PEDV were carried out between 1982 and 1990, and spe-
under laboratory conditions has been fastidious. At- cific antibodies were detected in Belgium, England, Ger-
tempts with different virus isolates were unsuccessful in many, France, the Netherlands, Switzerland, Bulgaria,
intestinal and tracheal explants from swine fetuses and and Taiwan but not in Austria and North America (De-
newborn pigs. Also, numerous cell types were employed Bouck et al. 1982; Hofmann and Wyler 1987; Möstl et al.
with or without treatments with trypsin and pancreatin 1990). The virus itself has been isolated in most swine-
and were nonpermissive for PEDV replication (Hess et al. raising countries in Europe and also in China (Qinghua
1980; Callebaut and DeBouck 1981; Witte et al. 1981). et al. 1992), Korea (Kweon et al. 1993), and Japan (Taka-
However, Vero (African green monkey kidney) cells were hashi et al. 1983; Kuwahara et al. 1988). No confirmation
found to support the serial propagation of PEDV. of the occurrence of PEDV in North or South America
Growth was successful in some Vero cell lines and not in has been given until now.
others, so that cells of different origin should be at- In recent years, severe outbreaks of PED with high
tempted. Viral growth depends on the presence of mortality figures have been reported from Asia. In the
trypsin in the cell culture medium. Cytopathic effects winter of 1996, a PED epizootic occurred in Japan on 108
consist of vacuolation and formation of syncytia. The mostly farrow-to-finish swine farms in 9 prefectures.
syncytia may contain over 100 nuclei. Growth kinetics Diarrhea and high mortality were encountered in
show peak titers of 105.5 plaque-forming units per milli- baby piglets: 39,509 suckling pigs died out of a total
liter 15 hours after inoculation (Hofmann and Wyler of 56,256 (T. Ogawa, pers. comm.—1997). Also in
1988, 1989). Vero cell–adapted PEDV has been success- Japan, outbreaks between September 1993 and June
fully propagated in other cell types such as MA104 (Ku- 1994 have been described, with 14,000 deaths and mor-
sanagi et al. 1992). tality between 30% and 80% in suckling pigs. During
PEDV or viral antigens have until now been detected these epi- zootics, adult pigs showed only a transient in-
in fecal material by electron microscopy (EM) and en- appetence and decreased milk secretion (Sueyoshi et al.
zyme-linked immunosorbent assay (ELISA). The sensi- 1995).
tivity of the ELISA test is much higher than that of EM In Korea, PED causes outbreaks of diarrhea in swine
(Callebaut et al. 1982). With ELISA, PED viral antigens of all ages. Of 71 viral enteric cases requested for diag-
were demonstrated in rectal swabs of pigs until 11 days nosis at the Veterinary Research Institute between Janu-
postinoculation (Carvajal et al. 1995b). ary 1992 and December 1993, 56.3% were identified as
Specific antibodies have been detected in sera from PED. PED occurred all year round but was more preva-
swine after natural or experimental infection with PEDV. lent during the cold season. The disease occurred nation-
They can be demonstrated by immunoelectron mi- wide, and 90% of the outbreaks were detected in piglets
croscopy, ELISA, ELISA blocking, indirect IF, IF block- less than 10 days old (Hwang et al. 1994).
ing, and seroneutralization performed in pigs or in Vero Recent serologic surveys or diagnostic studies have
cell cultures (Pensaert et al. 1981; Prager and Witte not been published in Europe, where epizootics of PED
1981a, b; Callebaut et al. 1982; Witte and Prager 1987; in suckling pigs have become rare. Outbreaks of diarrhea
Hofmann and Wyler 1989, 1990). Using ELISA, antibod- due to PEDV are mainly confined to feeder and fattening
ies are easier to detect against the viral peplomer protein pigs and to young breeding animals. Very likely, this is
than against the nucleocapsid protein (Knuchel et al. due to the enzootic character of the virus and the pres-
1990). ence of lactogenic immunity, which protects suckling
There are no indications that different serotypes of pigs. In a longitudinal study in 10 groups of multisource
PEDV exist. Isolates from Europe, Korea, and China are feeder pigs entering a fattening unit in September 1993,
serologically identical to the prototype CV777 strain none seroconverted, whereas the pigs in another 7
(Hess et al. 1980; Witte et al. 1981; Vannier and DeBouck groups, entering in February 1994, had seroconverted 4
1983; Q. Yongqing, pers. comm.—1997). Additionally, weeks after arrival (Van Reeth and Pensaert 1994). These
polypeptide bands detected by immunoblotting with the 7 groups showed diarrhea during the first week after en-
Korean isolate showed a molecular weight similar to that tering and PED was demonstrated in feces.
of CV777 (Kweon et al. 1993). Also, the nucleotide se- In Spain, PED was identified as the cause of an epi-
quence of the N gene of PEDV isolated in Korea showed zootic of acute watery diarrhea in 7 of 15 farms, and the
CHAPTER 16 PORCINE EPIDEMIC DIARRHEA Pensaert 181

diarrhea became persistent in a small number of sows on PEDV does not differ markedly from TGEV with re-
one of the farms (Carvajal et al. 1995a). In a Spanish gard to modes of transmission, but it appears to persist
serosurvey carried out in 1992–93, PEDV specific anti- more easily on a farm once the acute infection has
bodies were detected in 1513 of 5098 sows, and positive passed.
animals were found in 55.9% of 798 breeding farms.
Prevalence was highest on farms with 20 or more sows CLINICAL SIGNS
(Carvajal et al. 1995c).
In the Netherlands, a clinical and virological study of The main and often the only obvious clinical sign of PED
an acute outbreak of PEDV diarrhea in a combined is watery diarrhea. Acute outbreaks on breeding farms
breeding and finishing pig herd was described (Pijpers et involving neonatal pigs have become rare in regions or
al. 1993). The first signs of disease were observed in fat- countries where the virus has become widespread and
tening pigs, and the infection spread rapidly to sows and where the sow population is largely immune.
their suckling pigs, gilts, and weaners housed in separate Outbreaks in susceptible breeding herds may show
barns. Diarrhea was most striking in fattening pigs and much variation in morbidity and mortality. On some
pregnant sows. Diarrhea in suckling pigs and young farms, pigs of all ages become sick, with morbidity ap-
weaners was mild or not at all present. The virus became proaching 100%. The disease is then very similar to TGE,
enzootic and persisted in 6- to 10-week-old pigs and in except for a slower spread and a somewhat lower mortal-
gilts newly introduced to the farm for at least 1.5 years af- ity in baby piglets. Piglets up to 1 week of age may die
ter the original outbreak. This outbreak was clinically from dehydration after the diarrhea has lasted 3–4 days.
similar to the type 1 outbreaks observed in the early sev- Mortality averages 50% but may be as high as 80%. Older
enties. pigs recover after about 1 week. After the acute outbreak
The situation in Asia, where diarrhea with high pig has passed, diarrhea may persist in pigs on the farm
mortality occurs, differs from the current situation in Eu- around 2–3 weeks after weaning, and newly introduced
rope. These Asian epizootics appear to be so severe that pigs may systematically become sick. Acute outbreaks
they cannot be differentiated clinically from typical acute with high mortality figures in neonatal pigs have been
TGEV outbreaks, and they incur heavy economic losses. described in recent years in Japan and Korea (Ogawa,
The virus in this continent, as well as in Japan, appears to pers. comm.—1997; Y. S. Lyoo, pers. comm.—1996).
have found nonimmune populations. Presumably an On some farms in Europe, however, weaned pigs and
evolution to enzootic prevalence and to population im- even adult pigs have been severely affected while suck-
munity will follow, as it did in Europe. A serosurvey in ling pigs have had no or only mild diarrhea even in the
Korea in 1994 involving 469 sera from slaughtered pigs absence of immunity. Morbidity in baby pigs is then low.
in 7 provinces showed 17.6–79% of pigs with PEDV anti- No explanation can be given at this time for the high vari-
bodies, with an overall average of 45%, clearly suggesting ation in clinical signs observed on breeding farms. Sever-
that the virus was endemic in some areas of that country al isolates have been tested for variation in virulence for
(Kweon et al. 1994). piglets, but no variation has been found.
Presently in Europe, the virus appears to persist Much less variation is observed when an acute PED
rather easily in densely swine populated areas but studies outbreak occurs in multisource feeder pigs or during the
on the mechanism or modalities of this persistence have fattening period. All the pigs in the unit will show diar-
not been carried out. rhea within a week. The animals are somewhat anorectic,
PEDV is transmitted by feces from infected animals. depressed, and their feces are watery (no blood). A PEDV
The natural infection starts after oral uptake. The feces- infection toward the end of the fattening period results
oral route of transmission is probably the main, if not in more severe disease than with TGEV. The animals ap-
the only, one. Acute outbreaks of PED on susceptible pear to have more abdominal pain. Recovery occurs as a
farms often occur within 4-5 days after sale or purchase rule after 7–10 days. Mortality of 1–3% may be seen in
of pigs. Virus probably enters by infected pigs, by way of fattening pigs infected toward the end of the fattening
contaminated trucks, boots, or other virus-carrying period. They die acutely, usually in the early stages of di-
fomites. After an outbreak has occurred on a breeding arrhea or even prior to the appearance of diarrhea. A
farm, the virus may either disappear or become enzootic. common necropsy finding in these animals is acute back
The enzootic status can be established on a farm if suffi- muscle necrosis. The highest mortality is found on farms
cient litters of pigs are produced and weaned so that the with stress-sensitive pig breeds. In general, PEDV repli-
virus is maintained through infection of consecutive lit- cation starts more easily in the intestine of feeder and
ters of pigs that have lost their lactogenic immunity at fattening pigs than in baby pigs. Consequently, fattening
weaning. PEDV may be a cause of persistent weaning di- pigs are more susceptible to the virus, and 100% morbid-
arrhea in 5- to 8-week-old pigs on such breeding farms. ity during an outbreak is common.
182 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Compared to TGEV, PEDV spreads more slowly in rectal swabs of experimentally inoculated pigs until 11
throughout the premises of closed-breeding farms and days (Carvajal et al. 1995b).
also within and between fattening units. On breeding Pathogenetic features described in Korea and Japan
farms that have several separate units and on fattening are identical to those observed in Europe (Hwang et al.
farms with separate buildings, it may take 4–6 weeks be- 1994; Sueyoshi et al. 1995).
fore the virus has infected different groups. Some units
may even remain free of infection. LESIONS

PATHOGENESIS Lesions have been described in experimentally and natu-


rally infected piglets (Pospischil et al. 1981; Ducatelle et
The pathogenesis of PED has been studied in hysterecto- al. 1982a, b; Hwang et al. 1994; Sueyoshi et al. 1995).
my-derived, colostrum-deprived piglets. Piglets were They are confined to the small intestine, which is dis-
orally inoculated with the CV777 isolate at the age of 3 tended with yellow fluid. Microscopically, vacuolation
days (DeBouck et al. 1981a). Upon inoculation, the pigs and exfoliation of enterocytes on the small-intestinal vil-
became sick after 22–36 hours. Viral replication, as li were observed starting at 24 hours postinoculation,
demonstrated by IF and transmission EM, occurred in which coincided with the onset of diarrhea. From that
the cytoplasm of villous epithelial cells throughout the time on, shortening of the villi occurred. These findings
small intestine and also in the colon. were confirmed in scanning EM studies. Histochemical-
Infected epithelial cells were observed as early as ly, the enzymatic activity in the small intestine was found
12–18 hours postinoculation, and a maximum was to be markedly reduced (Ducatelle et al. 1981a). This
reached between 24 and 36 hours. Viral replication in pathology is very similar to that described for TGE
the small intestine resulted in cell degeneration leading (Pospischil et al. 1981; Sueyoshi et al. 1995). No
to villous shortening. A reduction of villous height:crypt histopathologic changes have been observed in the
depth ratio from the normal 7:1 value to 3:1 was ob- colon.
served. No cell degeneration was seen in the colonic ep- Ultrastructural changes occurred mainly in the cyto-
ithelial cells. Fluorescent cells were detectable until 5 plasm of enterocytes (Horvath and Moscari 1981;
days postinoculation. Pospischil et al. 1981), in which cell organelles had de-
The pathogenetic features of PEDV in the small in- creased, leaving electron translucent areas. Later, the mi-
testine of piglets are very similar to those of TGEV. How- crovilli and terminal web disappeared, and parts of the
ever, the events with TGEV are of a more rapid and dras- cytoplasm protruded into the intestinal lumen. The cells
tic nature, leading to a villous shortening that is more became flattened, the tight junction was lost, and cell re-
extensive and occurs within 18–24 hours postinocula- lease occurred into the gut lumen. Intracellular virus for-
tion. Since viral replication and progress of the infection mation was seen by budding through membranes of the
in the small intestine with PEDV occurs at a slower rate, endoplasmic reticulum (Ducatelle et al. 1981b). In the
a longer incubation period is observed. PEDV replication colon, some cellular changes were observed in entero-
in piglets has not been detected in cells outside the in- cytes containing virus particles, but no exfoliation was
testinal tract. seen.
The pathogenesis of PEDV in older swine has not
been studied in much detail, but fluorescence was found DIAGNOSIS
in the epithelial cells of the small intestinal and colonic
villi of conventional fattening swine after experimental, A diagnosis of PED cannot be made on a clinical basis
as well as natural, infection. Witte et al. (1981), however, only. Acute PED outbreaks in which diarrhea is observed
did not observe this but found the colon positive only in in pigs of all ages, baby pigs included, cannot be clinical-
1-day-old piglets and not in older specific-pathogen-free ly differentiated from TGE. In Europe, outbreaks with a
swine. rapidly spreading watery diarrhea in weaned pigs and
It is not clear how much the colonic infection adds to older animals on a breeding farm, in which no baby
the severity of clinical signs. Also, no pathogenetic ex- piglets are involved, point toward PED.
planation can be given for the sudden death accompa- An etiologic diagnosis can be made in the laboratory
nied by acute back muscle necrosis often observed in fin- by direct demonstration of PEDV and/or its antigens or
ishing pigs and adult pigs. A recent study proved that by detection of antibodies.
halothane-positive fattening swine of 40–70 kg showed A direct IF test and an immunohistochemical tech-
markedly increased concentrations of the skeletal mus- nique applied on sections of the small intestine of baby
cle enzymes creatine phosphokinase and lactate dehy- pigs with diarrhea are the most sensitive, rapid, and reli-
drogenase in their blood, starting 3 days after inocula- able methods. They can only be used on the intestines of
tion with PEDV. pigs killed during the acute phase of diarrhea, preferably
Using the ELISA tests, PEDV antigens can be detected within 3 days of onset. The results of these techniques
CHAPTER 16 PORCINE EPIDEMIC DIARRHEA Pensaert 183

are often not reliable on pigs that die naturally, because practical diagnostic circumstances (Prager and Witte
of the severe villous atrophy (DeBouck et al. 1981b; 1981a, b). The indirect IF test has also been found to be
Sueyoshi et al. 1995; Bernasconi et al. 1995). less sensitive than the IF-blocking test and the ELISA
PEDV particles can be demonstrated in the feces of (Prager and Witte 1981a, b; Witte and Prager 1987; Hof-
pigs with diarrhea by direct EM. Examination of feces for mann and Wyler 1990; Carvajal et al. 1995b).
coronaviruses is often difficult because the virus particles PEDV antibodies demonstrated in the serum with the
are not easy to detect if the spikes are lost or not clearly ELISA-blocking and IF-blocking tests have been shown to
visible. The highest percentage of positive fecal samples persist for at least 1 year. Reinfection of the intestine,
obtained in experimentally inoculated piglets was 73% in with development of diarrhea, may occur in seropositive
feces collected the first day after the onset of diarrhea. pigs that have recovered from a first infection 5 months
Furthermore, immunoelectron microscopy has to be ap- previously. However, such pigs will show a rapid booster
plied to differentiate PEDV from TGEV, since both virus- reaction (DeBouck and Pensaert 1984; Witte and Prager
es have the same morphology. 1987).
So far, field strains of PEDV from diarrheic feces need
to be adapted to cell culture before they can be routinely TREATMENT AND PREVENTION
cultivated. Therefore, cell cultures cannot be used rou-
tinely for diagnosis of PED (Hofmann and Wyler 1988). Definite recommendations with regard to measures to be
A number of ELISA techniques have been developed taken during a PED outbreak cannot be given at this
for detection of PEDV antigens in feces as well as for time. Antibiotic treatments are of no help. Pigs with di-
demonstration of specific antibodies in serum. They are arrhea should have free access to water to diminish de-
sensitive and reliable for diagnosis, particularly on a hydration. It is advisable to withhold feed, particularly in
group basis. For the antigen ELISAs, polyclonal and fattening swine. Since PEDV does not spread very quick-
monoclonal antibodies were used with pig-cultivated ly, preventive measures to temporarily prevent virus en-
virus (Callebaut et al. 1982; van Nieuwstadt and Zetstra trance into farrowing units with newly born piglets may
1991; Carvajal et al. 1995a). For the antibody ELISA, anti- be of help to postpone the infection of these piglets un-
gen consists of semipurified virus, either pig cell culti- til a later age, resulting in fewer deaths. In the meantime,
vated (Callebaut et al. 1982; Kweon et al. 1994; Carvajal artificial spread of the virus to pregnant sows will stimu-
et al. 1995b; De Arriba et al. 1995) or S and N viral pro- late a rapid lactogenic immunity and thus shorten the
tein extracted from infected Vero cells (Knuchel et al. outbreak on the farm. This artificial exposure can be ac-
1992). The antibody test has also been used for detection complished by use of fecal material from pigs with wa-
of immunoglobulins in sow’s milk. tery diarrhea or of intestinal contents from pigs that
PEDV antigens can be demonstrated in fecal swabs have died. This approach is similar to that used with
from 3 until 11 days after experimental inoculation, with TGEV. If persistence of the virus is diagnosed in consec-
peak excretion being at 4 and 5 days. The time periods of utive litters of weaned piglets after an outbreak has oc-
antigen excretion are shorter in naturally infected pigs. curred on a breeding farm, virus elimination can be at-
Fecal material should be collected from several pigs, tempted by interrupting consecutive infections through
preferably during the acute phase of diarrhea. The removing pigs immediately after weaning to another site
ELISA antigen test is of sufficient sensitivity to detect the for at least 4 weeks. Also, introduction of new pigs
virus in situations where it is endemic and is a cause of should be stopped temporarily.
persistent diarrhea in breeding farms and where the Sanitary measures should prevent introduction of
amount of virus is too low to be detected by any other the virus to the farm. Present epizootiologic knowledge
methods. indicates that virus introduction occurs mainly (if not
A serologic diagnosis can be made by demonstration only) by animal and human traffic (Bollwahn 1983).
of PEDV antibodies. Several methods have been report- In Europe, the disease caused by PEDV in 1997 was
ed: the indirect IF test and the IF-blocking test on PEDV- not of sufficient economic importance to initiate re-
positive cryostat sections of pig intestine, the ELISA, and search to develop a vaccine. Outbreaks in Asia have been
the ELISA-blocking test. Antibodies are detectable by so severe that attempts have been made to develop an at-
blocking ELISA at 7 days postinoculation and by indirect tenuated virus. Bernasconi et al. (1995) reported that cell
IF between 10 and 13 days postinoculation (Carvajal et culture adaptation of the CV777 virus made it strikingly
al. 1995b). With all tests, paired serum samples should different with regard to genomic sequences. Also, the vir-
be examined. The second (convalescent) serum sample ulence of this cell-culture-adapted virus was much lower
should be collected no sooner than 2–3 weeks after the for newborn cesarean-derived piglets, since only 5 of 21
onset of diarrhea. had a mild diarrhea at 40 hours postinoculation and
The antibodies demonstrated with the indirect IF histopathological changes were decreased in severity. A
technique are reported to be temperature labile, which Korean strain, the KPEDV strain, when passaged 93
appears to make the use of this method difficult under times in Vero cells, also showed a reduced pathogenicity
184 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

for neonatal pigs and was found to be safe for pregnant pathogenesis of an enteric infection in pigs experimen-
sows. Therefore, the possible use of such a cell-culture- tally induced by the coronavirus-like agent CV777. Vet
adapted virus as a candidate for a vaccine has been pro- Microbiol 6:157–165.
posed (C. H. Kweon, pers. comm.—1996). DeBouck, P.; Callebaut, P.; and Pensaert, M. 1981b. The di-
agnosis of coronavirus-like agent (CVLA) diarrhea in
REFERENCES suckling pigs. Curr Top Vet Med Anim Sci 13:59–61.
———. 1982. Prevalence of the porcine epidemic diarrhea
Bernasconi, C.; Guscetti, F.; Utiger, A.; Van Reeth, K.; Acker- (PED) virus in the pig population of different countries.
mann, M.; and Pospischil, A. 1995. Experimental infec- Proc Int Congr Pig Vet Soc 7:53.
tion of gnotobiotic piglets with a cell culture adapted Ducatelle, R.; Coussement, W.; Charlier, G.; DeBouck, P.;
porcine epidemic diarrhoea virus: Clinical, histopatho- and Hoorens, J. 1981a. Three-dimensional sequential
logical and immunohistochemical findings. In Proc 3d study of the intestinal surface in experimental porcine
Congr ESVV, Interlaken, Switzerland, pp. 542–546. CV777 coronavirus enteritis. Zentralbl Veterinärmed
Bollwahn, W. 1983. Epizootic viral diarrhoea of pigs. Pig (B) 28:483–493.
News Inf 4:141–144. Ducatelle, R.; Coussement, W.; Pensaert, M.; DeBouck, P.;
Bridgen, A.; Duarte, M.; Tobler, K.; Laude, H.; and Acker- and Hoorens, J. 1981b. In vivo morphogenesis of a new
mann, M. 1993. Sequence determination of the nucleo- porcine enteric coronavirus, CV777. Arch Virol
capsid protein gene of the porcine epidemic diarrhoea 68:35–44.
virus confirms that this virus is a coronavirus related to Ducatelle, R.; Coussement, W.; DeBouck, P.; and Hoorens, J.
human coronavirus 229E and porcine transmissible gas- 1982a. Pathology of experimental CV777 coronavirus
troenteritis virus. J Gen Virol 74:1795–1804. enteritis in piglets. I. Histological and histochemical
Callebaut, P.; and DeBouck, P. 1981. Some characteristics of study. Vet Pathol 19:46–56.
a new porcine coronavirus and detection of antigen and Ducatelle, R.; Coussement, W.; Charlier, G.; DeBouck, P.;
antibody by ELISA. In Proc 5th Int Congr Virol, Stras- and Hoorens, J. 1982b. Pathology of experimental
bourg, p. 420. CV777 coronavirus enteritis in piglets. II. Electron mi-
Callebaut, P.; DeBouck, P.; and Pensaert, M. 1982. Enzyme- croscopic study. Vet Pathol 19:57–66.
linked immunosorbent assay for the detection of the Egberink, H. F.; Ederveen, J.; Callebaut, P.; and Horzinek,
coronavirus-like agent and its antibodies in pigs with M. C. 1988. Characterization of the structural proteins
porcine epidemic diarrhea. Vet Microbiol 7:295–306. of porcine epizootic diarrhea virus, strain CV777. Am J
Carvajal, A.; Diego, R.; Lanza, I.; Rubio, P.; Carmenes, P.; Vet Res 49:1320–1324.
and Schwyzer, M. 1995a. Evaluation of an ELISA for the Hess, R. G.; Bollwahn, W.; Pospischil, A.; Heinritzi, K.; and
detection of porcine epidemic diarrhea virus (PEDV) in Bachmann, P. A. 1980. Neue Aspekte der Virusaetiologie
feces of naturally infected pigs. In Proc 3d Congr ESVV, bei Durchfallerkrankungen des Schweines: Vorkommen
Interlaken, Switzerland, pp. 516–519. von Infektionen mit dem Epizootischen Virusdiarrhoe-
Carvajal, A.; Lanza, I.; Diego, R.; Rubio, P.; and Carmenes, P. (EVD-) Virus. Berl Muench Tierärtzl Wochenschr
1995b. Evaluation of a blocking ELISA using monoclon- 93:445–449.
al antibodies for the detection of porcine epidemic diar- Hofmann, M., and Wyler, R. 1987. Serologische Unter-
rhea virus and its antibodies. J Vet Diagn Invest 7:60–64. suchung über das Vorkommen der Epizootischen Virus-
———. 1995c. Seroprevalence of porcine epidemic diar- diarrhoe der Schweine (EVD) in der Schweiz. Schweiz
rhea virus infection among different types of breeding Arch Tierheilkd 129:437–442.
swine farms in Spain. Preventive Vet Med 23:33–40. ———. 1988. Propagation of the virus of porcine epidem-
Chasey, D., and Cartwright, S. F. 1978. Virus-like particles ic diarrhea in cell culture. J Clin Microbiol
associated with porcine epidemic diarrhoea. Res Vet Sci 26:2235–2239.
25:255–256. ———. 1989. Quantitation, biological and physiochemical
De Arriba, M. L.; Carvajal, A.; Lanza, I.; Rubio, P.; and properties of cell culture–adapted porcine epidemic di-
Carmenes, P. 1995. Development of an ELISA for the de- arrhea coronavirus (PEDV). Vet Microbiol 20:131–142.
tection of antibody isotypes against porcine epidemic ———. 1990. Enzyme-linked immunosorbent assay for the
diarrhoea virus (PEDV) in sow’s milk. In Proc 3d Congr detection of porcine epidemic diarrhea coronavirus an-
ESVV, Interlaken, Switzerland, pp. 222–225. tibodies in swine sera. Vet Microbiol 21:263–273.
DeBouck, P., and Pensaert, M. 1980. Experimental infection Horvath, I., and Moscari, E. 1981. Ultrastructural changes
of pigs with a new porcine enteric coronavirus CV777. in the small intestinal epithelium of suckling pigs affect-
Am J Vet Res 41:219–223. ed with a transmissible gastroenteritis (TGE)–like dis-
———. 1984. Porcine epidemic diarrhea: Kinetics of ac- ease. Arch Virol 68:103–113.
tively and passively acquired serum antibodies and the Hwang, E. K.; Kim, J. H.; Jean, Y. H.; Bae, Y. C.; Yoon, S. S.;
effect of reinfection. Proc Int Congr Pig Vet Soc 8:53. Park, C. K.; Kweon, C. H.; Yoon, Y. D.; and Ackermann,
DeBouck, P.; Pensaert, M.; and Coussement, W. 1981a. The M. 1994. Current occurrence of porcine epidemic diar-
CHAPTER 16 PORCINE EPIDEMIC DIARRHEA Pensaert 185

rhoea in Korea. RDA J Agri Sci 36:587–596. microscopy and ultrahistology of intestinal changes in
Knuchel, M.; Ackermann, M.; Müller, H. K.; and Kihm, U. pigs infected with enzootic diarrhoea virus (EVD): Com-
1990. Characterization of the E2-glycoprotein and nu- parison with transmissible gastroenteritis (TGE) virus
cleoprotein of porcine epidemic diarrhoea virus (PEDV) and porcine rotavirus infections. Zentralbl Veter-
and application of their physico-chemical properties for inärmed (B) 28:564–577.
the detection of anti PEDV antibodies by ELISA. Proc Prager, D., and Witte, K. 1981a. Die serologische Diagnose
Int Congr Pig Vet Soc 11:214. der Epizootischen Virusdiarrhoe (EVD) des Schweines
———. 1992. An ELISA for detection of antibodies against mit Hilfe der indirekten Immunofluoreszenztechnik
porcine epidemic diarrhoea virus (PEDV) based on the (IIFT). Tierärztl Umsch 36:404–414.
specific solubility of the viral surface glycoprotein. Vet ———. 1981b. Die serologische Diagnose der Epizootis-
Microbiol 32:117–134. chen Virusdiarrhoe (EVD) des Schweines mit Hilfe der
Kusanagi, K.; Kuwahara, H.; Katoh, T.; Nunoya, T.; indirekten Immunofluoreszenztechnik (IIFT). II. An-
Ishikawa, Y.; Samjima, T.; and Tajima, M. 1992. Isola- tikorper-Antwort nach experimenteller Infektion.
tion and serial propagation of porcine epidemic diar- Tierärztl Umsch 36:477–480.
rhea virus in cell cultures and partial characterization of Qinghua, C., et al. 1992. Investigation on epidemic diarrhea
the isolate. J Vet Med Sci 54:313–318. in pigs in Qinghai region. Qinghai Xumu Shaoyi Zazhi
Kuwahara, H.; Nunoya, T.; Samejima, T.; and Tajima, M. 22:22–23.
1988. Passage in piglets of a coronavirus associated with Sueyoshi, M.; Tsuda, T.; Yamazaki, K.; Yoshida, K.; Nakaza-
porcine epidemic diarrhoea. J Jpn Vet Med Assoc wa, M.; Sato, K.; Minami, T.; Iwashita, K.; Watanabe,
41:169–173. M.; Suzuki, Y.; and Mori, M. 1995. An immunohisto-
Kweon, C. H.; Kwon, B. J.; Jung, T. S.; Kee, Y. J.; Hur, D. H.; chemical investigation of porcine epidemic diarrhoea. J
Hwang, E. K.; Rhee, J. C.; and An, S. H. 1993. Isolation of Comp Path 113:59–67.
porcine epidemic diarrhea virus (PEDV) in Korea. Kore- Takahashi, K.; Okada, K.; and Ohshima, K. 1983. An out-
an J Vet Res 33:249–254. break of swine diarrhoea of a new type associated with
Kweon, C. H.; Kwon, B. J.; Kang, Y. B.; and An, S. H. 1994. coronavirus-like particles in Japan. Jpn J Vet Sci
Cell adaptation of KPEDV-9 and serological survey on 45:829–832.
porcine epidemic diarrhea virus (PEDV) infection in Ko- Utiger, A.; Tobler, K.; Brigen, A.; and Ackermann, M. 1995.
rea. Korean J Vet Res 34:321–326. Identification of the membrane protein of porcine epi-
Möstl, K.; Horvath, E.; and Bürki, F. 1990. Erhebungen über demic diarrhea virus. Virus Genes 10:137–148.
porcine Coronaviren in Osterreich. II. Porcine epidemic Vannier, P., and DeBouck, P. 1983. Identification de la diar-
diarrhea virus (PEDV) der Schweine. Wien Tierärztl rhée épidemique porcine (DEP) en Bretagne. Rec Méd
Monatsschr 77:10–18. Vet 159:19–24.
Oldham, J. 1972. Pig Farming (Oct suppl), pp. 72–73. van Nieuwstadt, A. P., and Zetstra, T. 1991. Use of two en-
Park, J. Y.; Kim, C. J.; Shin, K. S.; Kim, W. Y.; Kang, S. Y.; zyme-linked immunosorbent assays to monitor anti-
Park, Y. H.; Han, H. J.; and Park, Y. H. 1995. Sequence body responses in swine with experimentally induced
analysis and cDNA probe hybridization of the nucleo- infection with porcine epidemic diarrhea virus. Am J Vet
capsid (N) protein gene of transmissible gastroenteritis Res 52:1044–1050.
virus (TGEV) and porcine epidemic diarrhea virus Van Reeth, K., and Pensaert, M. 1994. Prevalence of infec-
(PEDV). Korean J Vet Res 35:515–530. tions with enzootic respiratory and enteric viruses in
Pensaert, M. B., and DeBouck, P. 1978. A new coronavirus- feeder pigs entering fattening herds. Vet Rec
like particle associated with diarrhea in swine. Arch Vi- 135:594–597.
rol 58:243–247. Witte, K. H., and Prager, D. 1987. Der Nachweis von An-
Pensaert, M. B.; DeBouck, P.; and Reynolds, D. J. 1981. An tikorpern gegen das Virus der Epizootischen Virusdiar-
immunoelectron microscopic and immunofluorescent rhoe (EVD) des Schweines mit dem Immunofluo-
study on the antigenic relationship between the coron- reszenz-blockadetest (IFBT). Tierärztl Umsch
avirus-like agent CV777 and several coronaviruses. Arch 42:817–820.
Virol 68:45–52. Witte, K. H.; Prager, D.; Ernst, H.; and Neuhoff, H. 1981. Die
Pensaert, M. B.; Callebaut, P.; and DeBouck, P. 1982. Epizootische Virusdiarrhoe (EVD). Tierärztl Umsch
Porcine epidemic diarrhea (PED) caused by a coron- 36:235–250.
avirus: Present knowledge. Proc Int Congr Pig Vet Soc Wood, E. N. 1977. An apparently new syndrome of porcine
7:52. epidemic diarrhoea. Vet Rec 100:243–244.
Pijpers, A.; van Nieuwstadt, A. P.; Terpstra, C.; and Verheij- Yaling, Z.; Ederveen, K.; Egberink, H.; Pensaert, M.; and
den, J. H. M. 1993. Porcine epidemic diarrhoea virus as Horzinek, M. C. 1988. Porcine epidemic diarrhea virus
a cause of persistent diarrhoea in a herd of breeding and (CV777) and feline infectious peritonitis virus (FIPV)
finishing pigs. Vet Rec 132:129–131. are antigenically related. Arch Virol 102:63–71.
Pospischil, A.; Hess, R. G.; and Bachmann, P. A. 1981. Light
Porcine Parvovirus
17 W. L. Mengeling

Porcine parvovirus (PPV) causes reproductive failure of tivity, and antigenicity are remarkably resistant to heat, a
swine characterized by embryonic and fetal infection wide range of hydrogen ion concentrations, and en-
and death, usually in the absence of outward maternal zymes.
clinical signs. The disease develops mainly when
seronegative dams are exposed oronasally to the virus Replication
anytime during about the first half of gestation, and con- Replication of PPV in vitro is cytocidal and characterized
ceptuses are subsequently infected transplacentally be- by “rounding up,” pyknosis, and lysis of cells (Fig.
fore they become immunocompetent. There is no defini- 17.1A). Many of the cell fragments often remain at-
tive evidence that infection of swine other than during tached, eventually giving the affected culture a ragged
gestation is of any clinical or economic significance. The appearance. Intranuclear inclusions develop (Cartwright
virus is ubiquitous among swine throughout the world et al. 1969) but they are often sparsely distributed (Rond-
and is enzootic in most herds that have been tested. Di- huis and Straver 1972). Infected cultures may hemad-
agnostic surveys have indicated that PPV is the major in- sorb slightly (Cartwright et al. 1969) (Fig. 17.1B). Cyto-
fectious cause of embryonic and fetal death (Cartwright pathic changes are extensive when cell culture–adapted
and Huck 1967; Mengeling 1978b; Thacker and Leman virus is propagated under appropriate conditions. How-
1978; Vannier and Tillon 1979; Mengeling et al. 1991). ever, on initial isolation several serial passages of the
virus (Cartwright et al. 1969) or, better, the infected cul-
ETIOLOGY ture may be necessary before the effects are recognized.
The use of immunofluorescence (IF) microscopy greatly
PPV is classified in the genus Parvovirus (Latin parvus = increases the likelihood of detecting minimally infected
small) of the family Parvoviridae (Siegl 1976; Bachmann cultures (Lucas and Napthine 1971; Mengeling 1975).
et al. 1979). All isolates of PPV that have been compared Primary and secondary cultures of fetal or neonatal
have been found antigenically similar if not identical porcine kidney cells are most often used for propagation
(Cartwright et al. 1969; Johnson and Collings 1969; Mo- and titration of PPV, although other kinds of cultures
rimoto et al. 1972a; Johnson et al. 1976; Ruckerbauer et are also susceptible (Pirtle 1974). Replication is en-
al. 1978). PPV is also antigenically related to several oth- hanced by infection of mitotically active cultures (Mayr
er members of the genus (Cotmore et al. 1983; Men- et al. 1968; Cartwright et al. 1969; Bachmann 1972; Hal-
geling et al. 1986, 1988). However, its identity can be es- lauer et al. 1972). Many cells in such cultures are in the S
tablished by relatively stringent serologic tests such as phase (i.e., the DNA synthesis phase) of their cell cycle,
serum neutralization (SN) and hemagglutination inhibi- wherein the DNA polymerases of cell origin needed for
tion (HI). viral replication are available (Tennant 1971; Siegl and
Gautschi 1973a, b).
Biophysical and Biochemical Properties If either fetal or adult bovine serum is incorporated
The biophysical and biochemical properties of PPV have in the nutrient medium of cell cultures used to propagate
been extensively studied (Siegl 1976; Molitor et al. 1983; PPV, it should be pretested for viral inhibitors (Coackley
Berns 1984) and are summarized as follows. A mature and Smith 1972; Johnson 1973; Pini 1975). The same
virion has cubic symmetry, two or three capsid proteins, may apply to sera of several other species (Joo et al.
a diameter of approximately 20 nm, 32 capsomeres, no 1976d). Because replication of PPV is affected by mitotic
envelope or essential lipids, and a weight of 5.3 × 106 dal- activity, the effect of the serum on the cells is also espe-
tons. The viral genome is single-stranded deoxyribonu- cially important. In addition, cultures should be pretest-
cleic acid (DNA) with a molecular weight of 1.4 × 106 ed for PPV contamination (Lucas and Napthine 1971;
(i.e., about 26.5% of the weight of the complete virion). Mengeling 1975). Cultures are sometimes unknowingly
Buoyant densities (g/mL in cesium chloride) of com- prepared from infected tissues of fetal (Mengeling 1975)
plete infectious virions, incomplete “empty” virions, and and postnatal (Huygelen and Peetermans 1967; Bach-
extracted virion DNA are 1.38–1.395, 1.30–1.315, and mann 1969; Cartwright et al. 1969; Hafez and Liess
1.724 respectively. Viral infectivity, hemagglutinating ac- 1979) pigs. Moreover, PPV can be accidentally intro-

187
188 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

17.1. Cell cultures infected with


PPV. (A) Cytopathic effect, sec-
ondary fetal porcine kidney cells,
120 hours after infection (×250)
(Mengeling 1972). (B) Hemadsorp-
tion, secondary adult porcine thy-
roid cells, guinea pig erythrocytes,
22 hours after thyroid cells were
infected and then subcultured A B
(May-Grünwald-Giemsa; ×100).

duced into cultures in several ways (Hallauer et al. 1971), cumulate viral antigen in their cytoplasm (Fig. 17.2C). A
including the use of contaminated trypsin (Croghan and second wave of viral replication can be induced if these
Matchett 1973; Croghan et al. 1973). If contamination is cells are stimulated to enter the S phase of the cell cycle
detected before all cells are infected, the virus can be as, for example, by the addition of fresh culture medium.
eliminated by repeatedly subculturing the cells in the
presence of nutrient medium containing PPV antiserum Hemagglutination
(Mengeling 1978a). PPV agglutinates human, monkey, guinea pig, cat, chick-
Several investigators have used IF microscopy to fol- en, rat, and mouse erythrocytes. Erythrocytes of other
low the development of PPV in cell culture (Cartwright kinds of animals that have been tested are relatively or
et al. 1969; Lucas and Napthine 1971; Mengeling 1972; completely insensitive, or the results have been equivocal
Siegl et al. 1972; Bachmann and Danner 1976). In gener- (Darbyshire and Roberts 1968; Mayr et al. 1968;
al, the sequence of events is as follows. Viral antigen is Cartwright et al. 1969; Hallauer et al. 1972; Mengeling
detected in the cytoplasm of cells soon after infection if 1972; Morimoto et al. 1972a). Several parameters of the
the inoculum contains a high titer of virus and viral anti- hemagglutination (HA) test—such as the temperature of
gen. Most, if not all, of this early cytoplasmic fluores- incubation (Mayr et al. 1968; Mengeling 1972), the
cence is the result of antigen phagocytized from the in- species of erythrocyte used, and in the case of chicken
oculum (Mengeling 1972; Mengeling and Cutlip 1975). erythrocytes the genetic composition (Cartwright et al.
By sequential examinations, such antigen can be demon- 1969; Pini 1975; Ruckerbauer et al. 1978) and age (Mori-
strated first on the external surface of the cytoplasmic moto et al. 1972a) of the donor—may quantitatively af-
membrane and later within the cytoplasm, often rela- fect results. The HA test is most commonly conducted at
tively concentrated in a juxtanuclear location. The first room temperature, at approximately neutral pH, and
unequivocal evidence of viral replication is the appear- with guinea pig erythrocytes. Higher HA titers have been
ance of nascent viral antigen in the nucleus (Fig. 17.2A). recorded when the diluent used in the test was veronal
In at least some infected cells, nascent antigen next ap- buffer rather than phosphate-buffered saline (Rucker-
pears in the cytoplasm in sufficient quantity that both cy- bauer et al. 1978). Elution of virus (the hemagglutinin is
toplasm and nucleus are brightly fluorescent. Infected part of the virion) can be induced by suspending eryth-
cells commonly seen in the lung of fetuses that develop a rocytes in alkaline buffer, pH 9 (Hallauer et al. 1972).
high titer of antibody for PPV probably represent this
stage of replication (see Fig. 17.8C). Affected cells subse- Infectivity Titrations
quently round up, become pyknotic, and disintegrate Infectivity titrations are conducted in a standard manner
with release of virus and viral antigen (Fig. 17.2B). Other except that, because cytopathic changes at terminal dilu-
cells in the culture that are not at the appropriate stage to tions are often vague, endpoints of infectivity are often
support viral replication continue to phagocytize and ac- determined either by examining cell cultures for intranu-
CHAPTER 17 PORCINE PARVOVIRUS Mengeling 189

A B C
17.2. Secondary cultures of fetal porcine kidney cells infected with PPV and examined by IF
microscopy (×500). (A) 14 hours after infection, culture fixed and then reacted with fluorescent
antibodies (FA). (B) 24 hours after infection, culture reacted with FA and then fixed; only
extracellular antigen and antigen in cells with disrupted cytoplasmic and nuclear membranes are
identified. (C) 48 hours after infection, culture fixed and then reacted with FA.

clear inclusions after appropriate staining or by examin- tive than the HI test (Johnson and Collings 1971; Joo et
ing cell culture medium for viral hemagglutinin al. 1975). A microtechnique for application of the SN
(Cartwright et al. 1969). A titration procedure wherein test has been described (Joo et al. 1975).
infected cells are made evident by IF microscopy (Men- Immunodiffusion (Joo et al. 1978), a modified direct
geling 1972) and a plaque assay (Kawamura et al. 1988) complement-fixation test (Ruckerbauer et al. 1978), and
also have been described. enzyme-linked immunosorbent assay (Hohdatsu et al.
1988; Westenbrink et al. 1989) also have been used suc-
Serologic Tests cessfully to detect antibody for PPV.
The HI test is frequently used for detection and quantita-
tion of humoral antibody for PPV. Antibody sometimes EPIDEMIOLOGY
can be detected as early as 5 days after swine are exposed
to live virus, and it may persist for years (Johnson et al. Porcine parvovirus is ubiquitous among swine through-
1976). Sera examined by the HI test are usually pretreat- out the world. In major swine-producing areas such as
ed by heat inactivation (56˚C, 30 minutes) and by ad- the midwestern United States, infection is enzootic in
sorption with erythrocytes (to remove naturally occur- most herds, and with few exceptions sows are immune.
ring hemagglutinins) and kaolin (to remove or reduce In addition, a large proportion of gilts are naturally in-
nonantibody inhibitors of HA) (Mengeling 1972; Mori- fected with PPV before they conceive, and as a result they
moto et al. 1972a). Trypsin also has been used to remove develop an active immunity that probably persists
nonantibody inhibitors of HA (Cartwright et al. 1969). throughout life. Collectively, the seroepidemiological da-
Parameters of the HI test have been studied in detail ta indicate that exposure to PPV is common. They also
(Kim 1974; Joo et al. 1976c). emphasize the high risk of infection and reproductive
The SN test is occasionally used for detection and disease among gilts that have not developed immunity
quantitation of humoral antibody for PPV. Neutraliza- before conception. The most common routes of infec-
tion of infectivity is usually confirmed by the absence or tion for postnatal and prenatal pigs are oronasal and
reduction either of intranuclear inclusions or fluorescent transplacental respectively.
cells in cultures or of viral hemagglutinin in the culture Pigs nursing immune dams absorb a high titer of an-
medium (Mengeling 1972; Johnson 1973; Joo et al. tibody for PPV from colostrum. These titers decrease
1975). The SN test has been reported to be more sensi- progressively with time by dilution as pigs grow as well
190 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

as by biological degradation. They usually reach subde- lymph nodes of boars killed 5, 8, 15, 21, and 35 days af-
tectable levels in 3–6 months if sera are examined by the ter oronasal exposure. After day 8, isolation was accom-
HI test (Etoh et al. 1979; Paul et al. 1982). Sometimes plished by cocultivating lymph node fragments with fetal
passively acquired antibody persists for a longer interval. porcine kidney cells (Mengeling, unpublished data—
Moreover, levels of antibody too low to be detected by 1976). Irrespective of their immune status, boars can al-
the HI test may be detected by the SN test (Johnson et al. so function as a vehicle for mechanical dissemination of
1976). The primary significance of passively acquired PPV among susceptible females.
antibody is that it interferes with the development of ac-
tive immunity. High levels of such antibody can prevent CLINICAL SIGNS
infection, and lower levels can minimize dissemination
from infected pigs (Suzuki and Fujisaki 1976; Paul et al. Acute infection of postnatal pigs, including pregnant
1980). Consequently, some groups of gilts are not fully dams that subsequently develop reproductive failure, is
susceptible to infection and dissemination of virus until usually subclinical (Johnson and Collings 1969; Cutlip
either shortly before conception or during early gesta- and Mengeling 1975a; Fujisaki et al. 1975; Johnson et al.
tion. 1976; Joo et al. 1976a; Mengeling and Cutlip 1976). How-
Contaminated premises are probably major reser- ever, in young pigs and probably in older breeding stock
voirs of PPV. The virus is thermostable, is resistant to as well, the virus replicates extensively and is found in
many common disinfectants (Brown 1981), and may re- many tissues and organs with a high mitotic index. Viral
main infectious for months in secretions and excretions antigen is especially concentrated in lymphoid tissues
from acutely infected pigs. It was shown experimentally (Cutlip and Mengeling 1975a; Fujisaki et al. 1975) (Fig.
that although pigs transmitted PPV for only about 2 17.3A, B). Many pigs, irrespective of age or sex, have a
weeks after exposure, the pens in which they were initial- transient, usually mild, leukopenia sometime within 10
ly kept remained infectious for at least 4 months (Men- days after initial exposure to the virus (Johnson and
geling and Paul 1986). The ubiquity of PPV also raises Collings 1969, 1971; Joo et al. 1976a; Mengeling and Cut-
the possibility that some pigs are persistently infected lip 1976). PPV and other structurally similar viruses have
and at least periodically shed virus. However, shedding been identified in the feces of pigs with diarrhea (Dea et
beyond the interval of acute infection has not been al. 1985; Yasuhara et al. 1989). However, there is no ex-
demonstrated (Johnson et al. 1976). The possibility of perimental evidence to suggest that PPV either replicates
immunotolerant carriers of PPV as a result of early in extensively in the intestinal crypt epithelium or causes
utero infection has been suggested (Johnson 1973). enteric disease as do parvoviruses of several other
When gilts were infected with PPV before day 55 of ges- species (Cutlip and Mengeling 1975a; Brown et al. 1980).
tation, their pigs were born infected but without anti- PPV also has been isolated from pigs with lesions de-
body. Virus was isolated from kidneys, testicles, and scribed as vesiclelike. The etiologic role of PPV in such
seminal fluid of such pigs killed at various times after lesions has not been clearly defined (Kresse et al. 1985).
birth up to the time they were 8 months of age; at which The major and usually only clinical response to infec-
time the experiment was terminated (Johnson and tion with PPV is maternal reproductive failure. Patholog-
Collings 1971). Results of another study, wherein dams ic sequelae depend mainly on when exposure occurs dur-
were infected early in gestation and their pigs were born ing gestation. Dams may return to estrus, fail to farrow
infected but without antibody, also suggest an acquired despite being anestrus, farrow few pigs per litter, or far-
immunotolerance (Cartwright et al. 1971). A possible ex- row a large proportion of mummified fetuses. All can re-
ample of an infected, immunotolerant, sexually active flect embryonic or fetal death or both. The only outward
boar was reported (Johnson et al. 1976). sign may be a decrease in maternal abdominal girth
Boars may play a significant role in dissemination of when fetuses die at midgestation or later and their asso-
PPV at a critical time. During acute infection the virus is ciated fluids are resorbed. Other manifestations of ma-
shed by various routes, including semen, and the isola- ternal reproductive failure, namely, infertility, abortion,
tion of PPV from semen of naturally infected boars has stillbirth, neonatal death, and reduced neonatal vitality,
been reported (Cartwright and Huck 1967; Cartwright et also have been ascribed to infection with PPV
al. 1969; McAdaragh and Anderson 1975). Semen may (Cartwright and Huck 1967; Johnson 1969; Morimoto et
be contaminated externally, as for example with virus- al. 1972b; Narita et al. 1975; Forman et al. 1977). These
containing feces, or within the male reproductive tract. are normally only a minor component of the disease.
The virus was isolated from a testicle of a boar 5 days af- The presence of mummified fetuses in a litter can pro-
ter it was injected into the boar’s prepuce (Lucas et al. long both gestation (Narita et al. 1975) and the farrowing
1974) and from testicles of boars killed 5 and 8 days af- interval (Mengeling et al. 1975). Either may result in still-
ter they were infected oronasally (Mengeling, unpub- birth of apparently normal littermates, whether or not
lished data—1976). Virus was also isolated from scrotal they are infected.
CHAPTER 17 PORCINE PARVOVIRUS Mengeling 191

17.3. Cryostat-micro-
tome sections of tissues
from PPV-infected 8-
week-old pigs, examined
by IF microscopy
(×312.5). (A) Viral anti-
gen in germinal center,
tonsil. (B) Viral antigen
in osteogenic layer of
periosteum, rib: a =
connective tissue, b =
A B cortical bone, c =
marrow cavity.

There is no evidence that either fertility or libido of al consequences of infection at different stages of gesta-
boars is altered by infection with PPV (Biront and Bonte tion are summarized in Table 17.1.
1983; Thacker et al. 1987). When only part of a litter is infected transplacental-
ly, as is often the case, one or more littermates are fre-
PATHOGENESIS quently infected by subsequent intrauterine spread of
virus. The same would apply if initial infection were
Dams are susceptible to PPV-induced reproductive fail- through contaminated semen. As a result, any combina-
ure if infected anytime during about the first half of ges- tion or all of the sequelae indicated in Table 17.1 can de-
tation. This interval of maternal susceptibility is indicat- velop in the same litter. Intrauterine dissemination is
ed by the collective results of several experimental probably less common when early embryos are infected
studies (Joo et al. 1976a; Mengeling and Cutlip 1976; because they are quickly resorbed after death, effectively
Mengeling 1979; Mengeling et al. 1980a), by in-depth removing the intrauterine reservoir of virus (Mengeling
epidemiological investigations (Donaldson-Wood et al. et al. 1980a). In such cases there is no evidence at far-
1977; Gillick 1977), and by estimates of the time of rowing for the cause of fewer pigs per litter.
death of fetuses collected during epidemiological sur-
veys (Mengeling 1978b; Mengeling et al. 1991). Conse-
quences of maternal infection during this interval are Table 17.1. Consequences of infection with PPV at
embryonic and fetal death followed by resorption and different intervals of gestation
mummification respectively. Transplacental infection al- Interval of Gestation
so follows maternal exposure after midgestation, but fe- (days)a
tuses usually survive without obvious clinical effects in Infection Infection Description Consequences
utero. The likely reason is that transplacental infection of of of of
often requires 10–14 days (Mengeling et al. 1978, 1980a) Dam Conceptusb Conceptus Infection
or longer (Joo et al. 1976a), and by 70 days of gestation ≤56 10–30 Embryo Death and
most fetuses are able to develop a protective immuno- resorption
logic response to the virus. In general, fetuses experi- 30–70 Fetus Death and
mentally infected by transuterine inoculation of the mummification
>56 70–term Fetus Immune response
virus have died when infected before day 70 of gestation, and usually
but they have survived and produced antibody when in- survival in utero
fected later in gestation (Redman et al. 1974; Bachmann
et al. 1975; Cutlip and Mengeling 1975b; Mengeling and a
Intervals are approximations.
Cutlip 1975). A strain of PPV of slightly greater viru- b
Assuming transplacental infections 10–14 days after mater-
lence also has been reported (Choi et al. 1987). The usu- nal exposure.
192 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

The effect, if any, of PPV on the ovum before ovula- fetal transfer of the virus by replicating through these tis-
tion is unknown. The virus adheres tenaciously to the ex- sues. However, this route cannot be excluded, since only
ternal surface of the zona pellucida of the fertilized a small part of the total area of contact was examined.
porcine ovum (Wrathall and Mengeling 1979a, b), and Transfer of the virus within macrophages has been con-
although it apparently cannot penetrate this layer, spec- sidered (Paul et al. 1979). Whatever the route, maternal
ulation is that it could pose a threat to the embryo after viremia seems a likely prerequisite for transplacental in-
hatching (Wrathall and Mengeling 1979a). fection (Joo et al. 1976a; Mengeling and Cutlip 1976).
Despite strong circumstantial evidence (Cartwright
et al. 1971), a direct causal role of PPV-contaminated se- LESIONS
men in reproductive failure has not been established un-
equivocally (Lucas et al. 1974). The zona pellucida could Neither macroscopic nor microscopic lesions have been
protect the early embryo while local immunity is devel- reported for nonpregnant pigs (Cutlip and Mengeling
oping. Conversely, the virus may cause uterine changes 1975a; Brown et al. 1980). It is conceivable that cellular
incompatible with gestation (Wrathall and Mengeling infiltrations subsequently described for fetuses could be
1979c). In any event, a female infected through semen induced by infection during the perinatal interval.
provides a focus of infection for others. Macroscopic lesions have not been reported in preg-
With the possible exception of the uterine changes al- nant dams; however, microscopic lesions have been seen
luded to in the preceding paragraph, PPV-induced repro- in tissues of gilts killed after their fetuses were infected
ductive failure is caused by the direct effect of the virus by transuterine inoculation of virus. Gilts that were
on the conceptus. In the absence of an immune re- seronegative when their fetuses were infected at 70 days
sponse, the virus replicates extensively throughout these of gestation had focal accumulations of mononuclear
tissues. By the time the conceptus dies, most of its cells cells adjacent to the endometrium and in deeper layers of
contain large quantities of intracytoplasmic viral antigen the lamina propria when they were killed 12 and 21 days
that can be demonstrated by IF microscopy. The relative later. In addition, there were perivascular cuffs of plasma
lack of nuclear fluorescence at the time of death, com- cells and lymphocytes in the brain, spinal cord, and
pared to earlier stages of the disease, indicates that when choroid of the eye (Hogg et al. 1977). When fetuses were
the conceptus is severely affected, mitotic activity and infected earlier in gestation (35, 50, and 60 days) and
the associated conditions necessary for viral replication their dams were killed 7 and 11 days later, the lesions
are suppressed more than phagocytic activity. were similar. However, uterine lesions were more severe
Death of the conceptus probably results from the col- and also included extensive cuffing of myometrial and
lective damage by the virus to a variety of tissues and or- endometrial vessels with mononuclear cells (Lenghaus et
gans, including the placenta (Cutlip and Mengeling al. 1978). Only focal accumulations of lymphocytes were
1975b). However, in the absence of an immune re- seen in uteruses of gilts that were seropositive when their
sponse, changes in almost any vital organ are probably fetuses were infected (Cutlip and Mengeling 1975b).
sufficient to eventually cause death. One of the most Macroscopic changes of embryos are death followed
striking features of viral distribution is the extensive in- by resorption of fluids (Fig. 17.4) and then soft tissues
volvement of endothelium. This seems to preclude fur- (Fig. 17.5). Virus and viral antigen are widely distributed
ther development of the vascular network of the concep- in tissues of infected embryos and their placentas (Men-
tus. Preparation for cellular mitosis (i.e., the S phase) geling et al. 1980a), and it is probable that microscopic
results in concomitant viral replication and cell death. lesions of necrosis and vascular damage, subsequently
Damage to the fetal circulatory system is indicated by described for fetuses, also develop in advanced embryos.
edema, hemorrhage, and the accumulation of large There are numerous macroscopic changes in fetuses
amounts of serosanguineous fluids in body cavities. infected before they become immunocompetent (Fig.
Necrosis of the endothelium is microscopically evident 17.6). These include a variable degree of stunting and
(Lenghaus et al. 1978). sometimes an obvious loss of condition before other ex-
The mechanism of transplacental infection has been ternal changes are apparent; occasionally, an increased
investigated by using IF microscopy to identify infected prominence of blood vessels over the surface of the fetus
cells in maternal and fetal tissues at progressively longer due to congestion and leakage of blood into contiguous
intervals after maternal oronasal exposure (Mengeling et tissues; congestion, edema, and hemorrhage with accu-
al. 1978). Examination of tissues contiguous with the mulation of serosanguineous fluids in body cavities;
maternal-fetal junction revealed viral antigen in en- hemorrhagic discoloration becoming progressively dark-
dothelial and mesenchymal cells of the chorion, with in- er after death; and dehydration (mummification). Many
creasing involvement of these tissues at progressively lat- of these changes also apply to the placenta. Microscopic
er stages of gestation. Viral antigen was never detected lesions consist primarily of extensive cellular necrosis in
unequivocally in either uterine epithelium or trophecto- a wide variety of tissues and organs (Joo et al. 1977;
derm. Consequently, there was no evidence for maternal- Lenghaus et al. 1978) (Fig. 17.7A). Inflammation (Joo et
CHAPTER 17 PORCINE PARVOVIRUS Mengeling 193

17.4. Embryos from a gilt experimentally infected oronasally immediately after breeding and
killed 22 days later. Bar = 1 cm. (Top) Noninfected, clinically normal embryo (arrow) and
associated extraembryonic membranes; (bottom) PPV-infected, dead littermate embryo (arrow)
and associated extraembryonic membranes, recent death, no obvious resorption of soft tissues.
(Mengeling et al. 1980a.)

cephalitis characterized by perivascular cuffing with pro-


liferating adventitial cells, histiocytes, and a few plasma
cells was seen in the gray and white matter of the cere-
brum and in the leptomeninges of PPV-infected stillborn
pigs. These lesions were believed to be pathognomonic
for PPV infection (Narita et al. 1975). Similar lesions
have been observed in PPV-infected, live fetuses collected
late in gestation (Hogg et al. 1977; Joo et al. 1977) (Fig.
17.7B).
Both general types of microscopic lesions (i.e., necro-
sis and mononuclear cell infiltration) may develop in fe-
tuses infected near midgestation (Lenghaus et al. 1978)
when the immune response is insufficient to provide pro-
tection.

DIAGNOSIS

PPV should be considered in a differential diagnosis of


reproductive failure of swine whenever there is evidence
of embryonic or fetal death or both. The pathologic se-
quelae of maternal infection during gestation have been
17.5. Segment of uterus opened to show necrotic described (see the section on clinical signs). If gilts but
remnants of a partially resorbed PPV-infected embryo not sows are affected, maternal illness is not seen during
(arrows) and associated extraembryonic membranes of a gestation, there are few or no abortions or fetal develop-
gilt experimentally infected oronasally immediately after mental anomalies, and other evidence suggests an infec-
breeding and killed 22 days later; remnants are laden tious disease, then a tentative diagnosis of PPV-induced
with virus and viral antigen. Bar = 1 cm. (Mengeling et
reproductive failure can be made. The relative lack of
al. 1980a.)
maternal illness, abortions, and fetal developmental
anomalies differentiates PPV from most other infectious
al. 1977) and intranuclear inclusions (Lenghaus et al. causes of reproductive failure. However, a definitive di-
1978) also have been described. agnosis requires laboratory support.
In contrast, macroscopic changes have not been re- Several mummified fetuses (<16 cm in length) or
ported for fetuses infected after they become immuno- lungs from such fetuses, if sufficiently developed, should
competent for PPV. Microscopic lesions are primarily be submitted to the diagnostic laboratory. Larger mum-
endothelial hypertrophy (Hogg et al. 1977) and mononu- mified fetuses (i.e., more than about 70 days of gesta-
clear cell infiltrations consistent with an immune re- tional age) (Marrable and Ashdown 1967), stillborn pigs,
sponse (Hogg et al. 1977; Joo et al. 1977). Meningoen- and neonatal pigs are not recommended for submission
194 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

A B

17.6. PPV-infected fetuses. Bars = 5 cm. (A) Litter of a gilt experimentally infected oronasally
˜
on day 47 of gestation and killed 34 days later; fetuses from left (L) and right (R) horn of uterus,
numbered 1–4 from cervix toward ovary; fetuses L1 and L4 stunted but alive at necropsy, fetus L3
recently dead, others later stages. (B) Fetuses from litter of a naturally infected gilt, collected at
about 114 days of gestation, advanced stage of dehydration (mummification). (Mengeling et al.
1975.)

unless they are the only samples available. If infected, reliable and sensitive diagnostic procedure. Sections of
their tissues will usually contain antibody that interferes fetal tissues are prepared with a cryostat microtome and
with laboratory tests for either virus or viral antigen. are then reacted with standardized reagents (Mengeling
If females fail to farrow despite being anestrus and et al. 1975; Mengeling 1978b). The test can be complet-
are sent to an abattoir, their uteruses should be collected ed within a few hours. In the absence of a fetal antibody
and examined for affected fetuses. Sometimes only rem- response, antigen is seen throughout fetal tissues (Fig.
nants of fetal tissues remain when fetuses die early in the 17.8A, B); even when antibody is present, infected cells
middle third of gestation. Nevertheless, these are ade- usually can be detected in fetal lung (Fig. 17.8C).
quate samples if tested for viral antigen by IF microscopy Detection of viral hemagglutinin also has been rec-
(Mengeling and Cutlip 1975; Mengeling 1978b). The ab- ommended as a diagnostic technique (Joo et al. 1976b;
sence of affected fetuses or fetal remnants does not ex- Joo and Johnson 1977a). Tissues are triturated in diluent
clude PPV-induced reproductive failure. When all em- and then sedimented by centrifugation. The supernatant
bryos of a litter die and are completely resorbed after the fluid is tested for agglutinating activity for guinea pig
first few weeks of gestation, the dam may remain en- erythrocytes. This test requires a minimum of laborato-
docrinologically pregnant and not return to estrus until ry equipment and is effective in the absence of antibody.
after the expected time of farrowing (Rodeffer et al. Virus isolation is less suitable as a routine diagnostic
1975). procedure than either of the aforementioned tests. Infec-
Identification of viral antigen by IF microscopy is a tivity is slowly but progressively lost after fetal death
CHAPTER 17 PORCINE PARVOVIRUS Mengeling 195

A B
17.7. Tissues of PPV-infected fetuses of gilts experimentally infected oronasally. (A) Necrotic
focus in liver of live fetus of a gilt infected on day 40 of gestation and killed 42 days later; fetus
had numerous macroscopic lesions (H&E; ×400). (B) Perivascular cuffing with mononuclear cells
in cerebrum of live fetus, littermate of A; fetus had no macroscopic lesions (H&E; ×320). (Insert)
Viral antigen associated with endothelium of cerebral vessel of fetus of a gilt infected on day 46 of
gestation and killed 25 days later (IF microscopy; ×312.5). All fetuses were probably infected by
intrauterine spread of PPV from transplacentally infected littermates. (Photographs A and B
courtesy of T. T. Brown, Jr., National Animal Disease Center.)

(Mengeling and Cutlip 1975); as a result, isolation of collected at intervals reveal seroconversion for PPV coin-
virus from mummified fetuses that have died as a result cident with reproductive failure (Morimoto et al. 1972b;
of infection is sometimes unsuccessful (Mengeling Mengeling et al. 1975; Rodeffer et al. 1975). Because PPV
1978b). Moreover, the procedure is time-consuming, is ubiquitous, the presence of antibody in a single sam-
and contamination is a constant threat because of the ple is otherwise meaningless. However, a determination
stability of PPV in the laboratory (Cartwright et al. 1969) of relative amounts of antibody present as immunoglob-
and because cell cultures are sometimes unknowingly ulin M and G can indicate the recency of infection (Kim
prepared from infected tissues (Huygelen and Peeter- 1974; Joo et al. 1978). Detection of antibody in sera of fe-
mans 1967; Bachmann 1969; Cartwright et al. 1969; tuses and stillborn pigs and in sera collected from neona-
Mengeling 1975; Hafez and Liess 1979). IF microscopy is tal pigs before they nurse is evidence of in utero infec-
often used to determine whether PPV has been isolated tion, since maternal antibody does not cross the
in cell culture (Cartwright 1970; Johnson 1973; Men- maternal-fetal junction (Johnson and Collings 1969,
geling 1978b). 1971; Cartwright et al. 1971; Mengeling 1972; Chaniago
In general, serologic procedures are recommended et al. 1978). When serum is not available, body fluids col-
for diagnosis only when tissues from mummified fetuses lected from fetuses or their viscera that have been kept in
are not available for testing as previously described. Re- a plastic bag overnight at 4˚C have been used successful-
sults with maternal sera are of value if antibody is not ly to demonstrate antibody (Cropper et al. 1976; Joo et al.
detected, thus excluding PPV as a cause, and if samples 1976b).
196 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

A B C
17.8. Cryostat-microtome sections of lungs of PPV-infected fetuses examined by IF microscopy.
(A) Lung of mummified fetus reacted with FA plus nonimmune serum (×312.5). (B) Replicate
section reacted with FA plus PPV-immune serum (i.e., blocking control) (×312.5). (C) Lung of live
fetus with HI antibody titer of 640 reacted with fluorescent antibodies (FA) plus nonimmune
serum, two infected cells (arrow) (× 162.5). (Insert) Two similar infected cells in the same section
as C (×500). (Mengeling 1978b.)

TREATMENT AND PREVENTION susceptible period of gestation but after the disappear-
ance of passively acquired colostral antibody, which
There is no treatment for PPV-induced reproductive fail- could interfere with the development of active immunity
ure. (Paul and Mengeling 1986). These limits may define a
Gilts should be either naturally infected with PPV or very brief interval for effective vaccination of gilts that
vaccinated for PPV before they are bred. To promote nat- are bred before 7 months of age. Although inactivated
ural infection, a common practice is to arrange contact vaccine provides maximum safety, there is experimental
between seronegative gilts and seropositive sows, with evidence that PPV can be sufficiently attenuated so that
the expectation that one or more of the sows will be it is unlikely to cause reproductive failure even if inad-
shedding virus. Moving gilts to a potentially contaminat- vertently administered during gestation (Paul and Men-
ed area, either currently or recently inhabited by geling 1980). The apparent safety of MLV vaccine may be
seropositive swine, also can be recommended. Once in- due to its reduced ability to replicate in tissues of the in-
fection is started, the virus spreads rapidly among fully tact host and cause the level of viremia needed for
susceptible swine. Just how effective these procedures are transplacental infection (Paul and Mengeling 1984).
in increasing the incidence of natural infection is un- Moreover, it has been shown by transuterine inoculation
known. For whatever reasons, infection is common, and of both virulent and attenuated virus that a much larger
probably well over one-half of all gilts in areas where dose of attenuated virus is required to establish infection
PPV is enzootic are infected before they are bred for the of fetuses (Mengeling et al. 1984). Duration of immuni-
first time (Mengeling 1972). ty following vaccination is unknown; however, in one
The use of vaccine is the only way to ensure that gilts study antibody titers were maintained for at least 4
develop active immunity before conception. Both inacti- months after administration of an inactivated vaccine
vated (Suzuki and Fujisaki 1976; Ide et al. 1977; Joo and (Joo and Johnson 1977b). Low levels of antibody found
Johnson 1977b; Mengeling 1977; Fujisaki 1978; Fujisaki to be protective allow speculation that, once the immune
et al. 1978b; Mengeling et al. 1979, 1980b) and modified system has been primed with PPV, subsequent exposure
live-virus (MLV) vaccines (Paul and Mengeling 1980; Fu- to virulent virus during gestation is unlikely to result in
jisaki and Murikami 1982) have been developed. An in- transplacental infection even if antibody from vaccina-
activated vaccine has been tested under field conditions tion is no longer detected (Mengeling et al. 1979).
(Fujisaki 1978; Fujisaki et al. 1978a), and both types of Vaccination is recommended also for seronegative
vaccines were effective when tested under controlled lab- sows and boars. Seronegative sows are usually found on-
oratory conditions (Mengeling et al. 1979, 1980b; Paul ly in PPV-free herds; in such cases, inactivated vaccine is
and Mengeling 1980). indicated. Experience has shown that few herds can be
Vaccines should be administered several weeks be- expected to remain free of PPV even if access is carefully
fore conception to provide immunity throughout the controlled. Introduction of PPV into a totally susceptible
CHAPTER 17 PORCINE PARVOVIRUS Mengeling 197

herd can be disastrous (Donaldson-Wood et al. 1977). Choi, C. S.; Molitor, T. W.; Joo, H. S.; and Gunther, R. 1987.
Vaccination of boars should reduce their involvement in Pathogenicity of a skin isolate of porcine parvovirus in
dissemination of the virus. swine fetuses. Vet Microbiol 15:19–29.
Vaccines are used extensively in the United States and Coackley, W., and Smith, V. W. 1972. Porcine parvoviruses
in several other countries where PPV has been recog- in Western Australia. Aust Vet J 48:536.
nized as an economically important cause of reproduc- Cotmore, S. F.; Sturzenbecker, L. J.; and Tattersall, P. 1983.
tive failure. All federally licensed vaccines marketed in The autonomous parvovirus MVM encodes two non-
the United States are inactivated. structural proteins in addition to its capsid polypep-
tides. Virology 129:333–343.
REFERENCES Croghan, D. L., and Matchett, A. 1973. β-propiolactone
sterilization of commercial trypsin. Appl Microbiol
Bachmann, P. A. 1969. Vorkommen und Verbreitung von Pi- 26:832.
codna (Parvo)-Virus beim Schwein. Zentralbl Veter- Croghan, D. L.; Matchett, A.; and Koski, T. A. 1973. Isola-
inärmed (B) 16:341–345. tion of porcine parvovirus from commercial trypsin.
———. 1972. Porcine parvovirus infection in vitro: A study Appl Microbiol 26:431–433.
model for the replication of parvoviruses. I. Replication Cropper, M.; Dunne, H. W.; Leman, A. D.; Starkey, A. L.;
at different temperatures. Proc Soc Exp Biol Med and Hoefling, D. C. 1976. Prevalence of antibodies to
140:1369–1374. porcine enteroviruses and porcine parvovirus in body
Bachmann, P. A., and Danner, K. 1976. Porcine parvovirus fluids of fetal pigs from small vs. large litters. J Am Vet
infection in vitro: A study model for the replication of Med Assoc 168:233–235.
parvoviruses. II. Kinetics of virus and antigen produc- Cutlip, R. C., and Mengeling, W. L. 1975a. Experimentally
tion. Zentralbl Veterinärmed (B) 23:355–363. induced infection of neonatal swine with porcine par-
Bachmann, P. A.; Sheffy, B. E.; and Vaughan, J. T. 1975. Ex- vovirus. Am J Vet Res 36:1179–1182.
perimental in utero infection of fetal pigs with a porcine ———. 1975b. Pathogenesis of in utero infection of eight-
parvovirus. Infect Immun 12:455–460. and ten-week-old porcine fetuses with porcine par-
Bachmann, P. A.; Hoggan, M. D.; Kurstak, E.; Melnick, J. L.; vovirus. Am J Vet Res 36:1751–1754.
Pereira, H. G.; Tattersall, P.; and Vago, C. 1979. Par- Darbyshire, J. H., and Roberts, D. H. 1968. Some respirato-
voviridae: Second report. Intervirology 11:248–254. ry virus and mycoplasma infections of animals. J Clin
Berns, K. I. 1984. The Parvoviruses. New York: Plenum. Pathol 21(Suppl 2):61–92.
Biront, P., and Bonte, P. 1983. Porcine parvovirus (P.P.V.) in- Dea, S.; Elazhary, M. A. S. Y.; Martineau, G. P.; and Vaillan-
fection in boars. I. Possibility of a genital localization in court, J. 1985. Parvovirus-like particles associated with
the boar after oronasal infection. Zentralbl Veter- diarrhea in unweaned piglets. Can J Comp Med
inärmed 30:541–545. 49:343–345.
Brown, T. T., Jr. 1981. Laboratory evaluation of selected dis- Donaldson-Wood, C. R.; Joo, H. S.; and Johnson, R. H. 1977.
infectants as virucidal agents against porcine par- The effect on reproductive performance of porcine par-
vovirus, pseudorabies virus, and transmissible gastroen- vovirus infection in a susceptible pig herd. Vet Rec
teritis virus. Am J Vet Res 42:1033–1036. 100:237–239.
Brown, T. T., Jr.; Paul, P. S.; and Mengeling, W. L. 1980. Re- Etoh, M.; Morishita, E.; and Watanabe, Y. 1979. Transition-
sponse of conventionally raised weanling pigs to experi- al antibodies and spontaneous infection in porcine par-
mental infection with a virulent strain of porcine par- vovirus infection. Jpn J Swine Husb Res 16:237–239.
vovirus. Am J Vet Res 41:1221–1224. Forman, A. J.; Lenghaus, C.; Hogg, G. G.; and Hale, C. J.
Cartwright, S. F. 1970. Tests available for the detection of 1977. Association of a parvovirus with an outbreak of
some virus infections of pigs and their interpretation. foetal death and mummification in pigs. Aust Vet J
Vet Annu 11:77–82. 53:326–329.
Cartwright, S. F., and Huck, R. A. 1967. Viruses isolated in Fujisaki, Y. 1978. Incidence and control of stillbirth caused
association with herd infertility, abortions and still- by porcine parvovirus in Japan. Proc Int Congr Pig Vet
births in pigs. Vet Rec 81:196–197. Soc 5:KA 14.
Cartwright, S. F.; Lucas, M.; and Huck, R. A. 1969. A small Fujisaki, Y., and Murikami, Y. 1982. Immunity to infection
haemagglutinating porcine DNA virus. I. Isolation and with porcine parvovirus in pigs inoculated with attenu-
properties. J Comp Pathol 79:371–377. ated HT-strain. Natl Inst Anim Health (Tokyo)
———. 1971. A small haemagglutinating porcine DNA 22:36–37.
virus. II. Biological and serological studies. J Comp Fujisaki, Y.; Morimoto, T.; Sugimori, T.; and Suziki, H.
Pathol 81:145–155. 1975. Experimental infection of pigs with porcine par-
Chaniago, T. D.; Watson, D. L.; Owen, R. A.; and Johnson, R. vovirus. Natl Inst Anim Health Q (Tokyo) 22:205–206.
H. 1978. Immunoglobulins in blood serum of foetal Fujisaki, Y.; Ichihara, T.; Sasaki, N.; Shimizu, F.; Murakami,
pigs. Aust Vet J 54:30–33. Y.; Sugimori, T.; and Sasahara, J. 1978a. Field trials on
198 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

inactivated porcine parvovirus vaccine for prevention of 53:550–552.


viral stillbirth among swine. Natl Inst Anim Health Q Joo, H. S.; Donaldson-Wood, C. R.; and Johnson, R. H. 1975.
(Tokyo) 18:184–185. A microneutralization test for the assay of porcine par-
Fujisaki, Y.; Watanabe, Y.; Kodama, K.; Hamada, H.; Mu- vovirus antibody. Arch Virol 47:337–341.
rakami, Y.; Sugimori, T.; and Sasahara, J. 1978b. Protec- ———. 1976a. Observations on the pathogenesis of
tion of swine with inactivated porcine parvovirus vac- porcine parvovirus infection. Arch Virol 51:123–129.
cine from fetal infection. Natl Inst Anim Health Q ———. 1976b. Rapid diagnostic techniques for detection
(Tokyo) 18:185. of porcine parvovirus infection in mummified foetuses.
Gillick, J. C. 1977. An outbreak of swine foetal mummifica- Aust Vet J 52:51.
tion associated with porcine parvovirus. Aust Vet J ———. 1976c. A standardised haemagglutination inhibi-
53:105–106. tion test for porcine parvovirus antibody. Aust Vet J
Hafez, S. M., and Liess, B. 1979. Isolation of parvovirus 52:422–424.
from kidney cell cultures of gnotobiotic piglets. Zentral- Joo, H. S.; Donaldson-Wood, C. R.; Johnson, R. H.; and Wat-
bl Veterinärmed (B) 26:820–827. son, D. L. 1976d. Antibody to porcine, feline and rat par-
Hallauer, C.; Kronauer, G.; and Siegl, G. 1971. Parvovirus as voviruses in various animal species. Res Vet Sci
contaminants of permanent human cell lines. I. Virus 21:112–113.
isolations from 1960–1970. Arch Gesamte Virusforsch Joo, H. S.; Donaldson-Wood, C. R.; Johnson, R. H.; and
35:80–90. Campbell, R. S. F. 1977. Pathogenesis of porcine par-
Hallauer, C.; Siegl, G.; and Kronauer, G. 1972. Parvoviruses vovirus infection: Pathology and immunofluorescence
as contaminants of permanent human cell lines. III. Bi- in the foetus. J Comp Pathol 87:383–391.
ological properties of the isolated viruses. Arch Gesamte Joo, H. S.; Johnson, R. H.; and Watson, D. L. 1978. Serolog-
Virusforsch 38:369–382. ical procedures to determine time of infection of pigs
Hogg, G. G.; Lenghaus, C.; and Forman, A. J. 1977. Experi- with porcine parvovirus. Aust Vet J 54:125–127.
mental porcine parvovirus infection of foetal pigs re- Kawamura, H.; Fujita, T.; and Imada, T. 1988. Plaque for-
sulting in abortion, histological lesions and antibody mation and replication of porcine parvovirus in embry-
formation. J Comp Pathol 87:539–549. onic swine kidney cell line, ESK cells. Jpn J Vet Sci
Hohdatsu, T.; Baba, K.; Ide, S.; Tsuchimoto, M.; Nagano, H.; 50:803–808.
Yamagami, T.; Yamagishi, H.; Fujisaki, Y.; and Matumo- Kim, Y. H. 1974. Studies on hemagglutination and hemag-
to, M. 1988. Detection of antibodies against porcine glutination-inhibition reaction of porcine parvovirus.
parvovirus in swine sera by enzyme-linked immunosor- Bull AZABU Vet Coll 27:61–65.
bent assay. Vet Microbiol 17:11–19. Kresse, J. I.; Taylor, W. D.; Stewart, W. C.; and Eernisse, K. A.
Huygelen, C., and Peetermans, J. 1967. Isolation of a hemag- 1985. Parvovirus infection in pigs with necrotic and vesi-
glutinating picornavirus from a primary swine kidney cle-like lesions. Vet Microbiol 10:525–531.
cell culture. Arch Gesamte Virusforsch 20:260–262. Lenghaus, C.; Forman, A. J.; and Hale, C. J. 1978. Experi-
Ide, S.; Yamagishi, K.; Yoshimura, M.; Maniwa, E.; Yasuda, mental infection of 35, 50 and 60 day old pig foetuses
H.; and Igarashi, J. 1977. Reaction of pigs to injection with porcine parvovirus. Aust Vet J 54:418–422.
with a bivalent vaccine of Japanese B encephalitis virus Lucas, M. H., and Napthine, P. 1971. Fluorescent antibody
and porcine parvovirus. J Jpn Vet Med Assoc technique in the study of three porcine viruses: Trans-
30:322–325. missible gastroenteritis virus, vomiting and wasting dis-
Johnson, R. H. 1969. A search for Parvoviridae (Picornaviri- ease virus, and the parvovirus 59e/63. J Comp Pathol
dae). Vet Rec 84:19–20. 81:111–117.
———. 1973. Isolation of swine parvovirus in Queensland. Lucas, M. H.; Cartwright, S. F.; and Wrathall, A. E. 1974.
Aust Vet J 49:257–259. Genital infection of pigs with porcine parvovirus. J
Johnson, R. H., and Collings, D. F. 1969. Experimental in- Comp Pathol 84:347–350.
fection of piglets and pregnant gilts with a parvovirus. Marrable, A. W., and Ashdown, R. R. 1967. Quantitative ob-
Vet Rec 85:446–447. servations on pig embryos of known ages. J Agric Sci
———. 1971. Transplacental infection of piglets with a 69:443–447.
porcine parvovirus. Res Vet Sci 12:570–572. Mayr, A.; Bachmann, P. A.; Siegl, G.; Mahnel, H.; and
Johnson, R. H.; Donaldson-Wood, C. R.; Joo, H. S.; and Al- Sheffy, B. E. 1968. Characterization of a small porcine
lender, U. 1976. Observations on the epidemiology of DNA virus. Arch Gesamte Virusforsch 25:38–51.
porcine parvovirus. Aust Vet J 52:80–84. McAdaragh, J. P., and Anderson, G. A. 1975. Transmission
Joo, H. S., and Johnson, R. H. 1977a. Observations on rapid of viruses through boar semen. In Proc 18th Annu Meet
diagnosis of porcine parvovirus in mummified foetuses. Am Assoc Vet Lab Diagn, pp. 69–76.
Aust Vet J 53:106–107. Mengeling, W. L. 1972. Porcine parvovirus: Properties and
———. 1977b. Serological responses in pigs vaccinated prevalence of a strain isolated in the United States. Am J
with inactivated porcine parvovirus. Aust Vet J Vet Res 33:2239–2248.
CHAPTER 17 PORCINE PARVOVIRUS Mengeling 199

———. 1975. Porcine parvovirus: Frequency of naturally proteins of selected autonomous parvoviruses. J Gen Vi-
occurring transplacental infection and viral contamina- rol 69:825–837.
tion of fetal porcine kidney cell cultures. Am J Vet Res Mengeling, W. L.; Lager, K. M.; Zimmerman, J. K.; Samarik-
36:41–44. ermani, N.; and Beran, G. W. 1991. A current assess-
———. 1977. Diagnosing porcine parvovirus-induced re- ment of the relative role of porcine parvovirus as a cause
productive failure. In Proc 20th Annu Meet Am Assoc of fetal porcine death. J Vet Diagn Invest 3:33–35.
Vet Lab Diagn, pp. 237–244. Molitor, T. W.; Joo, H. S.; and Collect, M. S. 1983. Porcine
———. 1978a. Elimination of porcine parvovirus from in- parvovirus: Virus purification and structural and anti-
fected cell cultures by inclusion of homologous anti- genic properties of virion polypeptides. J Virol
serum in the nutrient medium. Am J Vet Res 45:842–854.
39:323–324. Morimoto, T.; Fujisaki, Y.; Ito, Y.; and Tanaka, Y. 1972a. Bi-
———. 1978b. Prevalence of porcine parvovirus-induced ological and physiochemical properties of porcine par-
reproductive failure: An abattoir study. J Am Vet Med vovirus recovered from stillborn piglets. Natl Inst Anim
Assoc 172:1291–1294. Health Q (Tokyo) 12:137–144.
———. 1979. Prenatal infection following maternal expo- Morimoto, T.; Kurogi, H.; Miura, Y.; Sugimori, T.; and Fu-
sure to porcine parvovirus on either the seventh or four- jisaki, Y. 1972b. Isolation of Japanese encephalitis virus
teenth day of gestation. Can J Comp Med 43:106–109. and a hemagglutinating DNA virus from the brain of
Mengeling, W. L., and Cutlip, R. C. 1975. Pathogenesis of in stillborn piglets. Natl Inst Anim Health Q (Tokyo)
utero infection: Experimental infection of 5-week-old 12:127–136.
porcine fetuses with porcine parvovirus. Am J Vet Res Narita, M.; Inui, S.; Kawakami, Y.; Kitamura, K.; and Mae-
36:1173–1177. da, A. 1975. Histopathological changes of the brain in
———. 1976. Reproductive disease experimentally induced swine fetuses naturally infected with porcine parvovirus.
by exposing pregnant gilts to porcine parvovirus. Am J Natl Inst Anim Health Q (Tokyo) 15:24–28.
Vet Res 37:1393–1400. Paul, P. S., and Mengeling, W. L. 1980. Evaluation of a mod-
Mengeling, W. L., and Paul, P. S. 1986. The relative impor- ified live virus vaccine for the prevention of porcine par-
tance of swine and contaminated premises as reservoirs vovirus-induced reproductive disease in pigs. Am J Vet
of porcine parvovirus. J Am Vet Med Assoc Res 41:2007–2011.
188:1293–1295. ———. 1984. Oronasal and intramuscular vaccination of
Mengeling, W. L.; Cutlip, R. C.; Wilson, R. A.; Parks, J. B.; swine with a modified live porcine parvovirus vaccine:
and Marshall, R. F. 1975. Fetal mummification associat- Multiplication and transmission of vaccine virus. Am J
ed with porcine parvovirus infection. J Am Vet Med As- Vet Res 45:2481–2485.
soc 166:993–995. ———. 1986. Vaccination of swine with inactivated
Mengeling, W. L.; Cutlip, R. C.; and Barnett, D. 1978. porcine parvovirus vaccine in the presence of passive
Porcine parvovirus: Pathogenesis, prevalence, and pro- immunity. J Am Vet Med Assoc 188:410–413.
phylaxis. Proc Int Congr Pig Vet Soc 5:KA 15. Paul, P. S.; Mengeling, W. L.; and Brown, T. T., Jr. 1979.
Mengeling, W. L.; Brown, T. T., Jr.; Paul, P. S.; and Gun- Replication of porcine parvovirus in peripheral blood
tekunst, D. E. 1979. Efficacy of an inactivated virus vac- lymphocytes, monocytes, and peritoneal macrophages.
cine for prevention of porcine parvovirus-induced re- Infect Immun 25:1003–1007.
productive failure. Am J Vet Res 40:204–207. ———. 1980. Effect of vaccinal and passive immunity on
Mengeling, W. L.; Paul, P. S.; and Brown, T. T., Jr. 1980a. experimental infection of pigs with porcine parvovirus.
Transplacental infection and embryonic death following Am J Vet Res 41:1368–1371.
maternal exposure to porcine parvovirus near the time Paul, P. S.; Mengeling, W. L.; and Pirtle, E. C. 1982. Dura-
of conception. Arch Virol 65:55–62. tion and biological half-life of passively acquired
Mengeling, W. L.; Paul, P. S.; Gutekunst, D. E.; Pirtle, E. C.; colostral antibodies to porcine parvovirus. Am J Vet Res
and Brown, T. T., Jr. 1980b. Vaccination for reproductive 43:1376–1379.
failure caused by porcine parvovirus. Proc Int Congr Pig Pini, A. 1975. Porcine parvovirus in pig herds in southern
Vet Soc 6:61. Africa. J S Afr Vet Assoc 46:241–244.
Mengeling, W. L.; Pejsak, Z.; and Paul, P. S. 1984. Biological Pirtle, E. C. 1974. Titration of two porcine respiratory virus-
assay of attenuated strain NADL-2 and virulent strain es in mammalian cell cultures by direct fluorescent anti-
NADL-8 of porcine parvovirus. Am J Vet Res body staining. Am J Vet Res 35:249–250.
45:2403–2407. Redman, D. R.; Bohl, E. H.; and Ferguson, L. C. 1974.
Mengeling, W. L.; Paul, P. S.; Bunn, T. O.; and Ridpath, J. F. Porcine parvovirus: Natural and experimental infections
1986. Antigenic relationships among autonomous par- of the porcine fetus and prevalence in mature swine. In-
voviruses. J Gen Virol 67:2839–2844. fect Immun 10:718–723.
Mengeling, W. L.; Ridpath, J. F.; and Vorwald, A. C. 1988. Rodeffer, H. E.; Leman, A. D.; Dunne, H. W.; Cropper, M.;
Size and antigenic comparisons among the structural and Sprecher, D. J. 1975. Reproductive failure in swine
200 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

associated with maternal seroconversion for porcine cular synthesis in rat embryo cells by Kilhan rat virus. J
parvovirus. J Am Vet Med Assoc 166:991–995. Virol 8:402–408.
Rondhuis, P. R., and Straver, P. J. 1972. Enige kenmerken Thacker, B., and Leman, A. D. 1978. Evaluation of gravid
van een klien, hemagglutinerend DNA-virus, geisoleer uteri at slaughter for porcine parvovirus infection. Proc
uit een verworpen varkensfoetus. Tijdschr Diergeneeskd Int Congr Pig Vet Soc 5:M-49.
97:1257–1267. Thacker, B. J.; Joo, H. S.; Winkelman, N. L.; Leman, A. D.;
Ruckerbauer, G. M.; Dulac, G. C.; and Boulanger, P. 1978. and Barnes, D. M. 1987. Clinical, virologic, and
Demonstration of parvovirus in Canadian swine and histopathologic observations of induced porcine par-
antigenic relationships with isolates from other coun- vovirus infection in boars. Am J Vet Res 48:763–767.
tries. Can J Comp Med 42:278–285. Vannier, P., and Tillon, J. P. 1979. Diagnostic de certitude de
Siegl, G. 1976. The Parvoviruses, 1st ed. Vienna, Austria: l’infection a parvovirus dans les trouble de la reproduc-
Springer-Verlag. tion de l’espèce porcine. Rec Med Vet 155:151–158.
Siegl, G., and Gautschi, M. 1973a. The multiplication of Westenbrink, F.; Veldius, M. A.; and Brinkhof, J. M. A. 1989.
parvovirus Lu III in a synchronized culture system. I. An enzyme-linked assay for detection of antibodies to
Optimum conditions for virus replication. Arch porcine parvovirus. J Virol Methods 23:169–178.
Gesamte Virusforsch 40:105–118. Wrathall, A. E., and Mengeling, W. L. 1979a. Effect of
———. 1973b. The multiplication of parvovirus Lu III in a porcine parvovirus on development of fertilized pig
synchronized culture system. II. Biochemical character- eggs in vitro. Br Vet J 135:249–254.
istics of virus replication. Arch Gesamte Virusforsch ———. 1979b. Effect of transferring parvovirus-infected
40:119–127. fertilized pig eggs into seronegative gilts. B Vet J
Siegl, G.; Hallauer, C.; and Novak, A. 1972. Parvoviruses as 135:255–261.
contaminants of permanent human cell lines. IV. Multi- ———. 1979c. Effect of inseminating seropositive gilts
plication of KBSH-virus in KB-cells. Arch Gesamte with semen containing porcine parvovirus. Br Vet
Virusforsch 36:351. 135:420–425.
Suzuki, H., and Fujisaki, Y. 1976. Immunizing effects of in- Yasuhara, H.; Matsui, O.; Hirahara, T.; Ohgtani, T.; Tanaka,
activated porcine parvovirus vaccine on piglets. Natl In- M. L.; Kodama, K.; Nakai, M; and Sasaki, N. 1989. Char-
st Anim Health Q (Tokyo) 16:81. acterization of parvovirus isolated from diarrheic feces
Tennant, R. W. 1971. Inhibition of mitosis and macromole- of a pig. Jpn J Vet Sci 51:337–344.
Porcine Reproductive and
18 Respiratory Syndrome
D. A. Benfield, J. E. Collins, S. A. Dee, P. G. Halbur,
H. S. Joo, K. M. Lager, W. L. Mengeling, M. P. Murtaugh,
K. D. Rossow, G. W. Stevenson, and J. J. Zimmerman

Previously unrecognized epidemics of an acute porcine syndrome,” and “porcine epidemic abortion and respira-
reproductive disease characterized by a marked increase tory syndrome.” Today PRRS is the name used almost ex-
of late-term abortions, stillborn and weak pigs, lowered clusively, and it is the name used hereafter in this chap-
farrowing rates, high death rates among weaned pigs, ter.
and delayed returns to estrus were first reported in the Although epidemiological data from even the very
United States in 1987. In many of the epidemics severe early cases of PRRS suggested that it was an infectious
respiratory disease in suckling and weaned pigs was also disease, it was not until several years later that the cause,
a prominent feature (Keffaber 1989; Loula 1991). Clini- PRRS virus (PRRSV), was identified with certainty
cally similar epidemics were recognized in Canada in the (Wensvoort et al. 1991; Collins et al. 1992). The origin of
autumn of 1987 (Dea et al. 1990), in Japan in 1989 PRRSV is still unknown, but it is intriguing that North
(Shimizu et al. 1994), in Germany in 1990 (Lindhaus and American and European isolates of the virus have
Lindhaus 1991), in the Netherlands, Spain, France, and marked genotypic and phenotypic differences, suggest-
the United Kingdom in 1991 (Wensvoort et al. 1991; ing that if they originated from a common ancestor, they
White 1991; Meredith 1992), in Denmark in 1992 developed along different evolutionary lines.
(Bøtner et al. 1994), and since 1992 in much of the rest PRRS continues to plague the swine industry both as
of the world wherever large numbers of swine are raised an endemic respiratory disease, often as part of what has
(Meredith 1995). been referred to as the “porcine respiratory disease com-
In 1991 the name “porcine reproductive and respira- plex,” and as a sporadic, acute reproductive disease most
tory syndrome” (PRRS) was proposed as the internation- strikingly manifested as an unusually high incidence of
al designation for such epidemics (European Commis- abortions. Recent severe epidemics of reproductive dis-
sion 1991), with the intent of supplanting an array of ease described in the United States as “acute” or “atypi-
different and somewhat confusing names that, until that cal” PRRS (Halbur and Bush 1997; Epperson and Holler
time, were being used in various parts of the world to de- 1997; Mengeling et al. 1997; Zimmerman et al. 1997a)
scribe the same syndrome. Among these were “mystery and caused by what may be new, more virulent strains of
swine disease” (often used in the United States before the PRRSV, suggest that PRRSV is continuously changing—
cause of PRRS was identified), “blue ear disease” (based a concept that is supported in part by base sequence (Ka-
on a transient, bluish discoloration of the ears of some pur et al. 1996; Murtaugh et al. 1997) and restriction en-
affected sows and gilts), “swine infertility and abortion zyme (Wesley et al. 1996; Mengeling et al. 1997) analysis.
If so, the potential for genotypic and related phenotypic
change, and the ability of PRRSV to persist in herds for
This chapter was compiled by the section editor from invited contribu- extended periods of time, pose serious challenges for the
tions of individuals recognized as being among the international experts in control and possible eradication of PRRS.
specific subject areas dealing with PRRS and PRRS virus. It was believed that
this approach would provide the most comprehensive and incisive treatment
of what is currently known about PRRS. Contributors and their correspond-
ETIOLOGY
ing subject areas are as follows: D. A. Benfield (Introduction, Etiology); J. E.
Collins and K. M. Lager (Diagnosis); S. A. Dee and H. S. Joo (Prevention and The first major step in establishing the etiology of PRRS
Control); P. G. Halbur and K. M. Lager (Lesions); W. L. Mengeling (Vaccines); was made by Collins et al. (1990), who reproduced the
M. P. Murtaugh (Immunity); K. D. Rossow (Pathogenesis); G. W. Stevenson respiratory facet of the syndrome by exposing gnotobi-
(Clinical Signs); and J. J. Zimmerman (Epidemiology). In addition, numerous
other individuals knowledgeable about PRRS or PRRS virus or both were
otic pigs to filtered homogenates of tissues from field
asked for their input. Every effort was made to blend the individual subject cases. About a year later Koch’s postulates were fulfilled
areas into a single coherent, highly informative, error-free document. when investigators at the Central Veterinary Research In-

201
202 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

stitute in Lelystad, Netherlands, caused the reproductive etiology, epidemiology) depending largely on the per-
facet of the syndrome with a virus they had isolated in sonal preference of the author, or authors, who con-
porcine alveolar macrophages (PAMs) and identified as tributed that information rather than on any generally
Lelystad virus (LV) (Terpstra et al. 1991; Wensvoort et al. accepted criteria.
1991). Soon thereafter, similar results were obtained by A number of studies (Benfield et al. 1992; Conzelman
American investigators with a virus they had isolated in et al. 1993; Dea et al. 1992; Wensvoort et al. 1992b; Meu-
an established cell line and identified as VR-2332 (Ben- lenberg et al. 1993a, b; Saito et al. 1996) have indicated
field et al. 1992; Collins et al. 1992). Additional studies that PRRSV is closely related biologically, structurally,
indicated that while LV and VR-2332 were sufficiently re- and genetically to equine arteritis virus (EAV), lactate de-
lated to be considered strains of PRRSV, they were also hydrogenase-elevating virus (LDV) of mice, and simian
genetically (Meng et al. 1995a; Murtaugh et al. 1995) and hemorrhagic fever virus (SHFV). On the basis of these
antigenically (Wensvoort et al. 1992a; Nelson et al. 1993) common properties, PRRSV, EAV, LDV, and SHFV
distinguishable—with LV the prototype of most strains (Plagemann and Moenning 1992; Plagemann 1996) have
of PRRSV subsequently isolated in Europe, and VR-2332 been grouped together in a newly created genus Ar-
the prototype of most strains of PRRSV subsequently terivirus, of the family Arterividae, order Nidovirales (Ca-
isolated in North America and Asia. vanagh 1997). Among the properties of arteriviruses
In addition to the substantial differences that have that are especially important from a clinical perspective
been identified between LV and VR-2332 there are also are their ability to (1) cause asymptomatic, persistent in-
differences among North American isolates of PRRSV fections as well as severe and often fatal diseases; (2)
(i.e., VR-2332-like isolates), including those isolated at replicate in macrophages; and (3) exhibit considerable
the same time from the same herd (Mengeling et al. genome plasticity (Plagemann 1996).
1997). Usually these are no more than what appear to be
minor sequence (base) changes that may have little or no Size and Morphology
effect on phenotypic expression. However, some North The causative agent of PRRS is an enveloped virus with a
American isolates have been shown to differ serological- diameter of 50–65 nm, a relatively smooth surface, and
ly (E. A. Nelson, unpublished data—1998) and in viru- a cubical, nucleocapsid core with a diameter of 25–35
lence (Halbur et al. 1995; Halbur et al. 1996b; Mengeling nm (Benfield et al. 1992; Dea et al. 1995; Wensvoort et al.
et al. 1996c). Despite the fact that most isolates can be 1992b) (Fig. 18.1). Occasionally, short (8–12 nm) granu-
differentiated on the basis of minor to marked sequence lar projections are observed on the surface of negatively
changes, and some on the basis of phenotypic expres- stained extracellular viral particles (Dea et al. 1995). In
sion, there is still no consensus as to exactly what prop- ultrathin sections of infected PAMs, PRRSV appears as a
erties define a “strain” versus an “isolate” of PRRSV. As a 45–65 nm spherical particle with a 30–35 nm nucleocap-
consequence, these two terms are often used inter- sid core and a smooth external lipid bilayer membrane
changeably in various subject areas of this chapter (e.g., (Dea et al. 1995; Wensvoort et al. 1992b).

18.1. A complete PRRSV particle


and (indicated by the open arrow) an
incomplete (coreless) PRRSV particle.
The insert shows a complete PRRSV
particle with the 35 nm nucleocapsid
core (black arrow) surrounded by an
envelope. Bars = 50 nm. (Courtesy of
D. Robison, South Dakota State
University.)
CHAPTER 18 PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME Benfield et al. 203

Density, Temperature, and pH Stability ese PRRSV isolates, antigenically related to VR-2332,
PRRSV has a buoyant density of 1.19 in cesium chloride, were shown to agglutinate mouse erythrocytes. Pretreat-
and glycerol-tartrate gradients; and 1.14–1.15 g/mL in ment of these isolates with detergent increased the
sucrose gradients (Benfield et al. 1992; Wensvoort et al. hemagglutinating activity, indicating possible release of
1992b; Bautista et al. 1996). an envelope glycoprotein responsible for hemagglutina-
The temperature stability of PRRSV is similar to that tion (Jusa et al. 1996).
of the porcine coronavirus (transmissible gastroenteritis
virus). PRRSV is stable for a long time (months to years) Genomic Organization
at temperatures of −70˚C and −20˚C. Approximately The genome of PRRSV is similar in organization to those
90% of PRRSV infectivity is lost within 1 week at 4˚C, but of other arteriviruses and the coronaviruses (Plagemann
low titers of infectious virus can still be detected for at and Moenning 1992; Conzelman et al. 1993; Meulenberg
least 30 days. PRRSV infectivity persists for 1–6 days at et al. 1993a, b; Plagemann 1996). Virions contain a 15.1
20–21˚C, 3–24 hours at 37˚C, and 6–20 minutes at 56˚C. kilobase (kb), linear, single-stranded, polyadenylated
The thermal stability of PRRSV in serum and tissues is RNA of positive polarity (Conzelman et al. 1993; Meu-
similar to that described for virus stored in media. lenberg et al. 1993a, b; Murtaugh et al. 1995). The base
PRRSV was isolated from 47%, 14%, and 7% of porcine sequence has been determined for the entire genome of
serum samples stored at 25˚C for 24, 48, and 72 hours, LV (Meulenberg et al. 1993a, b) and for the 3′ terminal
respectively. When serum was stored at 4˚C or −20˚C, 3.5 kb portion of the genome of other European and
PRRSV was isolated from 85% of the samples after 72 many North American isolates of PRRSV (Conzelman et
hours (Van Alstine et al. 1993). In another study, serum al. 1993; Mardassi et al. 1994a; Meng et al. 1995a, b;
and muscle collected from 14 pigs 5 days after they had Murtaugh et al. 1995; Saito et al. 1996; Suarez et al.
been exposed to PRRSV were tested for infectious virus 1996). The genome contains eight open reading frames
after subsequent storage at 4˚C. Infectious PRRSV was (ORFs) which encode specific viral proteins. ORF 1a and
isolated from all of the sera after 48 hours in storage and 1b, which span the 5′ terminal 12 kb part of the genome,
from muscle of 2 of the 14 pigs after 5 days in storage encode proteins involved in RNA replication and tran-
(Bloemraad et al. 1994). scription, including the RNA-dependent RNA poly-
PRRSV is stable at pH 6.5–7.5, but infectivity is rapid- merase. The remaining 3.5 kb located on the 3′ terminal
ly lost at pH below 6 and above 7.5 (Benfield et al. 1992; of the genome comprise ORFs 2 through 7, which encode
Bloemraad et al. 1994). the putative viral structural proteins. The order of the
Collectively, these studies indicate that PRRSV is genes is 5′-ORF 1a and 1b (genes encoding the RNA
thermal and pH labile and is not likely to survive in the polymerase)-ORFs 2 through 6 (genes encoding the
environment for extended periods of time. It can be as- membrane-associated proteins)-ORF 7 (gene encoding
sumed that the labile nature of PRRSV is part of the rea- the nucleocapsid protein)-3′ end. This sequence of gene
son why it is not consistently isolated from aborted fe- transcription and expression is the same in EAV, LDV,
tuses and stillborn pigs during epidemics of PRRS. toroviruses, and coronaviruses.
Serum and tissues collected for virus isolation (VI) Genomic transcription involves the formation of a 3′-
should be stored at − 20˚C or 4˚C to preserve the infec- coterminal nested set of six subgenomic messenger
tivity of PRRSV in the sample. RNAs for each of the ORFs 2 through 7. Some PRRSV
isolates produce seven subgenomic mRNAs. The extra
Sensitivity to Lipid Solvents and mRNA has a molecular mass between those of the mR-
Hemagglutinating Activity NAs transcribed from ORFs 2 and 3 (Meng et al. 1996).
PRRSV is inactivated by treatment with the lipid sol- The significance of this extra subgenomic mRNA is un-
vents, namely, chloroform and ether (Benfield et al. known, but the number of subgenomic mRNAs pro-
1992; Dea et al. 1992; Wensvoort et al. 1992b). Most ar- duced by PRRSV does not correlate with either pneu-
teriviruses, including PRRSV, are also highly unstable in movirulence or with RNA from defective-interfering
solutions containing low concentrations of detergents, particles (Meng et al. 1996). All isolates of LDV synthe-
which disrupt the envelope with concomitant release of size seven subgenomic mRNAs, none of which are pack-
the noninfectious core particles and loss of infectivity aged into virions (Plagemann and Moenning 1992;
(Plagemann 1996). Plagemann 1996). Each subgenomic mRNA carries a
North American and European PRRSV isolates (Ben- nonencoding leader at its 5′ end that is derived from the
field et al. 1992; Dea et al. 1992; Wensvoort et al. 1991) 5′ end of the viral genome. The ORFs of LDV and
and other arteriviruses (Plagemann and Moenning 1992; PRRSV are read in alternate frames, with ORFs 2, 5, and
Plagemann 1996) failed to agglutinate erythrocytes of 7 read in one frame, ORF 4 in another, and ORFs 3 and 6
any of the species tested. However, recently, two Japan- in the third frame (Plagemann 1996).
204 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Viral Proteins assembly, attachment, virulence, pathogenicity, and


Experiments in which the mRNAs have been cloned into antigenicity.
Escherichia coli, baculovirus, and Semliki Forest virus in-
dicate that each of the subgenomic mRNAs codes for one Biologic and Genetic Variability
protein. In cells infected with LV or VR-2332, at least of PRRSV Isolates
three viral proteins of 15, 19, and 26 kDa can be detect- There is ample biologic and genetic evidence to suggest
ed by radioimmunoprecipitation and western blotting the presence of different PRRSV strains or isolates. RNA
using polyclonal antisera from pigs previously infected synthesis is inherently error prone, and RNA genomes
with VR-2332 (Nelson et al. 1993; Nelson et al. 1995; have high mutation frequencies as a result of point mu-
Bautista et al. 1996). Similar proteins have been detected tations, deletions, additions, and substitutions (Holland
in lysates of cells infected with either LV or the Canadian 1995). The biologic and genetic differences of PRRSV are
IAF-exp91 isolate of PRRSV (Meulenberg et al. 1995; determined on the basis of (1) different clinical presen-
Meulenberg and Den Besten 1996; Mardassi et al. 1995). tations of the disease; (2) experimental evidence to indi-
These three proteins have also been observed in purified cate differences in pneumovirulence and reproductive
preparations of virions, suggesting that the 15, 19, and virulence; (3) antigenic differences defined by reactivity
26 kDa proteins are structural. The molecular weights of with polyclonal and monoclonal antibodies in serologic
these proteins correspond well with the reported molec- assays; and (4) differences in the RNA sequences.
ular weights of the major structural proteins of LDV and LV and VR-2332 produce remarkably similar repro-
EAV, which contain a nucleocapsid protein of 12–15 ductive and respiratory clinical syndromes. However, the
kDa, a nonglycosylated membrane-spanning protein of first biologic difference noted between the North Ameri-
16–19 kDa, and at least one glycosylated envelope pro- can and European PRRSV isolates was the frequent ob-
tein of 24–44 kDa (Plagemann and Moenning 1992). servation of blue discoloration of ears, teats, snout, ven-
Recent experiments using proteins expressed from tral cervical skin, vulva, and abdomen in outbreaks of
ORFs 2 through 7 of LV indicate that the products of PRRS in Europe, but not in North America (Goyal 1993).
ORFs 2 through 5 are glycosylated, whereas the products Comparative pathogenicity studies in colostrum-de-
of ORFs 6 and 7 are not (Meulenberg et al. 1995; Meu- prived, cesarean-derived pigs indicated that PRRSV iso-
lenberg and Den Besten 1996; Van Nieuwstadt et al. lates could be classified in high- and low-virulence
1996). The products of ORFs 2 through 4 were identified groups based on the severity of gross and microscopic
in preparations of purified virions using monoclonal an- lung lesions (Halbur et al. 1995; Halbur et al. 1996b). In
tibodies or polyclonal antisera that had been prepared by addition, some PRRSV isolates were more likely to in-
using either PRRSV proteins expressed in vitro or syn- duce rhinitis, encephalitis, and myocarditis (Halbur et al.
thetic peptides prepared from the sequence of each viral 1996b), and some U.S. isolates were more or less virulent
protein. These antibodies were then used to identify the than LV (Halbur et al. 1995). While the least virulent iso-
corresponding ORF product in lysates of either PRRSV- lates had greater sequence variation in ORFs 2 through 4
infected cell cultures or purified virions by radioim- than did other U.S. isolates to which they were com-
munoprecipitation and western blotting. Results showed pared, virulence could not be correlated with any specif-
that ORFs 2, 3, and 4 encode for N-glycosylated proteins ic changes in the sequence of the viral genome (Meng et
with molecular weights of 29–30, 45–50, and 31–35 al. 1995b). Similar studies done with high- and low-
kDa, respectively (Meulenberg et al. 1995; Meulenberg pneumovirulent PRRSV isolates in pregnant gilts sug-
and Den Besten 1996; Van Nieuwstadt et al. 1996). Mon- gested that PRRSV isolates differ in their effect on the re-
oclonal antibodies to the ORF 4 protein of LV neutralize productive tract. However, there was no clear indication
viral infectivity, indicating that at least part of this pro- that the virulence of a particular PRRSV isolate for the
tein is exposed at the surface of the virion (Van Nieuw- respiratory tract was a predictor of its virulence for the
stadt et al. 1996). reproductive tract (Mengeling et al. 1996c).
LV is the first arterivirus shown to contain six struc- Assessment of antigenic differences between PRRSV
tural proteins, consisting of four membrane glycopro- isolates has been difficult due to technical problems in
teins, a membrane-spanning protein (M), and a nucleo- producing monoclonal antibodies to the PRRSV struc-
capsid protein (N). Only three to four structural proteins tural proteins and the unreliability of monospecific poly-
have been identified on North American PRRSV isolates clonal serum for use in neutralization assays for the dif-
(Nelson et al. 1993; Nelson et al. 1995; Mardassi et al. ferentiation of serotypes. A comparison of 24 field sera
1995; Mardassi et al. 1996; Bautista et al. 1996) and the and 7 PRRSV isolates from Europe and North America
other arteriviruses EAV (De Vries et al. 1992) and LDV failed to reveal two-way cross-reactions between Euro-
(Harty and Plagemann 1988; Plagemann and Moenning pean and North American PRRSV isolates using the im-
1992; Faaberg and Plagemann 1995; Plagemann 1996). munoperoxidase macrophage assay (Wensvoort et al.
Currently, there is little information on the biologic 1992b). While the North American PRRSV isolates re-
function of each of these viral proteins in terms of viral acted with antisera against the European strains, the Eu-
CHAPTER 18 PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME Benfield et al. 205

ropean strains did not react with antisera from the U.S. RespPRRS/Repro®) and Prime Pac® PRRS. Both of these
field cases (Wensvoort et al. 1992a, b). In other studies, vaccines are modified-live viruses that have been attenu-
pigs seropositive for LV were found to be seronegative for ated in virulence by serial passage in monkey kidney
VR-2332, and vice versa (Bautista et al. 1993). Mono- cells. Genetic and antigenic differences between the vac-
clonal antibodies specific for epitopes on the 15 kDa nu- cine strains and virulent field strains of PRRSV have
cleocapsid protein indicate that there are conserved and been identified and used diagnostically. For example, the
variable epitopes between European and North Ameri- ORF 5 sequence of RespPRRS® contains several uncom-
can isolates (Nelson et al. 1993; Drew et al. 1995; Magar mon restriction sites. As a consequence, digestion of
et al. 1995). Nelson et al. (1993) suggested that the North RespPRRS® with selected restriction endonucleases gives
American and European isolates be classified into two a gel pattern distinct from that of at least most field iso-
antigenic subgroups on the basis of their reactivity with lates of PRRSV (Wesley et al. 1996). Likewise, Prime
nucleocapsid monoclonal antibodies. The monoclonal Pac® PRRS does not react with the SDOW 12 or 17 mon-
antibodies SDOW 12 and 17, which are specific for con- oclonal antibodies, indicating that the nucleocapsid pro-
served epitopes on the nucleocapsid protein of North tein of Prime Pac® PRRS is missing an epitope present
American and European isolates, are used as universal on a majority of field isolates of PRRS—including the
diagnostic reagents. Monoclonal antibodies to the N virulent field isolate from which it originated (R. A.
protein and a 45 kDa protein encoded by ORF 3 of the Hesse, unpublished data—1998; E. A. Nelson, unpub-
Humberside isolate of PRRSV react with European, but lished data—1998). The antigenic difference between
not U.S., isolates (Drew et al. 1995). Antigenic differ- Prime Pac® PRRS and its parent strain suggests a change
ences on the M protein of North American and Euro- associated with serial passage in vitro.
pean isolates of PRRSV were also demonstrated using
monoclonal antibodies to the M protein of a Canadian Cultivation and Morphogenesis
isolate of PRRSV, which reacts only with North Ameri- Most isolates of PRRSV replicate to titers of 105–107
can PRRSV isolates (Magar et al. 1997). median tissue culture infectious doses (TCID50) in PAMs,
The antigenic variation observed between the Euro- African green monkey kidney cells (MA-104), derivatives
pean and North American PRRSV isolates has been con- of MA-104 (CL-2621 and MARC-145), and CRL-11171
firmed by analysis of the nucleotide and amino acid se- cells (Wensvoort et al. 1991; Benfield et al. 1992; Dea et
quences of LV and VR-2332. Nucleotide sequence al. 1992; Kim et al. 1993; Goyal 1993; Halbur et al. 1995).
analysis of VR-2332 and LV indicated that the character- The virus replicates cytocidally in each cell type, causing
istics of ORFs 2 through 7 are very similar, but the pre- cells to become rounded, clumped, and pyknotic. Affect-
dicted protein structures indicate numerous differences ed cells eventually detach from the culture vessel. The cy-
in the molecular weights, isoelectric points, and predict- topathic effect usually develops within 1–4 days in PAM
ed glycosylation sites of the corresponding proteins. cultures and within 2–6 days in CL-2621 cell cultures
Amino acid sequences for VR-2332 and their similarities (Benfield et al. 1992; Pol et al. 1992). Virus replication is
to those of LV are 76% ORF 2, 72% ORF 3, 80% ORF 4, restricted to the cytoplasm, as demonstrated by fluores-
80% ORF 5, 91% ORF 6, and 74% ORF 7 (Murtaugh et al. cent antibody or immunoperoxidase staining (Benfield
1995). Despite significant sequence differences, the hy- et al. 1992; Dea et al. 1992; Wensvoort et al. 1991).
dropathicity profiles of VR-2332 and LV indicated that A few North American isolates of PRRSV have been
PRRSV tends to conserve the structure of the proteins, found to grow exclusively in either PAMs or CL-2621
and that the extensive amino acid differences in the pro- cells, but most grow in both cell types (Bautista et al.
teins account for the differences in serologic cross-reac- 1993; Goyal 1993). Conversely, most, if not all, European
tivity (Murtaugh et al. 1995). The amino acid homolo- isolates of PRRSV replicate better or exclusively in
gies among North American isolates are 90% or greater PAMs. Vaccine viruses replicate at levels 100–1000 times
for ORFs 2 through 7 (Kapur et al. 1996; Meng et al. lower on PAMs than on derivatives of a monkey kidney
1995a, b; Mardassi et al. 1994a). Among U.S. isolates, cell line (D. A. Benfield, unpublished data—1998). Al-
ORF 5 is the most variable and ORF 6 the most con- though data are lacking, it is probable that the use of CL-
served (Kapur et al. 1996). Sequence analysis of PRRSV 2621 or MARC-145 cells versus PAMs selects for the iso-
isolates from midwestern swine herds indicates the lation of vaccine viruses rather than field isolates of
viruses are evolving by random mutation and by intra- PRRSV.
genic recombinations between different PRRSV isolates There is limited information on the morphogenesis
(Kapur et al. 1996). of PRRS virions. Pol et al. (1992) have described the mor-
phogenesis of LV in PAMs. These investigators observed
Differences between Field Isolates viral nucleocapsids budding through the smooth endo-
and Vaccine Viruses plasmic reticulum (ER) by 6 hours after inoculation. By 9
There are currently two commercially available modi- hours after inoculation they noted enveloped viral parti-
fied-live-virus vaccines: RespPRRS® (recently renamed cles in the lumen of the smooth ER and in the Golgi re-
206 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

gion. Viral replication was restricted to the cytoplasm. entered and became endemic in a large proportion of the
Intact viral particles were released from infected cells by world’s domestic swine population. The original source
exocytosis as early as 9–12 hours after inoculation. of the virus and the circumstances under which it came
Pulse-chase experiments with a Canadian isolate of into the domestic swine population are not known. A
PRRSV propagated in MARC-145 cells indicated that vi- wildlife reservoir is the most logical explanation, with
ral assembly starts in the ER, where the matrix and enve- feral swine the most likely candidate species, but in
lope proteins are linked by disulfide bonds and then in- North America, preliminary data suggest that the virus
teract with the nucleocapsid protein that has previously has moved into feral swine from domestic swine, rather
accumulated in the cytoplasm of the perinuclear region than the reverse (J. J. Zimmerman, unpublished data—
of the cell. Thereafter, vesicles containing enveloped nu- 1998). Further complicating the puzzle, the prototypic
cleocapsids are derived from the ER and transported to European and North American PRRSV isolates are suffi-
the Golgi region, where budding takes place and some ciently different from each other, despite the temporal
envelope proteins undergo terminal glycosylation. At the proximity of their emergence, to suggest that either they
end of the transit from the ER to the Golgi region, the en- originated from two independent sources, or they devel-
velope protein has acquired its mature structure, which oped along different evolutionary lines over a period of
appears to influence the onset of virus release by exocy- at least several years. Although PRRSV has become ubiq-
tosis (Mardassi et al. 1996). uitous in domestic swine, the question of its origin re-
mains relevant because, until we know otherwise, we
EPIDEMIOLOGY cannot discount the possibility that new strains of
PRRSV may emerge from the original reservoir.
Currently available information suggests that PRRSV en-
tered the domestic swine population relatively recently Geographic Distribution and Prevalence
and spread rapidly thereafter. The earliest evidence of Although survey information is not available from all
PRRSV infection in domestic swine comes from a retro- countries, PRRSV appears to have become endemic in
spective serologic study of herds in Ontario, Canada. nearly all swine-producing areas of the world. Although
Carman et al. (1995) found that none of 50 herds sam- some countries claim to be free of PRRSV infection, only
pled in 1978 were serologically positive for antibodies Australia and Sweden have documented their virus-free
against PRRSV by enzyme-linked immunosorbent assay status. In Australia, a survey of 875 serum samples from
(ELISA) or indirect fluorescent antibody (IFA), but anti- 163 herds selected across all states revealed no evidence
bodies were detected in sera of 2 of 51 (3.9%) herds sam- of antibodies against PRRSV (Garner et al. 1996). In
pled in 1979 and in sera of 8 of 51 (15.7%) herds sampled Sweden, a serosurvey of 11,003 samples collected be-
in 1980. tween 1993 and 1996 also revealed no evidence of infec-
Apparently, PRRSV did not enter the United States tion (Elvander et al. 1997). However, given the demon-
for several years after the first infections in Canada. None strated transmissibility of PRRSV, it will not be easy to
of 1425 serum samples collected from 118 Iowa swine maintain these areas free of infection.
herds in 1980 were antibody positive by IFA (Zimmer- Reliable estimates of the prevalence of infection in
man et al. 1997c). One of 26 herds (3.8%) sampled in endemically infected areas are not generally available for
1985 was PRRSV-infected. Each successive year showed a several reasons. Most published studies are not based on
marked increase in prevalence. By 1988, 17 of 27 herds statistically valid population sampling procedures and,
(63.0%) and 313 of 658 (47.6%) pigs were seropositive. therefore, may lack accuracy. More recently, prevalence
The low prevalence in 1985 suggests that entry of the studies have become confounded by the use of modified-
virus into Iowa occurred during or shortly prior to 1985. live-PRRSV vaccines, the use of which results in serum
Similar to the Iowa data, the earliest evidence of infec- antibodies that are indistinguishable from those pro-
tion in the state of Minnesota comes from serum sam- duced against field isolates. Epidemiologic investigations
ples collected in 1986 (Yoon et al. 1992). are further complicated by the fact that vaccine virus is
The infection did not remain limited to North Amer- disseminated from vaccinated to naive pigs.
ica. In Asia, antibodies against PRRSV were retrospec- In the United States the best prevalence estimate is
tively documented in South Korea in serum from pigs based on a 1995 National Animal Health Monitoring
imported in October 1985 (Shin et al. 1993), and in System (NAHMS) study (U.S. Department of Agricul-
Japan in samples collected in June 1988 (Hirose et al. ture 1997). In the NAHMS study, 8038 serum samples
1995). In Europe, clinical outbreaks of PRRS were first were collected from 286 herds in 16 swine-producing
reported in Germany in November 1990 (European states. Among herds not using modified-live-PRRSV vac-
Commission 1991), but there is serologic evidence sug- cine, 129 of 217 (59.4%) herds were infected. Among
gestive of the presence of PRRSV in Germany as early as 6376 unvaccinated animals, 41.3% had antibody titers
1988. detectable by IFA. When animals were identified by sta-
Thus, in a period of approximately 10 years, PRRSV tus, 23.5% of unvaccinated breeding animals and 51.7%
CHAPTER 18 PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME Benfield et al. 207

of unvaccinated finishers were IFA positive. As in the ed boars through day 92 postexposure (Christopher-
United States, reports from other parts of the world in- Hennings et al. 1995a). Transmission of PRRSV to fe-
dicate that the prevalence of infection is high in endemi- males artificially inseminated with undiluted semen
cally infected regions. from experimentally infected boars has been demon-
Hirose et al. (1995) reported that 46.4% of pigs sam- strated (Yaeger et al. 1993), and transmission of PRRSV
pled in 1993 in Chiba Prefecture, Japan, were serological- to females using extended semen from experimentally
ly positive for PRRSV. In Belgium, Maes (1997) found infected boars has also been proven (Gradil et al. 1996).
that 50 of 50 herds and 96% of market hogs tested were Virus has also been recovered from mammary secretions
seropositive. And although PRRS was first reported in collected from gilts and sows challenged in the third
Germany in November 1990, Geue (1995) found that trimester of gestation (J. J. Zimmerman, unpublished
490 of 689 herds (71.1%) in the Nordfriesland district of data—1998). The prevalence of fecal shedding of
Germany were already infected by October 1992. One ex- PRRSV remains an open question. Yoon et al. (1993) re-
ception to the trend of rapid spread and high prevalence ported extensive fecal shedding by young pigs over a 35-
is the “Pays de la Loire” region of France. By implement- day observation period. In contrast, Rossow et al. (1994)
ing a rigorous program of serologic monitoring and found only intermittent shedding, and Wills et al.
strict control measures, prevalence was held below 2% (1997b) found no infectious virus in fecal samples col-
nearly 2.5 years after PRRSV had entered the region (Le lected from pigs over a 42-day postinoculation observa-
Potier et al. 1997). tion period.
Shedding of virus in saliva, urine, and perhaps feces
Transmission results in environmental contamination. In general,
Efficient transmission has been a hallmark of PRRS since PRRSV is a fragile virus that is quickly inactivated in the
it was first identified clinically. In the absence of vaccines environment; however, it can remain infectious for an ex-
or treatments effective in stopping shedding of virus or tended period of time under specific conditions of tem-
eliminating infection in chronic carrier animals, strate- perature and moisture. Bloemraad et al. (1994) found
gies for the prevention, control, and elimination of both temperature and pH to influence the stability of in-
PRRSV must be grounded upon a thorough understand- fectious virus. At 37˚C and a pH of 6.0, the half-life (in-
ing of the means by which transmission occurs. Swine activation of one-half of the virus population) of LV was
are susceptible to PRRSV by a number of routes of ex- estimated at 6.5 hours, but half-life declined to 0.65
posure, including oral, intranasal, intramuscular, in- hours at pH 5.0 and 1.28 hours at pH 8.5. At 4˚C and a
traperitoneal, and vaginal. The virus is highly infectious pH of 7.5, the half-life of LV was estimated to be 140
and swine are readily infected by inoculation of 10 or hours. The virus is quickly inactivated in the absence of
fewer PRRSV particles by either intranasal or intramus- moisture or in hostile solutions. Pirtle and Beran (1996)
cular routes (K. J. Yoon, unpublished data—1998). The examined the persistence of PRRSV in or on 16 fomites,
infectious dose by other routes has not been determined, including plastic, stainless steel, rubber, alfalfa, wood
but studies with LDV, a closely related virus of mice, are shavings, straw, corn, swine starter feed, denim cloth,
suggestive of what we might expect. For LDV, the mini- phosphate-buffered saline, saline G, well water, city wa-
mum infectious dose has been shown to vary consider- ter, swine saliva, urine, and fecal slurry. At 25–27˚C virus
ably depending on the route of exposure. Cafruny and did not persist on fomites beyond day zero, except for
Hovinen (1988) found the minimum infectious dose by phosphate-buffered saline through day 3, saline G
intraperitoneal or tail cartilage injections approached 1 through day 6, well water through day 8, and city water
virus particle; that is, similar to PRRSV. In contrast, ex- through day 11. From a practical perspective, standard
posure via mucosal surfaces such as ocular, vaginal, or cleaning and disinfection procedures should be effective
oral routes required a minimum infectious dose of for inactivation of PRRSV in facilities and on equipment.
103.3–105.3 virions, depending on the specific anatomical Although the data indicate that virus cannot persist in fe-
location (Cafruny and Hovinen 1988). Although we cal slurry, the potential for persistence of virus in la-
know PRRSV is highly infectious, additional work is goons has not been examined.
needed to further define the effect of route of exposure Infection produces a prolonged carrier state. Zim-
on minimum infectious dose. merman et al. (1992) reported transmission by direct
Infection results in the shedding of virus in saliva, contact from sows infected 99 days earlier to commin-
urine, semen, and mammary secretions. Wills et al. gled susceptible animals. Albina et al. (1994a, b) subse-
(1997b) detected PRRSV in saliva for up to 42 days and quently demonstrated transmission of PRRSV by pigs
in urine for up to 14 days. Swenson et al. (1994b) found infected 15 weeks earlier. More recently, isolation of
infectious virus in the semen of experimentally infected virus from oropharyngeal samples for up to 157 days af-
boars for as long as 43 days following exposure. Using a ter experimental inoculation was reported (Wills et al.
reverse transcription–polymerase chain reaction, viral 1997c). Overall, these data have provided definitive evi-
RNA was detected in the semen of experimentally infect- dence that PRRSV persists in swine for an extended peri-
208 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

od of time. Exactly how long animals remain potentially species, mallard ducks in particular, are susceptible to
infectious is not known. PRRSV (Zimmerman et al. 1997b). Mallards exposed to
Although PRRSV is highly infectious (i.e., has a low PRRSV in drinking water shed virus in feces for an ex-
minimum infectious dose), it is not highly contagious. tended period of time. At the termination of one experi-
Transmission usually occurs by close contact between ment, virus was recovered from fecal samples collected
animals. This finding is in sharp contrast to earlier from 8 of 20 birds 39 days after exposure. In a second ex-
thought. It was once suggested that aerosolized virus was periment, mallard-to-mallard transmission was demon-
the primary route of transmission of PRRSV, with strated by infecting ducks with feces from ducks
aerosol transmission postulated to occur routinely and shedding PRRSV. And finally, pigs were shown to be sus-
over distances of up to 20 kilometers. Since PRRSV is ceptible to mallard-derived virus. That is, pigs
present in the upper respiratory tract and oropharyngeal intranasally exposed to PRRSV isolated from mallard
area of infected pigs for an extended period of time, this feces became viremic, seroconverted by ELISA, and
was not an unreasonable hypothesis. However, Wills et transmitted the virus to sentinel pigs. Although it is
al. (1997a) found that transmission among penmates oc- unlikely that mallards are active participants in the
curred much more readily than transmission across a transmission of PRRSV to and among swine and swine
space of 40 inches or less. In further testimony to the herds, they could theoretically serve as a source of new
limited contagiousness of the virus, Torremorell et al. virus types, as in the case of the influenza viruses. At this
(1997) documented the transmission of PRRSV from juncture it would seem to be more important to further
acutely infected pigs to only 2 of 8 contact controls un- investigate the susceptibility of other birds and animals
der experimental conditions. These studies highlight our known to frequent swine facilities and potentially serve
limited understanding of the process of transmission it- as carriers.
self.
Given that the exact mechanism of transmission be- Transmission within Herds
tween pigs is uncertain, studies of LDV in mice provide Once infected, PRRSV tends to circulate within a herd in-
clues to the importance of certain types of interactions definitely. Rarely, spontaneous elimination of PRRSV
between animals. For LDV, it was observed that certain from commercial herds has been reported by producers
laboratory strains of young male mice rarely became in- or veterinary practitioners, but the circumstances under
fected when placed in the same cage as infected mice, which this occurs are not well defined. At this point, the
compared to approximately 50% infection rates when mechanisms that make endemicity possible are not en-
General Purpose (GP) Swiss male mice were placed in the tirely clear. However, the key components appear to be
same conditions (Notkins and Shochat 1963; Notkins et persistent PRRSV infection in clinically normal carrier
al. 1964). However, it was noted that GP Swiss mice were animals and the continual introduction of susceptible
more prone to fighting than were the other strains of animals, either through birth or purchase (Wills et al.
mice, and if the incisors of both inoculated and exposed 1997c).
6-month-old GP Swiss male mice were removed, trans- Typically, the virus is perpetuated by a cycle of trans-
mission rarely occurred. In contrast, transmission always mission from dams to pigs either in utero or postpartum,
occurred if the incisors of the inoculated mice were or by commingling naive animals with infected animals
present, regardless of whether or not the incisors of the in later stages of production. In neonatal pigs, maternal
exposed mice were removed. If the incisors of only the antibodies may provide some immunologic resistance to
inoculated mice were removed, transmission still oc- infection, but the protection is not absolute and is of
curred a majority of the time. Overall, the results sug- short duration. Under conditions in which susceptible
gested that, during the course of fighting, infectious and infected pigs are mixed (e.g., at weaning), a large
mice could transmit LDV by the injection of saliva or proportion of the population may quickly become in-
susceptible mice could contract the infection by the in- fected. Dee and Joo (1994a), for example, reported
gestion of blood or tissue from infectious mice. 80–100% of the pigs in 3 swine herds were infected by
As in the case of LDV, the social dominance behavior 8–9 weeks of age, and Maes (1997) found 96% of market
which occurs among pigs in direct contact is ideal for the hogs sampled from 50 herds to be seropositive.
transmission of PRRSV. The fighting which invariably However, it has become increasingly clear that the
occurs when pigs are mixed, and the low minimum in- pattern of infection in PRRSV-endemic herds often devi-
fectious dose and the persistence of virus in the oropha- ates from this description of rapid, uniform spread. And
ryngeal region for up to several months after infection, even within infected herds, marked differences in infec-
make the movement of pigs both within and between tion rates between groups, pens, or rooms of pigs are of-
farms the ideal mode of transmission. ten observed in the field. Houben et al. (1995b) found
Nonporcine hosts exist, but their role in the epidemi- transmission to vary even within litters, with some litter-
ology of PRRSV is uncertain. Although rodents are not mates seroconverting as early as 6–8 weeks and other in-
susceptible to PRRSV (Hooper et al. 1994), some avian dividuals as late as 10–12 weeks of age, and described a
CHAPTER 18 PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME Benfield et al. 209

slow spreading pattern. In some cases, litters of pigs CLINICAL SIGNS


reached 12 weeks of age, the end of the monitoring peri-
od, still free of PRRSV. Thus, it is possible for animals in Descriptions of the clinical signs of PRRS in swine herds
endemically infected herds to escape infection for an ex- are similar in North America (Keffaber 1989; Moore
tended period of time, as when Le Potier et al. (1997) ob- 1990; Bilodeau et al. 1991; Loula 1991; Sanford 1992)
served seroconversion in young sows on farms using in- and Europe (de Jong et al. 1991; Leyk 1991; Wensvoort et
herd gilt replacements. al. 1991; Anon. 1992; Busse et al. 1992; Gordon 1992;
Hopper et al. 1992; White 1992a, b). Clinical signs of
Transmission between Herds PRRS are extremely variable and influenced by strain of
Herd-to-herd transmission occurs but is generally recog- virus (Halbur et al. 1996b), immune status of the herd
nized too long after the fact to accurately determine the (Keffaber 1989; Wensvoort 1993), and management fac-
origin of the virus. Known sources of herd-to-herd trans- tors (Blaha 1992; White 1992a). Clinical disease in a herd
mission include infected carrier animals and the use of is primarily the consequence of acute viremia in individ-
PRRSV-contaminated semen. Potential but unproved uals (Pol et al. 1991; Terpstra et al. 1991; Collins et al.
sources include contaminated biologics or needles, wa- 1992) and transplacental transmission of virus from
terborne virus, airborne virus, nonporcine hosts, arthro- viremic dams to their fetuses (Terpstra et al. 1991), which
pods, and fomites. In the course of the PRRSV control occurs most efficiently in the third trimester of pregnan-
program in France, Le Potier et al. (1997) reported that cy (Christianson et al. 1993; Mengeling et al. 1994).
the infection was introduced to 56% of herds through in- Strains of PRRSV vary remarkably in virulence (Hal-
fected pigs, 20% through infected semen, 21% through bur et al. 1996b). Low-virulence strains can cause com-
fomites, and 3% through unidentified sources. pletely subclinical epidemic or endemic infections of
Area spread, that is, herd-to-herd transmission in the herds (Morrison et al. 1992), whereas highly virulent
absence of any apparent animal or human contact, is al- strains can cause severe clinical illness. In clinical epi-
so known to occur but is generally more restricted in dis- demics occurring in immunologically naive herds, all
tance than once believed. This may be attributed to the ages are susceptible, and acute infection is most consis-
fact that, until the existence of subclinical infections was tently evidenced by inappetence, fever, and dyspnea. For
recognized, the distance between clinical outbreaks was the next 1–4 months, many sows farrow prematurely,
used to measure virus travel. More recently, Le Potier et usually after 100 days of gestation. Fewer farrow after a
al. (1997) found that 45% of herds suspected to have be- prolonged gestation of 115–118 days. Affected litters
come infected through area spread were located within that are born early, full term, or late are composed of any
500 meters (0.3 miles) of the postulated source herd and or all of the following: stillborn pigs, mummified fetuses,
only 2% were 1 kilometer from the initial outbreak. late-term dead fetuses, variably sized weak-born pigs,
Area spread is most commonly attributed to aerosols and variably sized, apparently normal pigs. Preweaning
of infectious PRRSV traveling downwind from infected mortality is high. In endemically infected herds, pre-
herds. Of course, aerosol transmission is known to occur dominant signs are in nursery-grower pigs and include
in the case of many viral infections. Foot-and-mouth dis- reduced thrift, dyspnea, exacerbation of other endemic
ease virus (FMDV), in particular, is understood in such diseases, and increased mortality. Less commonly, there
detail that the behavior of aerosolized FMDV can be pre- is sporadic reproductive failure in subpopulations of sus-
dicted with a high degree of accuracy using mathemati- ceptible gilts.
cal models. Transmission of PRRSV in aerosols, howev-
er, has been difficult to achieve experimentally. Epidemic Infection of Herds
Torremorell et al. (1997) were able to transmit PRRSV In clinically evident epidemic infections in immunologi-
from a group of acutely infected pigs over a distance of cally naive herds, the first phase of PRRS lasts approxi-
just 1 meter in only one of two attempts. Wills et al. mately 2 weeks and is an acute systemic illness in 5–75%
(1997a) described similar results across a comparable of animals that is caused by initial viremia and that is
distance, and Robertson (1992) reported no success in most often characterized by anorexia and lethargy. It be-
isolating virus from the airspace in which acutely infect- gins in one or more stages of production and quickly
ed pigs were housed. Specific information is needed at spreads within 3–7 days to all stages of production. Indi-
present. In particular, we need to know the quantity of viduals are anorectic for 1–5 days, and the spread of the
virus shed by acutely infected animals in respired air and disease through a segregated group of pigs usually re-
the stability of aerosolized PRRSV under various condi- quires 7–10 days, giving rise to the descriptive term
tions of humidity and temperature. Until such data are “rolling inappetence.”
available, it will not be possible to fully evaluate the im- In addition to anorexia and lethargy in acutely ill an-
portance of aerosols in transmission. Even so, the avail- imals, there are also less consistent clinical signs that
able information makes it clear that aerosol transmission may be seen in animals of all ages. Frequently, ill animals
is much more limited than once thought. are lymphopenic and/or pyretic with rectal tempera-
210 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

tures from 39 to 41˚C (102 to 106˚F) and/or hyperpneic or completely mummified fetuses. Typically, pigs born
and dyspneic (“thumping”). A few (1–2%) exhibit tran- dead compose 0–100% of each affected litter and 7–35%
sient “blotchy” cutaneous hyperemia or cyanosis of ex- of the total pigs born in a farrowing group. As this 1- to
tremities, which is most visible on the ears, snouts, mam- 4-month phase progresses, the majority of abnormal
mary glands, and vulvas. In addition to these clinical pigs in each litter typically shifts from stillborn pigs and
features seen in all stages of production, there are fea- large partially mummified pigs to smaller more com-
tures that are unique to the different stages of produc- pletely mummified pigs to small weak-born pigs to pigs
tion. of normal size and vigor (Keffaber 1989; Loula 1991;
White 1992a). In some herds, the majority of abnormal
SOWS. During the phase of acute illness, abortions pigs are born alive, premature, weak, and small, but few
may occur in 1–3% of sows that are from 21 to 109 days are born dead (Gordon 1992).
of gestation (Hopper et al. 1992; White 1992a). Some re- Among sows farrowing abnormal litters, periparturi-
ports emphasize abortions in the last one-half to one- ent mortality may be 1–2% (Keffaber 1989; de Jong et al.
third of gestation; however, in these reports early loss of 1991). Typically, when these sows are bred again, they ex-
pregnancy was alternatively reported as an immediate hibit delayed returns to estrus and suffer low conception
increase in the number of sows with irregular returns to rates. These low conception rates, combined with the
heat as well as a subsequent reduction in farrowing rate previously mentioned increases in overt abortions, irreg-
associated with an increase in the number of sows that ular returns to estrus, and nonpregnant sows, result in a
were not pregnant (Keffaber 1989; Loula 1991). Also, in depression in farrowing rate for a complete reproductive
some herds, there is a 1–4% mortality in acutely ill sows cycle.
(Loula 1991) that is sometimes associated with lesions of
pulmonary edema or cystitis/nephritis (Hopper et al. BOARS. During the first phase of acute illness, in ad-
1992). Recently in the United States, a very virulent form dition to anorexia, lethargy, and respiratory clinical
of PRRS was described (sometimes referred to as “sow signs, boars may lack libido and have variable reduction
abortion and mortality syndrome”) with abortion rates in semen quality (de Jong et al. 1991; Feitsma et al. 1992;
of 10–50% and mortality rates of up to 10% in sows. Cen- Prieto et al. 1994). Changes in sperm occur 2–10 weeks
tral nervous system clinical signs such as ataxia, circling, after infection with virus and include reduced motility
and falling to one side were sometimes seen as complica- and acrosomal defects. Although transmission of
tions of abortion (Epperson and Holler 1997; Halbur PRRSV through semen occurs and may be a significant
and Bush 1997). Other inconsistently reported signs in portal of PRRSV entry into susceptible herds (Yaeger et
acutely ill sows include agalactia (Hopper et al. 1992), in- al. 1993; Swenson et al. 1994a), the impact of PRRS
coordination (de Jong et al. 1991), and a dramatic exac- viremia in boars on conception is not clear (Yaeger et al.
erbation of endemic diseases such as sarcoptic mange, 1993; Swenson et al. 1994a; Lager et al. 1996; Prieto et al.
atrophic rhinitis, or cystitis/pyelonephritis (White 1996a, c).
1992a).
Beginning approximately 1 week after the onset of SUCKLING PIGS. During the 1- to 4-month phase of
acute illness, a second phase of the disease begins. It is a late-term reproductive failure, high preweaning mortali-
consequence of transplacental transmission of the virus ty (up to 60%) occurs in both pigs born weak and those
and is characterized by late-term reproductive failure. It born with normal vigor. Nearly all of the premature
occurs in sows without previous clinical signs as well as weak pigs die within hours of birth. In the rest, mortali-
in those clinically affected during the first phase of the ty is greatest within the first week after birth but contin-
disease. The second phase initially overlaps the first, but ues to weaning and beyond. A variety of clinical signs are
it typically lasts much longer, usually for 1–4 months. reported in suckling pigs with PRRS. Most consistently
During the second phase, 5–80% of sows may have re- reported are listlessness, emaciation/starvation, splay-
productive failure at any time between 100 and 118 days legged posture, hyperpnea, dyspnea (“thumping”), and
of gestation. Most affected sows farrow prematurely, but chemosis. The chemosis that occurs in some pigs is se-
they may also farrow full term or late term, or they may vere, causing a characteristic swelling of the eyelids and
abort. For example, Gordon (1992) described a herd dur- ocular conjunctiva that some consider a nearly “diagnos-
ing an outbreak of PRRS in which the proportion of tic” lesion of PRRS when observed in pigs under 3 weeks
sows farrowing prematurely from days 110 to 114 of ges- of age (White 1992a). Watery diarrhea that is nonre-
tation rose from a normal of 20% to a peak of 76% by sponsive to antibiotic therapy is consistent in the United
week 4 of the outbreak. Affected litters contain variable Kingdom (Gordon 1992; Hopper et al. 1992; White
numbers of normal pigs, weak small pigs, and dead pigs 1992a) and less consistent elsewhere (Keffaber 1989;
that are fresh stillborn (intrapartum deaths), autolytic Leyk 1991). Also reported in some herds in piglets are
stillborn (prepartum deaths), and partially mummified tremors or paddling (Keffaber 1989; Loula 1991), slight
CHAPTER 18 PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME Benfield et al. 211

doming of foreheads (Gordon 1992), and thrombocy- ed with virus can shed virus to other pigs within their
topenia with consequent hemorrhage from navels, sites age-segregated cohort as these pigs become increasingly
of injections, and tails following docking, as well as ane- susceptible due to decline of maternal immunity. The
mia (Hopper et al. 1992; White 1992a). An increased clinical disease in these subpopulations of pigs is the
number of secondary bacterial infections, such as pol- same as that seen in weaned pigs in a generalized whole-
yarthritis, also contributes to morbidity and mortality. herd epidemic as described above.
Clinical signs in the breeding herd in endemically in-
WEANLING AND GROWER PIGS. Acute PRRSV in- fected herds are usually limited to susceptible gilts or re-
fection in weaned pigs in the nursery or grower pigs in placement boars that are exposed to PRRSV after intro-
finishing barns is characterized most consistently by duction into the herd (Grosse-Beilage and
anorexia, lethargy, hyperpnea, dyspnea (“thumping”), Grosse-Beilage 1992; Dee and Joo 1994b; Dee et al.
and cutaneous hyperemia as described above. Coughing 1996). Acute clinical disease in gilts or boars is as de-
is not a consistent clinical feature of uncomplicated scribed above. The reproductive manifestation of en-
PRRS in weaned pigs. Consistently reported in PRRSV- demic PRRS depends on the number of gilts infected and
infected weaned pigs is an “ill-thrift” that is characterized the stage of their reproductive cycle when infected, both
by rough hair coats and a variable reduction in average of which may vary widely (Torrison et al. 1994). If scat-
daily gain and feed efficiency which causes a large varia- tered gilts are infected in a somewhat continuous and
tion in size of pigs in age-matched groups (Moore 1990; random manner, then there may be scattered abortions,
White 1992b). Frequently associated with PRRS in nurs- irregular returns to estrus, nonpregnant gilts, and late-
ery-grower pigs is a higher than usual incidence of a term reproductive failure with abnormal litters that may
number of endemic diseases, which results in elevated only be recognized if records are evaluated on a parity-
mortality of up to 12–20% (Moore 1990; Loula 1991; Bla- specific basis (White 1992b). Alternatively, gilts may es-
ha 1992; Keffaber et al. 1992; White 1992a; Stevenson et cape exposure to PRRSV until there is a significant sub-
al. 1993). These endemic diseases include septicemic sal- population of susceptible gilts in various stages of
monellosis, Glasser’s disease, streptococcal meningitis, gestation. In this situation, endemic PRRS in the breed-
septicemia and polyarthritis, exudative dermatitis, acti- ing herd manifests as periodic mini-outbreaks of PRRS
nobacillary pleuropneumonia, mycoplasmal pneumo- in gilts and possibly first-parity sows. The clinical signs
nia, bacterial bronchopneumonia, atrophic rhinitis, in affected gilts/sows in these parity-specific mini-out-
postweaning colibacillosis, proliferative enteritis, swine breaks are identical to those in a whole-herd outbreak
dysentery, spirochetal colitis, and sarcoptic mange. This but on a smaller scale (Dee and Joo 1994b).
PRRSV-associated enhancement of endemic disease is
most significant in herds with lower health status and/or Conclusion and Caveat
poor management (Blaha 1992). However, most at- This description of the clinical signs of PRRS is the dis-
tempts to experimentally reproduce PRRSV enhance- tillation of observations reported by swine clinicians
ment of bacterial disease have been unsuccessful (Galina from around the world and presents the most consistent
et al. 1994; Carvalho et al. 1995; Pol et al. 1995; Solano et clinical features of a “classical” PRRS outbreak or en-
al. 1995; Van Alstine et al. 1996). demic herd infection and attempts to highlight signifi-
cant or unique clinical features of the disease that may be
Endemic Infection of Herds useful diagnostically. However, probably the only com-
Swine herds often remain infected with PRRSV for years pletely consistent clinical feature of PRRSV infection in
(Stevenson et al. 1992; Stevenson et al. 1993; Terpstra et pigs is that there is no single consistent feature of PRRSV
al. 1992) through various mechanisms, including pro- infection in pigs. For nearly every clinical sign described,
longed or persistent viremia and viral shedding in indi- there are exceptions. On a broader scope, there are ex-
vidual pigs (Albina et al. 1994a, b; Wills et al. 1995b; ceptions involving entire phases of production. For ex-
Benfield et al. 1997), continuous introduction of suscep- ample, there are reports of outbreaks in single-site farms
tible breeding stock (Dee and Joo 1994b), and replication where there was severe clinical disease in the grower pigs
of virus in susceptible weaned pigs (Stevenson et al. and no clinical signs in the breeding herd (Kuwahara et
1994). The clinical signs of endemic PRRS infection in al. 1994). Probably more than for any other disease in
herds are most often obvious in weaned pigs in the nurs- swine, clinicians need a thorough understanding of the
eries or early in the finishing barns when a limited out- pathogenesis and epidemiology of PRRSV in order to ac-
break occurs in a susceptible subpopulation of pigs (Kef- curately recognize the various clinical presentations of
faber et al. 1992; Stevenson et al. 1993). This can occur in PRRS in individuals and populations. Otherwise, one
two general nonexclusive ways. Infected pigs in continu- could erroneously “see” PRRS in nearly every set of clin-
ous-flow facilities can shed virus to negative younger sus- ical signs or fail to recognize PRRS in its many clinical
ceptible pigs. Alternatively, pigs born persistently infect- variations.
212 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

PATHOGENESIS PRRSV was first isolated in PAMs (Wensvoort et al.


1991) and has also been reported to replicate in mono-
The pathogenesis of PRRSV infection is based on PRRSV cytes (Voicu et al. 1994), in microglial cells (Molitor et al.
infection and replication within cells of the mono- 1996), in two MA-104 (embryonic monkey kidney cell)
cyte/macrophage lineage (Wensvoort et al. 1991; Voicu clones known as CL-2621 (Benfield et al. 1992) and
et al. 1994; Rossow et al. 1995; Rossow et al. 1996a; Moli- MARC-145 (Kim et al. 1993), and in CRL-11171 cells (Hal-
tor et al. 1996). Exposure of a mucosal surface to PRRSV bur et al. 1995). PRRSV replication can be selective be-
(Fig. 18.2) results in viremia within 12 hours after infec- tween these cell types (Bautista et al. 1993). PAMs from a
tion (Rossow et al. 1995), and PRRSV antigen is identi- pig less than 6 weeks old are most susceptible to PRRSV
fied in macrophages of the nasal mucosa, lung, and ton- infection, and within a PAM population, PRRSV has a
sil (Rossow 1996). Sows can be infected with PRRSV by preference for replication within immature macrophages
exposure of the nasal mucosa or uterine endometrium (Choi et al. 1994; Mengeling et al. 1995). The predilec-
(Christianson et al. 1992; Yaeger et al. 1993; Swenson et tion of PRRSV for PAMs from immature swine corre-
al. 1994c; Lager et al. 1996). PRRSV is also transmitted sponds with the severe disease most commonly de-
by inoculation (Mengeling et al. 1994) and presumably scribed in young pigs (Goyal 1993). Activation or
by bite wounds. Macrophages within a mucosal surface maturation of monocytes or PAMs treated with
are the site of primary PRRSV replication, with distribu- macrophage-colony stimulating factor may modulate
tion to regional lymphoid tissue and subsequent sys- surface marker expression and susceptibility to PRRSV
temic distribution to macrophages in multiple tissues replication (Choi et al. 1994). PRRSV antigen has been
and/or to monocytes (Rossow et al. 1995; Rossow et al. identified in macrophages of multiple tissues, mono-
1996a). cytes, endothelial cells, smooth muscle cells, and fibro-

18.2. Pathogenesis of PRRSV infection. (Courtesy of K. D. Rossow, South Dakota State University.)
CHAPTER 18 PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME Benfield et al. 213

blasts by in situ hybridization and immunohistochem- (Hopper et al. 1992; Goyal 1993; Zeman et al. 1993;
istry (Magar et al. 1993; Halbur et al. 1995; Rossow et al. Stevenson et al. 1994). Although coinfection with single
1996a; Sur et al. 1996). Replication of PRRSV in endoge- or multiple bacteria (Goyal 1993; Rossow 1996), and less
nous porcine cells has only been demonstrated in PAMs, frequently with other viruses (Larochelle et al. 1994), is a
monocytes, and microglial cells (Wensvoort et al. 1991; severe complicating factor of PRRSV infection, an in-
Voicu et al. 1994; Molitor et al. 1996). Resident crease in susceptibility to bacterial superinfections has
macrophages within a variety of tissues should be con- only been experimentally demonstrated once (Galina et
sidered susceptible to PRRSV infection (Rossow et al. al. 1994). Since the PRRSV-bacterial interaction is not
1995; Rossow et al. 1996a). Because macrophages are easily reproduced experimentally, there may be multiple
found in all tissues, there is no apparent limitation to the factors involved in susceptibility to secondary bacterial
types of tissue that can be infected by PRRSV. However, infections, including the PRRSV strain, swine genetic
macrophages residing in different tissues are constitu- composition, recruitment of susceptible macrophages
tively different because of their varied local environ- by bacterial coinfections, and environmental factors.
ment, and thus their permissiveness and the productivi- PRRSV infection in finishing pigs, unbred gilts or
ty of PRRSV replication vary (Rossow et al. 1995; Rossow sows, and boars is commonly characterized by a tran-
et al. 1996a). sient fever and inappetence (Goyal 1993; Christopher-
Microscopic lesions develop subsequent to PRRSV Hennings et al. 1995a; Christopher-Hennings et al. 1996;
infection of macrophages in multiple tissues, and the Rossow 1996). Mortality associated with PRRSV infec-
type and degree of inflammation depend on the virus tion of adult swine has been reported and may reach 5%.
strain, pig age, infection with other bacterial or viral Some PRRSV-infected boars demonstrate a loss of li-
agents, host genetic composition, and environmental bido, while most appear clinically unaffected (Feitsma et
stressors (Halbur et al. 1995; Rossow 1996). Interstitial al. 1992; Yaeger et al. 1993; Christopher-Hennings et al.
pneumonia, encephalitis, myocarditis, lymphadenopa- 1995a; Prieto et al. 1996b). PRRSV is identified in semen
thy, and arteritis (Rossow 1996) characterize microscop- of intact and vasectomized boars, indicating a similar
ic lesions resulting from PRRSV infection. Lesions result- route of shedding independent of testicular involvement
ing from PRRSV infection occur in other tissues less (Christopher-Hennings et al. 1996). PRRSV field and
frequently (Rossow 1996). Placental lesions have been vaccine strains can be shed in semen of infected or vacci-
observed only by electron microscopy and were charac- nated boars (Christopher-Hennings et al. 1995a; Molitor
terized by placental-uterus microseparation with epithe- and Shin 1995; Christopher-Hennings et al. 1996;
lial cell necrosis and desquamation (Stockhofe-Zur- Christopher-Hennings et al. 1997). The source of PRRSV
wieden et al. 1993). The severity of systemic disease will in the semen has not been identified. Changes in sperm
determine the clinical presentation of PRRSV infection. morphology and function have been described subse-
The influence of the immune response on the devel- quent to PRRSV infection (Christopher-Hennings et al.
opment and expression of PRRSV infection has not been 1997): a decrease in sperm motility, a decrease in num-
determined; however, anti-PRRSV antibody enhances ber of sperm with normal acrosomes, and an increase in
the phagocytosis of PRRSV by macrophages (antibody- sperm with distal cytoplasmic droplets. However, there
dependent enhancement [ADE]), resulting in increased also are reports describing no changes in sperm after
virus replication (Christianson et al. 1993; Yoon et al. PRRSV infection (Prieto et al. 1996b). PRRSV can be
1996). The significance of ADE in natural PRRSV infec- identified in semen prior to seroconversion and after the
tion has not been determined. end of viremia (Christopher-Hennings et al. 1995a).
PRRSV infection results in clinical or subclinical dis- Infection of sows in their third trimester results in
ease with resolution or persistent infection (Rossow et al. the typical clinical presentation of abortion or prema-
1995; Benfield et al. 1996). The clinical presentation of ture farrowing (Christianson et al. 1992; Mengeling et al.
disease is age dependent. PRRSV infection in neonatal 1994). Fetuses in all stages of gestation can support repli-
pigs is characterized by dyspnea, central nervous signs, cation of PRRSV when infected intraamniotically (Chris-
and mortality rates up to 100% (Collins et al. 1992; Goy- tianson et al. 1993; Lager and Mengeling 1995). Howev-
al 1993). er, fetal PRRSV infection is less likely to occur after
PRRSV infection in weaned pigs is characterized by intranasal infection of midgestation sows (Christianson
fever, pneumonia, lethargy, failure to thrive, and a et al. 1993; Mengeling et al. 1994). Postcoital intrauter-
marked increase in mortality from single to multiple con- ine inoculation of PRRSV at or near the time of concep-
current bacterial infections (Rossow 1996). Concurrent tion has little or no effect on reproductive performance,
bacterial infections drive the marked increases in mor- and 4- to 16-cell-stage porcine embryos infected with
tality associated with PRRSV infections in immature PRRSV develop normally and do not support PRRSV
swine, and the increased mortality subsequent to PRRSV replication (Lager et al. 1996; Prieto et al. 1996c). There
infection is clinically obvious from observations of in- are anecdotal reports of PRRSV-induced abortion in
creases in postweaning mortality from 1–2% to 10–15% first- and second-trimester sows. The pathogenesis of fe-
when all other factors apparently remain unchanged tal PRRSV infection has not been determined. A conse-
214 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

quence of late-gestation PRRSV infection is persistent productive tracts of dams and in fetuses are not as well
virus infection of surviving fetuses (Benfield et al. 1996). characterized and are less consistently observed.
Persistence resulting from RNA-virus infection is de-
fined as the continued presence of virus within a host for Neonatal Pigs
extended periods of time after acute infection (Ahmed et Gross and microscopic lesions in neonatal pigs often are
al. 1996). Evidence for persistence of PRRSV infection is quite remarkable. PRRSV-infected lungs are mottled tan
limited but is based on four different reports drawing and red and fail to collapse. It is often difficult to clearly
similar conclusions: (1) PRRSV RNA has been demon- demarcate affected and unaffected tissue. Lesions are
strated by the polymerase chain reaction (PCR) in se- most commonly observed in the cranioventral portion,
men, but not serum, collected from boars 92 days after and the extent of lung affected macroscopically may vary
they were exposed experimentally to PRRSV when a con- from 0% to 100% depending in part on the virulence of
current serum sample was negative for PRRSV RNA the strain and the time postinfection (Halbur et al. 1995;
(Christopher-Hennings et al. 1995a). (2) PRRSV naive, Halbur et al. 1996a, b). Lymph nodes are moderately to
specific-pathogen-free pigs were commingled with severely enlarged and tan in color (Halbur et al. 1995;
PRRSV seronegative pigs that had been exposed to Rossow et al. 1994; Rossow et al. 1995). Lymph nodes in
PRRSV 22 weeks previously (Albina et al. 1994a, b). The the cervical, cranial thoracic, and inguinal regions are the
PRRSV-exposed pigs were treated with exogenous gluco- most obvious at necropsy. Fluid-filled spaces in the
corticoids prior to commingling (Albina et al. 1994a, b). lymph nodes are inconsistently observed.
The PRRSV-naive pigs developed anti-PRRSV antibod- Microscopic examination reveals moderate-to-severe
ies, had clinical signs of PRRSV infection, and became multifocal interstitial pneumonia characterized by three
viremic within 1–2 weeks of exposure to the PRRSV car- hallmark changes: (1) alveolar septal infiltration by a
rier pigs (Albina et al. 1994a, b). (3) PRRSV has been iso- mixed population of mononuclear cells; (2) type 2 pneu-
lated from oropharyngeal scrapings up to 157 days mocyte hypertrophy and hyperplasia; and (3) marked ac-
postinoculation, 134 days after the last isolation of cumulation of mixed inflammatory and necrotic alveolar
PRRSV from serum (Wills et al. 1995b). (4) Sera of pigs exudate. Lymph nodes are reactive, with marked follicu-
exposed to PRRSV at 85–90 days of gestation contained lar hyperplasia, foci of follicular necrosis, increased
PRRSV RNA (PCR technique) at 110 days postfarrowing numbers of tingible-body macrophages, and karyorrhec-
(Benfield et al. 1996). Sentinel pigs cohoused with the tic debris within follicles (Halbur et al. 1995; Halbur et
PRRSV persistently infected pigs (98 days postfarrow- al. 1996a; Rossow et al. 1994; Rossow et al. 1995). The
ing) developed anti-PRRSV antibodies 14 days later follicular mitotic index is increased, and the paracortex is
(Benfield et al. 1996). expanded with mixed inflammatory cells. Other lesions
These experiments indicate that PRRSV persists in such as lymphoplasmacytic rhinitis, encephalitis, and
infected swine in a viable state that may not stimulate an- myocarditis are also observed in neonatal pigs.
tibody production and that PRRSV can be recovered
long after initial exposure from serum, the oropharyn- Nursery Pigs
geal cavity, and semen. Persistently infected pigs may be Lungs from affected pigs fail to collapse and have a vari-
an important factor for PRRSV survival and transmis- able amount of tan and red mottling (Fig. 18.3). Practi-
sion within a herd. Differentiation of PRRSV strains by tioners sometimes describe the gross lung lesions as
their ability to cause persistent infections or acute, re- “thymus-like” or “liver-like.” Even if gross lung lesions
solved infections has not been demonstrated, although are not apparent, microscopic examination usually re-
this difference has been verified for another arterivirus veals interstitial pneumonia similar to that seen in
(Gravell et al. 1986). neonatal pigs but often less severe and more multifocal.
The most consistent gross lesion, which has become the
LESIONS hallmark of PRRSV infection in nursery pigs, is the
markedly enlarged tan lymph nodes (Fig. 18.4). Other
PRRSV induces a multisystemic infection in pigs result- less consistent gross lesions include chemosis and in-
ing in the potential for all tissues to become infected with creased amounts of clear fluid in the abdomen, thoracic
virus. However, gross lesions are usually observed in on- cavity, and pericardial space.
ly a few organ systems (e.g., respiratory and lymphoid). Microscopic examination of brain, tonsil, lymph
Gross and microscopic lesions characteristic of PRRSV nodes, and heart is of diagnostic value. The majority of
infection are most remarkable in neonatal and nursery PRRSV-infected pigs have some degree of lymphohistio-
pigs. In older pigs, lesions attributed to PRRSV infection cytic meningoencephalitis and choroiditis characterized
are similar but much less remarkable. Under field condi- by perivascular cuffing, vasculitis, gliosis, and glial nod-
tions, most PRRSV-infected pigs are coinfected with one ule formation (Halbur et al. 1995; Rossow et al. 1994;
or more pathogens, which complicates the diagnosis of Rossow et al. 1995). Severity varies from very mild to se-
PRRS based on lesions. PRRSV-induced lesions in the re- vere enough to resemble pseudorabies virus infection.
CHAPTER 18 PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME Benfield et al. 215

18.3. Lateral view of a lung


from a pig infected 10 days
previously with a virulent strain of
PRRSV. The lung has failed to
collapse, and nearly the entire
lung is firm and mottled tan in
color. (Courtesy of P. G. Halbur,
Iowa State University.)

18.4. Ventral view of enlarged


lymph nodes (arrows) located at
the thoracic inlet in a pig infected
28 days previously with a virulent
strain of PRRSV. T = trachea,
H = heart. (Courtesy of W. L.
Mengeling and K. M. Lager,
National Animal Disease Center.)

Lymphoid tissues exhibit lymphoid hyperplasia and fo- complex” is commonly used to describe a syndrome that
cal follicular necrosis. Lymphoplasmacytic and histiocyt- has become more problematic since the mid-1990s.
ic myocarditis is often detected beyond 7 days postinfec- PRRSV is thought to be a major primary pathogen in the
tion. Myocardial inflammation is often most severe in complex. Mycoplasma hyopneumoniae, Pasteurella multo-
the subendocardial and perivascular areas of the my- cida, and swine influenza virus are common coinfectors,
ocardium. Mild-to-severe nonsuppurative rhinitis with resulting in dark red and tan lungs and cranioventral
epithelial metaplasia has also been attributed to PRRSV consolidation of 30–70% of the lung in grower-finisher
infection (Halbur et al. 1995). pigs. Coinfection with mycoplasma and bacteria results
in the presence of exudate in airways and clearer lines of
Grower-Finisher Pigs demarcation between affected and unaffected lung tis-
Gross lesions in grower-finisher pigs attributed to sue.
PRRSV are similar to, but less remarkable than, those in Microscopic examination reveals typical PRRSV-in-
nursery age pigs. Lymphadenopathy is consistently ob- duced interstitial pneumonia underlying the endemic
served. Lung lesions are often complicated by mixed in- pneumonia or bacterial bronchopneumonia. Encephali-
fections (Zeman et al. 1993). “Porcine respiratory disease tis and myocarditis lesions are similar to, but usually less
216 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

common and less severe than, those observed in younger lar hemorrhage (Lager and Halbur 1996). Interstitial
pigs. pneumonia characterized by mild septal infiltration by
mononuclear cells, type 2 pneumocyte hypertrophy and
Boars hyperplasia, and increased amounts of mixed inflamma-
The presence of PRRSV in boar semen and the transmis- tory and necrotic alveolar exudate is observed in some
sion of PRRSV by boar semen have been demonstrated cases (Lager and Halbur 1996; Rossow et al. 1996b).
(Swenson et al. 1994b; Christopher-Hennings et al. Necrotizing and lymphoplasmacytic pulmonary arteritis
1995b). Consistent or remarkable lesions in the adult is also of diagnostic value when observed (Lager and Hal-
boar attributable to PRRSV infection have not been ob- bur 1996; Rossow et al. 1996b). Bronchiolar bud necrosis
served following experimental infection (Swenson et al. and pulmonary hemorrhage have also been reported in
1994b). PRRSV has been isolated from the bulbourethral fetuses experimentally infected between 45 and 49 days
gland of a boar euthanized at 104 days after inoculation, of gestation (Lager and Ackermann 1994). Lymphoplas-
suggesting that infectious PRRSV may be shed from ac- macytic myocarditis, myocardial fibrosis, and lympho-
cessory sex glands (Christopher-Hennings 1995a). De- plasmacytic perivascular encephalitis are less frequently
tailed chronological and morphological studies to char- observed (Rossow et al. 1996b).
acterize PRRSV-induced lesions and antigen distribution
in the adult boar have not been reported to date. DIAGNOSIS

Sows A diagnosis of PRRS is based on subjective (history, clin-


Natural epidemics and experimental inoculation studies ical signs, gross and microscopic lesions) and objective
of PRRSV-induced reproductive failure are characterized (production record analysis, serology, virus detection)
primarily by late-term abortions, premature farrowings, factors. PRRS should be considered when there are clini-
and increased numbers of autolyzed or mummified fe- cal signs of respiratory disease occurring at any stage of
tuses, stillborn pigs, and weak-born pigs. There general- production, when reproductive failure occurs, and when
ly are no pathognomonic gross or microscopic lesions in herd performance is suboptimal. Mild or subclinical dis-
the pregnant animal; however, only a limited number of ease is common, so absence of clinical signs does not en-
naturally or experimentally infected sows have been ex- sure a PRRSV-free herd. The herd history, clinical signs,
amined histologically in detail. Microscopic examination and lesions associated with PRRS have been previously
of the uterus of uncomplicated PRRSV-induced abor- discussed and will not be repeated here.
tion may reveal mild-to-moderate lymphoplasmacytic
endometritis and myometritis (Christianson et al. 1993; Record Analysis
Lager and Halbur 1996). Endometrial edema is also com- Herd production records are becoming a useful adjunct
mon. Lymphoplasmacytic placentitis is less consistently to PRRS investigations. A review of production records
observed (Stockhofe-Zurwieden et al. 1993). Mild lym- in clinically active PRRS herds often reveals evidence of
phoplasmacytic encephalitis, mild multifocal histiocytic abortions, early farrowings, reduced numbers of live-
interstitial pneumonia, and lymphoplasmacytic my- born pigs, increased preweaning mortality, reduced fer-
ocarditis are inconsistently observed in sows and gilts. tility, and increased nonproductive sow days. The rate
and duration of farrowing rate reduction, if it occurs,
Fetuses vary with the speed at which the infection is transmitted
A spectrum of fetal lesions following transplacental in- within the herd. While PRRSV is generally highly infec-
fection have been reported. Fetal lesions are useful when tious, farm design, pig flow through the facilities, and
present; however, they are inconsistently observed and production system type influence herd transmission and
are not pathognomonic for PRRS. A typical PRRSV-in- the duration and severity of clinical signs. Thus, farrow-
fected litter may contain some normal fetuses, stillborn ing rates may be depressed for only 3–4 consecutive
pigs, fetuses that have been dead for several days in weeks on some farms, whereas on others they may de-
utero, resulting in brown discoloration and autolysis, cline for 6–8 weeks or more.
and fetuses that are covered with a sticky mixture of The relative proportion of irregular and delayed re-
meconium, blood, and amniotic fluids. The most consis- turns to estrus sometimes increase dramatically at the
tent gross lesion in fetuses is segmental to full-length he- expense of normal returns to estrus, but the explanation
morrhages in the umbilical cord. Perirenal and colonic for this is unclear since PRRSV seems to have minimal ef-
mesenteric edema is also commonly observed in PRRSV- fects on conception (Lager et al. 1996; Prieto et al.
infected fetuses (Lager and Halbur 1996). 1996a). All parities of sows are affected during epi-
Microscopic examination may reveal segmental demics of PRRS, whereas in endemically infected herds
necrotizing umbilical arteritis characterized by fibri- lower-parity sows are typically more at risk of PRRS re-
nosuppurative inflammation and necrosis of the tunica productive failure. Endemically infected PRRS herds may
intima and tunica media with intramural and perivascu- therefore show either no change in fertility or a paradox-
CHAPTER 18 PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME Benfield et al. 217

ical improvement in fertility with service number (i.e., influence of passively acquired maternal antibodies on
fertility would normally fall with number of services that the success of VI is unknown, so obtaining presuckle
a sow has after weaning) (Collins and Dial 1996; G. D. serum in addition to other samples should be attempted
Dial, unpublished data—1998). when investigating abortion outbreaks.
Specimens for VI must be refrigerated or frozen soon
Serology after collection. PRRSV is rapidly degraded by heat and
PRRSV antibodies are detected using a variety of sero- is preserved by refrigeration and freezing (Benfield et al.
logic tests, including IFA (Yoon et al. 1992), immunoper- 1992; Van Alstine et al. 1993; Bloemraad et al. 1994).
oxidase monolayer assay (IPMA) (Wensvoort et al. 1991; Care should be used to prevent inadvertent heating be-
Drew 1995), ELISA (Albina et al. 1992; Cho et al. 1996), fore and during shipment. For example, transport of
and serum virus neutralization assay (SVN) (Yoon et al. samples to the veterinary clinic on warm summer days or
1994). Advantages of the automated ELISA test are that excessive warming of blood samples during centrifuga-
large numbers of sera can be tested in a short time, and tion may subject PRRSV to heat inactivation and thereby
results are potentially more consistent among diagnostic preclude VI. The virus has a narrow range of pH stabili-
laboratories. An additional advantage is that ELISA can ty so sterility must be maintained to avoid pH alterations
detect North American and European PRRSV strains, caused by bacterial contamination. Specimens should be
whereas IFA and IPMA detect only those PRRSV isolates packaged in an insulated, leak-proof container with am-
that are antigenically close to the PRRSV isolate used in ple amounts of refrigerant and shipped by overnight
the test. The PRRS vaccination status and age of the pigs courier.
and the regional prevalence of PRRS must be considered Several laboratory techniques have been used to
when interpreting serologic test results because serolog- identify PRRSV antigen. Immunohistochemistry (IHC)
ic tests do not distinguish vaccinated from naturally in- (Halbur et al. 1994; Larochelle and Magar 1995) and IFA
fected pigs and because young pigs may possess passive- (Yoon et al. 1992; Lager and Ackermann 1994) tests de-
ly acquired antibodies for 5–10 weeks after infection tect PRRSV antigen in many tissues, although lung and
(Houben et al. 1995a). tonsil are preferred. Tissues should be submitted refrig-
The PRRSV antibody kinetics of the IFA, IPMA, and erated for IFA and fixed in 10% neutral buffered formalin
ELISA tests are similar. PRRSV antibodies are detected for IHC. Unfortunately, the IHC and IFA test results are
7–14 days after infection, reach maximal titers by 30–50 influenced by technician skill, so results should be con-
days, and then gradually decline to low or undetectable firmed by VI or PCR. In situ hybridization is a sensitive
levels by 4–6 months after infection. The SVN test is less method to identify viral nucleic acid (Larochelle et al.
sensitive than other serologic tests for detecting recent 1996; Sur et al. 1996; Haynes et al. 1997); however, it
infection with PRRSV because neutralizing antibodies finds greater application in research than in routine di-
develop slowly. However, it provides better long-term ev- agnosis.
idence for past infection because PRRSV neutralizing an- PCR is a highly sensitive test for detecting viral RNA.
tibodies persist for at least 1 year (Yoon et al. 1994). Use It is useful when VI is troublesome, such as when testing
of an IFA test that detects anti-PRRSV IgM antibodies semen (Christopher-Hennings et al. 1995b) and when
has been advocated because of its ability to detect recent virus has been partially degraded by autolysis (as in
infection (Park et al. 1995). Acute and convalescent necrotic fetal tissues). Nested PCR tests have been devel-
serum samples should be provided to a diagnostic labo- oped which appear as sensitive as conventional VI tech-
ratory when seeking evidence of recent infection. niques for detecting an acute virus infection in pigs
(Mardassi et al. 1994b; Van Woensel et al. 1994; Kono et
Virus Detection al. 1996; Gilbert et al. 1997). Moreover, these tests may
As with all diagnostic investigations, the first and most be more sensitive than VI during the convalescent stages
important step is to select the samples most likely to of infection. Automated PCR tests, which are less prone
yield useful laboratory information. In general, speci- to contamination, have enabled diagnostic laboratories
mens collected from younger, rather than older, pigs are to offer sensitive and specific PCR testing at much lower
preferred because PRRSV is found in greater amounts cost (Molitor et al. 1997). PCR samples should be col-
and for longer periods of time in young pigs. Virus can lected and handled like VI samples.
be isolated from serum for 4–6 weeks after infection of To date, PRRSV has only been propagated in vitro in
suckling, weanling, and grower pigs (Mengeling et al. a limited number of cell types. These are PAMs
1996d) and for 1–2 weeks after infection in boars (Wensvoort et al. 1991), the MA-104 cell line and its de-
(Christopher-Hennings et al. 1995a) and sows (Men- rivatives (the MARC-145 and CL-2621 cell lines; Kim et
geling et al. 1996c). Virus can typically be isolated from al. 1993; Benfield et al. 1992), the CRL-11171 cell line
bronchioalveolar lavage samples for at least several (Halbur et al. 1995), blood-derived monocytes (Voicu et
weeks following the cessation of detectable viremia al. 1994), and microglia (Molitor et al. 1996). Different
(Mengeling et al. 1995; Mengeling et al. 1996b, c, d). The strains or isolates of PRRSV vary in their ability to repli-
218 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

cate in different cell types (Bautista et al. 1993); there- REPRODUCTIVE DISEASE. Diagnosis of PRRSV in-
fore, the use of PAMs in conjunction with a cell line such fection is difficult to achieve by examining fetal tissues.
as MA-104 maximizes VI success. Heparin prevents Fetus and placenta seldom have microscopic lesions
PRRSV from infecting MA-104 cells, so heparin should (Rossow et al. 1996b), and if present the lesions are non-
not be added to cell cultures (Jusa et al. 1997). Blood diagnostic (Lager and Ackermann 1994). VI is trouble-
samples for VI should be collected in serum or EDTA some because typically only some of the fetuses and/or
tubes but not in tubes containing heparin. piglets of an affected sow are infected at the time of de-
Serum, buffy coat, lung, alveolar washings, lymph livery and because virus is degraded by in utero fetal au-
node, and tonsil are the specimens of choice for VI. For tolysis. Moreover, PRRSV-infected sows may abort in the
isolation, the sample is inoculated onto cell cultures and absence of in utero infection. For the same reasons, at-
cultures are observed for cytopathic effect. The presence tempts to identify PRRSV antigen in fetal tissues by IHC
of PRRSV can then be confirmed by IFA. Monoclonal an- and IFA are often unsuccessful. PRRS viral detection by
tibodies directed against the highly conserved nucleo- PCR has provided promising results presumably because
capsid protein are useful for immunofluorescence testing this technique does not depend on the presence of live
because they detect antigenically diverse PRRSV strains virus (K. D. Rossow, unpublished data—1998), but it
(Nelson et al. 1993). The cytopathic effect of PRRSV is does not circumvent the problem of identifying which fe-
usually obvious by 2–7 days after initial exposure of a tuses and pigs are infected at the time of sample collec-
susceptible cell culture to a diagnostic sample containing tion.
a field strain of the virus. However, it may escape notice Collecting pre- and postsuckle serum from several
if the sample contains little of the virus or if the particu- weak-born and live-born pigs per litter improves the
lar isolate replicates poorly in cell culture. Consequently, chances of successful virus detection and provides an op-
at least one additional cell culture passage is recom- portunity to measure PRRSV antibodies. The presence of
mended to increase the reliability of the isolation proce- PRRSV antibodies in presuckle sera indicates in utero in-
dure. fection. Unfortunately, PRRSV antibodies are present in
Diagnosis by VI is complicated by the use of PRRS only a minority of PRRSV-infected pigs at delivery (Men-
modified-live-virus vaccines because vaccinated pigs may geling et al. 1996c). Using PCR to identify PRRSV in sera
be viremic for up to several weeks and may transmit vac- of aborting sows often yields positive results, whereas VI
cine virus to penmates (Torrison et al. 1996). Therefore, from the same samples is seldom successful (J. E. Collins,
attempts have been made to distinguish PRRS field virus unpublished data—1998). Serologic examination of
from vaccine virus by restriction fragment length poly- acute and convalescent sow sera may provide evidence of
morphism (Wesley et al. 1996), genetic analysis, and seroconversion.
monoclonal antibody profiling (Nelson et al. 1997). Re- PCR has proven to be a highly sensitive test for de-
sults of these tests should be interpreted cautiously be- tecting PRRSV in semen (Christopher-Hennings et al.
cause vaccine and field isolates may in some instances 1995b; Shin et al. 1997). In most cases a positive PCR test
provide similar results. Also, only small regions of the correlates with a positive swine bioassay (Christopher-
PRRSV genome are sequenced for routine diagnostic Hennings et al. 1995b), indicating that viral RNA detect-
testing, so important differences between isolates might ed in semen represents infectious virus. Collecting at
go undetected (K. S. Faaberg, unpublished data—1998). least 3–5 successive semen samples at weekly intervals
has been recommended because boars may shed PRRSV
Diagnosis Summary intermittently over prolonged time periods (Christo-
The clinical manifestations of PRRS are variable and are pher-Hennings et al. 1995a). The cost of PCR testing se-
affected by many factors that confound the diagnosis of men samples can be reduced by pooling in groups of
this disease. A definitive diagnosis is dependent on the 3–5, although decreased test sensitivity may occur when
collection and submission of correct samples. semen contains low amounts of the virus.

RESPIRATORY DISEASE. Pigs should be evaluated IMMUNOLOGY


for gross and microscopic lesions of PRRS. Portions of
lung, heart, brain, lymph node, kidney, and tonsil should Pigs infected with PRRSV produce an immune response
be submitted refrigerated and fixed in 10% neutral that is easily detected by the presence of serum antibod-
buffered formalin. IFA, IHC, PCR, and VI on lung, tonsil, ies to the virus. The appearance of antibodies correlates
and lymph nodes can be attempted. Collecting alveolar with decreases in disease signs, viremia, and viral shed-
washings enhances VI in late stages of PRRSV infection ding. Animals that recover from infection are immune
and from older animals (Mengeling et al. 1995; Men- and are protected from subsequent challenge. The anti-
geling et al. 1996b, c, d). A variety of sampling strategies body response is detected by the appearance of specific
have been proposed for serologic detection of PRRSV IgM by 7 days and of specific IgG by 14 days after infec-
(Collins et al. 1996). tion (Vezina et al. 1996).
CHAPTER 18 PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME Benfield et al. 219

Humoral immune responses are monitored by a vari- to the difficulty in developing antigen-specific, major-
ety of assays, including IFA, IPMA, ELISA, and SVN. histocompatibility-complex-restricted T-cell prolifera-
Most serologic tests indicate that antibodies reach a tion and cytotoxicity assays in swine.
maximum titer by 5–6 weeks postinfection and persist Immunosuppression, perhaps due to destruction of
for the life of a commercial feeder pig. However, virus- PAMs during viral replication, has been suggested to ac-
neutralizing antibodies appear more slowly, usually be- count for reports of increased incidences of respiratory
ing detected first at 4–5 weeks postinfection, and reach- disease and loss of productivity on farms affected with
ing a maximal titer about 10 weeks postinfection (Yoon PRRS and for episodes of apparent vaccine failure. How-
et al. 1995b). The appearance of neutralizing antibodies ever, efforts to demonstrate immunosuppressive effects
seems to be delayed in boars, occurring at about 10 of PRRSV infection, or exacerbation of other diseases by
weeks and reaching a maximum at about 18 weeks (Nel- concurrent infection with PRRS, have generally been un-
son et al. 1994). successful (Grosse-Beilage 1995; Van Alstine et al. 1996).
The duration of antibody responses in sows and Likewise, it has been proposed that the presence of anti-
boars is not well described since most experiments are of PRRSV antibodies in animals might exacerbate the
relatively short duration. Sows that are seropositive se- severity of infection due to antibody-mediated entry of
crete antibodies in the colostrum (Eichhorn and Frost virus into macrophages via the Fc receptor. However,
1997). Noninfected piglets born to seropositive sows are there are few, if any, convincing data in support of this
seronegative at birth and acquire antibodies passively hypothesis.
from the colostrum. The development of licensed vaccines for prevention
Antibodies are presumed to play a role in protection of PRRS has led to widespread application and serologic
against reinfection. Antibody responses to the major testing in North America. The resulting “experiments of
proteins of the virion, including the nucleocapsid prod- nature” have revealed cases in which animals fail to
uct of ORF 7, the envelope glycoprotein product of ORF seroconvert following exposure to vaccine, potential lack
5, and the integral membrane protein encoded by ORF 6, of cross-protection, and potential examples of immuno-
as well as to the protein products of the ORF 2, ORF 3, logic nonresponsiveness following multiple exposures to
and ORF 4 genes, are readily detected. Antibodies to the an attenuated vaccine virus. Some of these examples are
envelope glycoprotein neutralize viral infectivity (S. Dea, predictable since genetic and environmental differences
unpublished data—1998) and immunization with re- in host animals influence the magnitude of an immune
combinant proteins from ORF 5 or ORF 3 appears to response. Ongoing genetic and antigenic variation
provide partial protection from infection (Plana Duran in the wild-type virus and immunologic selection due to
et al. 1997). Antinucleocapsid antibodies are not neu- vaccination make it likely that variants will arise which
tralizing, since the protein is not on the surface of the escape the protection afforded by currently available vac-
virion. Interestingly, antibodies to the ORF 4 proteins are cines.
reported to neutralize infectivity of LV (Van Nieuwstadt Our knowledge of PRRS immunology is at present
et al. 1996). Whether the ORF 4 protein is on the virion rather rudimentary and descriptive. There are many
surface and subject to neutralization in North American more questions than answers regarding the essential fea-
strains of PRRSV is not known. Antigenic variation is tures of a protective immune response. These include
well documented in the amino acid sequences of PRRSV the effect of antibodies on progression of viral infection
proteins based on DNA sequencing and is also reflected in macrophages, the existence and consequences of
in the specificities of monoclonal antibodies to PRRSV PRRS-mediated immunosuppression, and the immuno-
isolates. Thus, serologic diagnostics that rely on detec- logic responses in piglets which are born viremic. An-
tion of whole virus or virus-infected cells with swine swers to these questions will be important in the contin-
serum are more reliable than antibody tests that rely on uing effort to control and prevent PRRS.
monoclonal antibodies or purified recombinant viral
proteins (Yoon et al. 1995a). However, some monoclonal PREVENTION AND CONTROL
antibodies have a range of cross-reactivity that exceeds
that of many polyvalent sera (Nelson et al. 1993). Reducing the clinical and economic effects of PRRSV
Little is known about the cellular immune responses within infected swine farms is a challenge to veterinari-
to PRRSV infection. Lymphocytopenia appearing tran- ans throughout the world. Despite the high degree of
siently at 3–9 days after infection is a hallmark of PRRSV frustration experienced with PRRS since the disease was
infection. However, by 14 days after infection total pe- first reported, progress has been made in the under-
ripheral blood lymphocyte numbers are normal. T cells standing of its epidemiology and in the accuracy of the
positive for CD8 increase, consistent with the hypothesis diagnostic testing procedures. Although numerous ques-
that PRRS-specific cytotoxic T cells are produced tions still remain unanswered, this section provides an
(Shimizu et al. 1996). Nevertheless, the role of cellular overview of existing strategies known to be effective for
immunity in resistance to PRRS is unknown, due in part the control of PRRS, as well as a review of specific steps
220 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

necessary to prevent the introduction of the virus into a 1. Understanding of the pattern of viral spread
naive farm. throughout the system through the application of popu-
lation-based diagnostic strategies
Prevention 2. Proper development of replacement stock prior to in-
Despite the fact that PRRSV infection is widespread troduction into PRRSV-infected herds
throughout the world, noninfected herds still exist. 3. Prevention of the transmission of the virus from
While all routes of virus introduction into a naive herd sow to piglet through breeding-herd stabilization
are not completely understood at this time, the primary 4. Control of the spread of virus in the nursery or
source is the infected pig. Therefore, it is critical to rou- finisher populations through weaned-pig management
tinely isolate and test breeding stock before introducing
them to a PRRS-negative herd. Replacement stock to be The purpose of such a model is to provide a generally
added to naive herds should be obtained from known applicable systems approach for the control of PRRS-re-
negative sources that carry out a regular schedule of herd lated disease problems. The individual components of
monitoring. In addition, there should be clear and open the model are described in greater detail as follows.
communication between veterinarians responsible for
the health of the source and recipient herds, respective- POPULATION-BASED DIAGNOSTIC STRATEGIES.
ly, prior to purchase of replacement stock. In today’s industry, swine production systems are a series
Isolation facilities should be located on another farm of interdependent populations, consisting of the breed-
site and visited at the end of the working day. Following ing, gestating, and lactating populations, the nursery
the arrival of new stock, all animals should be blood-test- and finisher populations, and the replacement gilt pool.
ed 14 days following entry into the isolation building. Previously published reports have indicated that PRRSV
Isolation periods should be at least 30 days to allow suf- may be limited to an individual population (e.g., the
ficient time to obtain laboratory results prior to animal nursery) of a particular farm (Dee and Joo 1994a).
introduction. If diagnostic results indicate that incoming Therefore, it is critical to accurately determine the pat-
stock are infected with PRRSV, all animals of the isolat- tern of viral transmission within a farm on a population
ed group should be removed from the building and mar- basis.
keted. The isolation facility should then be cleaned using Serologic profiling using IFA or ELISA is the preferred
95˚C water, disinfected using phenolic- or formaldehyde- method of data collection. Randomly selected swine are
based products, and allowed to remain empty for a min- tested using cross-sectional sampling in order to assess
imum of 7 days. Finally, in accordance with strict isola- the prevalence of infection and the pattern of viral trans-
tion and testing policies, sound principles of biosecurity mission within each population. Increased sample sizes
previously described for other swine pathogens (e.g., may be required in larger herds, and statistical tables
transmissible gastroenteritis virus) should be practiced should be referenced to ensure that the correct number
at all times (Moore 1992). of samples are collected. An example of a testing proto-
col is provided in Table 18.1. Tables 18.2 and 18.3 pro-
Control vide examples of virus infection localized to the nursery
Following the identification of PRRSV and the develop- or widespread throughout all populations, respectively.
ment of diagnostic tests, practitioners throughout the The data presented in Table 18.2 are indicative of a
world have attempted to control the effects of PRRS. The chronically infected farm experiencing postweaning,
initial attempts were based on the use of strategies PRRS-related, respiratory disease, whereas the data pre-
known to be effective for controlling other diseases of sented in Table 18.3 are typical of an acute epidemic.
swine (e.g., segregated early weaning); however, results In addition to the technique of cross-sectional profil-
were inconsistent (Dee et al. 1993). The central compo- ing, serial testing of weaned pigs is helpful not only for
nent of PRRS control is the reduction of the spread of detecting the point of seroconversion to PRRSV but also
the virus within the breeding herd, thereby preventing for determining the role of other pathogens in the dis-
the infection of offspring prior to weaning. Recently, a ease process (e.g., Mycoplasma hyopneumoniae or swine
report describing the presence of seronegative subpopu- influenza virus). Using this strategy, a minimum of 10
lations in chronically infected breeding herds was pub- pigs are identified and sampled throughout the nursery
lished (Dee et al. 1996). This report concluded that in- and/or finishing periods at 4-week intervals. Although
consistent levels of virus exposure exist within this method requires a longer period of time to obtain re-
endemically infected herds, and the formation of sub- sults, it can be used in conjunction with cross-sectional
populations assisted in the maintenance of virus trans- profiling to enhance the accuracy of the sampling
mission in the breeding herd over time. process.
Recently, a model for the control of PRRS which fo- Ideally, the diagnostic evaluation should take place
cuses on the elimination of subpopulations was devel- before vaccination. Serologic assessment is much more
oped. The components of the model are as follows: difficult following vaccination, since tests for differenti-
CHAPTER 18 PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME Benfield et al. 221

Table 18.1. Serologic profiling protocol for evaluation of a PRRSV-infected herd


Key Points concerning Profiling:
1. Ideally, profiling should take place before vaccination.
2. Statistical tables should be consulted to ensure adequate sample sizes are collected.
3. Results are indicative of infection, not immunity.
4. Tissue samples from infected pigs should be submitted simultaneously for verification of viral antigen and microscopic evidence of an
infectious disease process.
Step 1: Diagnostic Program
Randomly select the following number of samples from the designated populations:
1. Breeding herd: n = 30 sows (5 samples/parity)
2. Recently weaned pigs (3–4 weeks old): n = 10
3. Pigs at the end of the nursery stage (8–10 weeks old): n = 10
4. Premarket finishers (5–6 months old): n = 10
5. Replacement gilts prior to entry into breeding herd: n = 10
Step 2: Monitoring Program
1. Breeding herd
Quarter 1 Month 1: From the initial sampling of 30 animals, select 10 monitor sows for serial testing.
Months 2 and 3: Retest monitor sows, along with 20 randomly selected animals monthly for a total of 30 samples each
testing period.
Quarters 2 through 4: Repeat quarter 1 protocol, selecting 10 new monitor sows.
2. Gilt pool: Randomly test 30 replacement gilts prior to entry into the breeding herd on a monthly basis.
3. Weaned-pig populations: Repeat sampling of 10 recently weaned pigs, 10 (8- to 10-week-old) nursery pigs, and 10 premarket finishers on
a quarterly basis.

Table 18.2. Cross-sectional serologic (ELISA) Table 18.3. Cross-sectional serologic (ELISA)
profile indicating infection in the nursery within a profile indicating active infection in all stages of
nonvaccinated herd production within a nonvaccinated herd
Sample Sample
No. Breeding 4 Weeks 8 Weeks 6 Months No. Breeding 4 Weeks 8 Weeks 6 Months
1 0.35* 0.18 1.95 0.50 1 2.53* 1.95 2.50 0.85
2 1.00 0.33 2.01 0.65 2 2.20 3.25 3.15 3.05
3 0.44 0.21 1.00 1.20 3 2.54 2.70 0.81 3.20
4 0.61 0.19 1.59 1.20 4 3.44 0.31 0.05 1.79
5 0.59 0.46 1.49 0.60 5 0.32 2.95 1.75 0.50
6 0.37 0.20 2.24 1.00 6 0.81 3.10 2.30 0.06
7 0.36 0.12 1.28 1.00 7 2.95 0.65 1.65 1.79
8 0.63 0.55 0.83 0.82 8 1.27 1.85 0.50 2.55
9 0.51 0.63 1.95 0.95 9 0.18 0.20 0.31 3.10
10 0.20 0.01 3.00 0.50 10 0.25 0.55 0.26 0.75
*ELISA S/P ratio (≥ 0.4 = positive). *ELISA S/P ratio (≥0.4 = positive).
Additional diagnostics: PRRSV isolated from tissues and sera Additional diagnostics: PRRSV isolated from tissues and
of nursery pigs. sera.
Clinical picture: Pigs healthy at weaning, developing evi- Clinical picture: Reproductive failure in late gestation, respi-
dence of respiratory disease 2–4 weeks postweaning. ratory disease in weaned pigs.
Production data: Nursery average daily gain = 0.25 kg; nurs- Production data: Stillborns = 35%; abortions = 10%; post-
ery mortality = 8.5%. weaning mortality = 10%.

ating antibodies produced against vaccine virus versus controlling subpopulation formation and the spread of
those produced against field virus are not available. Fi- the virus in the breeding herd. Episodes of recurrent re-
nally, in conjunction with serology, tissue submissions productive failure following direct introduction of naive
from affected pigs will assist in the verification of a replacement gilts into an actively infected herd have
PRRSV-related disease process. Microscopic evidence of been described (Dee and Joo 1994b). Therefore, it is im-
PRRSV infection and demonstration of the presence of portant to understand whether there are differences be-
viral antigen in tissue frequently support the serologic in- tween the PRRS status of incoming replacement stock
terpretation. and the recipient breeding herd, and to establish pro-
grams for animal introduction.
DEVELOPMENT OF REPLACEMENT STOCK. Isolation and acclimatization of replacement stock
Proper management of replacement stock is essential for have been used to reduce the risk of introducing new
222 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

pathogens to an existing herd, as well as to properly pre- tect noninfected herds from production and economic
pare animals for entry into a PRRS-positive system. In losses if they become exposed to the virus.
the past, isolation periods typically ranged from 21 to 30
days; however, due to the ability of PRRSV to induce pro- WEANED-PIG MANAGEMENT. For the control of
longed periods of viremia following infection, isolation chronic PRRS in the weaned-pig population, two prima-
and acclimatization periods need to be extended. There- ry strategies exist: partial depopulation and pig vaccina-
fore, development may begin early in the life of the ani- tion. Again, the success of either strategy depends on
mal and take place in facilities specifically designed for preventing the introduction of actively infected animals
this purpose (Dee et al. 1994). Animal introduction may into the nursery.
take place during the suckling period (i.e., 2 days of age), Partial depopulation consists of a strategic adjust-
at the time of weaning, or at 25 or 100 kg of body weight. ment in pig flow to prevent the lateral spread of
In order to accommodate the need for extended develop- PRRSV within chronically infected populations. Previ-
ment periods, it is recommended that larger groups of ous studies have indicated that the spread of the virus
younger animals be introduced less frequently through- within the nursery or finishing stage is maintained
out the year. through the transmission of the virus to recently
During the development period, a standard program weaned, naive piglets from older, previously infected
of immunization against PRRSV and exposure to previ- pigs (Dee and Joo 1994a). Partial depopulation has
ously infected swine is practiced to enhance the develop- been applied extensively to the nursery, and published
ment of protective immunity. Serologic profiling of ani- data have indicated highly significant improvements
mals following the completion of the development (p < .0001) in average daily gain and mortality (Dee et al.
program can assess the PRRS status of the replacement 1997c).
population prior to their addition to the breeding herd. The economic benefit of nursery depopulation has
Previously published summaries provide additional in- been demonstrated as well. In a recently published study,
formation on specific development programs (Dee improved profitability was demonstrated in 94% of the
1997a, b). study farms, despite the fact that PRRSV was not elimi-
In conclusion, improved understanding of the rela- nated in all cases (Dee et al. 1997b). Limitations of this
tionship between replacement stock and the control of strategy include the logistics involved in depopulating
PRRS-related reproductive disease has emphasized the large nurseries and the fact that depopulation may need
importance of this population. The challenge to the in- to be repeated periodically to maintain improvements in
dustry will be not only to improve these existing systems performance over extended periods of time.
but to apply their principles to the control of other in- Commercially available vaccines for the control of
fectious disease problems of swine. PRRS in pigs of 3–18 weeks of age became available in
the United States in June 1994. Prior to implementing a
BREEDING-HERD STABILIZATION. While replace- vaccination program, it is important to determine the
ment stock programs are critical for controlling the for- timing of virus exposure in order to immunize the ani-
mation of subpopulations, commercially available mod- mal prior to infection. Because transmission of modi-
ified-live-virus vaccines have been used to induce a fied-live-vaccine virus to negative swine can take place,
consistent immune response throughout the adult popu- the use of this product is not recommended in cases
lation. Currently, such products are only approved for where a seronegative status is desired (Torrison et al.
use in nonpregnant females 3–4 weeks prior to breeding; 1996).
however, they have been used extensively in pregnant Finally, if the suckling piglet population is acutely in-
sows and in boars in the field (Dee 1996). Use of vaccines fected, a series of management strategies have been out-
in this manner is at the discretion of the veterinarian, lined (Henry 1994). Emphasized within this report is
and it must be realized that vaccination of naive preg- limiting the movement of piglets between litters to the
nant sows during the third trimester of gestation may re- initial 24 hours of life, humanely destroying chronically
sult in fetal infection (Mengeling et al. 1996c). Only lim- infected offspring prior to weaning, and maintaining
ited published data generated following implementation strict all-in/all-out animal flow in the nursery.
of breeding-herd vaccination strategies exist at this time; In conclusion, although the application of this model
however, one study has indicated the ability of vaccina- has resulted in successful control of PRRS-related dis-
tion to reduce the spread of virus among members of the ease problems over a wide range of production systems,
adult population and prevent the infection of piglets pri- elimination of the virus is not easily attained at this time.
or to weaning (Dee et al. 1997a). The ability of the virus to establish persistent infections
Finally, although efficacious killed-virus vaccines are and the lack of regulatory control over the sale of infect-
not yet commercially available, such a product may be ed breeding stock are key issues that must be resolved
beneficial to control subpopulation formation, maintain prior to the initiation of large-scale eradication pro-
protective immunity within the breeding herd, and pro- grams.
CHAPTER 18 PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME Benfield et al. 223

VACCINES success a number of factors need to be considered when


designing a vaccination program. (1) Vaccine virus can
Several studies have established that vaccination against persist for weeks and perhaps months (Mengeling et al.
PRRS can result in protective immunity (Gorcyca et al. 1996c), and its persistence may be similar to that of vir-
1995; Gorcyca et al. 1996b; Hesse et al. 1996a, b; Men- ulent virus (Mengeling et al. 1996a, c; Wills et al. 1995a,
geling et al. 1996a; Plana Duran et al. 1995). Such stud- b). (2) Vaccine virus can be transmitted from vaccinated
ies have led to the extensive use, especially in the United pigs to naive pigs (Torrison et al. 1996; Bøtner et al.
States, of commercial vaccines as an aid in reducing the 1997; Mengeling et al. 1998). (3) Vaccine virus can cross
clinical consequences of infection with virulent PRRSV. the placenta to cause congenital infection (Mengeling et
Although both attenuated and inactivated vaccines have al. 1996c). (4) Vaccine virus can persist in boars and be
been described, the former are generally believed to be disseminated through semen (Christopher-Hennings et
more efficacious. As a result, virus inactivation has been al. 1997). (5) Vaccine-induced protective immunity is
used mostly as a method of preparing autogenous vac- slow to develop (Mengeling et al. 1996a; W. L. Men-
cines. geling, K. M. Lager, and A. C. Vorwald, unpublished da-
Currently, two attenuated vaccines are commercially ta—1998). (6) The time required to develop protective
available for use as an aid in the prevention and control immunity is likely to be strain related; that is, immunity
of PRRS. RespPRRS/Repro® has been marketed in may develop more quickly and more forcefully (W. L.
North America since 1994 for administration to pigs be- Mengeling, K. M. Lager, and A. C. Vorwald, unpublished
tween 3 and 18 weeks of age for the prevention of the data—1998) and last longer (Lager and Mengeling, un-
respiratory component of PRRS, and since 1996 for ad- published data—1998) against the homologous and
ministration to nonpregnant gilts and sows for the pre- antigenically similar strains than against more distantly
vention of the reproductive component of PRRS. (The related strains of PRRSV. (7) Some vaccinated pigs, gilts,
same vaccine is marketed in Europe under the name In- and sows may fail to seroconvert for PRRSV following
gelvac® PRRS MLV.) Prime Pac® PRRS has been market- vaccination (W. L. Mengeling, K. M. Lager, and A. C.
ed in the United States since 1996 for administration to Vorwald, unpublished data—1998). (8) Successful vacci-
nonpregnant gilts and sows for the prevention of the re- nation of adult females (as judged by seroconversion)
productive component of PRRS. may not ensure the complete absence of transplacental
Much of what is now known about vaccination infection and vertical transmission of virulent virus
against PRRS is based on controlled laboratory studies (Mengeling et al. 1996a). In addition, evidence has been
with RespPRRS/Repro® and with three experimental presented for the transmission of vaccine virus from vac-
vaccines developed at the National Animal Disease Cen- cinated sows to nonvaccinated sows, and from vaccinat-
ter (NADC) (Mengeling et al. 1996a). All of these vac- ed herds to nonvaccinated herds, with the subsequent
cines, as well as Prime Pac® PRRS, were derived (i.e., at- development of vaccine-virus-induced reproductive fail-
tenuated) by repeated passage of virulent, North ure (Bøtner et al. 1997).
American field strains of PRRS virus in cell culture. They With these issues in mind, a vaccination program
differ genetically from one another to some extent, but that emphasizes safety as well as efficacy might be de-
they are more closely related to each other than to LV. fined by the following. For at least several weeks after
Information from field use of PRRS vaccine is limited vaccination (the longer the better), vaccinated pigs, gilts,
mostly to RespPRRS/Repro®, which has been used ex- and sows would be isolated to minimize transmission of
tensively since it was first licensed and marketed in 1994 vaccine virus. Neither mature boars (used for natural ser-
under the name RespPRRS®. Although RespPRRS® was vice or semen collection) nor pregnant gilts and sows
not approved for use in gilts and sows for the prevention would be vaccinated. Gilts would be vaccinated twice be-
of the reproductive component of PRRS until 1996 fore breeding to increase the likelihood of seroconver-
(when the name was changed to RespPRRS/Repro®), it sion. The age of gilts at the time of the first vaccination
was often used “off label” for this purpose. would depend on the limitations of the particular opera-
The following discussion relative to the safety, effica- tion, but in general, the earlier the better so that the sec-
cy, and limitations of PRRS vaccines is based almost en- ond dose of vaccine, administered 1–2 months after the
tirely on information derived from laboratory studies first (preferably the latter), could be administered at least
with RespPRRS/Repro® and NADC vaccines and from 1 month before breeding. Moreover, PRRSV replicates
field use of RespPRRS/Repro®. The relative merits of the more extensively in young pigs and, as a consequence,
more recently introduced Prime Pac® PRRS remain to be may induce a better immune response. Assuming that
determined through independent laboratory and field sows are immune and their suckling pigs are protected
comparisons. by passively acquired antibodies, a booster vaccination
It is generally believed that, when used judiciously, at- could probably be administered safely during the nurs-
tenuated vaccines can be of value in preventing and con- ing interval. The objective of this schedule is to provide
trolling PRRS (Dee 1996). However, to achieve greatest the maximum time for the development of protective
224 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

immunity against the widest range of virus strains, to P. 1992. An enzyme linked immunosorbent assay
minimize the chance that vaccination interferes with (ELISA) for the detection of antibodies to porcine repro-
conception or gestation, and to maintain a high level of ductive and respiratory syndrome (PRRS) virus. Ann
maternal immunity. Rech Vet 23:167–176.
The temporal parameters for vaccinating young pigs Albina, E.; Madec, F.; Cariolet, R.; and Torrison, J. 1994a.
are less well defined than they are for vaccinating gilts Immune response and persistence of the porcine repro-
and sows. What is known is that passively acquired anti- ductive and respiratory syndrome virus in infected pigs
bodies can be protective and interfere with replication of and farm units. Vet Rec 134:567–573.
at least the homologous strain of PRRSV (Gorcyca et al. Albina, E.; Vannier, P.; Madec, F.; Cariolet, R.; and Torrison,
1996a; Morrison et al. 1996). Consequently, vaccine is J. 1994b. Persistence of the porcine epidemic abortion
likely to be less effective when it is administered too soon and respiratory syndrome (PEARS) virus in infected pigs
to pigs that suckle immune dams. However, a decision to and farm units. Proc Int Congr Pig Vet Soc 13:53.
delay vaccination until the disappearance of passively ac- Anon. 1992. Porcine reproductive and respiratory syn-
quired antibodies is complicated by the fact that protec- drome (PRRS or blue-eared pig disease). Vet Rec
tive immunity is slow to develop—which, in turn, em- 130:87–89.
phasizes the importance of early vaccination. Therefore, Bautista, E. M.; Goyal, S. M.; Yoon, I. J.; Joo, H. S.; and
to help ensure active and protective immunity as soon as Collins, J. E. 1993. Comparison of porcine alveolar
possible in young pigs, it may be necessary to administer macrophages and CL 2621 for the detection of porcine
two doses of vaccine, the first before (assuming at least reproductive and respiratory syndrome (PRRS) virus
some stimulation of the immune system), and the sec- and anti-PRRS antibody. J Vet Diagn Invest 5:163–165.
ond after, the disappearance of passively acquired anti- Bautista, E. M.; Meulenberg, J. J. M.; Choi, C. S.; and Moli-
bodies. tor, T. W. 1996. Structural polypeptides of the American
Once a vaccination program is selected, the next deci- (VR-2332) strain of porcine reproductive and respirato-
sion may be whether to use attenuated virus in herds that ry syndrome virus. Arch Virol 141:1357–1365.
appear free of virulent field virus. The fact that Prime Benfield, D. A.; Nelson, E.; Collins, J. E.; Harris, L.; Goyal, S.
Pac® PRRS is (with the advice of a veterinarian), and M.; Robison, D.; Christianson, W. T.; Morrison, R. B.;
RespPRRS/Repro® is not, recommended by the manu- Gorcyca, D.; and Chladek, D. 1992. Characterization
facturers for use in virus-free herds may be more a mat- of swine infertility and respiratory syndrome (SIRS)
ter of judgment than a reflection on the particular prop- virus (isolate ATCC VR-2332). J Vet Diagn Invest 4:127–
erties of the two vaccines. A possible compromise might 133.
be the use of attenuated virus in a PRRSV-free herd only Benfield, D.; Rowland, R.; Nelson, J.; Neiger, J.; Nelson, E.;
when there is reason to suspect imminent introduction Rossow, K.; and Collins, J. 1996. A model for the study of
of virulent virus with the potential for clinical disease persistent PRRSV infection. Conf Res Workers Anim
and financial loss. Dis Abstr 187.
Although vaccines, especially attenuated vaccines, Benfield, D. A.; Christopher-Hennings, J.; Nelson, E. A.;
have played a role in the prevention and control of PRRS Rowland, R. R. R.; Nelson, J. K.; Chase, C. C. L.; Rossow,
and are likely to continue to do so in the foreseeable fu- K. D.; and Collins, J. E. 1997. Persistent fetal infection of
ture, there is still much to be learned about their efficacy porcine reproductive and respiratory syndrome (PRRS)
and safety and about the minimal interval between vac- virus. In Proc 28th Annu Meet Am Assoc Swine Pract,
cination and exposure to virulent virus necessary for pro- pp. 455–458.
tective immunity. And because of strain differences Bilodeau, R.; Dea, S.; Sauvageau, R. A.; and Martineau, G. P.
there may be a need for multistrain vaccines to provide 1991. Porcine reproductive and respiratory syndrome in
the earliest protection against the widest range of het- Quebec [letter]. Vet Rec 129:102–103.
erologous strains. Also, inactivated vaccines (Plana Du- Blaha, T. 1992. Epidemiological investigations into PEARS
ran et al. 1995; Swenson et al. 1995), because of their in Germany: Consequences in fattening pigs. Proc Int
added level of safety, need to be further evaluated for Congr Pig Vet Soc 12:126.
their potential for the control of PRRS, either when used Bloemraad, M.; de Kluijver, E. P.; Petersen, A.; Burkhardt, G.
alone or as an adjunct to attenuated vaccines. E.; and Wensvoort, G. 1994. Porcine reproductive and
respiratory syndrome: Temperature and pH stability of
REFERENCES Lelystad virus and its survival in tissue specimens from
viraemic pigs. Vet Microbiol 42:361–371.
Ahmed, R.; Morrison, L. A.; and Knipe, D. M. 1996. Persis- Bøtner, A.; Nielsen, J.; and Bille-Hansen, V. 1994. Isolation
tence of viruses. In Field’s Virology, 3d ed. Ed. B. N. of porcine reproductive and respiratory syndrome
Fields, D. M. Knipe, and P. M. Howley. Philadelphia: (PRRS) virus in a Danish swine herd and experimental
Lipincott-Raven, pp. 219–249. infection of pregnant gilts with the virus. Vet Microbiol
Albina, E.; Leforban, Y.; Baron, T.; Duran, J. P.; and Vannier, 40:351–360.
CHAPTER 18 PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME Benfield et al. 225

Bøtner, A.; Strandbygaard, B.; Sorensen, K. J.; Have, P.; Rowland, R.; Yaeger, M.; Chase, C.; and Benfield, D.
Madsen, K. G.; Madsen, S.; and Alexandersen, S. 1997. 1996. Identification of PRRSV in semen and tissue of va-
Appearance of acute PRRS-like symptoms in sow herds sectomized and non-vasectomized boars. Conf Res
after vaccination with a modified live PRRS vaccine. Vet Workers in Anim Dis Abstr 182.
Rec 141:497–499. Christopher-Hennings, J.; Nelson, E. A.; Nelson, J. K.; and
Busse, F. W.; Alt, M.; Janthur, I.; Neumann, W.; and Schoss, Benfield, D. A. 1997. Effects of a modified-live virus vac-
A. P. 1992. Epidemiologic studies on porcine epidemic cine against porcine reproductive and respiratory syn-
abortion and respiratory syndrome (PEARS) in Lower drome in boars. Am J Vet Res 58:40–45.
Saxony of Germany. Proc Int Congr Pig Vet Soc Collins, J. E., and Dial, G. 1996. Clinical signs of porcine re-
12:115. productive and respiratory syndrome. In Proc Swine
Cafruny, W. A., and Hovinen, D. E. 1988. The relationship Health Summit, Nat Pork Producers Council, pp. 41–51.
between route of infection and minimum infectious Collins, J. E.; Benfield, D. A.; Goyal, S. M.; and Shaw, D. P.
dose: Studies with lactate dehydrogenase-elevating 1990. Experimental transmission of swine reproductive
virus. J Virol Methods 20:265–268. failure syndrome (mystery swine disease) in gnotobiotic
Carman, S.; Sanford, S. E.; and Dea, S. 1995. Assessment of pigs. Conf Res Workers in Anim Dis Abstr 2.
seropositivity to porcine reproductive and respiratory Collins, J. E.; Benfield, D. A.; Christianson, W. T.; Harris, L.;
syndrome (PRRS) virus in swine herds in Ontario: 1978 Hennings, J. C.; Shaw, D. P.; Goyal, S. M.; McCullough,
to 1982. Can Vet J 36:776–777. S.; Morrison, R. B.; Joo, H. S.; Gorcyca, D.; and Chladek,
Carvalho, L.; Segales, J.; and Pijoan, C. 1995. Effect of D. 1992. Isolation of swine infertility and respiratory
PRRSV on subsequent Pasteurella multocida challenge in syndrome virus (isolate ATCC VR-2332) in North Amer-
pigs. In Proc 2d Int Symp on Porcine Reproductive and ica and experimental reproduction of disease in gnoto-
Respiratory Syndrome, Copenhagen, Denmark, p. 20. biotic pigs. J Vet Diagn Invest 4:117–126.
Cavanagh, D. 1997. Nidovirales: A new order comprising Collins, J. E.; Dee, S.; Halbur, P.; Keffaber, K.; Lautner, B.;
Coronaviridae and Arteriviridae. Arch Virol 142:629–633. McCaw, M.; Rodibaugh, M.; Sanford, E.; and Yeske, P.
Cho, H. J.; Deregt, D.; and Joo, H. S. 1996. An ELISA for 1996. Laboratory diagnosis of porcine reproductive and
porcine reproductive and respiratory syndrome: Pro- respiratory syndrome (PRRS) virus infection. Swine
duction of antigen of high quality. Can J Vet Res Health and Prod 4(1):33–35.
60:89–93. Conzelman, K. K.; Visser, N.; Van Woensel, P.; and Thiel, H.
Choi, C.; Gustafson, K.; Chinsakchai, S.; Hill, H.; and Moli- J. 1993. Molecular characterization of porcine reproduc-
tor, T. 1994. Heterogeneity of porcine alveolar tive and respiratory syndrome virus, a member of the
macrophage subpopulations: Immune functions and arterivirus group. Virology 193:329–339.
susceptibility to PEARS virus. Proc Int Congr Pig Vet Soc Dea, S.; Bilodeau, R.; Sauvageau, R.; and Martineau, G. P.
13:97. 1990. Virus isolations from farms in Quebec experienc-
Christianson, W. T.; Collins, J. E.; Benfield, D. A.; Harris, L.; ing severe outbreaks of respiratory and reproductive
Gorcyca, D. E.; Chladek, D. W.; Morrison, R. B.; and Joo, problems. In Proc Mystery Swine Dis Comm Meet Livest
H. S. 1992. Experimental reproduction of swine infertil- Conserv Inst, Denver, Colo., pp. 67–72.
ity and respiratory syndrome in pregnant sows. Am J Vet Dea, S.; Bilodeau, R.; Athanassious, R.; Sauvageau, R.; and
Res 53:485–488. Martineau, G. P. 1992. Swine reproductive and respira-
Christianson, W. T.; Choi, C. S.; Collins, J. E.; Molitor, T. W.; tory syndrome in Quebec: Isolation of an enveloped
Morrison, R. B.; and Joo, H. S. 1993. Pathogenesis of virus serologically-related to Lelystad virus. Can Vet J
porcine reproductive and respiratory syndrome virus in- 33:801–808.
fection in mid-gestation sows and fetuses. Can J Vet Res Dea, S.; Sawyer, N.; Alain, R.; and Athanassious, R. 1995. Ul-
57:262–268. trastructural characteristics and morphogenesis of
Christopher-Hennings, J.; Nelson, E. A.; Hines, R. J.; Nelson, porcine reproductive and respiratory syndrome virus
J. K.; Swenson, S. L.; Zimmerman, J. J.; Chase, C. C. L.; propagated in the highly permissive MARC-145 cell
Yaeger, M. J.; and Benfield, D. A. 1995a. Persistence of clone. In Corona- and Related Viruses. Ed. P. J. Talbot
porcine reproductive and respiratory syndrome virus in and G. A. Levy. New York: Plenum Press, pp. 95–98.
serum and semen of adult boars. J Vet Diagn Invest Dee, S. A. 1996. The decision on using PRRS vaccine in the
7:456–464. breeding herd: When and how to use it. In Proc 23d An-
Christopher-Hennings, J.; Nelson, E. A.; Nelson, J. A.; Hines, nu Meet Allen D. Leman Swine Conf, pp. 143–146.
R. J.; Swenson, S. L.; Hill, H. T.; Zimmerman, J. J.; Katz, ———. 1997a. Gilt development and PRRS: A strategy for
J. B.; Yaeger, M. J.; Chase, C. C. L.; and Benfield, D. A. the US swine industry. Comp Cont Ed Food Anim Med
1995b. Detection of porcine reproductive and respirato- Management 19(9):228–233.
ry syndrome virus in boar semen by PCR. J Clin Micro- ——— 1997b. An overview of strategies to prepare naive
biol 33:1730–1734. gilts for impending PRRS virus challenge: A global per-
Christopher-Hennings, J.; Nelson, E.; Nelson, J.; Rossow, K.; spective. Swine Health and Production 5(6):231–239.
226 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Dee, S. A., and Joo, H. S. 1994a. Prevention of PRRS virus pigs in Sweden. Epidémiol Santé Anim 31–32:07.B.39.
spread in endemically infected swine herds by nursery Epperson, B., and Holler, L. 1997. An abortion storm and
depopulation. Vet Rec 135:6–9. sow mortality syndrome. In Proc 28th Annu Meet Am
———. 1994b. Clinical investigation of recurrent repro- Assoc Swine Pract, pp. 479–484.
ductive failure associated with PRRS virus infection in a European Commission, Directorate General for Agriculture.
sow herd. J Am Vet Med Assoc 205(7):1017–1018. 1991. The new pig disease: Conclusions reached at the
Dee, S. A.; Morrison, R. B.; and Joo, H. S. 1993. Eradication seminar. In The New Pig Disease: Porcine Respiratory
of PRRS virus using multi-site production and nursery and Reproductive Syndrome. A Report on the Semi-
depopulation. Swine Health and Production 1(5):20–23. nar/Workshop Held in Brussels on 29–30 Apr 1991, pp.
Dee, S. A.; Joo, H. S.; and Pijoan, C. 1994. Controlling the 82–86.
spread of PRRS virus in the breeding herd through man- Faaberg, K. S., and Plagemann, P. G. W. 1995. The envelope
agement of the gilt pool. Swine Health and Production proteins of lactate dehydrogenase-elevating virus and
3(2):64–69. their membrane topography. Virology 212:512–525.
Dee, S. A.; Joo, H. S.; Henry, S.; Tokach, L.; Park, B. K.; Moli- Feitsma, H.; Grooten, H. J.; and Schie, F. W. 1992. The effect
tor, T. W.; and Pijoan, C. 1996. Detecting subpopula- of porcine epidemic abortion and respiratory syndrome
tions after PRRS virus infection in large breeding herds (PEARS) on sperm production. Proc Int Congr Anim Re-
using multiple serologic tests. Swine Health and Produc- prod 12:1710–1712.
tion 4(4):181–184. Galina, L.; Pijoan, C.; Sitjar, M.; Christianson, W. T.;
Dee, S. A.; Joo, H. S.; Park, B. K.; Molitor, T. W.; and Bruna, Rossow, K.; and Collins, J. E. 1994. Interaction between
G. 1997a. Attempted elimination of PRRS virus from a Streptococcus suis serotype 2 and porcine reproductive
seedstock farm using breeding herd vaccination and and respiratory syndrome virus in specific pathogen-
nursery depopulation. Vet Rec (accepted for publica- free piglets. Vet Rec 134:60–64.
tion). Garner, M. G.; Gleeson, L. J.; Martin, R.; and Higgins, P.
Dee, S. A.; Joo, H. S.; Polson, D. D.; and Marsh, W. E. 1997b. 1996. Report on the national serological survey for PRRS
Evaluation of the effects of nursery depopulation on the in Australia. Pig Research and Development Corpora-
persistence of PRRS virus and the reproductivity of 34 tion Project No. BRS 1/1037. Animal and Plant Health
pig farms. Vet Rec 140:247–248. Branch, Bureau of Resource Sciences, PO Box E11,
Dee, S. A.; Joo, H. S.; Polson, D. D.; Park, B. K.; Pijoan, C.; Queen Victoria Terrace, Parkes ACT 260.
Molitor, T. W.; Collins, J. E.; and King, V. 1997c. An eval- Geue, A. 1995. Untersuchungen zur Pravalenz und Inzidenz
uation of nursery depopulation on 34 pig farms: Effect des Porcine Reproductive and Respiratory Syndrome
on PRRS virus persistence and productivity. Vet Rec (ac- (PRRS) in einem Kreis Schleswig-Holsteins. Fachbereich
cepted for publication). Veterinärmedizin, Freie Universität, Berlin, Germany.
de Jong, M. F.; Cromwijk, W.; and Van’t Veld, P. 1991. The Gilbert, S. A.; Larochelle, R.; Magar, R.; Cho, H. J.; and
new pig disease: Epidemiology and losses in the Nether- Deregt, D. 1997. Typing of porcine reproductive and res-
lands. In Report of the First EC Seminar/Workshop on piratory syndrome viruses by a multiplex PCR assay. J
“The New Pig Disease, Porcine Reproductive and Respi- Clin Microbiol 35:264–267.
ratory Syndrome,” pp. 9–19. Gorcyca, D.; Schlesinger, K.; Chladek, D.; Behan, W.; Pol-
De Vries, A. F.; Chirnside, E. D.; Horzinek, M. C.; and Rotti- son, D.; Roof, M.; and Doitchenoff, D. 1995. RespPRRS:
er, P. J. M. 1992. Structural proteins of equine arteritis A new tool for the prevention and control of PRRS in
virus. J Virol 66:6294–6303. pigs. In Proc 26th Annu Meet Am Assoc Swine Pract, pp.
Drew, T. W. 1995. Comparative serology of porcine repro- 1–22.
ductive and respiratory syndrome in eight European lab- Gorcyca, D. E.; Schlesinger, K. J.; Geeding, P. W.; Chladek,
oratories, using immunoperoxidase monolayer assay D. W.; and Short, J. A. 1996a. The effect of maternal im-
and enzyme-linked immunosorbent assay. Rev Sci Tech munity on respiratory disease syndrome caused by
Off Int Epiz 14:761–775. porcine reproductive and respiratory syndrome (PRRS)
Drew, T. W.; Meulenberg, J. J. M.; Sands, J. J.; and Paton, D. virus. Proc Int Congr Pig Vet Soc 14:61.
J. 1995. Production, characterization and reactivity of ———. 1996b. Protection against the reproductive disease
monoclonal antibodies to porcine reproductive and res- caused by porcine reproductive and respiratory syn-
piratory syndrome virus. J Gen Virol 76:1361–1367. drome (PRRS) virus by vaccinating with a modified live
Eichhorn, G., and Frost J. W. 1997. Study on the suitability virus PRRS vaccine prior to breeding. Proc Int Congr Pig
of sow colostrum for the serological diagnosis of Vet Soc 14:66.
porcine reproductive and respiratory syndrome (PRRS). Gordon, S. C. 1992. Effects of blue-eared pig disease on a
Zentralbl Veterinärmed (B) 44:65–72. breeding and fattening unit. Vet Rec 130:513–514.
Elvander, M.; Larsson, B.; Engvall, A.; Klingeborn, B.; and Goyal, S. M. 1993. Porcine reproductive and respiratory
Gunnarsson, A. 1997. Nation-wide surveys of syndrome: Review article. J Vet Diagn Invest 5:656–664.
TGE/PRCV, CSF, PRRS, SVD, L. pomona, and B. suis in Gradil, C.; Dubuc, C.; and Eaglesome, M. D. 1996. Porcine
CHAPTER 18 PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME Benfield et al. 227

reproductive and respiratory syndrome virus: Seminal challenge. In Proc 27th Annu Meet Am Assoc Swine
transmission. Vet Rec 138:521–522. Pract, pp. 103–105.
Gravell, M.; London, W. T.; Leon, M.; Palmer, A. E.; and Hesse, R. A.; Couture, L. P.; Lau, M. L.; Wunder, K. K.; and
Hamilton, R. S. 1986. Elimination of persistent simian Wasmoen, T. L. 1996b. Efficacy of Prime Pac PRRS in
hemorrhagic fever (SHF) virus infection in patas mon- controlling PRRS reproductive disease: Heterologous
keys (42244). Proc Soc Exp Biol Med 181:219–225. challenge. In Proc 27th Annu Meet Am Assoc Swine
Grosse-Beilage, E. 1995. Significance of PRRS virus infec- Pract, pp. 107–110.
tions for respiratory tract infections in swine: A litera- Hirose, O.; Kudo, H.; Yoshizawa, S.; Hiroike, T.; and
ture review. Dtsch Tierarztl Wochenschr 102:457–469. Nakane, T. 1995. Prevalence of porcine reproductive
Grosse-Beilage, E., and Grosse-Beilage, T. 1992. Epidemio- and respiratory syndrome virus in Chiba prefecture. J
logical investigations into PEARS in Germany: Influence Jpn Vet Med Assoc 48:650–653.
on reproduction. Proc Int Congr Pig Vet Soc 12:125. Holland, J. 1995. Replication error, quasispecies popula-
Halbur, P. G., and Bush, E. 1997. Update on abortion storms tions, and extreme evolution rates of RNA viruses. In
and sow mortality. Swine Health and Production Emerging Viruses. Ed. S. S. Morse. New York: Oxford
5(2):73. University Press, pp. 203–218.
Halbur, P. G.; Andrews, J. J.; Huffman, E. L.; Paul, P. S.; Hooper, C. C.; Van Alstine, W. G.; Stevenson, G. W.; and
Meng, X.-J.; and Niyo, Y. 1994. Development of a strep- Kanitz, C. L. 1994. Mice and rats (laboratory and feral)
tavidin-biotin immunoperoxidase procedure for the de- are not a reservoir for PRRS virus. J Vet Diagn Invest
tection of porcine reproductive and respiratory syn- 6:13–15.
drome virus antigen in porcine lung. J Vet Diagn Invest Hopper, S. A.; White, M. E.; and Twiddy, N. 1992. An out-
6:254–257. break of blue-eared pig disease (porcine reproductive
Halbur, P. G.; Paul, P. S.; Frey, M. L.; Landgraf, J.; Ernissee, and respiratory syndrome) in four pig herds in Great
K.; Meng, X.-J.; Lum, M. A.; Andrews, J. J.; and Rathje, J. Britain. Vet Rec 131:140–144.
A. 1995. Comparison of the pathogenicity of two U.S. Houben, S.; Callebaut, M. B.; and Pensaert, M. B. 1995a.
porcine reproductive and respiratory syndrome virus Comparative study of a blocking enzyme-linked im-
isolates with that of the Lelystad virus. Vet Pathol munosorbent assay and the immunoperoxidase mono-
32:648–660. layer assay for the detection of antibodies to the porcine
Halbur, P. G.; Paul, P. S.; Frey, M. L.; Landgraf, J.; Eernisse, reproductive and respiratory syndrome virus in pigs. J
K.; Meng, X.-J.; Andrews, J. J.; Lum, M. A.; and Rathje, J. Virol Methods 51:125–128.
A. 1996a. Comparison of the antigen distribution of Houben, S.; van Reeth, K.; and Pensaert, M. B. 1995b. Pat-
two U.S. porcine reproductive and respiratory syn- tern of infection with the porcine reproductive and res-
drome virus isolates with that of the Lelystad virus. Vet piratory syndrome virus on swine farms in Belgium.
Pathol 33:159–170. Zentralbl Veterinärmed (B) 42:209–215.
Halbur, P. G.; Paul, P. S.; Meng, X.-J.; Lum, M. A.; Andrews, Jusa, E. R.; Inaba, Y.; Kohno, M.; Mashimo, H.; and Hirose,
J. J.; and Rathje, J. A. 1996b. Comparative pathogenicity O. 1996. Hemagglutination with porcine reproductive
of nine U.S. porcine reproductive and respiratory syn- and respiratory syndrome virus. J Vet Med Sci
drome virus (PRRSV) isolates in a five-week-old cesare- 58:521–527.
an-derived, colostrum-deprived pig model. J Vet Diagn Jusa, E. R.; Inaba, Y.; Kouno, M.; and Hirose, O. 1997. Effect
Invest 8:11–20. of heparin on infection of cells by porcine reproductive
Harty, J. T., and Plagemann, P. G. W. 1988. Formalin inacti- and respiratory syndrome virus. Am J Vet Res
vation of the lactate dehydrogenase-elevating virus re- 58(5):488–491.
veals a major neutralizing epitope not recognized during Kapur, V.; Elam, M. R.; Pawlovich, T. M.; and Murtaugh, M.
natural infection. J Virol 62:3210–3216. P. 1996. Genetic variation in porcine reproductive and
Haynes, J. S.; Halbur, P. G.; Sirinarumitr, T.; Paul, P. S.; respiratory syndrome virus isolates in midwestern Unit-
Meng, X.-J.; and Huffman, E. L. 1997. Temporal and ed States. J Gen Virol 77:1271–1276.
morphologic characterization of the distribution of Keffaber, K. K. 1989. Reproductive failure of unknown etiol-
porcine reproductive and respiratory syndrome virus ogy. Am Assoc Swine Pract Newsl 1:1–10.
(PRRSV) by in situ hybridization in pigs infected with Keffaber, K.; Stevenson, G.; Van Alstine, W.; Kanitz, C.; Har-
isolates of PRRSV that differ in virulence. Vet Pathol ris, L.; Gorcyca, D.; Schlesinger, K.; Schultz, R.; Chladek,
34:39–43. D.; and Morrison, R. 1992. SIRS virus infection in nurs-
Henry, S. 1994. Clinical considerations in “acute” PRRS. In ery/grower pigs. Am Assoc Swine Pract Newsl 4:38–39.
Proc 25th Annu Meet Am Assoc Swine Pract, pp. Kim, H. S.; Kwang, J.; Yoon, I. J.; Joo, H. S.; and Frey, M. L.
231–235. 1993. Enhanced replication of porcine reproductive and
Hesse, R. A.; Couture, L. P.; Lau, M. L.; Dimmick, S. K.; and respiratory syndrome (PRRS) virus in a homogenous
Ellsworth, S. R. 1996a. Efficacy of Prime Pac PRRS in subpopulation of MA-104 cell line. Arch Virol
controlling PRRS reproductive disease: Homologous 133:477–483.
228 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Kono, Y.; Kanno, T.; Shimizu, M.; Yamada, S.; Ohashi, S.; Immunohistochemical detection of porcine reproduc-
Nakamine, M.; and Shirai, J. 1996. Nested PCR for de- tive and respiratory syndrome virus using colloidal gold.
tection and typing of porcine reproductive and respira- Can J Vet Res 57:300–304.
tory syndrome (PRRS) virus in pigs. J Vet Med Sci Magar, R.; Larochelle, R.; Dea, S.; Gagnon, C. A.; Nelson, E.
58:941–946. A.; Christopher-Hennings, J.; and Benfield, D. A. 1995.
Kuwahara, H.; Nunoya, T.; Tajima, M.; Kato, A.; and Same- Antigenic comparison of Canadian and US isolates of
jima, T. 1994. An outbreak of porcine reproductive and porcine reproductive and respiratory syndrome virus
respiratory syndrome in Japan. J Vet Med Sci using monoclonal antibodies to the nucleocapsid pro-
56:901–909. tein. Can J Vet Res 59:232–234.
Lager, K. M., and Ackermann, M. R. 1994. Pulmonary le- Magar, R.; Larochelle, R.; Nelson, E. A.; and C. Charreyre.
sions in fetuses exposed in utero to porcine reproductive 1997. Differential reactivity of a monoclonal antibody
and respiratory syndrome virus. J Vet Diagn Invest directed to the membrane protein of porcine reproduc-
6:480–483. tive and respiratory syndrome virus. Can J Vet Res
Lager, K. M., and Halbur, P. G. 1996. Gross and microscop- 61:69–71.
ic lesions in porcine fetuses infected with porcine repro- Mardassi, H.; Mounir, S.; and Dea, S. 1994a. Identification
ductive and respiratory syndrome virus. J Vet Diagn In- of major differences in the nucleocapsid genes of a Que-
vest 8:275–282. bec strain and European strains of porcine reproductive
Lager K. M., and Mengeling W. L. 1995. Pathogenesis of in and respiratory syndrome virus. J Gen Virol
utero infection in porcine fetuses with porcine repro- 75:681–685.
ductive and respiratory syndrome virus. Can J Vet Res Mardassi, H.; Wilson, L.; Mounir, S.; and Dea, S. 1994b. De-
59:187–192. tection of porcine reproductive and respiratory syn-
Lager, K. M.; Mengeling, W. L.; and Brockmeier, S. L. 1996. drome virus and efficient differentiation between Cana-
Effect of post-coital intrauterine inoculation of porcine dian and European strains by reverse transcription and
reproductive and respiratory syndrome virus on concep- PCR amplification. J Clin Microbiol 32:2197–2203.
tion in gilts. Vet Rec 138:227–228. Mardassi, H.; Mounir, S.; and Dea, S. 1995. Structural gene
Larochelle, R., and Magar, R. 1995. Comparison of im- analysis of a Quebec reference strain of porcine repro-
munogold silver staining (IGSS) with two immunoper- ductive and respiratory syndrome virus (PRRSV). In
oxidase staining systems for the detection of porcine re- Corona- and Related Viruses. Ed. P. J. Talbot and G. A.
productive and respiratory syndrome virus (PRRSV) Levy. New York: Plenum Press, pp. 277–281.
antigens in formalin-fixed tissues. J Vet Diagn Invest Mardassi, H.; Massie, B.; and Dea, S. 1996. Intracellular syn-
7:540–543. thesis, processing, and transport of proteins encoded by
Larochelle, R.; Sauvageau, R.; and Magar, R. 1994. Im- ORFs 5 to 7 of porcine reproductive and respiratory syn-
munohistochemical detection of swine influenza virus drome virus. Virology 221:98–112.
and porcine reproductive and respiratory syndrome Meng, X.-J.; Paul, P. S.; Halbur, P. G.; and Lum, M. A. 1995a.
virus in porcine proliferative and necrotizing pneumo- Phylogenetic analysis of the putative M (ORF 6) and N
nia from Quebec. Can Vet J 35:513–515. (ORF 7) genes of porcine reproductive and respiratory
Larochelle, R.; Mardassi, H.; Dea, S.; and Magar, R. 1996. syndrome virus (PRRSV): Implication for the existence
Detection of porcine reproductive and respiratory syn- of two genotypes of PRRSV in U.S.A. and Europe. Arch
drome virus in cell cultures and formalin-fixed tissues by Virol 140:745–755.
in situ hybridization using a digoxigenin-labeled probe. Meng, X.-J.; Paul, P. S.; Halbur, P. G.; and Morozov, I.
J Vet Diagn Invest 8:3–10. 1995b. Sequence comparison of open reading frames 2
Le Potier, M.-F.; Blanquefort, P.; Morvan, E.; and Albina, E. to 5 of low and high virulence United States isolates of
1997. Results of a control programme for the porcine re- porcine reproductive and respiratory syndrome virus. J
productive and respiratory syndrome in the French Gen Virol 76:3181–3188.
“Pays de la Loire” region. Vet Microbiol 55:355–360. Meng, X.-J.; Paul, P. S.; Morozov, I.; and Halbur, P. G. 1996.
Leyk, W. 1991. Observations in three affected herds in A nested set of six or seven subgenomic mRNAs is
North Rhine Westphalia. In Report of the First EC Sem- formed in cells infected with different isolates of porcine
inar/Workshop on “The New Pig Disease, Porcine Re- reproductive and respiratory syndrome virus. J Gen Vi-
productive and Respiratory Syndrome,” pp. 3–4. rol 77:1265–1270.
Lindhaus, W., and Lindhaus, B. 1991. Tatselhafte Mengeling, W. L.; Lager, K. M.; and Vorwald, A. C. 1994.
Sweinekrankheit. Praktische Tierarztl 25:423–425. Temporal characterization of transplacental infection of
Loula, T. 1991. Mystery pig disease. Agri-practice 12:23–34. porcine fetuses with PRRS virus. Am J Vet Res
Maes, D. 1997. Descriptive epidemiological aspects of the 55:1391–1398.
seroprevalence of five respiratory disease agents in ———. 1995. Diagnosis of porcine reproductive and respi-
slaughter pigs from fattening herds. Epidémiol Santé ratory syndrome. J Vet Diagn Invest 7:3–16.
Anim 31–32:05.B.19. ———. 1996a. An overview on vaccination for porcine re-
Magar, R.; Larochelle, R.; Robinson, Y.; and Dubuc, C. 1993. productive and respiratory syndrome. In Proc 23d Allen
CHAPTER 18 PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME Benfield et al. 229

D. Leman Swine Conf, pp. 139–142. Swine Pract, pp. 563–569.


———. 1996b. Alveolar macrophages as a diagnostic sam- Molitor, T. W.; Tune, K. A.; Shin, J.; Collins, J.; and Kapur, V.
ple for detecting natural infection of pigs with porcine 1997. Applications of TaqManTM PCR in the detection
reproductive and respiratory syndrome virus. J Vet Di- of porcine reproductive and respiratory syndrome
agn Invest 8:238–240. virus. In Proc 24th Allen D. Leman Swine Conf, pp.
Mengeling, W. L.; Lager, K. M.; Vorwald, A. C.; and Brock- 173–175.
meier, S. L. 1996c. Comparison among strains of Moore, C. 1990. Clinical presentation of mystery swine dis-
porcine reproductive and respiratory syndrome virus ease in the growing pig. In Proc Mystery Swine Dis
for their ability to cause reproductive failure. Am J Vet Comm Meet Livest Conserv Inst, Denver, Colo, pp.
Res 57:834–839. 41–49.
Mengeling, W. L.; Vorwald, A. C.; Lager, K. M.; and Brock- ———. 1992. Biosecurity and minimal disease herds. In
meier, S. L. 1996d. Diagnosis of porcine reproductive Veterinary Clinics of North America: Swine Reproduc-
and respiratory syndrome using infected alveolar tion. Ed. R. C. Tubbs and A. D. Leman. Philadelphia: W.
macrophages collected from live pigs. Vet Microbiol B. Saunders and Harcourt Brace Jovanovich, pp.
49:105–115. 461–474.
Mengeling, W. L.; Lager, K. M.; Vorwald, A. C.; Wesley, R. Morrison, R. B.; Collins, J. E.; Harris, L.; Chladek, D. W.;
D.; and Clouser, D. F. 1997. An update of research at the Gorcyca, D. E.; Joo, H. S.; Christianson, W.; Benfield, D.
National Animal Disease Center on current strains of A.; Marsh, W. E.; Goyal, S.; and Annelli, J. F. 1992. Sero-
porcine reproductive and respiratory syndrome (PRRS) epidemiologic investigation of porcine epidemic abor-
virus. In Proc 24th Allen D. Leman Swine Conf, pp. tion and respiratory syndrome (PEARS, PRRS, SIRS).
138–145. Proc Int Congr Pig Vet Soc 12:114.
Mengeling, W. L.; Lager, K. M.; and Vorwald, A. C. 1998. Morrison, R. B.; Gorcyca, D. E.; Spiess, D. E.; Molitor, T. W.;
Clinical effects of porcine reproductive and respiratory Murtaugh, M.; Schlesinger, K. J.; Chladek, D. W.; and
syndrome virus on pigs during the early postnatal inter- Harris, L. L. 1996. Influence of maternal immunity on
val. Am J Vet Res 59:52–55. infection with porcine reproductive and respiratory syn-
Meredith, M. J. 1992. Review of porcine reproductive and drome (PRRS) virus. Proc Int Congr Pig Vet Soc 14:60.
respiratory syndrome. Cambridge, U.K.: Pig Disease In- Murtaugh, M. P.; Elam, M. R.; and Kakach, L. T. 1995. Com-
formation Centre, pp. 1–24. parison of the structural protein coding sequences of
———. 1995. Porcine reproductive and respiratory syn- the VR-2332 and Lelystad virus strains of the PRRS
drome (PRRS). Pig Disease Information Centre, Cam- virus. Arch Virol 140:1451–1460.
bridge, U.K., pp. 5–10. Murtaugh, M. P.; Faaberg, K. S.; Yuan, S.; and Kapur, V.
Meulenberg, J. J. M., and Den Besten, A. P. 1996. Identifica- 1997. Interrelatedness of PRRS virus isolates in North
tion and characterization of a sixth structural protein America. In Proc 24th Allen D. Leman Conf, pp.
of Lelystad virus: The glycoprotein GP2 encoded by 146–149.
ORF2 is incorporated in virus particles. Virology Nelson, E. A.; Christopher-Hennings, J.; Drew, T.;
225:44–51. Wensvoort, G.; Collins, J. E.; and Benfield, D. A. 1993.
Meulenberg, J. J. M.; De Meijer, E. J.; and Moormann, R. J. Differentiation of US and European isolates of porcine
M. 1993a. Subgenomic RNAs of Lelystad virus contain reproductive and respiratory syndrome virus by mono-
a conserved leader-body junction sequence. J Gen Virol clonal antibodies. J Clin Microbiol 31:3184–3189.
74:1697–1701. Nelson, E. A.; Christopher-Hennings, J.; and Benfield, D. A.
Meulenberg, J. J. M.; Hulst, M. M.; De Meijer, E. J.; Moonen, 1994. Serum immune responses to the proteins of
P. L. J. M.; Den Besten, A.; De Kluyer, E. P.; Wensvoort, porcine reproductive and respiratory syndrome (PRRS)
G.; and Moorman, R. J. M. 1993b. Lelystad virus, the virus. J Vet Diagn Invest 6:410–415.
causative agent of porcine epidemic abortion and respi- ———. 1995. Structural proteins of porcine reproductive
ratory syndrome (PEARS) is related to LDV and EAV. Vi- and respiratory syndrome virus (PRRSV). In Corona-
rology 206:155–163. and Related Viruses. Ed. P. J. Talbot and G. A. Levy. New
Meulenberg, J. J. M.; Petersen-Den Besten, A.; De Kluyver, York: Plenum Press, pp. 321–323.
E. P.; Moorman, R. J. M.; Schaaper, W. M. M.; and Nelson, E. A.; Nelson, J. K.; Couture, L. P.; Lau, M. L.;
Wensvoort, G. 1995. Characterization of proteins en- Christopher-Hennings, J.; Chase, C. C. L.; and Hesse, R.
coded by ORFs 2 to 7 of Lelystad virus. Virology A. 1997. A single amino acid substitution allows for the
206:155–163. differentiation of the Prime Pac PRRS vaccine from field
Molitor, T., and Shin, J. 1995. Porcine reproductive and res- isolates of PRRSV. Conf Res Workers in Anim Dis Ab-
piratory syndrome in boars. In Proc 22d Allen D. Leman stract 202.
Swine Conf, pp. 101–102. Notkins, A. L., and Shochat, S. J. 1963. Studies on the mul-
Molitor, T. W.; Xiao, J.; and Choi, C. S. 1996. PRRS virus in- tiplication and the properties of the lactic dehydroge-
fection of macrophages: Regulation by maturation and nase agent. J Exp Med 117:735–747.
activation state. In Proc 27th Annu Meet Am Assoc Notkins, A. L.; Scheel, C.; and Scherp, H. W. 1964. Trans-
230 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

mission of the lactic dehydrogenase agent in normal and virus. Theriogenology 45:383–395.
partially endentulous mice. Nature 20(2):418–419. Prieto, C.; Suarez, P.; Martin-Rillo, S.; Simarro, I.; Solana,
Park, B. K.; Joo, H. S.; Dee, S. A.; and Pijoan, C. 1995. Evalu- A.; and Castro, J. M. 1996c. Effect of porcine reproduc-
ation of an indirect fluorescent IgM antibody test for the tive and respiratory syndrome virus (PRRSV) on devel-
detection of pigs with recent infection of porcine repro- opment of porcine fertilized ova in vitro. Theriogenolo-
ductive and respiratory syndrome virus. J Vet Diagn In- gy 46(4):687–693.
vest 7:544–546. Robertson, I. B. 1992. Porcine reproductive and respiratory
Pirtle, E. C., and Beran, G. W. 1996. Stability of porcine re- syndrome (blue eared pig disease): Some aspects of its
productive and respiratory syndrome virus in the pres- epidemiology. In Proc Soc Vet Epidemiol Prev Med, pp.
ence of fomites commonly found on farms. J Am Vet 24–38.
Med Assoc 208:390–392. Rossow, K. D. 1996. Pathogenesis of Porcine Reproductive
Plagemann, P. G. W. 1996. Lactate dehydrogenase-elevating and Respiratory Syndrome Virus. Ph.D. diss., Univ Min-
virus and related viruses. In Fields Virology, 3d ed. Ed. B. nesota, Minneapolis-St. Paul.
N. Fields, D. M. Knipe, P. M. Howley, et al. Philadelphia: Rossow, K. D,, Bautista, E. M.; Goyal, S. M.; Molitor, T. W.;
Lippincott-Raven, pp. 1105–1120. Murtaugh, M. P.; Morrison, R. B.; Benfield, D. A.; and
Plagemann, P. G. W., and Moenning, V. 1992. Lactate dehy- Collins, J. E. 1994. Experimental porcine reproductive
drogenase-elevating virus, equine arteritis virus, and and respiratory syndrome virus infection in one-, four-,
simian hemorrhagic fever virus: A new group of posi- and 10-week-old pigs. J Vet Diagn Invest 6:3–12.
tive-strand RNA viruses. Adv Virus Res 41:99–192. Rossow, K. D.; Collins, J. E.; Goyal, S. M.; Nelson, E. A.;
Plana Duran, J.; Bastons, A.; Urniza, M.; and Vayreda, M. Christopher-Hennings, J.; and Benfield, D. A. 1995.
1995. Vaccine against porcine reproductive and respira- Pathogenesis of porcine reproductive and respiratory
tory syndrome (PRRS). In Proc 2d Int Symp on Porcine syndrome virus infection in gnotobiotic pigs. Vet Pathol
Reproductive and Respiratory Syndrome, Copenhagen, 32:361–373.
Denmark, p. 37. Rossow, K. D.; Benfield, D. A.; Goyal, S. M.; Nelson, E. A.;
Plana Duran, J.; Climent, I.; Sarraseca, J.; Urniza, A.; Cortes, Christopher-Hennings, J.; and Collins, J. E. 1996a.
E.; Vela, C.; and Casal, J. I. 1997. Baculovirus expression Chronological immunohistochemical detection and lo-
of proteins of porcine reproductive and respiratory syn- calization of porcine reproductive and respiratory syn-
drome virus strain Olot/91: Involvement of ORF3 and drome virus infection in gnotobiotic pigs. Vet Pathol
ORF5 proteins in protection. Virus Genes 14:19–29. 33:551–556.
Pol, J. M.; van-Dijk, J. E.; Wensvoort, G.; and Terpstra, C. Rossow, K. D.; Laube, K. L.; Goyal, S. M.; and Collins, J. E.
1991. Pathological, ultrastructural, and immunohisto- 1996b. Fetal microscopic lesions in porcine reproductive
chemical changes caused by Lelystad virus in experi- and respiratory syndrome virus–induced abortion. Vet
mentally induced infections of mystery swine disease Pathol 33:95–99.
(synonym: porcine epidemic abortion and respiratory Saito, A.; Kanno, T.; Murakami, Y.; Muramatsu, M.; and Ya-
syndrome [PEARS]). Vet Q 13:137–143. maguchi, S. 1996. Characteristics of major structural
Pol, J.; Wagenaar, F.; Broekhuijsen-Davies, J.; and protein coding gene and leader-body sequence in subge-
Wensvoort, G. 1992. The morphogenesis of Lelystad nomic mRNA of porcine reproductive and respiratory
virus in porcine lung alveolar macrophages. Proc Int syndrome virus isolated in Japan. J Vet Med Sci
Congr Pig Vet Soc 12:127. 58:377–380.
Pol, J.; van Leengoed, L.; Stockhofe, N.; and Wensvoort, G. Sanford, E. 1992. Porcine epidemic abortion and respirato-
1995. Dual infections of PRRS/influenza and ry syndrome (PEARS): Establishment and spread be-
PRRS/APP in the respiratory tract. In Proc 2d Int Symp tween 1987 and 1992 in Ontario, Canada. Proc Int Con-
on Porcine Reproductive and Respiratory Syndrome, gr Pig Vet Soc 12:117.
Copenhagen, Denmark, p. 22. Shimizu, M.; Yamada, S.; Murakami, Y.; Morozumi, T.;
Prieto, C.; Suarez, P.; Sanchez, R.; Solana, A.; Simarro, I.; Kobayashi, H.; Mitani, K.; Ito, N.; Kubo, M.; Kimura, K.;
Martin-Rillo, S.; and Castro, J. R. 1994. Semen changes Kobayashi, M.; Yamamota, K.; Miura, Y.; Yamamoto, T.;
in boars after experimental infection with porcine epi- and Watanabe, K. 1994. Isolation of porcine reproduc-
demic abortion and respiratory syndrome (PEARS) tive and respiratory syndrome (PRRS) virus from Heko-
virus. Proc Int Congr Pig Vet Soc 13:98. Heko disease of pigs. J Vet Med Sci 56:389–391.
Prieto, C.; Sanchez, R.; Martin-Rillo, S.; Suarez, P.; Simarro, Shimizu, M.; Yamada, S.; Kawashima, K.; Ohashi, S.;
I.; Solana, A.; and Castro, J. M. 1996a. Exposure of gilts Shimizu, S.; and Ogawa, T. 1996. Changes of lympho-
in early gestation to porcine reproductive and respirato- cyte subpopulations in pigs infected with porcine repro-
ry syndrome virus. Vet Rec 138(22):536–539. ductive and respiratory syndrome (PRRS) virus. Vet Im-
Prieto, C.; Suarez, P.; Bautista, J. M.; Sanchez, R.; Rillo, S. munol Immunopathol 50:19–27.
M.; Simarro, I.; Solana, A.; and Castro, J. M. 1996b. Se- Shin, J.-H.; Kang, Y.-B.; Kim, Y.-J.; Yeom, S.-H.; Kweon, C.-
men changes in boars after experimental infection with H.; Lee, W.-Y.; Jean, Y.-H.; Hwang, E.-K.; Rhee, J.-C.; An,
porcine reproductive and respiratory syndrome (PRRS) S.-H.; Cho, I.-S.; Oh, J.-S.; Joo, H.-S.; Choi, C.-S.; and
CHAPTER 18 PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME Benfield et al. 231

Molitor, T. W. 1993. Sero-epidemiological studies on ratory syndrome (PRRS) virus–contaminated semen.


porcine reproductive and respiratory syndrome in Ko- Am Assoc Swine Pract Newsl 2:19–23.
rea. I. Detection of indirect fluorescent antibodies. RDA ———. 1995. Preliminary assessment of an inactivated
J Agri Sci 35(2):572–576. PRRS virus vaccine on the excretion of virus in semen.
Shin, J.; Torrison, J.; Choi, C. S.; Gonzalez, S. M.; Crabo, B. Swine Health and Production 3(6):244–247.
G.; and Molitor, T. W. 1997. Monitoring of porcine re- Terpstra, C.; Wensvoort, G.; and Pol, J. M. 1991. Experi-
productive and respiratory syndrome virus infection in mental reproduction of porcine epidemic abortion and
boars. Vet Microbiol 55:337–346. respiratory syndrome (mystery swine disease) by infec-
Solano, G.; Segales, J.; and Pijoan, C. 1995. Interaction be- tion with Lelystad virus: Koch’s postulates fulfilled. Vet
tween porcine reproductive and respiratory syndrome Q 13:131–136.
virus (PRRSV) and Haemophilus parasuis. In Proc 2d Int Terpstra, C.; Wensvoort, G.; and van Leengoed, L. A. M. G.
Symp on Porcine Reproductive and Respiratory Syn- 1992. Persistence of Lelystad virus in herds affected by
drome, Copenhagen, Denmark, p. 21. porcine epidemic abortion and respiratory syndrome.
Stevenson, G. W.; Van Alstine, W. G.; Kanitz, C. L.; and Kef- Proc Int Congr Pig Vet Soc 12:118.
faber, K. K. 1992. Isolation of SIRS virus from nursery Torremorell, M.; Pijoan, C.; Janni, K.; Walker, R.; and Joo,
pigs in two herds without current reproductive failure. H.-S. 1997. Airborne transmission of Actinobacillus pleu-
In Proc Annu Meet Livest Conserv Inst, pp. 253–259. ropneumonia and porcine reproductive and respiratory
———. 1993. Endemic porcine reproductive and respirato- syndrome virus in nursery pigs. Am J Vet Res
ry syndrome virus infection of nursery pigs in two swine 58:828–832.
herds without current reproductive failure. J Vet Diagn Torrison, J.; Vannier, P.; Albina, E.; Madec, F.; and Morri-
Invest 5:432–434. son, R. 1994. Incidence and clinical effect of PRRS virus
Stevenson, G. W.; Van-Alstine, W. G.; and Kanitz, C. L. in gilts on commercial swine farms. Proc Int Congr Pig
1994. Characterization of infection with endemic Vet Soc 13:511.
porcine reproductive and respiratory syndrome virus in Torrison, J.; Knoll, M.; and Weisman, B. 1996. Evidence of
a swine herd. J Am Vet Med Assoc 204:1938–1942. pig-to-pig transmission of a modified-live PRRS virus
Stockhofe-Zurwieden, N.; Navarre Camarro, J. A.; Grosse- vaccine. In Proc 27th Annu Meet Am Assoc Swine Pract,
Beilage, E.; Chevas, J.; and Pohlenz, J. 1993. Uterine and pp. 89–91.
placental alterations in pregnant sows associated with U.S. Department of Agriculture (USDA), Animal and Plant
the porcine epidemic abortion and respiratory syn- Health Inspection Service (APHIS), Veterinary Services
drome (PEARS). J Vet Med B 40:261–271. (VS). 1997. Prevalence of PRRS Virus in the United
Suarez, P.; Zardoya, R.; Martin, M. J.; Prieto, C.; Dopazo, J.; States. NAHMS Info Sheet N225.197.
Solana, A.; and Castro, J. M. 1996. Phylogenetic rela- Van Alstine, W. G.; Kanitz, C. L.; and Stevenson, G. W.
tionships of European strains of porcine reproductive 1993. Time and temperature survivability of PRRS virus
and respiratory syndrome virus (PRRSV) inferred from in serum and tissues. J Vet Diagn Invest 5:621–622.
DNA sequences of putative ORF-5 and ORF-7 genes. Van Alstine, W. G.; Stevenson, G. W.; and Kanitz, C. L.
Virus Res 42:159–165. 1996. Porcine reproductive and respiratory syndrome
Sur, J.-H.; Cooper, V. L.; Galeota, J. A.; Hesse, R. A.; Doster, virus does not exacerbate Mycoplasma hyopneumoniae in-
A. R.; and Osorio, F. A. 1996. In vivo detection of fection in young pigs. Vet Microbiol 49:297–303.
porcine reproductive and respiratory syndrome virus Van Nieuwstadt, A. P.; Meulenberg, J. J. M.; Van Essen-
RNA by in situ hybridization at different times postin- Zandgergen, A.; Den Besten, A. P.; Bende, R. J.; Moor-
fection. J Clin Microbiol 34:2280–2286. mann, R. J. M.; and Wensvoort, G. 1996. Proteins en-
Swenson, S. L.; Hill, H. T.; Zimmerman, J.; Evans, L. E.; coded by open reading frames 3 and 4 of the genome of
Landgraf, J. G.; Wills, R.; McGinley, M.; Brevik, A.; Lelystad virus (Arteriviridae) are structural proteins of
Ciszewski, D.; and Frey, M. L. 1994a. PEARS virus in ex- the virion. J Virol 70:4767–4772.
perimentally infected boars. Proc Int Congr Pig Vet Soc Van Woensel, P.; Van Der Wouw, J.; and Visser, N. 1994. De-
13:58. tection of porcine reproductive and respiratory syn-
Swenson, S. L.; Hill, H. T.; Zimmerman, J. J.; Evans, L. E.; drome virus by polymerase chain reaction. J Virol Meth-
Landgraf, J. G.; Wills, R. W.; Sanderson, T. P.; McGinley, ods 47:273–278.
M. J.; Brevik, A. K.; Ciszewski, D. K.; and Frey, M. L. Vezina, S. A.; Loemba, H.; Fournier, M.; Dea, S.; and Ar-
1994b. Excretion of porcine reproductive and respirato- chambault, D. 1996. Antibody production and blasto-
ry syndrome virus in semen after experimentally in- genic response in pigs experimentally infected with
duced infection in boars. J Am Vet Med Assoc porcine reproductive and respiratory syndrome virus.
204:1943–1948. Can J Vet Res 60:94–99.
Swenson, S. L.; Hill, H. T.; Zimmerman, J. J.; Evans, L. E.; Voicu, I. L.; Silim, A.; Morin, M.; and Elazhary, M. A. S. Y.
Wills, R. W.; Yoon, K. J.; Schwartz, K. J.; Althouse, G. C.; 1994. Interaction of porcine reproductive and respirato-
McGinley, M. J.; and Brevik, A. K. 1994c. Artificial in- ry syndrome virus with swine monocytes. Vet Rec
semination of gilts with porcine reproductive and respi- 134:422–423.
232 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Wensvoort, G. 1993. Lelystad virus and the porcine epi- transmission of porcine reproductive and respiratory
demic abortion and respiratory syndrome. Vet Res syndrome (PRRS) virus in boar semen. Swine Health
24:117–124. and Production 1(5):7-9.
Wensvoort, G.; Terpstra, C.; Pol, J. M.; ter Laak, E. A.; Yoon, I. J.; Joo, H. S.; Christianson, W. T.; Kim, H. S.;
Bloemraad, M.; De Kluyver, E. P.; Kragten, C.; van Collins, J. E.; Morrison, R. B.; and Dial, G. D. 1992. An
Buiten, L.; Den Besten, A.; Wagenaar, F.; et al. 1991. indirect fluorescent antibody test for the detection of
Mystery swine disease in the Netherlands: The isolation antibody to swine infertility and respiratory syndrome
of Lelystad virus. Vet Q 13:121–130. virus in swine sera. J Vet Diagn Invest 4:144-147.
Wensvoort, G.; De Kluyver, E. P.; Luijtze, E. A.; Den Besten, Yoon, I. J.; Joo, H. S.; Christianson, W. T.; Morrison, R. B.;
A.; Harris, L.; Collins, J. E.; Christianson, W. T.; and and Dial, G. D. 1993. Persistent and contact infection in
Chaldek, D. 1992a. Antigenic comparison of Lelystad nursery pigs experimentally infected with porcine repro-
virus and swine infertility and respiratory syndrome ductive and respiratory syndrome (PRRS) virus. Swine
(SIRS) virus. J Vet Diagn Invest 4:134–138. Health and Production 1(4):5-8.
Wensvoort, G.; De Kluyver, E. P.; Pol, J. M. A.; Wagenaar, F.; Yoon, I. J.; Joo, H. S.; Goyal, S. M.; and Molitor, T. W. 1994.
Moorman, R. J. M.; Hulst, M. M.; Bloemraad, R.; Den A modified serum neutralization test for the detection of
Besten, A.; Zetstra, T.; and Terpstra, C. 1992b. Lelystad antibody to porcine reproductive and respiratory syn-
virus, the cause of porcine epidemic abortion and respi- drome virus in swine sera. J Vet Diagn Invest 6:289-292.
ratory syndrome: A review of mystery swine disease re- Yoon, K. J.; Zimmerman, J. J.; McGinley, M. J.; Landgraf,
search at Lelystad. Vet Microbiol 33:185–193. J.; Frey, M. L.; Hill, H. T.; and Platt, K. B. 1995a. Failure
Wesley, R. D.; Mengeling, W. L.; Andreyev, V.; and Lager, K. to consider the antigenic diversity of porcine repro-
M. 1996. Differentiation of vaccine (Strain RespPRRS) ductive and respiratory syndrome (PRRS) virus isolates
and field strains of porcine reproductive and respiratory may lead to misdiagnosis. J Vet Diagn Invest 7:386-
syndrome virus by restriction enzyme analysis. In Proc 387.
27th Annu Meet Am Assoc Swine Pract, pp. 141–143. Yoon, K. J.; Zimmerman, J. J.; Swenson, S. L.; McGinley, M.
White, M. 1991. Blue ear disease of pigs. Vet Rec 128:574. J.; Eernisse, K. A.; Brevik, A.; Rhinehart, L. L.; Frey, M.
White, M. E. C. 1992a. The clinical signs and symptoms of L.; Hill, H. T.; and Platt, K. B. 1995b. Characterization of
blue-eared pig disease (PRRS). Pig Vet J 28:62–68. the humoral immune response to porcine reproductive
———. 1992b. PRRS: Clinical update. Pig Vet J 29:179–187. and respiratory syndrome (PRRS) virus infection. J Vet
Wills, R. W.; Zimmerman, J. J.; Yoon, K.; Swenson, S. L.; Diagn Invest 7:305-312.
McGinley, M. J.; Hill, H. T.; and Platt, K. B. 1995a. Yoon, K. J.; Wu, L. L.; Zimmerman, J. J.; Hill, H. T.; and
Porcine reproductive and respiratory syndrome virus: Platt, K. B. 1996. Antibody-dependent enhancement
Isolation from chronically infected swine. In Proc 26th (ADE) of porcine reproductive and respiratory syn-
Annu Meet Am Assoc Swine Pract, pp. 387–389. drome virus (PRRSV) infection in pigs. Viral Immunol
Wills, R.; Zimmerman, J.; Yoon, K.-J.; Swenson, S.; McGin- 9:51–63.
ley, M.; Hill, H.; Platt, K.; Christopher-Hennings, J.; and Zeman, D.; Neiger, R.; Yaeger, M.; Nelson, E.; Benfield, D.;
Nelson, E. 1995b. Porcine reproductive and respiratory Leslie-Steen, P.; Thomson, J.; Miskimins, D.; Daly, R.;
syndrome virus: A persistent infection. In Proc 2d Int and Minehart, M. 1993. Laboratory investigation of
Symp on Porcine Reproductive and Respiratory Syn- PRRS virus infection in three swine herds. J Vet Diagn
drome, Copenhagen, Denmark, p. 19. Invest 5:522–528.
Wills, R. W.; Zimmerman, J. J.; Swenson, S. L.; Yoon, K.–J.; Zimmerman, J.; Sanderson, T.; Eernisse, K.; Hill, H.; and
Hill, H. T.; Bundy, D. S.; and McGinley, M. J. 1997a. Frey, M. 1992. Transmission of SIRS virus from conva-
Transmission of porcine reproductive and respiratory lescent animals to commingled penmates under experi-
syndrome virus by direct, close, or indirect contact. mental conditions. Am Assoc Swine Pract Newsl
Swine Health and Production (in press). 4(4):27.
Wills, R. W.; Zimmerman, J. J.; Yoon, K.–J.; Swenson, S. L.; Zimmerman, J.; Epperson, W.; Wills, R. W.; and McKean, J.
Hoffman, L. J.; McGinley, M. J.; Hill, H. T.; and Platt, K. D. 1997a. Results of the recent survey of the member-
B. 1997b. Porcine reproductive and respiratory syn- ship of the AASP for outbreaks of sow abortion and
drome virus: Routes of excretion. Vet Microbiol 57:69- mortalilty. Swine Health and Production 5(2): 74–75.
81. Zimmerman, J. J.; Yoon, K. J.; Pirtle, E. C.; Wills, R. W.;
Wills, R. W, Zimmerman, J. J.; Yoon, K.–J.; Swenson, S. L.; Sanderson, T. J.; and McGinley, M. J. 1997b. Studies of
McGinley, M. J.; Hill, H. T.; Platt, K. B.; porcine reproductive and respiratory syndrome (PRRS)
Christopher–Hennings, J.; and Nelson, E. A. 1997c. virus infection in avian species. Vet Microbiol
Porcine reproductive and respiratory syndrome virus: A 55:329–336.
persistent infection. Vet Microbiol 55:231-240. Zimmerman, J. J.; Yoon, K.-J.; Wills, R. W.; and Swenson, S.
Yaeger, M. J.; Prieve, T.; Collins, J.; Christopher–Hennings, L. 1997c. General overview of PRRS virus: A perspective
J.; Nelson, E.; and Benfield, D. 1993. Evidence for the from the United States. Vet Microbiol 55:187–196.
Pseudorabies
19 (Aujeszky’s Disease)
J. P. Kluge, G. W. Beran, H. T. Hill, and K. B. Platt

The history of pseudorabies (PR) has been summarized host ranges. Other biological characteristics of the alpha
by Baskerville et al. (1973) and more recently by herpesviruses include lytic replication cycles of less than
Wittmann and Rziha (1989). The disease was first de- 24 hours and the ability to establish latent infections in
scribed in the United States in 1813 in cattle. The cattle sensory ganglia of the nervous system and lymphoid tis-
suffered from extreme pruritus and eventually died; con- sue of the tonsils (Wheeler and Osorio 1991).
sequently the disease was termed “mad itch.” The term The pig is the only natural host of PR virus, which ac-
“pseudorabies” was first used in Switzerland in 1849 be- counts for its ability to be subclinically and latently in-
cause the clinical signs in cattle were similar to those of fected. Other common farm animals that the virus in-
rabies. In 1902 Aujeszky established the etiologic agent fects are cattle, sheep, goats, cats, and on rare occasions
as nonbacterial, and in 1910 Schmiedhofer confirmed horses. Infection of these animals is lethal, although
that the agent was viral by filtration experiments. In there is one report documenting the survival of an in-
1934 Sabin and Wright identified the virus as a her- fected cow. Several wild animal species are also suscepti-
pesvirus that was immunologically related to herpes sim- ble to lethal PR virus infection. The most common of
plex and herpes B viruses. these species are the raccoon, opossum, rat, and mouse,
Prior to the 1960s the disease was important in East- all of which are frequently found around farms. Experi-
ern Europe but not considered of economic importance mental studies in nonhuman primates have shown that
in the United States. Subsequently, PR has emerged as an chimpanzees and Barbary apes are refractory to PR virus
important disease in the United States and most areas of infection but rhesus monkeys and marmosets are sus-
the world where swine are raised. Several reasons have ceptible. Reports of human infection have been limited
been postulated for the apparent increase in disease and are inadequately documented. None of these reports
severity, prevalence, and distribution. First, new and have been based on virus isolation. A report in 1987 de-
more virulent viral strains may have emerged. A second scribed three patients in Denmark and France who de-
reason involves the decline of hog cholera and the devel- veloped low-level transient seroconversion to PR virus
opment of effective hog cholera vaccines and specific di- following an illness that was possibly transmitted by
agnostic tests. Prior to fluorescent antibody techniques, cats. The possibility that the transient seroconversions
cases of PR may have been misdiagnosed as hog cholera. were due to a serologic cross-reaction induced by anoth-
Porcine hyperimmune serum was often used in conjunc- er herpesvirus was not considered. The most recent re-
tion with virulent hog cholera virus as part of the vacci- port of possible human infection by PR virus was de-
nation regimen. Shope (1935) demonstrated that most scribed in 1992 (Anusz et al.). Six of seven workers who
of the serum contained both PR and hog cholera viral an- were in direct contact with PR virus–infected cattle in
tibodies, which provided some passive immunity to both Poland developed a transient pruritus which occurred
diseases. The third reason commonly cited is the dra- first on the hands and subsequently spread to the shoul-
matic change in swine management systems during the ders and back.
past 30 years. The advent of total confinement systems PR virus consists of an enveloped nucleocapsid that
with large numbers of animals and the incorporation of surrounds a linear genome of about 145 kb of DNA. The
continuous farrowing have created an environment that PR virus genome is about 30 times the size of the small-
facilitates the maintenance and spread of a virus within est known DNA-containing viral pathogen of swine (i.e.,
herds. porcine parvovirus) and is large enough to code for
about 100 proteins. The overall dimension of the virus
ETIOLOGY ranges from 150 to 180 nm in diameter. The nucleocap-
sid is reported to be 105–110 nm in diameter. The nucle-
PR virus belongs to the alphavirus subfamily of the fam- ocapsid is composed of at least eight proteins ranging in
ily Herpesviridae. Alpha herpesviruses include viruses size from 22.5 to 142 kDa. Proteins with molecular
with both broad (as in the case of PR virus) and narrow weights of 142, 34, and 32 kDa are the most prominent

233
234 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

(Stevely 1975). The viral envelope contains at least nine binant-derived gD and gI (Marchioli et al. 1987; Kost et
structural proteins with molecular weights ranging from al. 1989).
50 to 130 kDa (Hampl et al. 1984; Wittmann and Rziha Only one serotype of PR virus is recognized, although
1989; Klupp et al. 1992). Eight of these proteins have distinct differences between some strains can be demon-
sugar residues attached to them and are referred to as strated by panels of monoclonal antibodies. PR virus
glycoproteins. strains can also be differentiated with a high degree of
Originally these glycoproteins were designated by Ro- certainty by biological and physical markers. Vaccine
man numerals or by their molecular mass (i.e., kilodal- and field strains of PR virus have been reliably differen-
tons). In recent years this system of protein nomencla- tiated by using the heat and trypsin inactivation markers
ture has been increasingly replaced by the letter system in combination with the mouse and rabbit virulence
used to designate the proteins of human herpesvirus markers. Genomic differences, as revealed by restriction
(i.e., herpes simplex virus 1 and 2). The old and new fragment length polymorphism (RFLP), can also be used
nomenclature of PR virus envelope glycoproteins is sum- to reliably differentiate PR virus strains. All of these
marized in Table 19.1. The reason for the change is to fa- characteristics have been shown to be stable in several
cilitate comparison of the biological properties of ho- PR virus strains after serial passage in pigs (Platt 1981;
mologous proteins of different herpesviruses. For Mengeling et al. 1983), which makes them useful in epi-
example, gE of PR virus (formerly referred to as gI) is the zootiological studies. Differentiation of virus strains may
counterpart of gE of herpes simplex virus 1. Other im- also help resolve questions of liability. However, in this
portant proteins that are encoded by the viral genome respect it must be remembered that although it is possi-
include the nonstructural glycoprotein gG (formerly ble to say with a high degree of certainty that strains are
known as gX) and thymidine kinase (TK). Genes that different, it is not possible to say with the same certainty
code for many of the above proteins are not essential for that virus strains with genomic similarities and identical
viral replication. For example, vaccine strains of PR virus biological markers are one and the same.
have been genetically engineered to be deficient in one or
more of the following proteins: gE, gC, gG, and TK. EPIDEMIOLOGY
The role that individual PR viral proteins play in vir-
ulence and in the induction of immunity has been par- A primary outbreak of PR in a naive immunologically
tially characterized. Virulence of PR virus is controlled unprotected herd can be a devastating event, with spread
synergistically by several genes, most notably the genes through the entire herd within 1 week and ending with
encoding glycoproteins gE, gD, gI, and TK (Wittmann more than 90% of suckling pigs dead, nursery pigs stunt-
and Rziha 1989; Kritas et al. 1994; Mulder et al. 1996). ed in their growth, and, depending on virus strains and
Glycoproteins gB, gC, and gD appear to be most impor- severity of exposure, with febrile respiratory disease in
tant with respect to the induction of immunity. This con- older hogs and abortion in those in gestation. PR virus is
clusion is based on observations that monoclonal anti- shed in nasal and oral secretions and is aerosolized in
bodies that represent multiple epitopes of gB neutralize droplets, which are moved rapidly by airflow to suscepti-
PR virus in vitro and are also active in antibody-depen- ble swine within and adjacent to shared airspaces; the
dent cell-mediated cytotoxicity. Specific gB monoclonal virus is also transmitted transplacentally and via vaginal
antibodies also confer protective immunity to mice that mucosa, semen, and milk. The outbreak ends with es-
are experimentally infected with PR virus. Monoclonal sentially every convalescent animal permanently latently
antibodies to gC also neutralize PR virus in vitro and infected (Davies and Beran 1980). If other respiratory in-
confer protective immunity to both mice and pigs. Pro- fectious agents, including Actinobacillus pleuropneumoni-
tective immunity has also been conferred to both mice ae or porcine influenza virus, are present in subclinical or
and swine injected with gD-specific monoclonal anti- low-grade form, they will exacerbate the outbreak fur-
body (Marchioli et al. 1988) or immunized with recom- ther.
A wide variety of factors may prevent this devastating
course in a primary-exposure episode. Vaccination of the
Table 19.1. Comparison of the old and new sys-
tem of nomenclature for the envelope proteins of PR
breeding herd with the genetically engineered gene-
virus deleted vaccines available today will protect the sows and
gilts from acute clinical signs. Depending upon severity
Old New
Nomenclature Nomenclature
of exposure to the field virus, these swine will still be in-
fected and develop latency as they did following in-
gI gE tranasal or intramuscular exposure to the vaccine virus
gIIa, b, and c gB
gIII gC
(Henderson et al. 1991). In the naive unvaccinated herd,
gp50 gD the course of clinical events will depend on the strain of
gp63 gI introduced virus; the stages of gestation in the breeding
gX gG herd; the air, water, and feed management system; the
CHAPTER 19 PSEUDORABIES (AUJESZKY’S DISEASE) Kluge, Beran, Hill, Platt 235

size and age segregation of the herd; the overall health raccoons, and skunks are moderately susceptible; and
maintenance of the herd; and the sanitary practices in rats and mice are less susceptible to direct exposure by
operation. Pigs nursing immunized sows or gilts will be ingestion of dead swine carcasses or indirect exposure by
protected from all but overwhelming exposures. inhalation of aerosolized virus or ingestion of contami-
Characteristically, the swine herd returns to normal nated feed or water. The incubation periods in these ani-
productivity following the primary episode, with the la- mals are commonly short, within 3 days; the clinical pe-
tently infected pigs being marketed while the breeding riods are characterized by rapidly progressing
herd remains latently infected. Reactivation of latent in- encephalitis with variable pruritus; and death is certain,
fections can occur at any time of stress (Davies and Be- usually within 2–3 days. The short incubation and clini-
ran 1980), but intensive vaccination, usually recom- cal periods usually restrict transmission to a single farm,
mended to be performed 3–5 times each year, reduces with swine exposed by ingestion of contaminated feed,
occurrence, levels, and duration of shedding. Only ex- water, or animal carcasses. Occasionally dogs drag car-
ceptionally—and usually only in small herds with casses of pigs that died of PR from one area to another,
turnover of the breeding stock and their replacement or rodents that died of PR are unknowingly milled in
with vaccinated gilts within 2 years or less—will the en- swine feed or are present in bedding from infected farms.
demicity be eliminated. Usually, a rigorous combination There is little field or experimental evidence that do-
of herd cleanups and protective interventions are need- mestic or synanthropic birds play any role in PR virus
ed, including intensive vaccination of the breeding herd transmission. Experimental oral exposures have been un-
(De Smet et al. 1992) and, if infected, of finisher pigs successful. Externally contaminated feed rapidly loses in-
with one or two doses of vaccine each between 10 and 16 fectivity (Beran 1991). Insect transmission has not been
weeks of age (De Smet et al. 1994). Intensive culling of demonstrated as playing an epizootiological role, al-
seropositive breeding stock and replacement by infec- though PR virus experimentally fed to houseflies re-
tion-free gilts, strong biosecurity and sanitation mea- mained viable in the gut with a half-life of 13 hours at
sures, all-in/all-out movement and segregation by age, 10˚C and 3 hours at 30˚C. Results of experimental expo-
and personnel access and movement restrictions (Thaw- sure via virus-contaminated flies onto corneas or abrad-
ley et al. 1982) are important. Progress in herd cleanups ed skin of susceptible swine have been variable, usually
must be monitored every 6–12 months and continued as negative (Zimmerman et al. 1989).
long as risk of reexposure is present. Interherd and intraherd PR virus transmission by air
Exposure and reexposure risks are by far the greatest or water or by contaminated fomites (especially move-
from proximate, infected, shedding, introduced swine. ment of feed or bedding from farms of undetermined PR
Epizootiological investigations of over 3000 newly rec- status) is hypothesized with some frequency. PR virus
ognized herd infections in the United States during the has a relatively low survivability compared to nonen-
period 1994–March 1997 reported that 20% were trans- veloped RNA viruses in the environment. It is most sta-
mitted by exposure to introduced, infected breeding ble between pH 6 and 8 in moist environments at cool,
swine and feeder pigs and that 2.0% were transmitted by nonfluctuating temperatures. It is inactivated in 1–7
contact with feral swine. Surveillance studies in the Unit- days at pH 4.3 or 9.7 at temperatures between 37˚C and
ed States have identified PR-seropositive feral swine in 11 4˚C (Davies and Beran 1981). It is highly susceptible to
of 13 sites investigated, with 34.8% of 1662 sera from the drying, especially in the presence of direct sunlight.
11 infected sites in Florida being positive by at least one Times required for infectious levels of PR virus to drop
test (Vander der Leek et al. 1993), and studies in Ger- to noninfectious levels when experimentally suspended
many have yielded 13/640 ELISA test positive findings in porcine saliva and held in contact with moist fomites
(Oslage et al. 1994). at 25˚C are summarized in Table 19.2 (Beran 1993;
So far as has been recognized, all livestock mammals Schoenbaum et al. 1993). Other studies at colder tem-
are susceptible to PR, with transmission from swine to peratures, usually below 4˚C, have shown long survival
other livestock occurring either directly onto mucous times, but PR virus is unstable at fluctuating tempera-
membranes or broken skin, leading to neurological dis- tures, even below freezing (Beran 1993; Davies and Be-
ease, usually with intense pruritus emanating from the ran 1981). Full advantage needs to be taken of these sur-
site of inoculation, or indirectly by aerosol inhalation, vival factors outside the living host in waste
leading to rapidly progressing encephalitic disease. management, in cleaning and disinfecting contaminated
These infected animals almost without exception die by premises, and in biosecurity of swine units.
the second clinical day without having shed PR virus Epizootiological evidence of airborne transmission
through routes which would endanger other swine; that even over distances up to many kilometers is occasional-
is, they are true dead-end hosts. Sheep are highly suscep- ly reported both in the United States and in Europe,
tible, and flocks in contact with swine often inadvertent- sometimes during major atmospheric events, such as fol-
ly serve as sentinels of active or reactivated latent infec- lowing the path of a tornado (Christensen et al. 1993;
tions in the herds. Cats are highly susceptible; dogs, Scheidt et al. 1991). Experimentally, infective levels of
236 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Table 19.2. Survival times of PR in contact with Genetically engineered modified-live-virus vaccines
fomites with compatible serologic tests to differentiate vaccine-
Survival in Saliva at and field-infection-induced antibodies have proven es-
Fomites (at 25˚C) Infectious Levels (Days) sential in area control efforts and in infection cleanups in
Nonfomite control test <4 all but the smallest herds of swine. Although commonly
Steel <4 used only in swine, inactivated vaccines and some of the
Concrete <4 TK gene-deleted live-virus vaccines have been shown to
Plastic <3 be safe for cattle and sheep. While the genetically engi-
Rubber <2
Denim cloth <1 neered vaccines afford relatively long protection of swine
Loam soil <7 from clinical PR, they effectively reduce transmission for
Green grass <2 only a few months, decreasing occurrence level and du-
Shelled corn <4 ration of shedding by reactivation of latent infections.
Pelleted hog feed <3
For a period of weeks to a few months, swine vaccinated
Meat and bone meal <2
Alfalfa hay <1 with these vaccines have enhanced resistance to infection
Straw bedding <4 by field viruses, especially following intranasal vaccina-
Sawdust bedding <2 tion, but infections with reduced shedding and develop-
Swine feed <2 ment of latency must be expected in vaccinates exposed
Well water <7
Chlorinated water <2 to field viruses (Mengeling et al. 1992). Intensive vacci-
Anaerobic lagoon effluent <2 nation programs must be accompanied by rigorous mon-
Swine unit pit effluent <1 itoring with culling and turnover of infected herds, by
preventing exposure of uninfected sectors of the herd to
infected swine, usually in the breeding herd, and by ef-
virus have persisted in aerosols up to 7 hours at 55% or fective biosecurity (Van Oirschot et al. 1996).
higher relative humidity. Droplets of infective salivary or
nasal mucus can carry infective doses over short dis- CONTROL AND ERADICATION
tances and when coalesced into droplet nuclei can be
moved over long distances within the experimental sur- Area control and elimination of infection are being pro-
vival time (Schoenbaum et al. 1990). gressively undertaken by major swine-producing coun-
All of these factors of survival and transmission of tries worldwide. The United Kingdom (Taylor 1989) and
PR virus are classified together into area spread (US- Denmark (Anderson et al. 1989) have achieved major
DA/APHIS/VS 1994–1997). During the period success. The U.S. eradication program was initiated in
1994–March 1997, it was concluded that movement of 1989 with a 10-year projected target for achievement.
contaminated feed or bedding was the source in 1% of Working on a state-by-state basis, this program progress-
the outbreaks investigated. In 38% of the outbreaks in- es through five stages according to a program standards
vestigated, the source could not be differentiated as to in- document that is revised annually by a broad-based com-
terherd movements of companion and wild animals, mittee representing national and state agencies, swine
trucks and other vehicles, people, insects, or air and wa- producers, and researchers (USDA/APHIS/VS 1997).
ter movement. Finally, 39% of these epizootiological in- Stage I is a time of preparation and formation of state
vestigations failed to identify exposure sources. plans and movement regulations. Stage II is a time of
In a cost-benefit study of the U.S. PR eradication pro- surveillance testing for prevalence and location of infect-
gram, productivity and economic impacts were calculat- ed herds and of implementing movement controls and
ed for PR-infected and noninfected herds, including cleanup of infected herds. It is the active period of iden-
preweaning, nursing, grower, and finisher pig mortality, tifying the infected herds statewide and of eliminating
breeding-herd mortality, feed conversion, labor, and vet- infection and preventing transmission to uninfected
erinary and biologic/pharmaceutic expenses. The study herds. Stage III is a time of intense surveillance and
calculated that there was a $6.00/hundredweight reduc- mandatory cleanup of all identified infected herds in the
tion of profitability due to PR herd infection (Anderson state. Area prevalence must be below 1% of herds. Stage
et al. 1989; Miller et al. 1996). IV is a time of continuing surveillance and is reached on-
Recombination has been demonstrated between field ly when no newly infected herds have been identified in
strains of PR virus and between field strains and live-vac- the past 12 months. Vaccination is prohibited except by
cine virus strains, in both experimental and field studies. special permission. Stage V is the certified free state, with
Only in a small number of field studies in Europe have surveillance at a maintenance level and entry of swine in-
prevailing field strains been identified as having recom- to the state only from other Stage IV or V states. Vacci-
binant vaccine virus components (Henderson et al. 1991; nation is prohibited.
Mayfield 1990). The PR eradication program in the United States is
CHAPTER 19 PSEUDORABIES (AUJESZKY’S DISEASE) Kluge, Beran, Hill, Platt 237

based on active partnership of swine producers, state of infected vaccinated breeding swine in the herd with
governments, and the federal government in organiza- the active surveillance based on differential serologic
tion and funding. All three are active in education, both testing until culling and turnover of the breeding herd
for the swine industry and the public. Practicing veteri- has been completed.
narians are active participants in case finding and As of March 1, 1997, the U.S. National Pseudorabies
cleanup of infected herds. Case finding is based on re- Control Board has granted Stage V status to 23 states
porting by veterinarians, by on-farm area testing, by cir- plus Puerto Rico, Stage IV status to 7 states plus U.S. Vir-
cle testing of herds within 1–2 km of identified infected gin Islands, split Stage III/IV status to 1 state, Stage III
herds, and by tracing back from slaughter surveillance status to 13 states, split Stage II/III status to 5 states, and
findings. On-farm area testing and slaughter surveillance Stage II status to 1 state (USDA/APHIS/VS 1994-1997).
are performed by statistical sampling, commonly based
on testing breeding swine at 95% probability of detecting CLINICAL SIGNS
infection in herds in which at least 10%, 20%, or 30% of
swine are seropositive (Table 19.3). The clinical signs associated with PR are primarily de-
A system of qualified negative herds, of qualified pendent on the strain of virus, the infectious dose, and,
negative vaccinated herds, and of monitored feeder pigs most important, the age of the swine affected. Like her-
is based on statistical testing at intervals varying from pesviruses of other animal species, younger swine are the
monthly to yearly. Vaccination is controlled on a state- most severely affected by PR virus. The virus has a
by-state basis; only genetically engineered vaccines with predilection for respiratory and nervous tissue; thus the
deletion of at least the gene encoding glycoprotein gE, majority of clinical signs are associated with dysfunction
with companion differential serologic tests, are permit- of these two organ systems. Generally, nervous signs are
ted, except in special conditions. more commonly observed in suckling and weaned pigs,
Herd cleanup plans are individually developed and while respiratory signs are observed in finisher pigs and
implemented in infected herds, adapting four basic adult swine.
plans. Cleaning and disinfection of individual swine The response to a PR viral infection can differ
units or entire premises are essential components. In de- markedly between herds. The disease may manifest itself
population-repopulation, the entire herd is removed, the as a rapidly spreading disease affecting all ages of swine
premises are cleaned and disinfected, and the farm is re- on the farm, or it may be a completely inapparent infec-
populated with clean stock. In herds with low prevalence tion detected only when a serologic evaluation of the
of infection (usually with fewer than 20% seropositive herd is made. PR is more often inapparent when the in-
breeding stock and all immature swine negative), testing fection occurs while neonatal pigs are not present, as in a
of the entire breeding herd, removal of the positive pigs, period between farrowing groups in a group-type farrow-
and active continuing surveillance testing may be effec- ing operation. Inapparent herd infections rarely occur
tive. Offspring segregation involves segregated rearing of when PR virus is introduced into a herd for the first time
progeny of infected breeding herds with rapid removal where neonatal pigs are present, because pigs of this age
of the breeding stock following weaning and monitoring are so highly susceptible. Inapparent infections in breed-
of all gilts raised as replacements. Active surveillance in- ing herds or separate finishing barns may be mild PR res-
cludes two negative tests 90 days apart and again at 12 piratory infections that are ignored or misdiagnosed as
months to declare the herd free of infection. Offspring some other disease, such as swine influenza.
segregation with intensive vaccination of all breeding The first clinical signs observed in a farrow-to-finish
stock is permitted in some programs, allowing retention herd will vary depending on the age group first infected.
Typically, the first clinical signs will be a few gilts or sows
Table 19.3. Size of the sample required to detect aborting; or finisher pigs coughing and becoming listless
reactor animals by population size for 95% and 99% and anorectic; or rough-haired suckling pigs that become
certainty of detection listless, anorectic, and within 24 hours ataxic and con-
Number of Reactor Animals in the Population vulsive. If any of these scenarios occurs, immediate di-
agnosis is imperative, because early vaccination in the
≥10% ≥20% ≥30%
face of an outbreak can limit losses.
Population Size 95% 99% 95% 99% 95% 99%
≤100 25% 36% 13% 19% 9% 13% Neonatal Pigs
101–200 27% 40% 13% 20% 9% 13% The incubation period in suckling pigs is usually quite
201–300 28% 41% 14% 20% 9% 13% short, ranging from 2 to 4 days. Before severe clinical
301–500 28% 42% 14% 21% 9% 13% signs are noted, suckling pigs will become listless,
501–1000 29% 43% 14% 21% 9% 13%
anorectic, and febrile (41˚C). Some pigs will develop cen-
>1000 29% 44% 14% 21% 9% 13%
tral nervous system (CNS) signs within 24 hours of the
238 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

onset of clinical signs, which progress from trembling, and lose considerable body weight. The duration of clin-
hypersalivation, incoordination, ataxia, and nystagmus ical signs is usually 6–10 days, and recovery is rapid once
to opisthotonos, to severe epileptiform-like seizures. Af- the fever disappears and the appetite returns. Although
fected pigs may sit like dogs because of posterior paraly- compensatory gain negates some of the lost body
sis, while others will circle or become recumbent and weight, grower-finisher pigs will lose at least a week in
paddle. Vomiting and diarrhea may also occur, but nei- the production cycle. Losses may be dramatically in-
ther of these clinical signs is consistent. Pigs with CNS creased if a PR virus infection is superimposed on an A.
signs usually die within 24–36 hours of onset. Mortality pleuropneumoniae infection. It has been reported that PR
in suckling pigs is quite high, often approaching 100%. In virus inhibits the function of alveolar macrophages (Igle-
herds with dams of varying immune status to PR virus, sias et al. 1989), thus reducing the capacity of these de-
clinical signs may be seen in some litters, or pigs within a fense cells to process and destroy bacteria.
litter, while neighboring litters or littermates remain nor-
mal. If susceptible sows or gilts are infected close to Adult Swine
term, pigs may be born weak and show clinical signs im- Sows and boars develop clinical signs, primarily respira-
mediately and die within the first day or two of life. tory in nature, very similar to those described above for
grower-finisher pigs. Pregnant gilts often abort, and in a
Weaned Pigs (3–9 Weeks) farrow-to-finish operation this may be the first clinical
Younger pigs in this age group tend to have clinical signs sign observed. Pregnant females infected with PR virus
similar to those described for suckling pigs. However, the in the first trimester may resorb the fetuses and return to
clinical signs are less severe and fewer pigs develop the estrus. Reproductive failure due to PR in the second or
severe CNS involvement, which invariably leads to coma third trimester is usually manifested by abortion or still-
and death. Mortality in 3- to 4-week-old pigs may ap- born or weak pigs if the time of infection is close to term.
proach 50% in severe outbreaks. Older pigs in this age If the gilt or sow is very close to farrowing when infected,
group become listless, anorectic, and febrile (41–42˚C) pigs may be born with PR, in which case they die within
within 3–6 days of exposure. Often, respiratory signs are a day or two. The PR virus can cross the placenta and in-
present, characterized by sneezing, nasal discharge, and fect and kill fetuses in utero, resulting in abortion. Fortu-
dyspnea, and progress to a severe cough. Pigs displaying nately, reproductive failure usually has a low incidence,
these clinical signs will lose significant body condition occurring in 20% or fewer of the pregnant females on a
and weight. Duration of clinical signs is usually only farm. Mortality in gilts, sows, and boars infected with PR
5–10 days; most pigs make a rapid full recovery once the virus rarely exceeds 2%.
fever and anorexia disappear. Pigs that develop CNS
signs often die, as do pigs with a PR viral respiratory in- PATHOGENESIS
fection that develop a secondary or concurrent bacterial
infection such as Pasteurella multocida or Actinobacillus The pathogenesis is variable depending on viral strain,
pleuropneumoniae. However, the mortality in pigs 5–9 age of the pig, size of inoculum, and route of infection.
weeks of age, if cared for properly and treated for sec- There is an increased resistance to development of
ondary infections, will usually not exceed 10% and is of- clinical signs with age; strains of low virulence may not
ten lower. The more severely affected pigs that survive, produce clinical signs in adult animals, and the viral
especially those developing CNS signs, will often be replication may be limited locally to the site of introduc-
stunted and sometimes show lasting signs, such as head tion. To produce clinical disease experimentally, a mini-
tilt. These pigs reach market weight 1–2 months after the mum dose of virus is required; however, under field con-
rest of the group. ditions very low quantities of virus may cause pigs to
seroconvert and even become latent carriers although no
Grower-Finisher Swine clinical signs can be detected in the herd. Experimental
Respiratory signs have become the hallmark of PR in infection by intranasal inoculation requires about 10
grower-finisher pigs. Morbidity is usually very high, ap- TCID50 in pigs under 2 weeks of age, about 103 TCID50 in
proaching 100%, but in uncomplicated cases mortality is 6-week-old pigs, and about 104 TCID50 in swine 4 months
low, 1–2%. CNS signs do occur, but only sporadically, of age or older (McCullough 1989).
and they may vary from mild muscle tremors to violent Experimentally, pigs develop infection after inocula-
convulsions. Typically, clinical signs appear in 3–6 days tion via the following routes: intramuscular, -venous,
and are characterized by a febrile response (41–42˚C), -cerebral, -gastric, -nasal, -tracheal, -conjunctival, -uter-
depression, anorexia, and mild to severe respiratory ine, -testicular, and oral. Intranasal inoculation results in
signs. A rhinitis develops causing sneezing and a nasal clinical signs and lesions similar to the natural disease,
discharge that progress to pneumonia, resulting in a and under field conditions the oral-nasal routes are the
harsh cough and labored breathing, especially when the most common.
pigs are forced to move about. These pigs become gaunt Under natural conditions the primary site of replica-
CHAPTER 19 PSEUDORABIES (AUJESZKY’S DISEASE) Kluge, Beran, Hill, Platt 239

tion is epithelium in the nasopharynx and tonsil (Masic This fact should be considered when administering
et al. 1965). From these sites there is lymphatic spread to steroids to animals in an infected herd that is in the
regional lymph nodes and replication in the nodes. The midst of an eradication program. Vaccination with live
virus also spreads via nerves from the primary site of in- or killed vaccines cannot be relied upon to prevent the
fection to the CNS. Examples include spread within axo- development of latency in postvaccination exposed
plasm of the trigeminal nerve to the medulla and pons swine. Some live vaccines have the potential to induce la-
and spread along olfactory nerves and the glossopharyn- tent CNS infection.
geal nerve to the medulla. After replication in neurons in
the medulla and pons, there may be spread to other por- LESIONS
tions of the brain. As noted earlier, with low-virulence
strains and moderate-virulence strains in older animals, Gross Lesions
viral spread may be limited to those locations. Gross lesions are often absent or are minimal and unde-
Strains of higher virulence will follow a course simi- tected. When they are present, they aid in arriving at a
lar to the above, but in addition there is widespread virus tentative diagnosis when combined with herd history
distribution in the body, and older swine will have clini- and clinical signs. Serous to fibrinonecrotic rhinitis is
cal signs and may have gross lesions (McFerran and Dow common but may be missed unless the head is split to ex-
1965; Sabó et al. 1968, 1969; Wittmann et al. 1980). pose the entire nasal cavity (Csontos and Széky 1966;
Probably all strains have a tropism for the upper respira- Olander et al. 1966). The lesions may extend to the lar-
tory tract and the CNS. Strains of higher virulence pro- ynx and even down the trachea. Necrotic tonsillitis is fre-
duce a brief viremia and virus can be demonstrated in quently seen along with swollen and hemorrhagic lymph
the serum and also associated with buffy-coat cells. High- nodes of the oral cavity and upper respiratory tract
virulence strains can be isolated from alveolar (Narita et al. 1984a). When present, lesions in the lower
macrophages (Iglesias et al. 1992), epithelium of termi- respiratory tract range from pulmonary edema to scat-
nal bronchioles, hepatocytes, lymphoid cells of the tered small foci of necrosis, hemorrhage, and/or pneu-
spleen and lymph nodes, adrenal cortical cells, tro- monia (Becker 1964; Corner 1965).
phoblasts and embryos from the gravid uterus, and Keratoconjunctivitis is common and often more obvi-
luteal cells of the ovary. The virus has been isolated from ous in white breeds because of the discoloration caused
semen but probably originated from preputial lesions, by excessive lacrimation and periocular deposits of exu-
since attempts to isolate virus from the testes and acces- date (Schneider and Howarth 1973).
sory sex glands have failed (Gueguen and Aynaud 1980; Typical herpetic yellow-white foci (2–3 mm) of necro-
Medveczky and Szabó 1981). The observed decrease in sis may be scattered through the liver and spleen (Fig.
sperm quality is probably the result of fever and systemic 19.1) and may be seen just beneath the serosal surface.
disease. Embryos are resistant to infection as long as the These lesions are most frequently seen in very young pigs
zona pellucida is intact; thus, associated virus can be that lack passive immunity.
washed from embryos, and embryo transplantation can Sows that have recently aborted may have a mild en-
be used as a method of salvaging genetic material from dometritis, and the wall of the uterus is often thickened
an infected herd (Bolin et al. 1983; Bolin and Bolin and edematous (Kluge and Maré 1978; Hsu et al. 1980).
1984). If the placenta is available for examination, a necrotic
Virus excretion precedes or coincides with the onset placentitis is often observed. Aborted fetuses may appear
of clinical disease; in clinically inapparent infection, ex- fresh, macerated, or occasionally mummified. Some pigs
cretion begins after an incubation period of 2–5 days. PR in an affected litter may be normal and others weak or
virus may be recovered from nasal exudate or secretions dead at birth. Infected fetuses or neonatal pigs frequent-
at levels of approximately 102 TCID50 per swab for 1 to ly have the previously described necrotic foci in liver and
more than 2 weeks and for similar periods but in lesser spleen as well as hemorrhagic necrotic foci in lungs and
quantities from tonsillar scrapings, vaginal secretions, tonsils (Csontos et al. 1962; Kluge and Maré 1976;
prepuce, milk, and occasionally urine (Wittmann and Wohlgemuth et al. 1978). A history of abortion along
Rziha 1989). Active immunity from vaccination or prior with the presence of necrotic foci is strongly suggestive
infection decreases the period of viral shedding. As with of a tentative diagnosis of PR. The only gross lesion re-
other herpesviruses, infection with PR virus results in a ported in the male reproductive tract is scrotal edema.
high percentage of latent infections in host pigs (Rziha et Necrotic enteritis in the lower jejunum and ileum has
al. 1982). The virus persists in ganglia such as the trigem- been reported in young pigs (Narita et al. 1984c).
inal ganglia and in tonsil (Cheung 1995). Latently infect-
ed pigs often have detectable viral recrudescence during Microscopic Lesions
times of stress, such as farrowing, crowding, and trans- Microscopic lesions are most frequently reported in the
port. Experimentally, corticosteroid injection results in CNS and persist for many weeks (12–24 weeks postin-
subsequent viral recrudescence and nasal shed of virus. fection) (Olander et al. 1976). They may also be present
240 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

19.1. Spleen and liver with


multiple small focal areas of
necrosis.

in pigs that failed to develop clinical signs, but they are and thoracic regions. Meninges over affected areas of
often absent in aborted fetuses. Nonsuppurative menin- brain and cord may be thickened because of infiltrates of
goencephalitis and ganglioneuritis are the characteristic mononuclear cells. Similar lesions have been reported in
lesions (Dow and McFerran 1962; Sabó et al. 1969; ganglia and ganglia cells (cerebrospinal, cardiac, and
Baskerville 1972). Lesions are present in both the gray celiac ganglia, Meissner’s and Auerbach’s plexuses, and
and the white matter, and the distribution is dependent the ganglion cell layer of the retina). Intranuclear inclu-
on the route of entry into the CNS. Affected areas are sion bodies may be present in neurons, astrocytes, and
characterized by predominantly mononuclear cell oligodendroglia, but in our experience they are much
perivascular cuffing (Fig. 19.2) and glial nodules. A few more common in lesions outside the nervous system.
granulocytes may be mixed with the mononuclear cells, In tonsils, necrosis begins in the subepithelial area
and pyknosis and karyorrhexis of the latter cells are of- and then spreads to the epithelium and deeper into the
ten prominent. The endothelium of affected vessels ap- lymphoid tissue (Narita et al. 1984c). Intranuclear inclu-
pears normal. Neuronal necrosis may be focal and the sion bodies are common in crypt epithelial cells adjacent
neurons surrounded by mononuclear cells, or affected to necrotic foci. The upper respiratory tract lesions con-
neurons may be diffusely scattered. Similar lesions may sist of mucosal epithelial necrosis and submucosal infil-
be present in the spinal cord, especially in the cervical trations of mononuclear cells (Baskerville 1971, 1973).

19.2. Brain with perivascular


cuffing.
CHAPTER 19 PSEUDORABIES (AUJESZKY’S DISEASE) Kluge, Beran, Hill, Platt 241

Lung lesions consist of necrotic bronchitis, bronchioli- droplets, knobbed cystic acrosomes, double heads, and
tis, and alveolitis. Peribronchial mucous gland epitheli- detached heads. These changes may be the result of
um may be necrotic. There is often hemorrhage and fib- pyrexia and not the result of viral infection of spermato-
rin exudation because of involvement of connective genic epithelium.
tissue and endothelium. Lesions often are patchy in ma- Enteric lesions consist of focal necrosis of the mucos-
jor airways, and healing by fibrosis is often observed in al epithelium and may involve the underlying muscularis
areas adjacent to acute lesions. Intranuclear inclusion mucosa and tunica muscularis (Narita et al. 1984b). In-
bodies are frequently present in the epithelial lining of tranuclear inclusion bodies may be present in degenerate
the airways, connective tissue cells, and cells sloughed in- crypt epithelial cells.
to alveolar spaces. There may be necrotizing vasculitis of arterioles,
Focal necrotic lesions are similar regardless of the tis- venules, and lymphatic vessels around tonsils and sub-
sue involved. They are most frequently found in spleen, maxillary lymph nodes (Narita et al. 1984a). Endothelial
liver (Fig. 19.3), lymph nodes, and adrenal glands. nuclei are pyknotic and karyorrhectic, and the vessel
Necrotic foci are randomly distributed and surrounded walls are infiltrated by neutrophils. Intranuclear inclu-
by a few inflammatory cells, or the latter may be absent. sion bodies often are present in affected endothelial cells.
Parenchymal cells at the margins of necrosis commonly Two types of intranuclear inclusion bodies may be
contain intranuclear inclusion bodies (Fig. 19.4). In mac- observed (Corner 1965): a homogenous basophilic body
erated fetuses microscopic necrotic foci often are visible that fills the entire nucleus and an eosinophilic body that
even though cellular detail cannot be discerned since the has a definite halo between it and the marginated chro-
pyknotic nuclei stain with hematoxylin. matin. In either case, specificity of the inclusion must be
Uterine infection is characterized by multifocal to dif- determined by demonstrating viral particles or antigen
fuse lymphohistocytic endometritis and vaginitis and by electron microscopy or immunohistochemistry.
necrotic placentitis with coagulative necrosis of chorion-
ic fossae (Bolin et al. 1985). Intranuclear inclusion bodies DIAGNOSIS
are present in degenerate trophoblasts associated with
the necrotic lesions (Kluge and Maré 1978; Hsu et al. The diagnosis of PR is usually made on a herd basis us-
1980). Depending upon the stage of infection, corpora ing a combination of herd history, clinical signs, gross le-
lutea may be necrotic and contain neutrophils, lympho- sions, microscopic lesions, serology, and virus detection
cytes, plasma cells, and macrophages (Bolin et al. 1985). by either the fluorescent antibody tissue section (FATS)
In the male reproductive tract there may be degener- test or virus isolation (VI). Classic clinical signs will often
ation of seminiferous tubules and necrotic foci in the tu- lead to a presumptive diagnosis that is often supported if
nica albuginea of the testicles (Corner 1965; Larsen et al. gross lesions (focal hepatic and splenic necrosis and
1980; Hall et al. 1984). Boars with exudative periorchitis necrotic tonsillitis) are observed in neonatal pigs. PR is
have necrotic and inflammatory lesions in the serosa cov- more difficult to diagnose if only grower-finisher pigs or
ering the genital organs. Spermatozoa abnormalities in- adult swine are involved. A PR outbreak in these age
clude tail abnormalities, retained distal cytoplasmic groups can easily be misdiagnosed as swine influenza if

19.3. Liver with focal area of


necrosis.
242 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

19.4. Liver with intranuclear


inclusion bodies in hepatocytes at
margins of necrotic focus.

the disease is manifested only by respiratory signs. If, In developing countries or in remote areas where di-
however, a few individuals develop CNS signs, it is easier agnostic laboratories are not available, the supernatant
to make a presumptive diagnosis of PR. PR virus is rela- fluid of a 10% brain suspension can be injected intra-
tively easy to demonstrate in infected swine if samples muscularly into the hindleg of a rabbit. Typical signs of
are taken at the right time and handled properly. Serolo- intense pruritus and self-mutilation at the site of injec-
gy is not the test of choice when trying to diagnose an tion within 48–96 hours will confirm the diagnosis of
acute PR virus infection because of the delay required for PR. This method should only be used as a last resort due
humoral antibodies to develop. Microscopic lesions can to concern for the experimental animal.
confirm a viral etiology and are helpful in finalizing the
diagnosis (see the section on microscopic lesions). Fluorescent Antibody Tissue Section Test
The FATS test is a rapid, reliable method of detecting PR
Virus Isolation virus in tissue. The tissue of choice is tonsil, but brain or
Brain, spleen, and lung are the organs of choice for VI. pharyngeal smears may also be used. In the pig, the ton-
The tissues should be collected from two or three acutely sil is easily identified, for it is in the dorsal area of the an-
affected pigs. Tissues should be refrigerated, preferably terior pharynx and it has visible crypts. The advantages
not frozen unless dry ice is used, and transported to the of the FATS test are that it can be completed in 1 hour
laboratory as soon as possible. If only grower-finisher and the results in neonatal pigs with typical clinical signs
and/or adult swine are affected and none have died, are comparable to VI results. In grower-finisher pigs and
nasal swabs collected in cold phosphate-buffered cell cul- in adults, the FATS test is not as sensitive as VI. If clinical
ture media with antibiotics may be utilized. If cell cul- signs and history are suggestive of PR, a negative FATS
ture fluid or a balanced salt solution is not available, a test result in older animals must be confirmed by VI.
cold sterile saline solution may be used. The sample is in-
oculated onto cell cultures, and the cultures are observed Serodiagnosis
for the presence of characteristic cytopathic effects. The Numerous serologic tests have been developed to assay
identity of the suspected isolate can be confirmed by uti- serum for PR antibodies. The most widely utilized sero-
lizing a fluorescent antibody test. Usually, PR virus will logic assays are serum virus neutralization (SVN), latex
be isolated within 2–5 days. However, most laboratories agglutination (LA), automated latex agglutination,
will conduct two blind passages 1 week apart before re- screening enzyme-linked immunosorbent assay (ELISA),
porting the specimen as negative. Tissues from aborted particle concentration fluorescence immunoassay, and
fetuses can also be submitted for VI; however, if the the differential ELISA for glycoproteins gE, gC, and gG.
abortion was due to a febrile response in the dam rather Other serologic tests have been developed, such as com-
than a direct effect of the virus on the fetuses, VI results plement-fixation, immunodiffusion, countercurrent im-
will be negative. Caution needs to be taken when inter- munoelectrophoresis, and indirect immunofluores-
preting these results, and additional diagnostic tests may cence; all have limitations of sensitivity, require
be required to confirm a diagnosis of a PR virus–induced considerable time to conduct, or are difficult to perform.
abortion. Consequently, they have received limited acceptance.
CHAPTER 19 PSEUDORABIES (AUJESZKY’S DISEASE) Kluge, Beran, Hill, Platt 243

The SVN test is a reliable yet time-consuming test vaccines with different deletions. Countries with PR
and requires 48 hours to perform. The SVN test is con- eradication programs will eventually need to conduct
sidered the standard and is the test used to compare new slaughter-plant surveillance once the prevalence of PR is
test procedures. It can be used to titrate PR viral anti- low. This surveillance will be possible only if a universal
body in serum, and it is still the most widely used test for gene deletion is used in all vaccines.
this purpose.
The ELISA has replaced the SVN test in laboratories TREATMENT: VACCINES
that test large numbers of sera for PR antibody, primari-
ly because of the short time required to conduct the test Modified-live-virus (MLV), inactivated, and gene-deleted
and its superior sensitivity. Several commercial ELISA vaccines have been developed for the control of PR and
kits are available worldwide that allow large numbers of are available in most countries in which PR is endemic.
sera to be tested in 3–4 hours. These vaccines have proven to be quite effective in reduc-
The LA test is the simplest of these three tests to per- ing or preventing clinical signs of PR and thus have re-
form, and it is commercially available worldwide. The LA duced the economic impact of the disease.
test can be performed on a single serum sample in 10 Vaccinated swine that subsequently become infected
minutes. Specificity problems can be minimized by di- have less invasion of tissues, usually limited to the upper
luting the test serum 1:40 instead of 1:4 without signifi- respiratory system, and do not transmit virus to the fetus
cantly affecting sensitivity. The high sensitivity of the LA in utero. Vaccinated swine shed lesser amounts of virus,
test makes it an excellent test for quick screening of sera in most studies at least 1000-fold less; and for a shorter
for PR antibodies. Generally, the LA test will detect sero- period of time, in most studies a 4- to 7-fold reduction.
conversion in 6–7 days, whereas the ELISA requires 7–8 Latent infections may not be prevented in swine infected
days and the SVN requires 8–10 days. The automated subsequent to vaccination. However, recent studies have
format of the LA test has replaced the screening ELISA in demonstrated that certain strains of MLV vaccines will
some high-volume laboratories. colonize the latency target tissue, the trigeminal ganglia,
Interpretation of serologic results can be difficult, es- of vaccinated animals and block the establishment of la-
pecially in young pigs. Maternal antibodies can be pres- tency by a superinfecting challenge of wild-type PR virus
ent for up to 4 months of age. The half-life of the mater- (Schang 1994). The efficiency of a particular vaccine in
nal antibodies that pigs receive in the colostrum of blocking a wild-type virus is dependent on the strain,
PR-immune sows is approximately 18 days. It takes 18 dose, and route of inoculation. MLV vaccines that have
days for the antibody titer to be divided in half (i.e., be extensive deletions of not only glycoproteins gE and gG
reduced from 1:16 to 1:8). If pigs from an immune sow but also other gene deletions for attenuation such as TK
are tested too early, they could be identified as being in- were less efficient at colonizing the trigeminal ganglia.
fected when the antibody is of maternal origin and not Prevention of latency by wild-type virus was also en-
the result of exposure and active infection. hanced by MLV vaccines that were of higher titer and ad-
The most important technological breakthroughs in ministered intranasally. Blocking or reducing the state of
PR control and eradication programs have been the de- latency by a wild-type virus is of paramount importance
velopment of the gene-deleted PR vaccines and their ac- in herds undergoing PR cleanup.
companying differential ELISA. These tests detect a spe- The MLV vaccines replicate principally at the site of
cific antibody produced against one of the viral injection and regional lymph nodes. They have been
glycoproteins. There are vaccines and companion diag- shown to be shed in nasal secretions and tonsillar mucus,
nostic serology tests for gE, gC, and gG. Animals vacci- but at levels too low for there to be any practical risk of
nated with a gene-deleted vaccine will lack antibody transmission to swine or any other animals. These vac-
against the specific glycoprotein encoded for by the cines vary in virulence in animal species other than
deleted gene, allowing a vaccinated pig to be differentiat- swine. Appropriate precautions must be taken when us-
ed from an infected pig. Once the pig is infected, it will ing MLV vaccines in species other than swine, and ex-
develop antibody against all of the PR viral proteins treme care must be taken that syringes and needles used
(antigens) and be positive on all differential serology with PR MLV vaccines are not then used when inoculat-
tests. Some countries have restricted commercial compa- ing other animals with other vaccines. This hazard and
nies as to which gene can be deleted from the vaccine, the concern for the potential for recombination of MLV
but in other countries different vaccines have different with wild-type virus have led to the improvement of
deletions. These vaccines would not be compatible in a killed PR vaccines. The employment of new adjuvants
single herd, because a pig vaccinated with two different has made the efficacy of some of these killed vaccines
gene-deleted vaccines would have antibodies specific for quite competitive with MLV vaccines, especially when
all the glycoproteins and be falsely identified as a PR multiple doses are used. However, killed vaccines do not
virus–infected pig. Therefore, it is important to know colonize latency target tissues and thus do not prevent or
which vaccine is used and to avoid vaccinating pigs with reduce latency.
244 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Killed vaccines have been developed from wild-type aerosol infection of pigs with a strain of Aujeszky’s dis-
strains and from the Bucharest and Bartha MLV vaccine ease virus. Res Vet Sci 12:590–592.
strains. Genetically engineered PR vaccines and some of ———. 1972. Aujeszky’s disease encephalitis in pigs pro-
the conventionally attenuated vaccines have deletions in duced by different modes of infection. Res Vet Sci
nonessential glycoprotein genes such as gE, gC, gG, and 14:223–228.
gI. The absence of these glycoproteins makes them use- ———. 1973. The histopathology of experimental pneu-
ful as negative immunological markers and has been uti- monia in pigs produced by Aujeszky’s disease virus. Res
lized in serologic tests designed to identify virus-infected Vet Sci 14:223–228.
vaccinated swine. Baskerville, A.; McFerran, J. B.; and Dow, C. 1973. Au-
The PR vaccines which have a gE deletion have be- jeszky’s disease in pigs. Vet Bull (London) 43:465–480.
come the vaccines of choice throughout the world be- Becker, C. H. 1964. Zur Bedeutung der Lunge für die pathol-
cause of the superiority of the gE differential serologic ogisch-anatomische Diagnose der Aujeszkyschen
tests (White et al. 1996). The gG gene-deleted vaccines Krankheit des Schweines. Monatsh Veterinärmed
are very immunogenic but the companion gG differen- 19:5–11.
tial ELISA lacks both sensitivity and specificity. Many an- Beran, G. W. 1991. Transmission of Aujeszky’s disease
imal health regulatory bodies have mandated that all PR virus. In Proc First Int Symp on Eradication of Pseudora-
vaccines must have a gE gene deletion. bies (Aujeszky’s) Virus, St. Paul, Minn., pp. 93–111.
The TK gene has also been deleted from genetically ———. 1993. Understanding the transmission of
engineered PR vaccines. TK facilitates PR infections of pseudorabies virus. Vet Med 88:70–79.
neurons, permitting viral replication. Since the natural Bolin, C. A.; Bolin, S. R.; Kluge, J. P.; and Mengeling, W. L.
endogenous level of TK in differentiated neural tissue is 1985. Pathologic effects of intrauterine deposition of
low, the virulence of a PR vaccine can be reduced by re- pseudorabies virus on the reproductive tract of swine in
moving the TK gene so that the virus can no longer en- early pregnancy. Am J Vet Res 46:1039–1042.
code for the production of this enzyme. Bolin, S. R., and Bolin, C. A. 1984. Pseudorabies virus infec-
The recombination of gene-deleted vaccines that tion of six- and ten-day-old porcine embryos. Theri-
might lead to a virulent virus that still contains a negative ogenology 22:101–106.
immunological marker is a concern (Henderson et al. Bolin, S. R.; Turek, J. J.; Runnels, L. J.; and Gustafson, D. P.
1991). Studies demonstrated that when pigs were inocu- 1983. Pseudorabies virus, porcine parvovirus, and
lated with two different gene-deleted vaccines, recombi- porcine enterovirus interactions with the zona pellucida
nation did occur. It is therefore imperative that only one of the porcine embryo. Am J Vet Res 44(6):1036–1039.
type of gene-deleted vaccine be used in a single herd an- Cheung, A. K. 1995. Investigation of pseudorabies virus
imal to avoid potential recombination between vaccine DNA and RNA in trigeminal ganglia and tonsil tissues of
viruses. Recombination potentially could also occur latently infected swine. Am J Vet Res 56 (1):45–50.
when gene-deleted MLV vaccines are used in the face of Christensen, L. S.; Mortensen, S.; Botner, A.; Strandby-
an outbreak with field PR virus, resulting in a gene-delet- gaard, B. S.; Ronsholt, L.; Henriksen, C. A.; and Ander-
ed virulent virus as a result of recombination between sen, J. B. 1993. Further evidence of long distance air-
the field strain and the gene-deleted vaccine strain. This borne transmission of Aujesky’s disease (pseudorabies)
apparently does not occur or occurs very infrequently, virus. Vet Rec 132:317–321.
for gene-deleted MLV vaccines have been widely used in Corner, A. H. 1965. Pathology of experimental Aujeszky’s
PR outbreaks, and as yet, there are no reports of a viru- disease in piglets. Res Vet Sci 6:337–343.
lent PR virus with a deletion of a gene for one of the gly- Csontos, L., and Széky, A. 1966. Gross and microscopic le-
coproteins. sions in the nasopharynx of pigs with Aujeszky’s dis-
ease. Acta Vet Acad Sci Hung 16:175–186.
REFERENCES Csontos, L.; Hejj, L.; and Szabó, I. 1962. A contribution to
the aetiology of Aujeszky’s disease in the pig: Foetal
Anderson, J. B.; Bitsch, V.; Christensen, L. S.; Hoff-Jor- damage and abortion due to the virus. Acta Vet Acad Sci
gensen, R. and Petersen, K. 1989. The control and eradi- Hung 12:17–23.
cation of Aujeszky’s disease in Denmark: Epidemiologi- Davies, E. B., and Beran, G. W. 1980. Spontaneous shedding
cal aspects. In Vaccination and Control of Aujeszky’s of pseudorabies virus from a clinically recovered post
Disease. Ed. J. T. Van Oirschot. Brussels and Luxem- parturient sow. J Am Vet Med Assoc 176:1345–1347.
bourg: ECSC, EEC, EAEC; Boston: Kluwer Academic ———. 1981. Influence of environmental factors upon the
Publishers, pp. 175–183. survival of Aujeszky’s disease virus. Res Vet Sci
Anusz, Z.; Szweda, W.; Popkko, J.; and Trybala, E. 1992. Is 31:32–36.
Aujeszky’s disease a zoonosis? Przegl Epidemiol De Smet, K.; De Wahle, K.; Pensaert, M.; Goovaerts, D.; and
46(3):181–186. De Ridder, E. 1992. Field experiments to evaluate the
Baskerville, A. 1971. The histopathology of pneumonia by feasibility of eradication of Aujeszky’s disease virus by
CHAPTER 19 PSEUDORABIES (AUJESZKY’S DISEASE) Kluge, Beran, Hill, Platt 245

different vaccination schedules. Vet Quart 14:22–28. bies virus glycoprotein gp50 as a vaccine for Aujeszky’s
De Smet, K.; De Wahle, K.; and Pensaert, M. 1994. Influence disease in mice and swine: Expression by vaccinia virus
of vaccine medium and vaccination schedules on the in- and Chinese hamster ovary cells. J Virol 61:3977–3982.
duction of active immunity against Aujeszky’s disease in Marchioli, C. C.; Yancey, R. J., Jr.; Timmins, J. G.; Post, L. E.;
maternally immune pigs. Res Vet Sci 56:89–94. Young, B. R.; and Povendo, D. A. 1988. Protection of
Dow, C., and McFerran, J. B. 1962. The neuropathology of mice and swine from pseudorabies virus–induced mor-
Aujeszky’s disease in the pig. Res Vet Sci 3:436–442. tality by administration of pseudorabies virus–specific
Gueguen, B., and Aynaud, J. M. 1980. Étude de lexcretion du mouse monoclonal antibodies. Am J Vet Res
virus de la maladie d’Aujeszky par les voies genitales du 49:860–864.
porc. Rec Med Vet 156:307–312. Masic, M.; Ercegan, M.; and Petrovic, M. 1965. Die Bedeu-
Hall, L. B.; Kluge, J. P.; Evans, L. E.; and Hill, H. T. 1984. The tung der Tonsillen für die Pathogenese und Diagnose
effect of pseudorabies (Aujeszky’s) virus infection on der Aujeszkyschen Krankheit bei Schweinen. Zentralbl
young mature boars and boar fertility. Can J Comp Med Veterinärmed 12:389–405.
48:192–197. Mayfield, J. E. 1990. Vaccine strains and field strains her-
Hampl, H.; Ben-Porat, T.; Ehrlicher, L.; Habermehl, K. O.; pesvirus I recombine in swine to produce virulent prog-
and Kaplan, A. S. 1984. Characterization of the enve- eny virus. In 15th Int Herpesvirus Workshop, Washing-
lope proteins of pseudorabies virus. J Virol 52:583–590. ton, D.C., p. 433(Abstr).
Henderson, L. M.; Levings, R. L.; Davis, A. J.; Katz, J. B.; and McCullough, S. J. 1989. Vaccination and control of Au-
Strutz, D. R. 1991. Recombination of pseudorabies virus jeszky’s disease in Northern Ireland. In Vaccination and
vaccine strains in swine. Am J Vet Res 52:820–825. Control of Aujeszky’s Disease. Ed. J. T. Van Oirschot.
Hsu, F. S.; Chu, R. M.; Lee, R. C.; and Chu, S. H. 1980. Pla- Brussels and Luxembourg: ECSC, EEC, EAEC; Boston:
cental lesions caused by pseudorabies virus in pregnant Kluwer Academic Publishers, pp. 231–238.
sows. J Am Vet Med Assoc 177:636–641. McFerran, J. B., and Dow, C. 1965. The distribution of the
Iglesias, G.; Pijoan, C.; and Molitor, T. 1989. Interactions of virus of Aujeszky’s disease (pseudorabies virus) in ex-
pseudorabies virus with swine alveolar macrophages: Ef- perimentally infected swine. Br Vet J 126:173–179.
fects of virus infection on cell functions. J Leukocyte Bi- Medveczky, I., and Szabó, I. 1981. Isolation of Aujeszky’s
ol 45:410–415. disease virus from boar semen. Acta Vet Acad Sci Hung
Iglesias, G.; Trujano, M.; Lokensgard, J.; and Molitor, T. 29:29–35.
1992. Study of the potential involvement of pseudora- Mengeling, W. L.; Paul, P. S.; Pirtle, E. C.; and Wathen, M.
bies virus in swine respiratory disease. Can J Vet Res W. 1983. Restriction endonuclease analysis of the
56(1):74–77. pseudorabies (Aujeszky’s disease) virus before and after
Kluge, J. P., and Maré, C. J. 1976. Gross and microscopic le- serial passage in-vivo and in-vitro. Arch Virol
sions of prenatal and neonatal pseudorabies (Aujeszky’s 78:213–220.
disease) in swine. Proc Int Congr Pig Vet Soc 4:G3. Mengeling, W. L.; Lager, K. M.; Volz, M.; and Brockmeier, S.
———. 1978. Natural and experimental in utero infection L.; 1992. Effect of various vaccination procedures on
of piglets with Aujeszky’s disease (pseudorabies) virus. shedding, latency, and reactivation of attenuated and
Am Assoc Vet Lab Diagn 21:15–24. virulent pseudorabies virus in swine. Amer Jour Vet Res
Klupp, B. G.; Visser, N.; and Mettenleiter, T. C. 1992. Identi- 53: 2164–2173.
fication and characterization of pseudorabies virus gly- Miller, G. Y.; Tsai, J. S.; and Forster, D. L. 1996. Benefit-cost
coprotein H. J Virol 66(5):3048–3055. analysis of the national pseudorabies virus eradication
Kost, T. A.; Jones, E. V.; Smith, K. M.; Reed, A. I.; Brown, A. program. J Am Vet Med Assoc 208:208–213.
L.; and Miller, T. J. 1989. Biological evaluation of glyco- Mulder, W.; Pol, J.; Kimman, T.; Kok, G.; Priem, J.; and
protein mapping to two distinct mRNAs within the Peeters, B. 1996. Glycoprotein D-negative pseudorabies
BamHl fragment 7 of pseudorabies virus: Expression of virus can spread transneuronally via direct neuron-to-
the coding regions by vaccinia virus. Virology neuron transmission in its natural host, the pig, but not
171(2):365–376. after additional inactivation of gE or gI. J Virol
Kritas, S. K.; Pensaert, M. B.; and Mettenleiter, T. C. 1994. 70(4):2191–2000.
Role of envelope glycoproteins gI, gp63, and gIII in the Narita, M.; Haritani, M.; and Moriwaki, M. 1984a. Necro-
invasion and spread of Aujeszky’s disease virus in the ol- tizing vasculitis in piglets infected orally with the virus
factory nervous pathway of the pig. J Gen Virol of Aujeszky’s disease. Jpn J Vet Sci 46:119–122.
75(9):2319–2327. Narita, M.; Kubo, M.; Fukusho, A.; Haritani, M.; and Mori-
Larsen, R. E.; Shope, R. E.; Leman, L. D.; and Kurtz, H. J. waki, M. 1984b. Necrotizing enteritis in piglets associat-
1980. Semen changes in boars after experimental infec- ed with the virus of Aujeszky’s disease. Vet Pathol
tions with pseudorabies virus. Am J Vet Res 41:733–739. 21:450–452.
Marchioli, C. C.; Yancey, R. J., Jr.; Petrovskis, E. A.; Tim- Narita, M.; Inui, S.; and Schimizu, Y. 1984c. Tonsillar
mins, J. G.; and Post, L. E. 1987. Evaluation of pseudora- changes in pigs given pseudorabies (Aujeszky’s disease)
246 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

virus. Am J Vet Res 45:247–251. dle-Western swine and the possible role of rats in herd-
Olander, H. J.; Saunders, J. R.; Gustafson, D. P.; and Jones, to-herd infections. J Exp Med 62:101–117.
R. K. 1966. Pathologic findings in swine affected with a Stevely, Y. W. S. 1975. Virus induced proteins in pseudora-
virulent strain of Aujeszky’s virus. Pathol Vet 3:64–82. bies infected cells. II. Proteins of the virion and nucleo-
Olander, H. J.; Gustafson, D. P.; and Qureshi, S. R. 1976. capsid. J Virol 16:944–950.
Residual lesions in swine convalescent from pseudora- Taylor, K. C. 1989. Epidemiological aspects of Aujeszky’s
bies virus infection. Proc Int Congr Pig Vet Soc 4:G13. disease control in Great Britain. In Vaccination and Con-
Oslage, U.; Dahle, J.; Muller, T.; Kramer, M.; Beier, D.; and trol of Aujeszky’s Disease. Ed. J. T. Van Oirschot. Brus-
Leoss, B. 1994. Pravalenz von Antikorpern genen die sels and Luxembourg: ECSC, EEC, EAEC; Boston: Kluw-
Viren der europäischen Schweinepest, der Aujeszky er Academic Publishers, pp. 185–196.
schen Krankheit und des porcine reproductive and res- Thawley, D. G.; Gustafson, D. P.; and Beran, G. W. 1982.
piratory syndrome@ (PRRS) bei Wildschweinen in den Swine pseudorabies eradication guidelines. South St.
Bundeslanderm Sachsen-Anhalt und Brandenburg. Paul, Minn.: Livest Conserv Inst, pp. 1–11.
Dtsch Tierärztl Wochenschr 101:33–38. U.S. Department of Agriculture/APHIS/VS, 1994–1997.
Platt, K. B. 1981. Genetic stability of the thermal, trypsin, Pseudorabies Eradication Quarterly Reports. Washing-
rabbit and mouse markers of Aujeszky’s disease ton, D.C.: USDA/APHIS/VS.
(pseudorabies) virus in the pig. Vet Microbiol ———. 1997. Pseudorabies Eradication: State-Federal-In-
6:225–232. dustry Program Standards. APHIS 91-55-035 (effective
Rziha, H. J.; Doller, P. C.; and Wittmann, G. 1982. Detection January 1, 1997), pp. 1–28.
of Aujeszky’s disease virus and viral DNA in tissues of Vander der Leek, M. L.; Becker, H. N.; Pirtle, E. C.;
latently infected pigs. In Current Topics in Veterinary Humphrey, P.; Adams, C. L.; All, B. D.; Erickson, G. A.;
Medicine and Animal Science. Ed. G. Wittmann and S. Belden, R. C.; Frankenberger, W. B.; and Gibbs, E. P.
A. Hall, 17:205–211. 1993. Prevalence of pseudorabies (Aujeszky’s disease)
Sabó, A.; Rajcani, J.; and Blaskovič, D. 1968. Studies on the virus antibodies in feral swine in Florida. J Wildlife Dis
pathogenesis of Aujeszky’s disease. 1. Distribution of 29:403–409.
the virulent virus in piglets after peroral infection. Acta Van Oirschot, J. T.; Kaashoek, M. J.; Rijsewijk, F. A.; and
Virol 12:214–221. Stegeman, J. A. 1996. The use of marker vaccines in
———. 1969. Studies on the pathogenesis of Aujeszky’s eradication of herpesviruses. J Biotech 44:75–81.
disease. III. The distribution of virulent virus in piglets Wheeler, J. G., and Osorio, F. A. 1991. Investigation of sites
after intranasal infection. Acta Virol 13:407–414. of pseudorabies virus latency, using polymerase chain
Schang, L. M.; Kutish, G. F.; and Osorio, F. A. 1994. Corre- reaction. Am J Vet Res 52(11):1799–1803.
lation between precolonization of trigeminal ganglia by White, A. K.; Ciacci-Zanella, J.; Galeota, J.; Ele, S.; and Oso-
attenuated strains of pseudorabies virus and resistance rio, F. A. 1996. Comparison of the abilities of serologic
to wild-type virus latency. J Virol 68:8470–8476. tests to detect pseudorabies-infected pigs during the la-
Scheidt, A. B.; Ruelf, L. R.; Grant, R. H.; Teclaw, R. F.; Hill, tent phase of infection. Am J Vet Res 57:608–611.
M. A.; Meyer, K. B.; and Clark, L. K. 1991. Epizootic of Wittmann, G., and Rziha, H. J. 1989. Herpesvirus diseases
pseudorabies among ten swine herds. J Am Vet Med of cattle, horses, and pigs. In Developments in Veteri-
Assn 199:725–730. nary Virology. Boston: Kluwer Academic Publishers, pp.
Schneider, W. J., and Howarth, J. A. 1973. Clinical course 230–325.
and histopathologic features of pseudorabies virus-in- Wittmann, G.; Jakubik, J.; and Ahl, R. 1980. Multiplication
duced keratoconjunctivitis in pigs. Am J Vet Res and distribution of Aujeszky’s disease (pseudorabies)
34:393–401. virus in vaccinated and non-vaccinated pigs after in-
Schoenbaum, M. A.; Zimmerman, J. J.; Beran, G. W.; and tranasal infection. Arch Virol 66:227–240.
Murphy, D. P. 1990. Survival of pseudorabies virus in Wohlgemuth, K.; Leslie, P. F.; Reed, D. E.; and Smidt, D. K.
aerosol. Am J Vet Res 51:331–333. 1978. Pseudorabies virus associated with abortion in
Schoenbaum, M. A.; Freund, J. D.; and Beran, G. W. 1993. swine. J Am Vet Med Assoc 172:478–479.
Survival of pseudorabies virus in the presence of select- Zimmerman, J. J.; Berry, W. J.; Beran, G. W.; and Murphy,
ed diluents and fomites. Am J Vet Med Assn D. P. 1989. Influence of temperatures and age on the re-
198:1393–1397. covery of pseudorabies (Aujeszky’s disease) virus from
Shope, R. E. 1935. Experiments on the epidemiology of house flies, Musca domestica. Am J Vet Res
pseudorabies. II. Prevalence of the disease among Mid- 50:1471–1474.
Rabies
20 L. G. Morehouse

Rabies is a highly infectious viral disease of humans and bies virions from the purified virus. By treatment with
animals that dates to antiquity. The dramatic clinical sodium deoxycholate and combined velocity and equi-
signs of this disease were described in the writings of librium gradient centrifugation, the viral coat and the
Plutarch and Celsus in A.D. 100 (Kelser 1947). The earli- nucleocapsid are separated. The latter contains all the
est reference to the disease in Asia is in the pre-Mosaic RNA and is made up of 96% protein and 4% RNA. The
Eshnunna Code (ca. 2000 B.C.), which predates the eigh- five structural proteins and conclusions regarding their
teenth-century B.C. code of Hammurabi of ancient Baby- function and arrangement within the virion have been
lon. reviewed (Cox 1982).
Despite the ancient history of the disease, many as-
pects of rabies are not clearly understood. Although al- EPIDEMIOLOGY
most all warm-blooded animals are susceptible, the long-
held belief in the fatal termination of the disease may Traditionally, the disease has been said to exist in two
not be applicable to all species. In fact, recovery from hu- epidemiologic forms: the urban type, spread mainly by
man rabies (U.S. Dept. HEW 1970) and recovery of dogs dogs; and the wildlife type, seen principally in skunks,
from experimental rabies infection (Nillson and Cortes foxes, wolves, jackals, coyotes, weasels, and bats (Bisseru
1975; D. C. Blenden, pers. comm.—1979; Fekadu et al. 1972). A marked shift to wildlife rabies occurred in the
1983) have been reported. Evidence also indicates that United States during the 1960s and 1970s. By 1964, 75%
some animal species are more resistant to the infection of the rabies in the United States was in wildlife, and less
than others (Bisseru 1972; Botros et al. 1979); the discus- than 10% was reported from dogs (U.S. Dept. HEW
sion in this chapter suggests that swine fall in this cate- 1964). A more recent report confirms this trend, with
gory. Nonetheless, the overwhelming mortality associat- 88.4% of recorded cases in wildlife and only 2.7% in dogs
ed with rabies infection makes it one of the most feared (U.S. Dept. HHS 1989).
diseases of humans and animals. With this increased percentage of rabies occurring in
wild animals, farm animals have been placed at greater
ETIOLOGY risk of exposure. The true incidence of rabies in swine is
difficult to assess. In the United States 854 cases were re-
Rabies is caused by a filterable virus of the rhabdovirus ported over the 17-year period from 1938 to 1955
group (Melnick and McCombs 1966). The external di- (Schoening 1956), representing 6.6% of cases reported in
mensions of the rabies particle are 750 × 180 nm. Its all farm animals. Cases of rabies on record in swine in
surface has a honeycomb appearance with projections the United States dropped to 3–8 annually, and in Cana-
6–7 nm long (Witkor 1971). The particle is cylindrical, da and Mexico to 10–21 annually (U.S. Dept. HEW
with one round or conical end and one planar or concave 1969–83). This trend has continued through 1988 (U.S.
end (Murphy 1975). The virus is classified as a ribonu- Dept. HHS 1989). In a review of the global rabies situa-
cleic acid (RNA)–containing virus. The RNA is a single- tion, however, specific reference has been made to con-
stranded molecule with a molecular weight of 4.6 × 106 firmed cases of porcine rabies in 25 different countries
daltons per nucleocapsid (Witkor 1971). Sokol et al. on five continents (Bisseru 1972). Yates et al. (1983) de-
(1969) have separated the different components of ra- scribe rabies outbreaks in 15 swine herds in western
Canada over a 14-year period.
Epidemiologic studies are now aided by monoclonal
antibody analysis of various strains of rabies virus from
Some of the material presented in this chapter is based on experience en- different animals and geographic regions (Smith et al.
countered in an outbreak of rabies in a herd of Missouri swine. The author 1984). Major antigenic groups of rabies virus in Canada
acknowledges contributions of his colleagues Dr. L. D. Kintner, Dr. S. L. Nel-
have been determined by antinucleocapsid monoclonal
son, and Dr. B. L. Moseley of the University of Missouri; the Veterinary Pub-
lic Health Unit, Jefferson City, Mo.; and Veterinary Services Laboratories, antibodies, allowing classification of antigenic groups by
APHIS, USDA, Ames, Iowa. geographic area (Webster et al. 1986).

247
248 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

CLINICAL SIGNS integrity of the nerve is necessary for CNS invasion, thus
confirming the case for a neural pathway for the virus.
The typical course of the disease is one of sudden onset, The rabies virus may enter the host via many routes,
loss of coordination, dullness, and later prostration. but the importance of oral and respiratory transmission
Death will likely ensue within 72 hours from the appear- is uncertain (Johnson 1971). Kucera et al. (1985) report
ance of clinical signs (Merrimann 1966; Morehouse et al. three routes of entry from the eye to the brain: the ocu-
1968). Peculiar twitching and preoccupation with the lomotor parasympathetic nerve, the preopticoretinal
nose, similar to that observed in swine with rings recent- pathway, and the ophthalmic nerve. The mechanism by
ly placed in their noses, has been noted. This may be fol- which the virus gains access to the CNS has been studied
lowed by prostration, rapid chewing movements, exces- by a number of workers. A progression of rabies infec-
sive salivation, and generalized clonic muscular spasms, tions in hamsters has been demonstrated, starting in
which become less marked as the disease progresses un- striated muscle cells near the site of inoculation, with in-
til muscular activity is manifested only by fine tremors. fection of myocytes and shedding of the virus into extra-
Affected swine may be unable to squeal, and elevation of cellular spaces. A working hypothesis was proposed that
body temperature may be absent (Morehouse et al. the route of passive centripetal passage of the rabies
1968). virus to the CNS is via peripheral nerve axoplasm from
Oldenberger (1963) reported a paralytic form of ra- invasion at unmyelinated distal end organs to progeny
bies in swine in the USSR, with a duration of illness of release at synaptic junctions in the spinal cord (Murphy
5–6 days. Gigstad (1971) reported clinical signs of rabies et al. 1973a). An infection of myocytes at the site of in-
in experimentally infected swine that included an inter- oculation with the rabies virus in skunks has been re-
mittent weakness in the rear legs, weakness in the shoul- ported (Charlton and Casey 1979). Immunofluorescence
ders, incoordination, posterior paralysis, prostration,
in muscle fibers remote from the inoculation site oc-
paddling, and death. Merrimann (1966) documented
curred only after infection of lumbar cord and dorsal
one case of the furious form of rabies in a pig, but this
root ganglia, suggesting infection as a result of centrifu-
pattern of behavior was observed in only 1 of 17 affected
gal neural migration of virus.
pigs. Hazlett and Keller (1986) reported an outbreak of
In the development of infection within the CNS, neu-
porcine rabies in a closed feeder barn where clinical signs
rons of spinal cord, brain stem, hippocampus, septal nu-
were not observed.
Although most reports suggest that swine die within clei, and limbic cortex appear especially vulnerable.
72–96 hours following development of clinical signs, Meningeal, ependymal, vascular endothelial, and glial
there is indication of a wide variation in the incubation cells show no antigen (Johnson 1971). However, Jackson
period. Merrimann’s report (1966) suggests an incuba- and Reimer (1989) have reported infection of ependymal
tion period of approximately 70 days for one group of cells lining lateral ventricles in mice following intracere-
swine. Morehouse et al. (1968) reported that since there bral exposure and postulate that virus entry occurs at
was a progression of rabies cases in a drove of swine over least in part by a cerebrospinal fluid pathway. Murphy et
a 2-month period, the incubation period would have al. (1973b) proposed a mechanism of viral spread to in-
been extremely variable if the disease resulted from a si- clude ascending infection within the CNS and centrifugal
multaneous exposure of a number of swine. Reichel and peripheral neural spread, with passive intraneuronal
Möckelmann (1963) reported an outbreak of rabies in movement of subviral entities interspersed with active
swine where the exposure time from rabid foxes was doc- viral replication at cell surfaces, such that progeny virus
umented. Incubation periods of 9, 56, and 123 days were might invade contiguous neurons or move within inter-
recorded for the affected swine. Gigstad (1971) reported cellular spaces to involve neural elements elsewhere.
incubation periods in experimentally infected swine that Charlton and Casey (1979) suggested direct transneu-
varied from 12 days when exposed intracerebrally to 98 ronal transfer of virus from perikarya and dendrites to
days when exposed intramuscularly. adjacent axon terminals as a mechanism in dissemina-
tion of rabies in the CNS of striped skunks.
PATHOGENESIS Whether or not factors such as the mechanism of
virus spread via the nerves, significance of oral and res-
The spread of rabies virus along peripheral or cranial piratory transmission, selective cellular vulnerability,
nerves appears to be the major avenue of access to the and host cell–virus relationships that might allow for la-
central nervous system (CNS) following intramuscular tent infection can result in a variability in species suscep-
or subcutaneous exposure to the virus. Dean et al. (1963) tibility remains an unanswered question in the patho-
published convincing evidence that fixed (attenuated) genesis of rabies. This is particularly true in the case of
and street (virulent) rabies viruses are ordinarily trans- swine, where the lack of detailed case reports or experi-
mitted from the site of inoculation to the CNS by way of mental studies and the relatively low reported number of
the peripheral nerves. Johnson (1971) emphasized that cases in this species compound the problem.
CHAPTER 20 RABIES Morehouse 249

LESIONS cord was characterized by perivascular cuffing that in-


volved a high percentage of the larger vessels in both
Necropsy examination of rabid swine will likely fail to re- gray and white matter. Figure 20.2 shows marked
veal gross changes (Merrimann 1966; Morehouse et al. perivascular cuffing of a vessel in the medulla.
1968; Hazlett and Koller 1986). Histopathologic changes
may be observed in the brain and spinal cord but may be
variable. This is true for many species, where changes
may range from indiscernible, except for early necrosis of
neurons and specific cytoplasmic inclusions in affected
nerve cells, to a diffuse encephalitis (Smith et al. 1972).
The variability in neuropathologic response in other
species infected with rabies seems true also for swine. In
one text the authors indicate that neuronal degeneration
may be very slight in pigs (Jubb et al. 1985). Merrimann
(1966) reported the presence of Negri bodies in only one
of three brains from rabid swine, although they were
positive to the fluorescent rabies antibody test and had
shown clinical signs of the disease. Gigstad (1971) re-
ported an extensive nonsuppurative encephalitis in four
pigs that had died following experimental inoculation
with rabies virus. One animal had been inoculated intra-
muscularly and died 42 days postinoculation, while the
other three had been inoculated intracerebrally and died
12–18 days postinoculation. Negri bodies were not ob-
served. Hazlett and Koller (1986) reported only one Ne-
gri body in 10 sections of brain from a rabid pig.
Neuropathologic changes were observed in pigs dy-
ing from rabies, varying from a mild reaction in the brain
consisting primarily of a mild vasculitis and focal gliosis
to marked changes throughout the brain and spinal
cord. Negri bodies were not observed (Morehouse et al.
1968). The changes included a diffuse meningitis of the
brain and spinal cord, which was most severe in the lep-
tomeninges covering the folia of the cerebellum (Fig. 20.1. Diffuse leptomeningitis of the cerebellar folia
20.1). The inflammatory response in the brain and spinal (H&E; ×135).

20.2. Marked perivascular cuffing and neuronal degeneration in the medulla (H&E; ×100).
250 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Neuronal destruction observed throughout the brain


and spinal cord was manifested by degeneration of neu-
rons in the Purkinje cell layer of the cerebellum. Phago-
cytosis of pyramidal cells was apparent (Fig. 20.3). The
inflammatory change associated with the vessels in Fig-
ure 20.3 was primarily intramural in nature, with the Vir-
chow-Robin space appearing clear. Degenerating neu-
rons in other areas of the hippocampus were shrunken
and stained heavily with eosin (Fig. 20.4). There was
necrosis of neurons in the pons with accompanying
satellitosis (Fig. 20.5) and neuronophagia (Fig. 20.6).
Large motor neurons in the ventral horn of some seg-
ments of the spinal cord were undergoing degeneration
(Fig. 20.7).

DIAGNOSIS

Animal Inoculation Test


The intracerebral inoculation of laboratory animals with
brain material from animals suspected of being rabid is
the oldest method of diagnosing rabies. Symptoms in
weanling mice may be observed within 6–14 days, and
the diagnosis of rabies is confirmed by the fluorescent
antibody (FA) test (or the demonstration of Negri bodies
in the mouse brains). For a negative result it is common
to wait at least 21 days, although some workers have re-
ported at least a 27-day incubation period in mice receiv- 20.3. Neuronal degeneration in the pyramidal
ing brain material from rabid swine (Morehouse et al. cell layer of the hippocampus (H&E; ×100).

20.4. Necrosis of neurons in the hippocampus (H&E; ×100).


CHAPTER 20 RABIES Morehouse 251

20.5. Satellitosis of a neuron in the pons 20.7. Degenerating neuron in the ventral horn of the
(H&E; ×100). spinal cord (H&E).

20.6. Phagocytosis of a dying neuron in the pons (H&E; ×400).


252 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

1968; R. Dillman, pers. comm.—1973; M. Keiffer, pers. unpredictable behavior of the disease when it occurs as a
comm.—1973). Additional confirmation includes a neu- herd problem. Blenden (pers. comm.—1979) inoculated
tralization test in mice, using specific rabies immune two weanling pigs by the intramuscular route with street
serum. rabies virus. No clinical signs were observed, although
high titers of antibody were found in cerebrospinal fluid
Histopathologic Examination and blood. Whether natural resistance of swine to rabies
Negri (1903) established the first definitive pathologic results in aberrant clinical signs, thus confusing the diag-
diagnosis of rabies when he described the eosinophilic nosis, remains open to question.
cytoplasmic inclusions in nerve cells and dendrites. A These observations do emphasize problems that may
number of stains are available for demonstrating these be encountered in the clinical and laboratory diagnosis
inclusion bodies (Gradwohl 1943). Bosch (1966) de- of rabies in swine, and they point to the value of careful
scribed the use of acidified phenolphloxine for staining neuropathologic examination and other laboratory pro-
Negri bodies in paraffin sections. Negri bodies, however, cedures in differentiating rabies from other encephali-
are not found in all rabid animals. Runnells et al. (1965) tides of swine described in this volume.
stated that Negri bodies are not present in about 25% of
rabid animals, and in many of the remaining 75% they TREATMENT AND PREVENTION
are so few in number as to be overlooked. Lennette et al.
(1965) reported that in a series of 365 rabies-positive The extremely limited literature and experience with ra-
specimens, only 239 (65.8%) were detected by the pres- bies in swine give no indication of effective treatment for
ence of Negri bodies; the variability of their presence in this disease, nor is there any indication of vaccine devel-
swine rabies has been reported by Merrimann (1966), opment for this species. However, producers and veteri-
Morehouse et al. (1968), Gigstad (1971), Dhillon and narians should be aware of the unpredictable behavior of
Dhingra (1973), and Hazlett and Koller (1986). swine affected with this disease when it occurs as a herd
problem. Preventive measures consist principally of lim-
Fluorescent Antibody Test iting exposure, particularly where rabies is endemic in
Goldwasser and Kissling (1958) were the first to apply wildlife populations adjacent to areas where swine are
the FA technique to the diagnosis of rabies. It has proved maintained.
to be a rapid and accurate test. McQueen et al. (1960)
demonstrated its dependability for diagnosing rabies in REFERENCES
brain tissue that was Negri-body negative. Lennette and
Emmons (1971) have emphasized that fluorescent anti- Beauregard, M.; Boulanger, P.; and Webster, W. A. 1965.
gen will be present and detectable in the brain at any The use of fluorescent antibody staining in the diagno-
stage of the disease in which the animal could possibly sis of rabies. Can J Comp Med 29:141–147.
have transmitted rabies. Blenden (1978) described FA Bisseru, B. 1972. Rabies. London: William Heinemann
identification of rabies virus antigen in the skin of mice Medical Books, p. 17.
before clinical signs developed; he later reported its val- Blenden, D. C. 1978. Identification of rabies virus antigen in
ue for early detection of rabies virus antigen (Blenden et the skin by immunofluorescent staining. Zoonoses 1,
al. 1983). Several authors, however, have found swine PAHO/WHO Zoonoses Center, pp. 7–11.
specimens negative on FA in cases later diagnosed posi- Blenden, D. C.; Bell, J. L.; Tsao, A. L.; and Usnok, O. U. 1983.
tive by mouse inoculation (Beauregard et al. 1965; More- Immunofluorescent examination of skin of rabies in-
house et al. 1968; Dhillon and Dhingra 1973). fected animals as a means of early detection of rabies
antigen. J Clin Microbiol 18:631–636.
Other Methods Bosch, R. 1966. Acidified phenol-phloxine for staining Negri
Bourgon and Charlton (1987) demonstrated rabies anti- bodies in paraffin section. Stain Technol 41(4):
gen in paraffin-embedded tissue by the peroxidase-an- 250.
tiperoxidase method and found it superior to the FA Botros, B. A. M.; Lewis, J. C.; and Kerkor, M. 1979. A study
technique for detection of antigen and preservation of to evaluate nonfatal rabies in animals. J Trop Med Hyg
morphologic detail. Perrin and Sureau (1987) described 82:137–141.
an experimental kit for rapid rabies enzyme immunodi- Bourgon, A. R., and Charlton, K. M. 1987. The demonstra-
agnosis (RREID). They found it to be specific, conve- tion of rabies antigen in paraffin-embedded tissues us-
nient, and a useful tool for epidemiologic studies and for ing a peroxidase anti-peroxidase method. Can J Vet Res
laboratories without ultraviolet microscopes; it was not 51(1):117–120.
as sensitive as the FA technique. Charlton, K. M., and Casey, G. A. 1979. Experimental rabies
in skunks—immunofluorescence light and electron mi-
Comment croscope studies. Lab Invest 41:36–44.
Gigstad (1971) concluded from experimental studies Cox, J. H. 1982. The structural proteins of rabies virus.
that swine may have a relatively high natural resistance Comp Immun Microbiol Infect Dis 5:21–25.
to the rabies virus and this might be responsible for the Dean, D. J.; Evans, W. M.; and McClure, R. C. 1963. Patho-
CHAPTER 20 RABIES Morehouse 253

genesis of rabies. Bull WHO 29:803. 1973b. Comparative pathogenesis of rabies and rabies-
Dhillon, S. S., and Dhingra, P. N. 1973. Rabies in swine. Vet like viruses—Infection of the central nervous system
Med Small Anim Clin 68:1044. and centrifugal spread of virus to peripheral tissues. Lab
Fekadu, M.; Shadduck, J. H.; Chandler, T. W.; and Baer, G. Invest 29:1–16.
M. 1983. Rabies virus in the tonsils of a carrier dog. Negri, A. 1903. Beitrag zum Studium der Aetiologie der
Arch Virol 78:37–47. Zollswith. Z Hyg Infektionskr 43:507. Cited in Infectious
Gigstad, D. C. 1971. Experimental rabies in swine. Dev Stud Diseases of Domestic Animals, 3d ed. Ed. W. A. Hagan
Lab Invest, APHIS, USDA, p. 20. and D. W. Bruner. Ithaca, N.Y.: Comstock, p. 735.
Goldwasser, R. A., and Kissling, R. E. 1958. Fluorescent an- Nillson, M. R., and Cortes, J. de A. 1975. Spontaneous
tibody staining of street and fixed rabies virus antigens. recovery from rabies of a dog infected experimentally.
Proc Soc Exp Biol Med 98:219. Rev Fac Med Vet Zootech Univ São Paulo 12:229–
Gradwohl, R. B. H. 1943. Clinical Laboratory Methods and 233.
Diagnosis, 3d ed., vol. 1. St. Louis: C. Y. Mosby. Oldenberger, A. A. 1963. Peculiarities in the trend of rabies
Hazlett, M. F., and Koller, M. A. 1986. Porcine rabies in a in swine. Veterinariia 40:30.
closed feeder barn. Can Vet J 27:116–118. Perrin, P., and Sureau, P. 1987. A collaborative study of an
Jackson, A. C., and Reimer, D. L. 1989. Pathogenesis of ex- experimental kit for rapid rabies enzyme immunodiag-
perimental rabies in mice: An immunohistochemical nosis (RREID). Bull WHO 65(4):489–493.
study. Acta Neuropathol 78(2):159–165. Reichel, K., and Möckelmann. 1963. Tollwut bein Schwein.
Johnson, R. T. 1971. The pathogenesis of experimental ra- Tierärztl Umsch 18:445. Abstr Vet Bull 34(189):143.
bies. In Rabies. Ed. Y. Nagano and F. Davenport. Balti- 1964.
more, London, Tokyo: University Park Press, pp. 59–72. Runnells, R. A.; Monlux, W. S.; and Monlux, A. W. 1965.
Jubb, K. F. V.; Kennedy, P. C.; and Palmer, N. 1985. Patholo- Principles of Veterinary Pathology, 7th ed. Ames: Iowa
gy of Domestic Animals, vol. 1. New York: Academic State Univ Press.
Press, p. 295. Schoening, H. 1956. Rabies. In Animal Diseases. Yearbook
Kelser, R. A. 1947. Rabies. In Diseases Transmitted from An- of Agriculture. Washington, D.C.: USDA, p. 199.
imals to Man, 3d ed. Ed. T. G. Hull. Springfield, Ill.: Smith, H. A.; Jones, T. C.; and Hunt, R. D. 1972. Veterinary
Charles C. Thomas, p. 338. Pathology, 4th ed. Philadelphia: Lea & Febiger, p. 353.
Kucera, P.; Dolivo, M.; Coulon, P.; and Flamand, A. 1985. Smith, J. S.; Sumner, J. W.; Roumillat, L. F.; Baer, G. M.; and
Pathways of the early propagation of virulent and aviru- Winkler, W. G. 1984. Antigenic characteristics of iso-
lent rabies strains from the eye to the brain. J Virol lates associated with a new epizootic of racoon rabies in
55:158–162. the United States. J Infect Dis 149:769–774.
Lennette, E. H., and Emmons, R. W. 1971. The laboratory Sokol, F.; Schlumberger, D.; Witkor, T. J.; Koprowski, H.;
diagnosis of rabies: Review and perspective. In Rabies. and Hummeler, K. 1969. Biochemical and biological
Ed. Y. Nagano and F. Davenport. Baltimore, London, studies on the nucleocapsid and RNA of rabies virus. Vi-
Tokyo: University Park Press, p. 77. rology 38:651.
Lennette, E. H.; Woodie, J. D.; Nakamura, K.; and Magoffin, U.S. Department of Health, Education, and Welfare (HEW).
R. L. 1965. The diagnosis of rabies by fluorescent anti- 1964. Communicable Disease Center Zoonoses Surveil-
body method (FRA) employing immune hamster serum. lance, Annual Rabies Summary. Public Health Serv,
Health Lab Sci 2(12):24. Washington, D.C.
McQueen, J. L.; Lewis, A. L.; and Schneider, N. J. 1960. ———. 1970. Communicable Disease Center Zoonoses
Diagnosis by fluorescent antibody. I. Its evaluation in Surveillance. Annual Rabies Summary. Public Health
a public health laboratory. Am J Public Health Serv, Washington, D.C.
50:1743. ———. 1969–83. Communicable Disease Center Zoonoses
Melnick, J. L., and McCombs, R. M. 1966. Classification and Surveillance. Annual Rabies Summary. Public Health
nomenclature of animal viruses. Prog Med Virol 8:400. Serv, Washington, D.C.
Merrimann, G. M. 1966. Rabies in Tennessee swine. J Am U.S. Department of Health and Human Services (HHS).
Vet Med Assoc 148:809. 1989. Rabies Surveillance Annual Summary for 1988.
Morehouse, L. G.; Kintner, L. D.; and Nelson, S. L. 1968. Ra- Atlanta: Centers for Disease Control.
bies in swine. J Am Vet Med Assoc 153:56. Webster, W. A.; Casey, G. A.; and Charlton, K. M. 1986. Ma-
Murphy, F. A. 1975. Rabies virus: Morphology and mor- jor antigenic groups of rabies virus in Canada deter-
phogenesis. In The Natural History of Rabies. Ed. G. M. mined by antinucleocapsid monoclonal antibodies.
Baer. New York, London: Academic Press, p. 33. Comp Immunol Microbiol Infect Dis 9(1):59–69.
Murphy, F. A.; Bauer, S. P.; Harrison, A. K.; and Winn, W. C. Witkor, T. J. 1971. Nature and properties of the rabies virus
1973a. Comparative pathogenesis of rabies and rabies- in rabies. In Rabies. Ed. Y. Nagano and F. M. Davenport.
like viruses—Viral infection and transit from inocula- Baltimore, London, Tokyo: University Park Press,
tion site to the central nervous system. Lab Invest p. 37.
28:361–376. Yates, W. D. H.; Rehmtulla, A. J.; and McIntosh, D. W. 1983.
Murphy, F. A.; Harrison, A. K.; Winn, W. C.; and Bauer, S. P. Porcine rabies in western Canada. Can Vet J 24:162–163.
Rotavirus and Reovirus
21 P. S. Paul and G. W. Stevenson

PORCINE ROTAVIRUS
Rotaviruses are important enteric pathogens in neonates ETIOLOGY
of many species, including pigs. The enteropathogenici-
ty of porcine rotaviruses has been demonstrated by their Virus Morphology
ability to produce severe gastroenteritis and villous atro- Rotavirus particles are nonenveloped and have a distinc-
phy under experimental conditions in gnotobiotic and tive wheel-like appearance when examined by electron
colostrum-deprived pigs in the absence of other microscopy. Rota is “wheel” in Latin—hence, they were
pathogens. They are also frequently detected in pigs with classified as rotavirus. Complete rotavirus particles have
diarrhea. Subclinical infections with porcine rotaviruses a well-defined outer rim, a feature that distinguishes
are equally common and suggest that other factors such them from the other two genus members, reovirus and
as host, agent, and environment may be important in the orbivirus, of family Reoviridae. Three forms of virus par-
pathogenesis of disease by porcine rotaviruses. ticles are visible by negative-staining electron mi-
Rotaviruses were first detected in calves (Mebus et al. croscopy. The double-capsid virus particles have an out-
1969) and were subsequently identified in humans and er capsid layer, an inner capsid layer, and an icosahedral
other animals (Estes et al. 1983). Serologic evidence for core and measure approximately 75 nm in diameter (Fig.
rotavirus infection in pigs was first shown using bovine 21.1A). The single-capsid virus particles are approxi-
rotavirus as an antigen (Woode and Bridger 1975). Sub- mately 60 nm in diameter and lack the outer capsid layer
sequently, porcine rotaviruses were detected by other re- (Fig. 21.1B). Cores are approximately 52 nm in diameter
searchers (Rodger et al. 1975; Lecce et al. 1976; McNulty and contain viral genome and RNA-dependent RNA
et al. 1976; Woode et al. 1976; Chasey and Lucas 1977; polymerase. Only double-capsid virus particles are infec-
Bohl et al. 1978; Tzipori and Williams 1978). Porcine ro- tious.
taviruses have now been detected in many of the swine-
producing countries (Lecce et al. 1976; McNulty et al. Classification
1976; Woode et al. 1976; Bohl et al. 1978; Lecce and King Rotaviruses are classified in the family Reoviridae under
1978; Bridger 1980; Corthier et al. 1980; Saif et al. 1980; the genus Rotavirus. Early studies demonstrated that all
Bohl et al. 1982; Askaa et al. 1983; Nilsson et al. 1984; rotaviruses, regardless of species of origin are antigeni-
Utrera et al. 1984; Theil et al. 1985; Dea et al. 1986; Fer- cally related (Woode et al. 1976; Thouless et al. 1977;
rari et al. 1986; Fu and Hampson 1987; Kim et al. 1987; Estes et al. 1983). The common group antigen is con-
Nagesha and Holmes 1988; Paul et al. 1988a; Atii et al. tained in viral structural protein VP6 on the inner capsid
1990; Bellinzoni et al. 1990; Magar and Larochelle 1992; (Estes and Cohen 1989). It became clear, however, that
Akopian et al. 1992; He et al. 1992; Gouvea et al. 1994; rotaviruses are quite diverse antigenically, for rotaviruses
Ciarlet and Liprandi 1994; Ciarlet et al. 1995, 1996; that resembled classical rotaviruses but lacked common
Pongsuwanna et al. 1996). Rotaviruses, regardless of group antigen were detected (Bridger 1980; Saif et al.
species of origin, were initially shown to share a com- 1980; Askaa and Bloch 1981; McNulty et al. 1981;
mon group antigen (Woode et al. 1976; Thouless et al. Bridger et al. 1982). Such rotaviruses have been termed
1977; Estes et al. 1983). It was later shown that ro- non–group A rotaviruses, novel rotaviruses, rotavirus-
taviruses are quite diverse serologically (Bridger 1980; like viruses, pararotaviruses, atypical rotaviruses, and
Saif et al. 1980; Askaa and Bloch 1981; McNulty et al. antigenically distinct rotaviruses (Bridger 1980; Saif et
1981). To date seven distinct serogroups of rotaviruses al. 1980; Bridger et al. 1982; Pedley et al. 1983; Snodgrass
have been described (Pedley et al. 1986; Bridger 1987). et al. 1984; Theil et al. 1985; Pedley et al. 1986; Bridger
Group A rotaviruses appear to be the most common and 1987). Recent studies show that some epitopes on VP6
best studied. Therefore, the majority of the information are conserved and are shared among different rotavirus
presented here pertains to porcine group A rotaviruses. groups (Tsunemitsu et al. 1992c).
In recent years, however, information on other groups of Serogroups with common group antigen on the inner
rotaviruses has begun to emerge and will be included capsid were identified by serologic tests such as im-
here where appropriate.
255
256 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

21.1. Rotavirus particles in feces viewed by negative-staining electron microscopy (×130,000).


(A) Double-shelled virus particles with intact outer capsids have characteristic smooth outlines.
(B) Single-shelled particles lack outer capsids and have spiked outlines.

munofluorescence, enzyme-linked immunosorbent as- used to differentiate rotaviruses into groups (Pedley et al.
say (ELISA), or immune electron microscopy (Bridger 1986). RNA segments cluster in four regions, I–IV (Fig.
1980; Saif et al. 1980; Pedley et al. 1983; Theil and Saif 21.2). Group A rotaviruses have 4:2:3:2 segments in re-
1985; Chasey et al. 1986; Pedley et al. 1986). Seven anti- gions I, II, III, and IV respectively. This pattern is 4:2:2:3
genically distinct groups (A–G) of rotaviruses have now for group B rotaviruses, and 4:3:2:2 for group C ro-
been reported, four (A, B, C, and E) of which affect pigs taviruses. In group E rotaviruses the pattern is similar to
(Bridger 1980; Saif et al. 1980; Bohl et al. 1982; Pedley et that seen with group B rotaviruses except segments 7–11
al. 1983; Chasey et al. 1986; Pedley et al. 1986). Group A migrate equidistant from each other (Pedley et al. 1986).
rotaviruses represent the earliest recognized rotaviruses, There appears to be a correlation between electro-
which appear to be most commonly associated with gas- pherotype and serogroup; however, it is emphasized that
troenteritis and are best characterized. Group B ro- serologic and nucleic acid–based assays are more reli-
taviruses, also referred to as atypical rotaviruses or ro- able.
tavirus-like viruses, have been detected in pigs, cattle, Rotaviruses within group A have been further divided
and humans (Bridger 1980; Bridger et al. 1982; Pedley et into subgroups and serotypes. Two subgroups, SI and SII,
al. 1983; Theil et al. 1985). Group C rotaviruses, also des- within group A rotaviruses have been reported (Green-
ignated as pararotaviruses, have been detected in pigs, berg et al. 1983a). Evidence indicates that viruses of ad-
cattle, and humans (Saif et al. 1980; Bohl et al. 1982; ditional subgroups may be prevalent in nature (Svensson
Pedley et al. 1983; Espejo et al. 1984; Bridger et al. 1986; et al. 1988). Serotypes within a serogroup are defined by
Tsunemitsu et al. 1991). Group E rotaviruses have been plaque reduction or fluorescent focus reduction assays
detected only in pigs in the United Kingdom (Chasey et using hyperimmune serum (Bohl et al. 1984; Hoshino et
al. 1986), whereas rotaviruses of groups D, F, and G have al. 1984; Paul et al. 1988a). A minimum of a 20-fold dif-
been detected in chickens and turkeys (Pedley et al. 1986; ference in the neutralization titer between the homolo-
Bridger 1987). gous and the heterologous rotavirus is required for a
Genomes of rotaviruses, when subjected to elec- virus to be considered a different serotype.
trophoresis in a polyacrylamide gel, yield a characteristic Serotyping is complex and sometimes gives ambigu-
electrophoretic migration commonly referred to as an ous results, due to involvement of two surface proteins,
electropherotype. Rotaviruses of serogroups identified VP4 and VP7, which induce neutralizing antibodies
thus far have unique electropherotypes, which have been (Greenberg et al. 1983b). The genes coding for these sur-
CHAPTER 21 ROTAVIRUS AND REOVIRUS Paul, Stevenson 257

face proteins segregate independently (Hoshino et al.


1985). A majority of hyperimmune antisera prepared
against the purified virions contain antibodies to VP7;
therefore, classic neutralization assays using hyperim-
mune antisera recognize serotypes based on VP7. A new
classification scheme has been proposed that takes into
account typing of both VP4 and VP7 surface proteins
(Estes and Cohen 1989). VP7 types are designated as G
types (for glycoprotein), and VP4 types are designated as
P types (for protease-sensitive protein).
A virus is designated by group/strain/subgroup/G
type/P type. At least 10 G (VP7) serotypes of porcine ro-
taviruses have been detected (Bohl et al. 1984; Hoshino
et al. 1984; Nagesha and Holmes 1988; Paul et al. 1988a;
Ruiz et al. 1988; Paul et al. 1990a, b, 1991; Gouvea et al.
1994; Bellinzoni et al. 1990; Ciarlet and Liprandi 1994;
Rosen et al. 1994): G1, G2, G3, G4, G5, G6, G8, G9, G10,
and G11. Information on G type and other serotype des-
ignations of selected rotavirus strains is presented in
Table 21.1. At least four P (VP4) types among porcine
group A rotaviruses have been detected (Estes 1996);
21.2. Electrophoretic patterns (electropherotypes) of however, information on P type on most porcine ro-
A/OSU, B/IA1146, and C/Cowden strains of groups tavirus strains is not available at this time. Monoclonal
A, B, and C rotaviruses in polyacrylamide gel stained antibodies (Kang et al. 1989; Nagesha et al. 1989; Kang et
with silver. Each rotavirus group has a distinct
al. 1993), nucleic acid probes (Johnson et al. 1990; Paul
electropherotype. Double-stranded RNA segments cluster
et al. 1990a, c; Rosen et al. 1992), and polymerase chain
in four regions, I–IV. The numbers of bands in the four
regions are 4:2:3:2, 4:2:2:3, and 4:3:2:2 for rotavirus reaction (PCR) methods (Winiarzyk and Paul 1997) have
groups A, B, and C respectively. become available, which are allowing typing of porcine
rotaviruses by G and P types in selected laboratories. Re-
cent studies suggest that serotypic diversity also exists
among group C rotaviruses, as evidence has been pre-

Table 21.1. Serogroup and serotype designations of selected porcine rotavirus isolates
Serotypea
Serogroup Strain(s) G (VP7) P (VP4)b Reference
A C60, C80, and C95 1 ? Bellinzoni et al. 1990; Ciarlet and Liprandi 1994
C134 2 Bellinzoni et al. 1990
CRW-8 3 7 Nagesha and Holmes 1988
ISU-65 3 ? Paul et al. 1988a, 1991
Gottfried 4 Bohl et al. 1984; Hoshino et al. 1984
SB-IAc 4 7 Hoshino et al. 1984, 1988
OSU 5 7 Bohl et al. 1984; Hoshino et al. 1984
6 6 Gouvea et al. 1994
8 Gouvea et al. 1994
ISU-64 9 7 Paul et al. 1988a, 1991
10 Gouvea et al. 1994
YM 11 Ruiz et al. 1988
YM-like 11 Rosen et al. 1994
B Ohio Theil et al. 1985
NIAD-1 Pedley et al. 1983
IA1146 Paul, unpublished data—1990; Lyoo et al. 1993
C Cowden Bohl et al. 1982
IA850 Proescholdt 1991
E DC-9 Pedley et al. 1986
a
Based on classification scheme proposed by Estes and Cohen (1989).
b
Although four P (VP4) types of porcine rotavirus have been identified by nucleic methods (Estes 1966), only two P (VP4) types
(listed here) have been conclusively identified by serologic methods.
c
Natural reassortant.
258 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

sented for the existence of at least two serotypes of IA, with a G4 and P7 type, has been described (Hoshino
group C porcine rotaviruses (Tsunemitsu et al. 1992b). et al. 1984; Midthun et al. 1987).
The buoyant density of the double-shelled, single-
Physicochemical and Biological Properties shelled, and core particles in cesium chloride is 1.36
Rotaviruses are extremely stable and are resistant to en- g/mL, 1.38 g/mL, and 1.44 g/mL respectively (Bridger
vironmental conditions such as temperature, pH, chemi- and Woode 1976; Bican et al. 1982). Some strains of
cals, and disinfectants. Rotaviruses are stable at pH 3–9. porcine rotavirus possess a hemagglutinin (Eiguchi et al.
In fecal preparations rotaviruses are resistant to temper- 1987; P. S. Paul, unpublished data—1990) and aggluti-
atures of 60˚C for 30 minutes and are resistant to tem- nate human type O, guinea pig, and rat erythrocytes.
peratures of 18–20˚C for at least 7–9 months (Woode The cellular receptor for group A rotaviruses has been
1986). Cell culture–adapted rotaviruses vary in suscepti- identified as monosialoganglioside or a family of mono-
bility to heat. Strains A/OSU and C/Cowden were inacti- sialogangliosides with sialic acid as an important epitope
vated by heat at 56˚C after 30 minutes, whereas the for virus binding (Rolsma et al. 1994).
A/Gottfried strain was resistant to heat at 56˚C (Terrett
et al. 1987). In the presence of organic material ro- Cultivation
taviruses are inactivated by 2% acid glutaraldehyde, 70% Rotaviruses have been difficult to cultivate in cell cul-
ethanol, 3.7% formaldehyde, 10% povidone-iodine, 67% tures. Porcine rotaviruses were first adapted to grow in
chloramine T, and 0.5% triclosan (Sattar et al. 1983; porcine primary kidney cell cultures by pretreatment of
Woode 1986). Rotaviruses are resistant to treatments by virions with trypsin or pancreatin (Theil et al. 1977).
ether, chloroform, and genetron. Calcium chloride, but Viruses were later successfully propagated in African
not magnesium chloride, stabilizes infectivity. Calcium monkey kidney cell line MA-104 (Bohl et al. 1984). Use of
ion chelators such as ethylenediaminetetraacetic acid roller cultures of MA-104 and addition of proteolytic en-
(EDTA) at 5 mM concentration remove the outer layer zymes, trypsin or pancreatin, were essential in isolation
and destroy infectivity. of rotaviruses. Trypsin enhances the growth of bovine
The rotavirus genome is double-stranded RNA and rotaviruses as much as 1000-fold (Almeida and Hall
has 11 segments. The total genomic size of rotavirus is 1978). Many strains of group A porcine rotaviruses have
estimated at approximately 18,522 base pairs by nucleic now been cultivated in roller cultures of the MA-104 cell
acid sequence and electron microscopy (Rixon et al. line by pretreatment of virions with trypsin (10 µg/mL
1984; Estes and Cohen 1989). The inner capsid is com- for 30 minutes) or pancreatin before infection. Alterna-
posed of two major structural proteins: VP2 with a mol- tively, trypsin or pancreatin (0.5–1.0 µg/mL) is added to
ecular mass (Mr) of 94 kDa and VP6 with a Mr of 41 kDa serum-free culture medium after virus adsorption. Cell
(Estes and Cohen 1989). The VP6 protein contains the culture–adapted rotavirus strains produce a cytopathic
major group and subgroup antigens. Minor subgroup- effect characterized by rounding of cells followed by re-
specific epitopes are also located on VP2 (Greenberg et moval of cells from the surface. Viral antigen can be
al. 1983a; Svensson et al. 1990). Other capsid proteins in- demonstrated in cytoplasm of virus-infected cells by im-
clude VP1 with a Mr of 125 kDa and VP3 with a Mr of 88 munofluorescence (Fig. 21.3) or immunochemical meth-
kDa. The outer capsid has two major surface proteins— ods. Rotaviruses form plaques under agarose in the pres-
VP4 with a Mr of 88 kDa and VP7 with a Mr of 37 kDa— ence of neutral red.
and several other proteins, including VP5 with a Mr of One strain of group C rotaviruses has been propagat-
60 kDa and VP8 with a Mr of 28 kDa (Estes and Cohen ed in porcine primary kidney cell cultures (Terrett and
1989). The VP4 protein is nonglycosylated, contains Saif 1987). Roller cultures and incorporation of high
hemagglutinin, and is important for infectivity and viru- concentrations of pancreatin in culture medium were es-
lence (Greenberg et al. 1983a; Kalica et al. 1983; Offit et sential for virus propagation (Terrett and Saif 1987). Af-
al. 1986a). Proteolytic cleavage of VP4 into VP5 and VP8 ter nine passages in primary kidney cells, the group C ro-
is important for viral infectivity. It is coded in gene seg- tavirus was adapted to grow in the MA-104 cell line (Saif
ment 4. The VP7 is the second most abundant viral et al. 1988). A porcine intestinal cell line has been suc-
structural protein, is glycosylated, and is coded in either cessfully used to propagate C/Cowden and an Iowa
gene segment 8 or 9 depending upon the strain of virus. strain, C/IA850, of group C rotaviruses (Proescholdt
Proteins analogous to VP6 and VP7 have been described 1991). Roller cultures and addition of high concentra-
for group C rotaviruses (Jiang et al. 1990). Both VP4 and tions of pancreatin were required for virus growth.
VP7 induce neutralizing antibodies (Greenberg et al. Groups B and E rotaviruses and some strains of group A
1983b; Offit and Blavat 1986), are important in induc- rotaviruses still cannot be serially propagated in cell cul-
tion of immunity (Offit et al. 1986b), and segregate in- tures. Group B porcine rotaviruses produce syncytia in
dependently (Hoshino et al. 1985). Thus, theoretically, monolayers of MA-104 following centrifugation of virus
reassortant rotavirus strains containing any combina- (Theil and Saif 1985). Viral antigen is present in inocu-
tion of P and G types are possible. One such strain, SB- lated cells without evidence for productive infection.
CHAPTER 21 ROTAVIRUS AND REOVIRUS Paul, Stevenson 259

acquired and appears to last 3–5 weeks (Tzipori et al.


1980a). Piglets born to gilts are more susceptible to ro-
tavirus infection than those born to sows (Askaa et al.
1983; Svensmark et al. 1989a). Pigs usually become in-
fected between 7 and 41 days of age and are infrequent-
ly infected at less than 7 days of age (Bohl et al. 1978). In
one cohort study (Fu and Hampson 1987), pigs became
infected at an average age of 19 days, with a range of
13–39 days. The virus is shed in feces and this is the ma-
jor source of infection; the greatest amount of the virus
is shed during the acute stages of infection. The duration
of group A porcine rotavirus shedding in feces is about
7.4 days, with a range of 1–14 days. Virus shedding is af-
fected by the level of passive immunity and serogroup of
porcine rotavirus. In group B rotavirus infections, lower
amounts of rotavirus are shed for a shorter duration
(Bridger 1980; Theil et al. 1985).
Recurrent infections with porcine rotaviruses with in-
termittent virus shedding have been detected (Debouck
and Pensaert 1983). These possibly represent infections
of different serogroups or serotypes. Adult animals do
21.3. Immunofluorescent staining of MA-104 cell not generally shed rotavirus (Debouck and Pensaert
line infected with group A porcine rotavirus showing 1983; Fu and Hampson 1987); however, occasionally
cytoplasmic accumulation of rotaviral antigens (× 400).
they may shed rotavirus near farrowing (Benfield et al.
1982) and may serve as sources of infection for suscepti-
ble pigs.
Such cultures with cytoplasmic viral antigen have proven Limited data are available on the relative prevalence
useful in a cell culture immunofluorescent assay (CCIF) of rotaviruses, especially of different serogroups and
for the detection of antibodies (Theil and Saif 1985). serotypes. Rotavirus infections account for about 14% of
diarrheas in pigs (Janke et al. 1989, 1990; Paul et al.
EPIDEMIOLOGY 1990c). In one study (Atii et al. 1990) porcine rotavirus
was detected in 43 of 96 (44.8%) samples from diarrheic
Porcine rotavirus is ubiquitous in swine herds wherever pigs but not in any of 41 samples from nondiarrheic
swine are raised. Serologic surveys show that a high pigs. There was also a higher infection rate (30.2%) in
percentage (77–100%) of the adult swine are seropositive piglets aged 1–3 weeks compared to pigs aged 4–6 weeks
for porcine groups A, B, and C rotaviruses (Bridger and (14.6%) (Atti et al. 1990). Group A rotavirus is by far the
Brown 1985; Terrett et al. 1987). Antibodies to group E predominant porcine rotavirus detected in diarrheic pigs
rotaviruses are also common in the United Kingdom (Janke et al. 1989; Paul et al. 1990c). Janke et al. (1989)
(Chasey et al. 1986). It is difficult to raise pigs free of ro- examined 90 rotavirus-positive samples and found that
taviruses under normal husbandry conditions. 60 contained group A, 9 contained group B, 11 contained
There appears to be an age-related variation in sero- group C, and 10 contained a mixture of these groups. In
logic prevalence, for antibodies to group B and C were nursing pigs the majority were group A; whereas in
lowest in pigs 3–8 weeks of age, whereas all pigs of this weaned pigs 38% were group A, 19% were group B, and
age had antibodies to the group A rotaviruses (Bridger 24% were group C. In another study, 89% of the ro-
and Brown 1985). Similarly, antibodies to group C ro- tavirus-positive samples were positive for group A ro-
taviruses were detected in 100% of adults, 59% of wean- tavirus (Paul et al. 1990c). In a study in Thailand, 26 of
lings, and 86% of nursing pigs tested from Ohio (Terrett the 557 fecal samples were positive for rotavirus by poly-
et al. 1987). acrylamide gel electrophoresis of RNA: 23 were group A,
Infection with porcine rotaviruses is enzootic in 1 was group B, and 2 were group C (Pongsuwanna et al.
swine herds. The virus spreads among pigs via the fecal- 1996). Group A rotaviruses with P types 6 and 7 are most
oral route. Following natural infection with a rotavirus, abundant (Zaberezhny et al. 1994). In one study, out of
pigs develop circulating and secretory antibodies and are 26 rotavirus-positive diarrheic field samples, 17 samples
immune to infection with homologous rotaviruses. Anti- (65.4%) had P type 7; 5 (15.4%) had P type 6; and 4
bodies are transferred to newborns through colostrum (15.4%) were negative for both P6 and P7 (Zaberezhny et
and milk; the duration of persistence of these passively al. 1994). Rotaviruses with G types 1, 2, 3, 4, 5, 6, 8, 9, 10,
acquired antibodies varies with the initial antibody level and 11 have been detected, with G types 3, 4, 5, 8, and 11
260 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

being predominant (Rosen et al. 1994; Gouvea et al. herds and may result in consistent yet differing patterns
1994; Winiarzyk and Paul 1997). Additional studies are of rotavirus-associated disease.
needed to fully determine the relative prevalence of ro- Naturally occurring rotavirus-associated diarrheal
tavirus serogroups and serotypes. disease is most often reported in 7- to 41-day-old suckling
Contaminated environment plays an important role pigs (Bohl et al. 1978; Roberts et al. 1980; Askaa et al.
in maintenance of rotaviruses in swine herds. Porcine ro- 1983; Debouck and Pensaert 1983; Svensmark et al.
taviruses are stable at extreme environmental conditions 1989b) or within 7 days following weaning (Woode et al.
and are resistant to commonly used disinfectants. 1976; Bohl et al. 1978; Lecce and King 1978; Tzipori et al.
Porcine rotaviruses can be detected in dry feces, dust, 1980a). The age of onset is often consistent in a given
and sewage of farrowing and weaner barns (Fu et al. herd. Where rotavirus is not complicated by concurrent
1989). Porcine rotaviruses have survived in swine barns infection with other enteric pathogens, diarrhea in suck-
kept empty for 3 months (Fu et al. 1989). ling pigs is usually mild and limited to 2–3 days’ dura-
tion. Feces are yellow or white, watery to creamy, and
CLINICAL SIGNS variably flocculant. Dehydration is usually mild and
mortality is less than 15% of clinically ill pigs. Morbidity
Numerous rotavirus inoculation studies in nonimmune is variable but is often 10–20%. Diarrhea sometimes pro-
gnotobiotic or colostrum-deprived pigs fed a liquid diet gresses to younger pigs in a farrowing room as more pigs
have resulted in consistent production of clinical dis- are affected. Dehydration is more severe in the younger
ease. Disease is most severe in pigs inoculated when 1–5 pigs. Gilt litters may be affected at a younger age with
days of age (Woode et al. 1976; Pearson and McNulty more severe disease than are sow litters. Diarrheic suck-
1977; Tzipori and Williams 1978; Debouck and Pensaert ling pigs frequently shed rotavirus along with other en-
1979; McAdaragh et al. 1980; Narita et al. 1982a). After teric pathogens such as transmissible gastroenteritis
an incubation period of 12–24 hours, pigs become (TGE) virus (Bohl et al. 1978), coccidia (Roberts et al.
anorexic and listless and occasionally vomit. Severe, pro- 1980), or enterotoxigenic Escherichia coli (Bohl et al.
fuse diarrhea begins 1–4 hours later. Feces are watery 1978). Diarrhea is generally more severe and morbidity
and yellow to white and contain variable amounts of and mortality are greater in combined infections.
flocculent material. Diarrhea continues for 3–5 days, The role of rotaviruses in diarrheic syndromes in re-
then feces progressively return to normal in 7–14 days. cently weaned pigs is unclear. Rotaviruses have been im-
Pigs become dehydrated and may die 2–5 days following plicated as a primary cause of severe diarrhea in recently
the onset of diarrhea; mortality may reach 50–100%. Di- weaned pigs resulting in 10–50% mortality (Woode
arrhea and dehydration are less severe in pigs innoculat- 1986); however, rotavirus inoculation studies in weaned
ed when 7–21 days old (Crouch and Woode 1978; Theil pigs do not support such conclusions. Where rotavirus
et al. 1978; Shaw et al. 1989a). Disease is limited to tran- shedding has been associated with severe diarrhea in
sient diarrhea of 1–1.5 days’ duration in pigs inoculated weaned pigs, it has been in combination with TGE virus
when 28 days old (Tzipori et al. 1980b; Lecce et al. 1982). (Bohl et al. 1978) or with hemolytic colony types of en-
Mortality decreases as the age at inoculation increases; terotoxigenic E. coli (Bohl et al. 1978; Tzipori et al. 1980a;
death is unusual in pigs older than 14 days. No clinical Lecce et al. 1982). Inoculation studies in weaned pigs
disease results when 21- to 28-day-old pigs that have con- with rotavirus and hemolytic colony types of enterotoxi-
sumed a dry diet for more than 3 days are inoculated genic E. coli suggest a subservient yet potentially impor-
with rotavirus (Tzipori et al. 1980b; Stevenson 1990; tant role for rotaviruses in severe diarrheic postweaning
Stevenson et al. 1990). syndromes in pigs (Tzipori et al. 1980b; Lecce et al.
Rotavirus inoculation studies conducted in nonim- 1982). Inoculation of weaned pigs with rotavirus or he-
mune pigs likely represent the more severe limits of ro- molytic E. coli alone results in mild transient diarrhea or
tavirus-induced clinical disease. Naturally occurring ro- no clinical disease; whereas inoculation of weaned pigs
tavirus infection in pigs often results in less severe with rotavirus followed by hemolytic E. coli results in ef-
clinical disease or is subclinical. Rotavirus is endemic in ficient hemolytic E. coli colonization and severe protract-
most swine herds; that is, nearly all gilts and sows are to ed diarrhea typical of that reported in natural outbreaks.
some degree immune. A commensurate degree of immu- Differences in virulence among group A rotavirus
nity is passed to suckling pigs via colostrum and milk strains have been reported in humans (Kapikian and
(Askaa et al. 1983). Levels of specific rotavirus antibod- Chanock 1990) and calves (Bridger and Pocock 1986).
ies in colostrum and milk decline dramatically in the first Differences in the outer capsid protein VP4 have been
few days of lactation, and rotavirus replication occurs in implicated as the cause of the differing virulence among
pigs when the oral challenge level of virus exceeds the human group A rotavirus strains (Kapikian and Chanock
consumed-milk-borne passive immunity. Management 1990). It is not clear whether there are differences in vir-
practices that impact either the level of passive immuni- ulence among serotypes or strains of swine group A ro-
ty or the level of oral virus challenge differ among swine taviruses. Inoculation studies in pigs with serotype 4
CHAPTER 21 ROTAVIRUS AND REOVIRUS Paul, Stevenson 261

(Bohl et al. 1984; Benfield et al. 1988), serotype 5 (Theil The degree of villous atrophy and the distribution of at-
et al. 1978; Janke et al. 1988), and a new serotype repre- rophic villi in the small intestine vary relative to the age
sented by strain ISU-64 (Stevenson 1990; Stevenson et al. of the pig and are primary factors determining the sever-
1990) have resulted in similar clinical disease and similar ity of clinical disease. In general, villous atrophy is more
lesions. One inoculation study suggested a difference in severe and extensive in younger pigs.
virulence between two strains of group A rotavirus in Numerous rotavirus inoculation studies in 1- to 7-
pigs (Woode et al. 1976); whereas other studies have day-old pigs have demonstrated that virus replication is
shown no difference in virulence among group A ro- usually most extensive in the distal one-half to two-
tavirus strains (Debouck and Pensaert 1979; Collins et al. thirds of the small intestine (Pearson and McNulty 1977;
1989). Genes encoding VP3, VP4, VP7, and NS28 pro- Theil et al. 1978; Narita et al. 1982a; Collins et al. 1989;
teins are believed to play an important role in viral viru- Stevenson 1990). Rotaviral antigen may be demonstrat-
lence (Hoshino et al. 1985). Little is known regarding ed in the cytoplasm in a few epithelial cells on villous tips
natural disease in swine caused by groups B, C, and E ro- in the duodenum and in nearly all villous epithelial cells
taviruses. Inoculation studies in gnotobiotic pigs with in the jejunum and ileum by 12–48 hours postinocula-
group C rotaviruses resulted in clinical disease typical of tion (Fig. 21.4). Villous atrophy is most severe by 24–72
group A rotaviruses (Bohl et al. 1982; Snodgrass et al. hours postinoculation, when villi may be as short as one-
1984). In contrast, inoculation studies in gnotobiotic tenth normal length in the jejunum and ileum. In con-
pigs with group B rotaviruses (Bridger et al. 1982; Theil trast, rotavirus inoculation studies in 21- to 24-day-old
et al. 1985) and group E (Chasey et al. 1986) resulted in pigs revealed the most extensive virus replication in the
less severe diarrhea of shorter duration than that ob- proximal one-half to two-thirds of the small intestine
served with groups A or C. (Shaw et al. 1989a; Stevenson 1990). Virus-infected ep-
ithelial cells were most often on the distal half of villi
PATHOGENESIS (Fig. 21.4), resulting in moderate villous atrophy. The
most atrophic villi were one-third normal length.
Rotaviruses replicate predominately in the cytoplasm of Diarrhea begins slightly before or concurrent with
differentiated small-intestinal villous epithelial cells and villous atrophy and has been attributed to multiple path-
to a lesser extent in the cytoplasm of M cells overlying ogenic mechanisms. Most proven mechanisms are relat-
Peyer’s patches (Buller and Moxley 1988) and in cecal or ed to decreased small-intestinal digestive and absorptive
colonic epithelial cells (Theil et al. 1978; Collins et al. functions caused by the loss of differentiated entero-
1989). Rotavirus replication results in small-intestinal cytes. Decreased small-intestinal disaccharidase activi-
villous epithelial cell dysfunction and death; lysis or ties (primarily lactase) result in the retention of disac-
desquamation of infected cells leads to villous atrophy. charides (primarily lactose) in the lumen of the small

A B
21.4. Rotavirus antigen in the cytoplasm of villous epithelial cells as viewed by the indirect
fluorescent antibody method (× 90). (A) Ileum from 1-day-old gnotobiotic pig 16 hours post-
inoculation with porcine rotavirus. Nearly all villous epithelial cells contain viral antigen.
(B) Midjejunum from a 27-day-old weaned conventional pig 3 days postinoculation.
Viral antigen is in epithelial cells on villous tips.
262 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

intestine. The excess disaccharides result in hyperosmo- villous atrophy and to sluggish crypt epithelial cell
larity of the intestinal contents, which causes an osmot- turnover. Complete replacement of villous epithelial
ic diarrhea (Davidson et al. 1977; Graham et al. 1984). cells requires 8–10 days in newborn pigs and 2–3 days in
Decreased small-intestinal Na+K+-ATPase activity and 21-day-old pigs (Moon 1971).
glucose-coupled Na+ absorption result in decreased Na+ The pathogenesis of groups B, C, and E rotaviruses in
absorption at villous tips. The normal secretion of Na+ pigs has been studied much less extensively than that of
and water from crypts into the small-intestinal lumen is group A rotaviruses. The limited information gained
not balanced by adequate absorption at villous tips and through inoculation studies in pigs with group B ro-
may result in net Na+ and water flux toward the small-in- taviruses (Pedley et al. 1983; Theil et al. 1985), group C
testinal lumen (Davidson et al. 1977; Graham et al. rotaviruses (Bohl et al. 1982; Snodgrass et al. 1984), and
1984). Recent studies in children have also suggested group E rotaviruses (Chasey et al. 1986) suggests a patho-
that prostaglandins, which are produced as a result of ro- genesis similar to that described for group A rotaviruses.
tavirus-induced enteric inflammation, may enhance the Virus replication was most extensive in the distal small
severity of rotavirus-associated diarrhea (Yamashiro et intestine for all rotavirus groups; however, villous atro-
al. 1989). Children with rotavirus-associated diarrhea phy was more severe with groups A and C than with
had higher serum and fecal levels of two prostaglandins groups B and E.
than did clinically normal children. Furthermore, oral as- The pathogenesis of combined infections of ro-
pirin therapy, which decreased serum and fecal tavirus and other commonly recognized en-
prostaglandin levels, also shortened the duration of di- teropathogens has been examined in a limited number of
arrhea. Similar studies in pigs have not been reported. studies. Inoculation of 3-day-old germ-free pigs with
Several interrelated factors may be involved in caus- group A rotavirus followed 24 hours later by K99 pilus-
ing the death of rotavirus-inoculated pigs. Diarrhea producing enterotoxigenic E. coli caused more severe di-
causes dehydration and electrolyte imbalances and may arrheal disease than did inoculation of pigs with either
lead to exhaustion of extracellular fluid reserves. Malab- agent separately. Villous atrophy was no more severe in
sorption results in malnutrition and may lead to energy pigs infected with both agents than in pigs inoculated
deficiency and hypothermia. Observed higher mortality with rotavirus alone (Benfield et al. 1988). Inoculation of
in neonatal pigs is most likely related to more severe and 28-day-old pigs with rotavirus followed by hemolytic
extensive villous atrophy, coupled with decreased extra- strains of enterotoxigenic E. coli resulted in more severe
cellular fluid and energy reserves, compared to slightly diarrheal disease than was seen in pigs inoculated with
older pigs. Suboptimal ambient temperatures have been either agent separately (Tzipori and Williams 1978; Lec-
shown to increase mortality in rotavirus-infected pigs. ce et al. 1982). It was concluded that rotavirus infection
Inoculation of newborn pigs with a human strain of enhances colonization of the small intestine by hemolyt-
group A rotavirus resulted in no mortality when pigs ic strains of enterotoxigenic E. coli. Inoculation of 7-day-
were maintained at 35˚C and resulted in 70–90% mortal- old germ-free pigs with rotavirus and TGE virus resulted
ity when pigs were maintained at 26˚C (Steel and Torres- in more severe diarrheal disease than in pigs inoculated
Medina 1984). Differences in mortality were attributed with rotavirus alone (Woode and Crouch 1978). In con-
to increased energy demands at lower ambient tempera- trast, simultaneous inoculation of 1-day-old pigs with ro-
tures combined with rotavirus-induced malnutrition. tavirus and enterovirus resulted in less severe diarrheal
Mortality may also be affected by virus dose. The mini- disease and less severe villous atrophy than was seen in
mum infective dose of two strains of porcine group A ro- pigs inoculated with rotavirus alone (Janke et al. 1988). It
tavirus has been shown to be as low as 1 plaque-forming was concluded that nonlethal enterovirus infection of
unit of virus (Graham et al. 1987; Shaw et al. 1989b). In villous epithelial cells interfered with subsequent ro-
another study, as few as 90 viral particles were infectious tavirus infection.
and caused diarrhea in 4-day-old pigs (Payment and
Morin 1990). Virus titration studies using a porcine LESIONS
group A rotavirus strain and done in a limited number of
neonatal colostrum-deprived pigs suggested higher mor- The lesions caused by rotaviruses in pigs are limited to
tality in pigs inoculated with lower dilutions (higher the small intestines and are the sum of the degenerative
virus dose) of inoculum (Shaw et al. 1989b). and functional consequences of rotavirus-induced vil-
The duration of rotavirus-induced diarrhea is deter- lous epithelial cell destruction and the adaptive and re-
mined by the time needed for the restoration of an ade- generative responses of the small intestine.
quate number of differentiated villous epithelial cells in Gross lesions are visible slightly before or concurrent
the small intestine. Complete restoration of morpholog- with the onset of diarrhea and are most severe in 1- to 14-
ically normal villi in 3-day-old pigs requires 6–10 days; day-old pigs (Pearson and McNulty 1977; Theil et al.
whereas only 2–4 days are required in 21-day-old pigs. 1978; Benfield et al. 1988; Janke et al. 1988; Collins et al.
Recovery is slower in younger pigs owing to more severe 1989; Stevenson 1990). The stomachs usually contain
CHAPTER 21 ROTAVIRUS AND REOVIRUS Paul, Stevenson 263

food and the distal one-half to two-thirds of the small in- postinoculation. The time required for complete regen-
testine is thin walled and flaccid and dilated with a large eration of normal villi depends on the age of the pig.
volume of watery, flocculant, yellow or gray fluid. Fol- Scanning electron microscopy has allowed detailed
lowing 24 hours of diarrhea, increased small-intestinal study of the surface topography of affected small-intesti-
motility may result in less distension and a less thin- nal mucosa in numerous rotavirus inoculation studies
walled, more nearly normal appearance. The lacteals in (McAdaragh et al. 1980; Torres-Medina and Underdahl
the distal two-thirds of the intestine contain no chyle, 1980; Collins et al. 1989; Stevenson 1990). Degenerate
and the associated mesenteric lymph nodes are small villous epithelial cells lyse and/or detach, resulting in vil-
and tan. The cecum and colon are likewise dilated with lous erosion and shortening (Fig. 21.6A, B). Within a few
similar contents. Gross lesions are less severe or are ab- hours, eroded villous tips are covered with a continuous
sent in pigs that are 21 days of age or older (Shaw et al. layer of epithelial cells and there are few remain-
1989a; Stevenson 1990). ing swollen and degenerate epithelial cells (Fig.
Light microscopic lesions have been described in nu- 21.6C).
merous rotavirus inoculation studies in suckling pigs Ultrastructural studies in rotavirus-inoculated or nat-
(Woode et al. 1976; Pearson and McNulty 1977; Crouch urally infected pigs have revealed lesions in the cyto-
and Woode 1978; Theil et al. 1978; McAdaragh et al. plasm of virus-infected villous epithelial cells that are
1980; Benfield et al. 1988; Janke et al. 1988; Collins et al. typical of those described for rotaviruses in many other
1989; Stevenson 1990). Villous epithelial cell degenera- mammalian and avian species (Chasey 1977; Saif et al.
tion begins in cells on villous tips and in groups of cells 1978; Pearson and McNulty 1979; Narita et al. 1982b).
on lateral villi by 16–18 hours postinoculation. Degener- Virus-infected cells contain within their cytoplasm mul-
ative cells are swollen and have rarified cytoplasm, tiple, variably sized, electron-dense granular viroplasms
swollen nuclei, and irregular brush borders. Degenera- that often have dense subviral cores or single-shelled par-
tive cells are frequently partially detached from adjacent ticles on the periphery. Single-shelled viral particles ob-
cells or from the basement membrane. By 16–24 hours tain the outer capsid by budding through the mem-
postinoculation, sloughing of cells results in significant branes of the rough endoplasmic reticulum (Fig. 21.7).
villous atrophy, which is most severe by 24–72 hours Mature, 75–78 nm double-shelled virus particles accu-
postinoculation (Fig. 21.5). The tips of atrophic villi may mulate in the cysternae of the endoplasmic reticulum
be eroded or may be covered by attenuated, nearly squa- and are released by cell lysis. Other degenerative changes
mous epithelial cells, and there is a variable amount of in virus-infected cells include cell swelling, mitochondri-
cellular debris in the lamina propria. Contact of the ex- al swelling, nuclear swelling, dilatation of the cytocavity
posed lamina propria of adjacent villi results in villous network, and fragmentation of microvilli. Macrophages
fusion, which may be observed for 24–168 hours in the lamina propria contain cellular membrane pro-
postinoculation. Crypt epithelial hyperplasia results in files, virus particles, viroplasm, and other cellular debris
significantly deeper crypts beginning 48–72 hours in phagosomes.

A B
21.5. Ileum from 3-day-old gnotobiotic pigs (H&E; × 35). (A) Normal villi in an uninoculated
control pig. (B) Severe villous atrophy present 18 hours postinoculation.
264 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

A B C
21.6. Scanning electron micrograph of the ileum of 3-day-old gnotobiotic pigs. (A) Normal villi
in an uninoculated control pig (× 70). (B) Sloughing, degenerate villous epithelial cells and exposed
lamina propria on severely atrophic villi, 18 hours postinoculation (×165). (C) The tips of atrophic
villi are covered by a continuous layer of sometimes swollen and degenerate epithelial cells, 24
hours postinoculation (× 230).

21.7. Ultrastructure of a swollen


and degenerate rotavirus-infected
villous epithelial cell containing
dense granular viroplasm (arrow-
head). Virus particles form and
obtain outer capsids through
budding into the rough
endoplasmic reticulum (small
arrow). Many double-shelled virus
particles are within the cysternae
of the endoplasmic reticulum
(large arrows) (× 23,800).

DIAGNOSIS hours after the start of diarrhea. This is especially critical


for the detection of certain groups of rotaviruses, for the
Rotavirus should be considered as a cause of diarrhea in amount of virus shed is lower and the duration of virus
neonatal pigs 1–8 weeks of age. Fecal samples or intesti- shedding is shorter in pigs infected with group B ro-
nal contents should be collected in acute phases of dis- taviruses (Bridger 1980; Theil et al. 1985; P. S. Paul, un-
ease and submitted for diagnosis. Highest concentra- published data—1990).
tions of porcine rotavirus are shed during the first 24
CHAPTER 21 ROTAVIRUS AND REOVIRUS Paul, Stevenson 265

A number of methods have been employed for the entiation of rotavirus groups and serotypes (Dimitrov et
detection of rotaviruses, including electron microscopy, al. 1985; Johnson et al. 1990; Paul et al. 1990a, c; Rosen
immune electron microscopy, immunohistochemistry, et al. 1990a, b; Koromyslov et al. 1990; Ojeh et al. 1993;
immunofluorescence on frozen sections or impression Gouvea et al. 1994; Rosen et al. 1994; Zaberezhny et al.
smears of small intestines, ELISA, virus isolation, latex 1994).
agglutination, dot blot hybridization, RNA electro- Serologic tests are of little value in the diagnosis of
pherotyping, and PCR (Paul et al. 1988b; Johnson et al. rotavirus infection because antibodies are common in
1990; Rosen et al. 1990a, b; Koromyslov et al. 1990; most swine herds; however, antibody titers and isotypes
Sanekata et al. 1991; Ojeh et al. 1992; Magar and shed light on the immune status of animals. Antibodies
Larochelle 1992; Jiang et al. 1992; Ojeh et al. 1993; Rosen to rotavirus can be detected by a number of serologic
et al. 1994; Zaberezhny et al. 1994). Electron microscopy tests. The indirect immunofluorescence test has often
has been extensively used for the detection of rotavirus- been used to detect antibodies to porcine rotavirus
es and remains a reference method for their detection. (Theil and Saif 1985; Chasey et al. 1986). Monolayers of
This method allows detection of different serogroups of MA-104 cells are inoculated with group A, B, or C ro-
rotaviruses. The morphology of the virus particles de- tavirus. The virus is allowed to adsorb onto cells in the
tected by negative-staining electron microscopy is affect- case of group A rotaviruses or is centrifuged onto cells in
ed by the staining methodology utilized and the the case of group B or C rotaviruses. Cells are fixed with
serogroup of rotavirus. Negative staining with phospho- an acetone-methanol mixture and used as antigen in an
tungstic acid (PTA) of neutral pH results in predomi- indirect immunofluorescence test. Alternatively, cryostat
nantly single-shelled capsids; whereas PTA of pH 4.5 or intestinal sections from pigs infected with noncultivable
uranyl acetate gives double-shelled particles (Nakata et rotaviruses may be used as the source of antigens. Anti-
al. 1987; Suzuki et al. 1987). Core particles have been de- bodies to serogroups B, C, and E have been detected by
tected in group B rotaviruses (Bridger et al. 1982; Theil et this method. Indirect ELISA has also been used to detect
al. 1985). Immune electron microscopy allows their dif- antibodies to rotaviruses. Antibodies detected by the
ferentiation into serogroups. above methods are usually directed against the common
Immunohistochemistry employing immunogold group antigen. Antibodies to group C have also been de-
staining with protein A gold and specific antisera has tected by blocking ELISA using monoclonal antibodies
been used to detect group A rotaviruses in formalin-fixed (Tsunemitsu et al. 1992a). ELISA combined with isotype-
paraffin-embedded sections of small intestines (Magar specific monoclonal antibodies (Paul et al. 1985, 1989)
and Larochelle 1992). has been used to detect classes of antibodies to rotavirus
ELISA is frequently used for the detection of rotaviral (Paul et al. 1986). Plaque reduction and fluorescent focus
antigens in fecal samples or intestinal contents. It is reduction neutralization assays have been used to detect
more sensitive than the latex agglutination test (Goyal et neutralizing antibodies (Hoshino et al. 1984; Paul et al.
al. 1987) but less sensitive than electron microscopy 1988a). The hemagglutination inhibition test has also
(Benfield et al. 1984; Benfield 1990). Human-based diag- been developed for the detection of antibodies to
nostic kits may be used for the detection of porcine porcine rotaviruses (Eiguchi et al. 1987).
group A rotaviruses. Monoclonal antibody capture
ELISA has been developed for the detection of group C TREATMENT
rotaviruses (Ojeh et al. 1992; Tsunemitsu et al. 1992a).
Electropherotyping of viral RNA is often used for the No known therapeutic agents are available for the specif-
detection and differentiation of rotavirus groups. Ro- ic treatment of porcine rotavirus infections. General sup-
taviruses of different serogroups have distinct electro- portive therapy, management procedures, and antibi-
pherotypes (Fig. 21.2), which only provide a tentative di- otics are recommended to minimize mortality due to
agnosis of a serogroup and should be confirmed by rotaviruses and secondary bacterial infections. Elec-
serologic or nucleic acid–based methods. For electro- trolyte solutions containing glucose-glycine fed ad libi-
pherotyping, viral RNA can be isolated in a crude form tum minimize dehydration and weight loss induced by
directly from feces and analyzed by polyacrylamide gel rotavirus infection (Bywater and Woode 1980; Bywater
electrophoresis (Paul et al. 1988b) or purified by CF-11 1983). Ambient environment should be optimized, min-
cellulose chromatography (Theil et al. 1981). RNA bands imizing draft and temperature fluctuations. Antibiotic
in gels are visualized by ethidium bromide or silver-stain- therapy may be used to reduce losses from secondary
ing methods. bacterial infections. In herds with a persistent problem
Terminal oligonucleotide fingerprinting has been of postweaning diarrhea with high mortality, a change in
used in differentiating porcine rotaviruses and has been weaner diet and weaning procedure should be consid-
useful in molecular epidemiological studies (Clarke and ered. Scheduled feeding of a high-energy weaner diet has
McCrae 1981). Nucleic acid probes and the PCR method been successfully used to reduce morbidity and mortali-
offer attractive alternatives for the detection and differ- ty (Tzipori et al. 1980a; Hampson and Beban 1985).
266 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

PREVENTION testinal tract of suckling pigs. Secretory IgA is the pre-


dominant Ig in milk of lactating sows and is the primary
Management Ig providing passive protection.
Rotavirus infection persists endemically in most swine The route of immunization of sows influences the
herds. Rotaviruses survive in dust and organic matter classes of Igs produced. Primary immunization by the
and may persist as subclinical infections in adult swine oral route is the best method known to date for induction
(Benfield et al. 1982). They are therefore difficult, if not of sIgA in milk. Parenteral routes, however, may be used
impossible, to eliminate. Studies have shown that in- to boost sIgA in animals previously primed by oral im-
creasing concentrations of rotavirus in the environment munization. The level of attenuation of a rotavirus also
cause more severe disease at a younger age (Lecce et al. influences its ability to induce lactogenic immunity. In-
1978). Thus, management practices should be directed activated vaccines are not immunogenic but modified-
at reducing the virus load for susceptible pigs and boost- live-virus vaccines are immunogenic. The best immunity
ing passive immunity. Virus load may be reduced by san- is induced by virulent virus because it induces optimal
itation and limiting contact between young susceptible intestinal immune responses (Saif and Fernandez 1996).
pigs and older pigs more likely to be shedding virus. The Induction of active immunity against postweaning diar-
floors in farrowing and nursing houses should be con- rhea in the presence of passive milk antibodies is prob-
structed to minimize feces buildup during use and to fa- lematic. Oral immunization is required for optimal prim-
cilitate cleaning. All-in/all-out management practices ing of the sIgA response, yet passively acquired milk
should be followed and rooms should be thoroughly antibodies will likely interfere in replication of immuniz-
cleaned and disinfected between groups. Recommended ing rotavirus. Immunization by the parenteral route is
disinfectants include formaldehyde and chlorine-based not as likely to result in protective immunity.
disinfectants such as chlorox or chloramine T (Multi- Immunity to rotaviruses, in general, is serotype spe-
chlor). The farrowing interval should be minimized to cific (Bohl et al. 1982; Losonsky et al. 1986). Pigs orally
prevent virus buildup and infection of later-farrowed lit- immunized with the A/OSU strain were resistant to in-
ters. Mixing animals of different ages should be discour- fection and were protected from gastroenteritis follow-
aged because it promotes virus transmission from older ing challenge with the A/OSU strain, but not from chal-
to younger pigs. To enhance passive immunity, replace- lenge with the A/Gottfried strain (Bohl et al. 1982).
ment gilts should be exposed to the feces of older sows. Similarly, pigs immunized with the A/Gottfried strain
Attention to lactation diet, feed intake, sow comfort, and were protected from challenge with A/Gottfried, but not
farrowing-crate design is important to ensure adequate from the A/OSU strain. This study demonstrated feasi-
milk supply and effective suckling necessary for success- bility for the development of a modified-live-rotavirus
ful immunity. vaccine. Two surface proteins, VP4 and VP7, are impor-
tant in induction of neutralizing antibodies and in pro-
Immunoprophylaxis tection (Greenberg et al. 1983b; Offit et al. 1986b; Hoshi-
Pigs recovered from a natural infection with rotavirus are no et al. 1988). In addition to serotype-specific epitopes,
immune to infection with homologous rotavirus for at these proteins also possess heterotypic epitopes, which
least a short time; the duration of immunity is unknown. are conserved among different serotypes. Heterotypic
The mechanisms of immunity have not been completely immunity is not as effective as that induced by homotyp-
elucidated. Protection is generally correlated with the ic strains. Reassortant rotaviruses containing the VP4
presence of antibodies in secretions. Antibodies in the and VP7 genes from two different serotypes may be used
intestinal lumen produced by local lymphoid cells bathe to induce immunity to both serotypes (Offit et al. 1986b;
susceptible villous epithelial cells and protect them from Hoshino et al. 1988). The reassortant rotavirus SB-1A,
infection (Chen et al. 1995; Saif and Fernandez 1996). containing the VP4 gene from the A/OSU strain and the
Secretory IgA (sIgA) is more effective than IgG and IgM VP7 gene from the A/Gottfried strain, induced neutral-
because sIgA is more resistant than IgG and IgM to pro- izing antibodies for both the OSU and Gottfried strains
teolytic degradation in the intestinal tract and IgA is the and protected orally immunized pigs from challenge
predominant immunoglobulin (Ig) in secretions. Circu- with both the OSU and Gottfried strains (Hoshino et al.
lating serum antibodies do not provide protection 1988). Protection, however, was greater against the
against rotavirus infections (Offit and Clark 1985). Stud- Gottfried strain than against the OSU strain.
ies in mice indicate that cellular immunity may also be Modified-live-virus and inactivated-virus vaccines are
important in protection against rotavirus infections (Of- commercially available for immunization of sows as well
fit and Dudzik 1988, 1989). as nursing pigs. Modified-live-virus vaccines are admin-
Piglets suckling immune sows are protected from ro- istered orally, orally and intramuscularly, or intramuscu-
tavirus gastroenteritis during the nursing period. This larly. Inactivated-virus vaccines are administered intra-
passive immunity is related to the continuous presence muscularly in sows and intraperitoneally in nursing pigs.
of specific virus-neutralizing antibodies in the gastroin- Not all of the commercially available vaccines have been
CHAPTER 21 ROTAVIRUS AND REOVIRUS Paul, Stevenson 267

evaluated by independent investigators. Two separate rotavirus serotypes (Table 21.1) may require their incor-
studies of a modified-live-rotavirus vaccine for post- poration into vaccines if they are found to be a signifi-
weaning diarrhea resulted in conflicting data. In one cant cause of gastroenteritis in swine. The VP4 and VP7
study, immunization of nursing pigs improved the genes of porcine rotavirus have been cloned and se-
weight gain and reduced the severity of disease (Wester- quenced (Gorziglia et al. 1986, 1988, 1990; Nishikawa
camp 1986). In the second study, neither improvement and Gorziglia 1988), and VP4 of one strain has been ex-
in weight gain nor curtailment of virus shedding was ob- pressed in baculovirus vector. VP4 protein and viruslike
served (Hoblet et al. 1986). Bovine rotavirus vaccine is particles are immunogenic. Recombinant proteins and
not effective in preventing rotaviral diarrhea in pigs (Lec- reassortant rotaviruses offer potential alternatives as im-
ce and King 1979). Recent identification of new porcine munogens for rotavirus gastroenteritis.

PORCINE REOVIRUS
Reoviruses have been detected from healthy pigs as well inner capsid is about 45–50 nm in diameter. Reovirus
as from pigs with respiratory and reproductive diseases replicates in the cytoplasm.
(Kasza 1970; Kirkbride and McAdaragh 1978; McFerran
1986). Their role in these swine diseases is unclear. Re- Physicochemical and Biological Properties
oviruses are associated with tenosynovitis in chickens Reoviruses have a segmented (10 segments), double-
(Van der Heide 1977; Robertson and Wilcox 1986) and stranded, RNA genome. The density of a complete (ma-
systemic illness affecting the majority of organs in ro- ture) virion in cesium chloride is 1.36 g/cm2. Porcine re-
dents (Tyler and Fields 1990) and thus have the potential oviruses are resistant to ether, chloroform, and trypsin.
to be significant pathogens in swine. They are stable at acidic pH 3 and are sensitive to 0.1%
sodium deoxycholate (Hirahara et al. 1988). Porcine re-
ETIOLOGY oviruses are susceptible to heat at 50˚C for 1 hour but are
stabilized by the presence of 1 molar MgCl2. They pos-
Virus Morphology sess a hemagglutinin that agglutinates human group O
Reovirus was the first genus identified in the family Re- and porcine erythrocytes at 4˚C, 22˚C, and 37˚C, but not
oviridae. The other two genuses of importance to ani- erythrocytes from guinea pig, rat, hamster, cattle, dog,
mals are Rotavirus and Orbivirus. “Reo” is an acronym for cat, or chicken. Mammalian reoviruses share a group
“respiratory and enteric orphan.” Reovirus particles are antigen that can be detected by complement fixation, im-
nonenveloped and icosahedral in shape with a fuzzy out- munofluorescence, and immunodiffusion (Sabin 1959).
er rim; they measure 75 nm in diameter (Fig. 21.8). The Avian reoviruses are not antigenically related to mam-
malian reoviruses but possess a group antigen that is
shared by avian reoviruses. All mammalian isolates can
be divided serologically into three types: types 1, 2, and
3. Reoviruses of different types can be distinguished by
serum neutralization and hemagglutination inhibition
tests.

Cultivation
Reoviruses can be cultivated in a wide variety of cell cul-
tures from many species: cell cultures of porcine, bovine,
feline, simian (African monkey kidney cell lines Vero and
MA-104), human (HeLa cell line), and canine (canine
thyroid adenocarcinoma cell line) origin (Kasza 1970;
Hirahara et al. 1988; P. S. Paul, unpublished data—
1990). Reovirus replication is slow and the majority
(80%) of the nascent rotavirus population remains cell
associated (McFerran 1986).
The cytopathic effect of reoviruses varies somewhat,
dependent on the cell line used. In general, cells round
21.8. Electron micrograph of reovirus particles viewed up, become granular, and slough off the surface.
by negative-staining electron microscopy (×115,000). Eosinophilic intracytoplasmic inclusion bodies can easi-
268 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

ly be seen in cultures stained with May-Greenwald- ly as 24 hours postinfection, and excretion may continue
Giemsa stain. for 14 days. The virus can also be isolated from boar se-
men (McAdaragh and Anderson 1975). The virus was
EPIDEMIOLOGY isolated from fecal tissues and placenta of sows inoculat-
ed intravenously or intramuscularly with type 3 reovirus.
Porcine reovirus infections appear to be widespread in The virus was recovered only from fetal tissues of sows
swine herds, and antibodies to all three types have been inoculated at 30–60 days of gestation, from both placen-
detected in pigs (Harkness et al. 1971; Zindel et al. 1980). tal and fetal tissues of sows inoculated at 50–70 days of
In a study by Zindel et al. (1980), pigs in 54% of the swine gestation, and from only placenta of sows inoculated af-
herds surveyed were seropositive for all three reovirus ter 70 days of gestation (McAdaragh and Anderson
types. Passively acquired antibodies to reoviruses persist 1975). Hemagglutination-inhibiting antibodies may be
in neonatal pigs for about 11 weeks, at which time pigs detected at 7 days postinfection and reach peak titers be-
become susceptible to infection (Watt 1978). Reoviruses tween 11 and 21 days postinfection (McFerran 1986).
are spread via fecal-oral and respiratory routes. Follow-
ing experimental infection, virus is shed in nasal secre- LESIONS
tions and feces for 6 and 14 days, respectively. Virus is
spread by contact exposure (McFerran et al. 1971). A limited number of reovirus inoculation studies in
swine have revealed few gross lesions and only mild mi-
CLINICAL SIGNS croscopic lesions. Although intravenous or intramuscu-
lar inoculation of sows between 40 and 85 days of gesta-
Reoviruses have been isolated from clinically healthy tion with reovirus resulted in litters containing a mixture
pigs as well as pigs with respiratory or enteric disease of mummified fetuses, stillborn pigs, small weak pigs,
(Kasza 1970; McFerran and Connor 1970; Robl et al. and normal pigs, no specific gross or histopathologic le-
1971; Elazhary et al. 1978; McFerran 1986) and from sions were reported (McAdargh and Robl 1976). Oral in-
aborted fetuses (Kirkbride and McAdaragh 1978). oculation of 1-week-old cesarean-derived colostrum-de-
Experimental studies have not consistently resulted prived pigs with enteric-origin reovirus resulted in focal
in reproduction of disease. In a majority of studies, in- villous atrophy in the jejunum and ileum (Elazhary et al.
tranasal, intraperitoneal, or intracerebral inoculation of 1978). Aerosol exposure of 4-week-old specific-
conventional and gnotobiotic pigs of 1–6 weeks of age pathogen-free (SPF) pigs to porcine-origin type 1 re-
with porcine reovirus type 1 or human reovirus type 1 ovirus resulted in no gross lesions; however, there were
did not result in clinical disease except for a transient consistent microscopic lesions in the lungs consisting of
febrile reaction (Kasza 1970; McFerran and Connor multifocal aggregates of lymphocytes and macrophages
1970; Baskerville et al. 1971; McFerran et al. 1971; Watt in alveoli and alveolar septae and mild peribronchiolar
1978). Virus could be isolated from respiratory and ali- nodular lymphocytic hyperplasia (Baskerville et al.
mentary tracts for up to 2 weeks postinfection. In one 1971). Intranasal inoculation of 70 kg SPF pigs with a
study, a rise in body temperature and diarrhea were de- porcine respiratory isolate of type 3 reovirus resulted in
tected in cesarean-derived colostrum-deprived 1-week- lobular atalectasis and vesicular emphysema and peri-
old pigs inoculated orally with type 1 reovirus; however, bronchiolar nodular lymphocytic hyperplasia, which
no clinical disease was detected in gnotobiotic pigs varied in intensity between lobules. Additional studies
(Elazhary et al. 1978). Mild respiratory disease charac- are needed to clearly characterize the clinical disease and
terized by pyrexia, sneezing, inappetance, and listless- lesions in swine caused by porcine reoviruses.
ness was reproduced in cesarean-derived colostrum-de-
prived pigs and conventional pigs inoculated DIAGNOSIS
intranasally with or exposed via aerosol to reovirus type
1 (Hirahara et al. 1988). Intravenous or intramuscular in- A number of methods may be employed for the detec-
oculation of seronegative sows between 40 and 85 days tion of reoviruses, including electron microscopy, im-
of gestation with reovirus resulted in term litters con- mune electron microscopy, immunofluorescence, virus
taining a mixture of mummified, stillborn, weak, and isolation, and RNA electropherotyping. Virus isolation
normal pigs. has commonly been used for diagnosis. A wide variety of
cells may be used; primary swine kidney cells and
PATHOGENESIS African monkey kidney cell lines such as MA-104 are ex-
tremely susceptible and are commonly available. Typing
Reoviruses replicate mainly in intestinal and respiratory of reovirus is achieved by virus neutralization and
tracts and can be isolated from nasal and fecal swabs. hemagglutination inhibition tests with reference antisera
The virus is excreted in nasal secretions and feces as ear- to three reovirus types.
CHAPTER 21 ROTAVIRUS AND REOVIRUS Paul, Stevenson 269

TREATMENT AND PREVENTION G.; and Theil, K. W. 1978. Rotavirus as a cause of diar-
rhea in pigs. J Am Vet Med Assoc 172:458–463.
No methods are available for treatment or prevention of Bohl, E. H.; Saif, L. J.; Theil, K. W.; Agnes, A. G.; and Cross,
porcine reovirus infections, and possibly none are war- R. F. 1982. Porcine pararotavirus: Detection, differentia-
ranted until the clinical significance of reovirus infec- tion from rotavirus, and pathogenesis in gnotobiotic
tions in swine is documented. pigs. J Clin Microbiol 15:312–319.
Bohl, E. H.; Theil, K. W.; and Saif, L. J. 1984. Isolation and
REFERENCES serotyping of porcine rotaviruses and antigenic compar-
ison with other rotaviruses. J Clin Microbiol 19:105–111.
Akopian, T. A.; Lunin, V. G.; Kruglyak, V. A.; Ruchadze, G. Bridger, J. C. 1980. Detection by electron microscopy of cali-
G.; Bakhutashvili, V. I.; Naroditsky, B. S.; and Ticho- civiruses, astroviruses, and rotavirus-like particles in the
nenko, T. I. 1992. Nucleotide sequence of the cDNA for feces of piglets with diarrhea. Vet Rec 107:532–533.
porcine rotavirus VP7 gene (strain K). Virus Genes ———. 1987. Novel rotaviruses in animals and man. In
6:393–396. Novel Diarrhea Viruses. CIBA Found Symp 128.
Almeida, J. D., and Hall, T. 1978. The effect of trypsin on the Chichester: Wiley, pp. 6–23.
growth of rotavirus. J Gen Virol 40:213–218. Bridger, J. C., and Brown, J. F. 1985. Prevalence of antibody
Askaa, J., and Bloch, B. 1981. Detection of porcine rotavirus to typical and atypical rotaviruses in pigs. Vet Rec
by EM, ELISA and CIET. Acta Vet Scand 22:32–38. 116:50.
Askaa, J.; Bloch, B.; Bertelsen, G.; and Rasmussen, K. O. Bridger, J. C., and Pocock, D. H. 1986. Variation of virulence
1983. Rotavirus-associated diarrhea in nursing piglets of bovine rotaviruses. J Hyg 96:257–264.
and detection of antibody against rotavirus in Bridger, J. C., and Woode, G. N. 1976. Characterization of
colostrum, milk, and serum. Nord Vet Med 35:441–447. two particle types of calf rotavirus. J Gen Virol
Atii, D. J.; Ojeh, C. K.; and Durojaiye, O. A. 1990. Detection 31:245–250.
of rotavirus antigen in diarrhoeic and non-diarrhoeic Bridger, J. C.; Clarke, I. N.; and McCrae, M. A. 1982. Char-
piglets in Nigeria. Rev Elev Med Vet Pays Trop acterization of an antigenically distinct porcine ro-
42:494–496. tavirus. Infect Immun 35:1058–1062.
Baskerville, A.; McFerran, J. B.; and Conner, T. 1971. The Bridger, J. C.; Pedley, S.; and McCrae, M. A. 1986. Group C
pathology of experimental infection of pigs with type 1 rotaviruses in humans. J Clin Microbiol 23:760–763.
reovirus of porcine origin. Res Vet Sci 12:172–174. Buller, C. R., and Moxley, R. A. 1988. Natural infection of
Bellinzoni, R. B.; Mattion, N. M.; Matson, D. O.; Blackhall, porcine ileal dome M cells with rotavirus and enteric
J.; La Torre, J. L.; Scodeller, E. A.; Urasawa, S.; Taniguchi, adenovirus. Vet Pathol 25:516–517.
K.; and Estes, M. K. 1990. Porcine rotaviruses antigeni- Bywater, R. J. 1983. Diarrhea treatments—Fluid replace-
cally related to human rotavirus serotypes 1 and 2. J Clin ment and alternatives. Ann Rech Vet 14:556–560.
Microbiol 28:633–636. Bywater, R. J., and Woode, G. N. 1980. Oral fluid replace-
Benfield, D. A. 1990. Diagnosis of rotavirus infections in ment by a glucose glycine electrolyte formulation in E.
food animals. In Proc Commer Diagn Technol Anim coli and rotavirus diarrhea in pigs. Vet Rec 106:75–78.
Health Monit. Ames: Nat Vet Serv Lab and Iowa State Chasey, D. 1977. Different particle types in tissue culture
Univ Vet Diagn Lab, pp. 181–193. and intestinal epithelium infected with rotavirus. J Gen
Benfield, D. A.; Stotz, I.; Moore, R.; and McAdaragh, J. P. Virol 37:443–451.
1982. Shedding of rotavirus in feces of sows before and Chasey, D., and Lucas, M. 1977. Detection of rotavirus in ex-
after farrowing. J Clin Microbiol 16:186–190. perimentally infected piglets. Res Vet Sci 22:124–125.
Benfield, D. A.; Stotz, I. J.; Nelson, E. A.; and Groon, K. S. Chasey, D.; Bridger, J. C.; and McCrae, M. A. 1986. A new
1984. Comparison of a commercial enzyme-linked im- type of atypical rotavirus in pigs. Arch Virol
munosorbent assay with electron microscopy, fluores- 89:235–243.
cent antibody, and virus isolation for the detection of Chen, W. K.; Campbell, T. , Vancott, J.; and Saif, L. J. 1995.
bovine and porcine rotavirus. Am J Vet Res Enumeration of isotype-specific antibody-secreting cells
45:1998–2002. derived from gnotobiotic piglets inoculated with
Benfield, D. A.; Francis, D. H.; McAdaragh, J. P.; Johnson, D. porcine rotaviruses. Vet Immunol Immunopathol
D.; Bergeland, M. E.; Rossow, K.; and Moore, R. 1988. 45:265–284.
Combined rotavirus and K99 Escherichia coli infection in Ciarlet, M., and Liprandi, F. 1994. Serological and genomic
gnotobiotic pigs. Am J Vet Res 49:330–337. characterization of two porcine rotaviruses with
Bican, R.; Cohen, J.; Charpiliene, A.; and Scherrer, R. 1982. serotype G1 specificity. J Clin Microbiol 32:269–272.
Purification and characterization of bovine rotavirus Ciarlet, M.; Ludert, J. E.; and Liprandi, F. 1995. Comparative
cores. J Virol 43:1113–1117. amino acid sequence analysis of the major outer capsid
Bohl, E. H.; Kohler, E. M.; Saif, L. J.; Cross, R. F.; Agnes, A. protein (VP7) of porcine rotaviruses with G3 and G5
270 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

serotype specificities isolated in Venezuela and Argenti- Ferrari, M.; Gualandi, G. L.; and Gelmetti, D. 1986. Isola-
na. Arch Virol 140:437–451. tion of cytopathic strains of rotavirus from pigs. Micro-
Ciarlet, M.; Hidalgo, M.; and Liprandi, F. 1996. Cross–reac- biologica (Bologna) 9:287–294.
tive, serotype– and monotype–specific neutralization Fu, Z. F., and Hampson, D. J. 1987. Group A rotavirus excre-
epitopes on VP7 of serotype G3 and G5 porcine ro- tion patterns in naturally infected pigs. Res Vet Sci
tavirus strains. Arch Virol 141:601–614. 43:297–300.
Clarke, I. M., and McCrae, M. A. 1981. A sensitive method Fu, Z. F.; Hampson, D. J.; and Blackmore, D. K. 1989. Detec-
for the production of diagnostic fingerprints of genome tion and survival of group A rotavirus in a piggery. Vet
segments of field isolates of rotavirus. J Viro Methods Rec 125:576–578.
3:261–269. Gorziglia, M.; Aguirre, Y.; Hoshino, Y.; Esparza, J.; Blumen-
Collins, J. E.; Benfield, D. A.; and Duimstra, J. R. 1989. tals, I.; Askaa, J.; Thompson, M.; Glass, R. I.; Kapikian,
Comparative virulence of two porcine group–A ro- A. Z.; and Chanock, R. M. 1986. VP7 serotype–specific
tavirus isolates in gnotobiotic pigs. Am J Vet Res glycoprotein of OSU porcine rotavirus: Coding assign-
50:827–835. ment and gene sequence. J Gen Virol 67:2445–2454.
Corthier, G.; Cohen, J.; and Scherrer, R. 1980. Isolation of Gorziglia, M.; Nishikawa, K.; Green, K.; and Taniguchi, K.
pig rotavirus in France: Identification and experimental 1988. Gene sequence of the VP7 serotype specific glyco-
infections. Ann Rech Vet 11:45–48. protein of Gottfried porcine rotavirus. Nucleic Acids Res
Crouch, C. F., and Woode, G. N. 1978. Serial studies of virus 16:775.
multiplication and intestinal damage in gnotobiotic Gorziglia, M.; Nishikawa, K.; Hoshino, Y.; and Taniguchi, K.
piglets infected with rotavirus. J Med Microbiol 1990. Similarity of the outer capsid protein VP4 of the
11:325–334. Gottfried strain of porcine rotavirus to that of asympto-
Davidson, G. P.; Gall, D. G.; Petric, M.; Butler, D. G.; and matic human rotavirus strains. J Virol 64:414–418.
Hamilton, J. R. 1977. Human rotavirus enteritis induced Gouvea, V.; Santos, N.; and Carmo Timenetsky, M. 1994.
in conventional piglets. J Clin Invest 60:1402–1409. Identification of bovine and porcine rotavirus G types
Dea, S.; Elazhary, M. A. S. Y.; and Roy, R. S. 1986. Distinct by PCR. J Clin Microbiol 32:1338–1340.
serotypes of porcine rotavirus associated with diarrhea Goyal, S. M.; Rademacher, R. A.; and Pomeroy, K. A. 1987.
in suckling piglets in southern Quebec. Can J Vet Res Comparison of electron microscopy with three commer-
50:130–132. cial tests for the detection of rotavirus in animal feces.
Debouck, P., and Pensaert, M. 1979. Experimental infection Diagn Microbiol Infect Dis 6:249–254.
of pigs with Belgian isolates of the porcine rotavirus. Graham, D. Y.; Sackman, J. W.; and Estes, M. K. 1984.
Zentralbl Veterinärmed (B)26:517–526. Pathogenesis of rotavirus-induced diarrhea. Prelimi-
———. 1983. Rotavirus excretion in suckling pigs followed nary studies in miniature swine piglets. Dig Dis Sci
under field circumstances. Ann Rech Vet 14:447–448. 29:1028–1035.
Dimitrov, D. H.; Graham, D. Y.; and Estes, M. K. 1985. De- Graham, D. Y.; Dufour, G. R.; and Estes, M. K. 1987. Mini-
tection of rotaviruses by nucleic acid hybridization with mal infective dose of rotavirus. Arch Virol 92:261–271.
cloned DNA of simian rotavirus SA11 genes. J Infect Dis Greenberg, H. B.; Flores, J.; Kalica, A. R.; Wyatt, R. G.; and
152:293–300. Jones, R. 1983a. Gene coding assignments for growth re-
Eiguchi, Y.; Yamagishi, H.; Fukusho, A.; Shimizu, Y.; and striction, neutralization and sub–group specificities of
Matumoto, M. 1987. Hemagglutination and hemagglu- the W and DS-1 strains on human rotovirus. J Gen Virol
tination-inhibition tests with porcine rotavirus. Kitasato 64:313–320.
Arch Exp Med 60:167–172. Greenberg, H. B.; Valdesuso, J.; Van Wyke, K.; Midthun, K.;
Elazhary, M. A. S. Y.; Morin, M.; Derbyshire, J. B.; Lagace, Walsh, M.; McAuliffe, V.; Wyatt, R. G.; Kalica, A. R.; Flo-
A.; Berthiaume, L.; and Corbeil, M. 1978. The experi- res, J.; and Hoshino, Y. 1983b. Production and prelimi-
mental infection of piglets with porcine reovirus. Res nary characterization of monoclonal antibodies direct-
Vet Sci 25:16–20. ed at two surface proteins of Rhesus rotavirus. J Virol
Espejo, R. T.; Puerto, F.; Soler, C.; and Gonzalez, N. 1984. 47:267–275.
Characterization of a human pararotavirus. Infect Im- Hampson, D. J., and Beban, R. L. G. 1985. Diet and control
mun 44:112–116. of porcine post-weaning diarrhea. NZ Vet J 33:190.
Estes, M. 1996. Rotaviruses and their replication. In Fields Harkness, J. W.; Chapman, M. S.; and Derbyshire, J. H.
Virology, 3d ed. Philadelphia: Lippincott–Raven, pp. 1971. A survey of antibodies to some respiratory viruses
1625–1656. in the sera of pigs. Vet Rec 88:441–447.
Estes, M. K., and Cohen, J. 1989. Rotavirus gene structure He, K. W.; Lin, J. H.; Ding, Z. D.; He, J. H.; Jiang, J. Y.; and
and function. Microbiol Rev 53:410–449. Hou, J. B. 1992. Serotyping and antigenic comparison of
Estes, M. K.; Palmer, E. L.; and Obijeski, J. D. 1983. Ro- some animal rotaviruses isolated in China. Zentralbl
taviruses: A review. Curr Top Microbiol Immunol Veterinärmed 39:299–302.
105:123–184. Hirahara, T.; Yasuhara, H.; Matsui, O.; Kodama, K.; Naki,
CHAPTER 21 ROTAVIRUS AND REOVIRUS Paul, Stevenson 271

M.; and Sasaki, N. 1988. Characteristics of reovirus type Kang, S. Y.; Benfield, D. A.; Gorziglia, M.; and Saif, L. J.
1 from the respiratory tract of pigs in Japan. Jpn J Vet Sci 1993. Characterization of the neutralizing epitopes of
50:353–361. VP7 of the Gottfried strain of porcine rotavirus. J Clin
Hoblet, K. H.; Saif, L. J.; Kohler, E. M.; Theil, K. W.; Bech- Microbiol 31:2291–2297.
Nielson, S.; and Stitzlein, G. A. 1986. Efficacy of an oral- Kapikian, A. Z., and Chanock, R. M. 1990. Rotaviruses. In
ly administered modified-live porcine-origin rotavirus Virology, 2d ed. New York: Raven Press, pp. 1353–1404.
vaccine against postweaning diarrhea in pigs. Am J Vet Kasza, L. 1970. Isolation and characterization of a reovirus
Res 47:1697–1703. from pigs. Vet Rec 87:681–686.
Hoshino, Y.; Wyatt, R. G.; Greenberg, H. B.; Flores, J.; and Kim, Y. H.; Lee, O. S.; Choi, C. U.; and An, S. H. 1987. Epi-
Kapikian, A. Z. 1984. Serotype similarity and diversity demiological studies on porcine rotavirus infection in
of rotaviruses of mammalian and avian origin as stud- piglets. Res Rep Rural Div Adm 29:129–133.
ied by plaque-reduction neutralization. J Infect Dis Kirkbride, C. A., and McAdaragh, J. P. 1978. Infectious
149:694–702. agents associated with fetal and early neonatal death
Hoshino, Y.; Sereno, M. M.; Midthun, K.; Flores, J.; and abortion in swine. J Am Vet Med Assoc
Kapikian, A. Z.; and Chanock, R. M. 1985. Independent 172:480–483.
segregation of two antigenic specificities (VP3 and VP7) Koromyslov, G. F.; Artjushin, S. K.; Zaberezhny, A. D.; and
involved in neutralization of rotavirus infectivity. Proc Grashuk, V. N. 1990. Development of hybridization
Nat Acad Sci (USA) 82:8701–8704. probes on the basis of recombinant DNA to identify
Hoshino, Y.; Saif, L. J.; Sereno, M. M.; Chanock, R. M.; and porcine parvoviruses and rotaviruses. Arch Exp Veter-
Kapikian, A. Z. 1988. Infection immunity of piglets to inärmed 44:897–900.
either VP3 or VP7 outer capsid protein confers resis- Lecce, J. G., and King, M. W. 1978. Role of rotavirus (reo-
tance to challenge with a virulent rotavirus bearing the like) in weanling diarrhea of pigs. J Clin Microbiol
corresponding antigen. J Virol 62:744–748. 8:454–458.
Janke, B. H.; Morehouse, L. G.; and Solorzano, R. F. 1988. ———. 1979. The calf reo-like virus (rotavirus) vaccine: An
Single and mixed infections of neonatal pigs with ro- ineffective immunization agent for rotaviral diarrhea of
taviruses and enteroviruses: Clinical signs and micro- piglets. Can J Comp Med 43:90–93.
scopic lesions. Can J Vet Res 52:364–369. Lecce, J. G.; King, M. W.; and Mock, R. 1976. Reovirus-like
Janke, B. H.; Nelson, J. K.; Benfield, D. A.; and Nelson, E. A. agent associated with fatal diarrhea in neonatal pigs. In-
1989. Relative prevalence of typical and atypical strains fect Immun 14:816–825.
among rotaviruses from diarrheic pigs in conventional Lecce, J. G.; King, M. W.; and Dorsey, W. E. 1978. Rearing
swine herds. In Proc 32d Annu Meet Am Assoc Vet Lab regimen producing piglet diarrhea (rotavirus) and its
Diagn, Las Vegas, p. 24. relevance to acute infantile diarrhea. Science
———. 1990. Relative prevalence of typical and atypical 199:776–778.
strains among rotaviruses from diarrheic pigs in con- Lecce, J. G.; Balsbaugh, R. K.; and Clare, D. A. 1982. Ro-
ventional swine herds. J Diagn Invest 2:308–311. tavirus and hemolytic enteropathogenic Escherichia coli
Jiang, B. M.; Saif, L. J.; Kang, S. Y.; and Kim, J. H. 1990. Bio- in weanling diarrhea in pigs. J Clin Microbiol
chemical characterization of the structural and non- 16:715–723.
structural polypeptides of a porcine group C rotavirus. J Losonsky, G. A.; Vonderfecht, S. L.; Eiden, J.; Wee, S.; and
Virol 64:3171–3178. Yolken, R. H. 1986. Homotypic and heterotypic anti-
Jiang, B. M.; Qian, Y.; Tsunemitsu, H.; Green, K. Y.; and Saif, bodies for prevention of experimental rotavirus gas-
L. J. 1992. Analysis of the gene encoding the outer cap- troenteritis. J Clin Microbiol 24:1041–1044.
sid glycoprotein (VP7) of group C rotaviruses by North- Lyoo, Y. S.; Mummidi, S.; Stout, B.; and Paul, P. S. 1993.
ern and dot blot hybridization. Virol 184:433–436. Molecular cloning and partial sequence analysis of a
Johnson, M. E.; Paul, P. S.; Gorziglia, M.; and Rosenbusch, porcine group B rotavirus gene. Proc Conf Res Workers
R. 1990. Development of specific nucleic acid probes for Anim Dis, p. 13.
the differentiation of porcine rotavirus serotypes. Vet Magar, R.; and Larochelle, R. 1992. Immunohistochemical
Microbiol 24:307–326. detection of porcine rotavirus using immunogold silver
Kalica, A. R.; Flores, J.; and Greenberg, H. B. 1983. Identifi- staining (IGSS). J Vet Diagn Invest 4:3–7.
cation of the rotaviral gene that codes for the hemagglu- McAdaragh, J. P., and Anderson, G. A. 1975. Transmission
tinin and protease-enhanced plaque formation. Virolo- of viruses through boar semen. In Proc 18th Annu Meet
gy 125:194–205. Am Assoc Vet Lab Diagn, pp. 69–76.
Kang, S.; Saif, L. J.; and Miller, K. L. 1989. Reactivity of VP4- McAdaragh, J. P., and Robl, M. G. 1976. Experimental re-
specific monoclonal antibodies to a serotype 4 porcine ovirus infections of pregnant sows. Proc Int Congr Pig
rotavirus with distinct serotypes of human (sympto- Vet Soc 4:DD1.
matic and asymptomatic) and animal rotaviruses. J Clin McAdaragh, J. P.; Bergeland, M. E.; Meyer, R. C.; Johnshoy,
Microbiol 27:2744–2750. M. W.; Stotz, I. J.; Benfield, D. A.; and Hammer, R. 1980.
272 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Pathogenesis of rotaviral enteritis in gnotobiotic pigs: A Offit, P. A., and Clark, H. F. 1985. Protection against ro-
microscopic study. Am J Vet Res 41:1572–1581. tavirus–induced gastroenteritis in a murine model by
McFerran, J. B. 1986. Reovirus infection. In Diseases of passively acquired gastrointestinal but not circulating
Swine, 6th ed. Ames: Iowa State Univ Press, pp. antibodies. J Virol 54:58–64.
390–394. Offit, P. A., and Dudzik, K. I. 1988. Rotavirus-specific cyto-
McFerran, J. B., and Connor, T. 1970. A reovirus isolated toxic T lymphocytes cross-react with target cells infected
from a pig. Res Vet Sci 11:388–390. with different rotavirus serotypes. J Virol 62:127–131.
McFerran, J. B.; Baskerville, A.; and Connor, T. 1971. Exper- ———. 1989. Rotavirus-specific cytotoxic T lymphocytes
imental infection of pigs with a human strain of type 1 appear at the intestinal mucosal surface after rotavirus
reovirus. Res Vet Sci 12:174–175. infection. J Virol 63:3507–3512.
McNulty, M. S.; Pearson, G. R.; McFerran, J. B.; Collins, D. Offit, P. A.; Blavat, G.; Greenberg, H. B.; and Clark, H. F.
S.; and Allan, G. M. 1976. A reovirus-like agent (ro- 1986a. Molecular basis of rotavirus virulence: Role of
tavirus) associated with diarrhea in neonatal pigs. Vet gene segment 4. J Virol 57:46–49.
Microbiol 1:55–63. Offit, P. A.; Clark, H. F.; Blavat, G.; and Greenberg, H. B.
McNulty, M. S.; Allan, G. M.; Todd, D.; McFerran, J. B.; and 1986b. Reassortant rotaviruses containing structural
McCracken, R. M. 1981. Isolation from chickens of a ro- proteins vp3 and vp7 from different parents induce an-
tavirus lacking the rotavirus group antigen. J Gen Virol tibodies protective against each parental serotype. J Vi-
55:405–413. rol 60:491–496.
Mebus, C. A.; Underdahl, N. R.; Rhodes, M. B.; and Ojeh, C. K.; Tsunemitsu, H.; Simkins, R. A.; and Saif, L. J.
Twiehaus, M. J. 1969. Calf Diarrhea (Scours): Repro- 1992. Development of a biotin-streptavidin-enhanced
duced with a Virus from a Field Outbreak. Univ Nebras- enzyme-linked immunosorbent assay which uses mono-
ka Agri Exp Stn Res Bull 233. clonal antibodies for detection of group C rotaviruses. J
Midthun, K.; Valdesuso, J.; Hoshino, Y.; Flores, J.; Kapikian, Clin Microbiol 30:1667–1673.
A. Z; and Chanock, R. M. 1987. Analysis by RNA-RNA Ojeh, C. K.; Parwani, A. V.; Jiang, B. M.; Theil, K. W.; Rosen,
hybridization assay of intertypic rotaviruses suggests B. I.; and Saif, L. J. 1993. Characterization of field iso-
that the gene reassortment occurs in vivo. J Clin Micro- lates of porcine group C rotaviruses using gene 5 (VP6)
biol 25:295–320. and gene 8 (VP7) cDNA probes. J Vet Diagn Invest
Moon, H. W. 1971. Epithelial cell migration in the alimen- 5:434–438.
tary mucosa of the suckling pig. Proc Soc Exp Biol Med Paul, P. S.; Van Deusen, R. A.; and Mengeling, W. L. 1985.
137:151–154. Monoclonal precipitating antibodies to porcine im-
Nagesha, H. S., and Holmes, I. H. 1988. New porcine ro- munoglobulin M. Vet Immunol Immunopathol
tavirus serotype antigenically related to human ro- 8:311–328.
tavirus serotype 3. J Clin Microbiol 26:171–174. Paul, P. S.; Mengeling, W. L.; Saif, L. J.; and Van Deusen, R.
Nagesha, H. S.; Brown, L. E.; and Holmes, I. A. 1989. Neu- A. 1986. Detection of classes of antibodies to transmis-
tralizing monoclonal antibodies against three serotypes sible gastroenteritis virus and rotavirus of swine using
of porcine rotavirus. J Virol 63:3545–3549. monoclonal antibodies to porcine immunoglobulins.
Nakata, S.; Petrie, B. L.; Calomeni, E. P.; and Estes, M. K. Proc Int Congr Pig Vet Soc 9:190.
1987. Electron microscopy procedure influences detec- Paul, P. S.; Lyoo, Y. S.; Andrews, J. J.; and Hill, H. T. 1988a.
tion of rotaviruses. J Clin Microbiol 25:1902–1906. Isolation of two new serotypes of porcine rotavirus from
Narita, M.; Fukusho, A.; Konno, S.; and Shimizu, Y. 1982a. pigs with diarrhea. Arch Virol 100:139–143.
Intestinal changes in gnotobiotic piglets experimentally Paul, P. S.; Lyoo, Y. S.; Woode, G. N.; Zheng, S.; Greenberg,
inoculated with porcine rotavirus. Nat Inst Anim Health H. B.; Matsui, S.; Schwartz, K. J.; and Hill, H. T. 1988b.
Q (Tokyo) 22:54–60. Isolation of a bovine rotavirus with a “super–short”
Narita, M.; Fukusho, A.; and Shimizu, Y. 1982b. Electron RNA electrophoretic pattern from a calf with diarrhea. J
microscopy of the intestine of gnotobiotic piglets infect- Clin Microbiol 26:2139–2143.
ed with porcine rotavirus. J Comp Pathol 932:589–597. Paul, P. S.; Mengeling, W. L.; Malstrom, C. E.; and Van
Nilsson, O.; Martinsson, K.; and Persson, E. 1984. Epidemi- Deusen, R. A. 1989. Production and characterization of
ology of porcine neonatal steatorrhea in Sweden. I. monoclonal antibodies to porcine immunoglobulin
Prevalence and clinical significance of coccidial and ro- gamma, alpha, and light chains. Am J Vet Res
tavirus infections. Nord Vet Med 36:103–110. 50:471–479.
Nishikawa, K., and Gorziglia, M. 1988. The nucleotide se- Paul, P. S.; Brooks, M. A.; and Lyoo, Y. S. 1990a. Characteri-
quence of the VP3 gene of porcine rotavirus OSU. Nu- zation of serotypic and genetic diversity among porcine
cleic Acids Res 16:11847. rotaviruses. Proc Int Congr Pig Vet Soc 11:276.
Offit, P. A., and Blavat, G. 1986. Identification of the two ro- Paul, P. S.; Johnson, M. E.; and Gorziglia, M. 1990b. Nucle-
tavirus genes determining neutralization specificities. J ic acid probes for the detection and differentiation of ro-
Virol 57:376–378. tavirus serotypes. Proc Int Congr Pig Vet Soc 11:212.
CHAPTER 21 ROTAVIRUS AND REOVIRUS Paul, Stevenson 273

Paul, P. S.; Proescholdt, T.; and Hill, H. 1990c. The preva- ferentiation of two serotypes of porcine rotavirus. Vet
lence of rotavirus groups in pigs with diarrhea. In Proc Microbiol 24:327–339.
Commer Diagn Technol Anim Health Monit. Ames: Nat Rosen, B. I.; Parwani, A. V.; Gorziglia, M.; Larralde, G.; and
Vet Serv Lab and Iowa State Univ Vet Diagn Lab, p. 236. Saif, L. J. 1992. Characterization of full-length and poly-
Paul, P. S.; Gorziglia, M.; and Johnson, M. 1991. Antigenic merase chain reaction–derived partial-length Gottfried
and genetic characterization of ISU-64 strain of porcine and OSU gene 4 probes for serotypic differentiation of
rotavirus. Unpublished. porcine rotaviruses. J Clin Microbiol 30:2644–2652.
Payment, P., and Morin, E. 1990. Minimal infective dose of Rosen, B. I.; Parwani, A. V.; Lopez, S.; Flores, J.; and Saif, L.
the OSU strain of porcine rotavirus. Arch Virol J. 1994. Serotypic differentiation of rotaviruses in field
112:277–282. samples from diarrheic pigs by using nucleic acid probes
Pearson, G. R., and McNulty, M. S. 1977. Pathological specific for porcine VP4 and human and porcine VP7
changes in the small intestine of neonatal pigs infected genes. J Clin Microbiol 32:311–317.
with a pig reovirus–like agent (rotavirus). J Comp Pathol Ruiz, A. M.; Lopez, I. V.; Lopez, S.; Espejo, R. T.; and Arias,
87:363–375. C. F. 1988. Molecular and antigenic characterization of
———. 1979. Ultra structural changes in small intestinal porcine rotavirus YM, a possible new rotavirus serotype.
epithelium of neonatal pigs infected with pig rotavirus. J Virol 62:4331–4336.
Arch Virol 59:127–136. Sabin, A. B. 1959. Reoviruses: A new group of respiratory
Pedley, S.; Bridger, J. C.; Brown, J. F.; and McCrae, M. A. and enteric viruses formerly classified as ECHO type 10
1983. Molecular characterization of rotaviruses with is described. Science 130:1387–1389.
distinct group antigens. J Gen Virol 64:2093–2101. Saif, L. J., and Fernandez, F. M. 1996. Group A rotavirus vet-
Pedley, S.; Bridger, J. C.; Chasey, D.; and McCrae, M. A. erinary vaccines. J Infec Dis 174(Suppl 1):S98–106.
1986. Definition of two new groups of atypical ro- Saif, L. J.; Theil, K. W.; and Bohl, E. H. 1978. Morphogene-
taviruses. J Gen Virol 67:131–137. sis of porcine rotavirus in porcine kidney cell cultures
Pongsuwanna, Y.; Taniguchi, K.; Chiwakul, M.; Urasawa, T.; and intestinal epithelial cells. J Gen Virol 39:205–217.
Wakasugi, F.; Jayavasu, C.; and Urasawa, S. 1996. Sero- Saif, L. J.; Bohl, E. H.; Theil, K. W.; Cross, R. F.; and House,
logical and genomic characterization of porcine ro- J. A. 1980. Rotavirus-like, calicivirus-like, and 23-nm
taviruses in Thailand: Detection of a G10 porcine ro- virus-like particles associated with diarrhea in young
tavirus. J Clin Microbiol 34:1050–1057. pigs. J Clin Microbiol 12:105–111.
Proescholdt, T. 1991. Cultivation of Porcine Group C Ro- Saif, L. J.; Terrett, L. A.; Miller, K. L.; and Cross, R. F. 1988.
tavirus in Porcine Intestinal Cell Line. MS thesis, Iowa Serial propagation of porcine group C rotavirus (pararo-
State Univ, Ames. tavirus) in a continuous cell line and characterization of
Rixon, F. P.; Taylor, P.; and Desselberger, U. 1984. Rotavirus the passaged virus. J Clin Microbiol 26:1277–1282.
RNA segments sized by electron microscopy. J Gen Virol Sanekata, T.; Kishimoto, E.; Sato, K.; Honma, H.; Otsuki, K.;
65:233–239. and Tsubokura, M. 1991. Detection of porcine rotavirus
Roberts, L.; Walker, E. J.; Snodgrass, D. R.; and Angus, K. W. in stools by a latex agglutination test. Vet Microbiol
1980. Diarrhea in unweaned piglets associated with ro- 27:245–251.
tavirus and coccidial infections. Vet Rec 107:156–157. Sattar, S. A.; Raphael, R. A.; Lochnan, H.; and Springthorpe,
Robertson, M. D., and Wilcox, G. E. 1986. Avian reovirus. V. S. 1983. Rotavirus inactivation by chemical disinfec-
Vet Bull 56:155–174. tants and antiseptics used in hospitals. Can J Microbiol
Robl, M. G.; McAdaragh, J. P.; Phillips, C. S.; and Bicknell, 19:1464–1469.
E. J. 1971. Experimental swine pneumonia caused by re- Shaw, D. P.; Morehouse, L. G.; and Solorzano, R. F. 1989a.
ovirus type III. Vet Med 66:903–909. Experimental rotavirus infection in three-week-old pigs.
Rodger, S. M.; Craven, J. A.; and Williams, I. 1975. Demon- Am J Vet Res 50:1961–1965.
stration of reovirus-like particles in intestinal contents ———. 1989b. Rotavirus replication in colostrum-fed and
of piglets with diarrhea. Aust Vet J 51:536. colostrum-deprived pigs. Am J Vet Res 50:1966–1970.
Rolsma, M. D.; Gelberg, H. B.; and Kuhlenshmidt, M. S. Snodgrass, D. R.; Herring, A. J.; Campbell, I.; Inglis, J. M.;
1994. Assay for evaluation of rotavirus-cell interactions: and Hargreaves, F. D. 1984. Comparison of atypical ro-
Identification of an enterocyte ganglioside fraction that taviruses from calves, piglets, lambs, and man. J Gen Vi-
mediates group A porcine rotavirus recognition. J Virol rol 65:909–914.
68:258–268. Steel, R. B., and Torres-Medina, A. 1984. Effects of environ-
Rosen, B. I.; Saif, L. J.; Jackwood, D. J.; and Gorziglia, M. mental and dietary factors on human rotavirus infection
1990a. Serotypic differentiation of group A rotaviruses in gnotobiotic piglets. Infect Immun 43:906–911.
with porcine rotavirus gene 9 probes. J Clin Microbiol Stevenson, G. W. 1990. Pathogenesis of a New Porcine
28:2526–2533. Serotype of Group A Rotavirus in Neonatal Gnotobiotic
Rosen, B. I.; Saif, L. J.; Jackwood, D. J.; and Gorziglia, M. and Weaned Conventional Pigs. Ph.D. diss, Iowa State
1990b. Hybridization probes for the detection and dif- Univ, Ames.
274 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Stevenson, G. W.; Paul, P. S.; and Andrews, J. J. 1990. Patho- Bridger, J. C.; Snodgrass, D. R.; and Herring, J. A. 1977.
genesis of a new serotype of porcine group A rotavirus Serological relationships between rotaviruses from dif-
in the small intestine mucosa of neonatal gnotobiotic ferent species as studied by complement fixation and
and weaned conventional pigs. In Proc Am Assoc Vet neutralization. Arch Virol 53:287–294.
Lab Diagn, p. 12. Torres–Medina, A., and Underdahl, N. R. 1980. Scanning
Suzuki, H.; Chen, G. M.; Hung, T.; Beards, G. M.; Brown, D. electron microscopy of intestine of gnotobiotic piglets
W. G.; and Flewett, T. H. 1987. Effects of two negative infected with porcine rotavirus. Can J Comp Med
staining methods on the Chinese atypical rotavirus. 44:403–411.
Arch Virol 94:305–308. Tsunemitsu, H.; Saif, L. J.; Jiang, B.; Shimizu, M.; Hiro, M.;
Svensmark, B.; Jorsal, S. E.; Nielsen, K.; and Willeberg, P. Yamaguchi, H.; Ishiyama, T.; and Hirai, T. 1991. Isola-
1989a. Epidemiological studies of piglet diarrhoea in in- tion, characterization, and serial propagation of a
tensively managed Danish sow herds. I. Pre-weaning di- bovine group C rotavirus in a monkey kidney cell line
arrhoea. Acta Vet Scand 30;43–53. (MA104). J Clin Microbiol 29:2609–2613.
Svensmark, B.; Nielsen, J.; Dalsgaard, K.; and Willeberg, P. Tsunemitsu, H.; Jiang, B.; and Saif, L. J. 1992a. Detection of
1989b. Epidemiological studies of piglet diarrhoea in in- group C rotavirus antigens and antibodies in animals
tensively managed Danish sow herds. III. Rotavirus in- and humans by enzyme-linked immunosorbent assays. J
fection. Acta Vet Scand 30:63–70. Clin Microbiol 30:2129–2134.
Svensson, L.; Grahnquist, L.; Petterson, C.; Grandien, M.; Tsunemitsu, H.; Jiang, B.; Yamashita, Y.; Oseto, M.; Ushiji-
Stintzing, G.; and Greenberg, H. B. 1988. Detection of ma, H.; and Saif, L. J. 1992b. Evidence of serologic di-
human rotaviruses which do not react with subgroup I versity within group C rotaviruses. J Clin Microbiol
and II specific monoclonal antibodies. J Clin Microbiol 30:3009–3012.
26:1238–1240. Tsunemitsu, H.; Ojeh, C. K.; Jiang, B.; Simkins, R. A.; Weil-
Svensson, L.; Padilla–Noriega, L.; Taniguchi, K.; and Green- nau, P. A.; and Saif, L. J. 1992c. Production and charac-
berg, H. B. 1990. Lack of cosegregation of the subgroup terization of monoclonal antibodies to porcine group C
II antigens on genes 2 and 6 in porcine rotaviruses. J Vi- rotaviruses cross–reactive with group A rotaviruses. Vi-
rol 64:411–413. rol 191:272–281.
Terrett, L. A., and Saif, L. J. 1987. Serial propagation of Tyler, K. L., and Fields, B. N. 1990. Reoviruses. In Virology,
porcine group C rotavirus (pararotavirus) in primary 2d ed. New York: Raven Press, pp. 1307–1328.
porcine kidney cell cultures. J Clin Microbiol Tzipori, S., and Williams, I. H. 1978. Diarrhea in piglets in-
25:1316–1319. oculated with rotavirus. Aust Vet J 54:188–192.
Terrett, L. A.; Saif, L. J.; Theil, K. W.; and Kohler, E. M. 1987. Tzipori, S.; Chandler, D.; Smith, M.; Makin, T.; and Hen-
Physicochemical characterization of porcine pararo- nessy, D. 1980a. Factors contributing to postweaning di-
tavirus and detection of virus and viral antibodies using arrhea in a large intensive piggery. Aust Vet J
cell culture immunofluorescence. J Clin Microbiol 56:274–278.
25:268–272. Tzipori, S.; Chandler, D.; Makin, T.; and Smith, M. 1980b.
Theil, K. W., and Saif, L. J. 1985. In vitro detection of Escherichia coli and rotavirus infections in four-week-old
porcine rotavirus-like virus (group B rotavirus) and its gnotobiotic piglets fed milk of dry food. Aust Vet J
antibody. J Clin Microbiol 21:844–846. 56:279–284.
Theil, K. W.; Bohl, E. H.; and Agnes, A. G. 1977. Cell culture Utrera, V.; Mazzali De Ilja, R.; Gorziglia, M.; and Esparza, J.
propagation of porcine rotavirus (reovirus–like agent). 1984. Epidemiological aspects of porcine rotavirus in-
Am J Vet Res 38:1765–1768. fection in Venezuela. Res Vet Sci 36:310–315.
Theil, K. W.; Bohl, E. H.; Cross, R. F.; Kohler, E. M.; and Van der Heide, L. 1977. Viral arthritis/tenosynovitis: A re-
Agnes, A. G. 1978. Pathogenesis of porcine rotaviral in- view. Avian Pathol 6:271–284.
fection in experimentally inoculated gnotobiotic pigs. Watt, R. G. 1978. Virological study of two commercial pig
Am J Vet Res 39:213–220. herds with respiratory disease. Res Vet Sci 24:147–
Theil, K. W.; McCloskey, C. M.; Saif, L. J.; Redman, D. R.; 153.
Bohl, E. H.; Hancock, D. D.; Kohler, E. M.; and Moor- Westercamp, D. H. 1986. Field trial of porcine rotavirus vac-
head, P. D. 1981. Rapid, simple method of preparing ro- cine to combat postweaning scours in baby pigs. Mod
taviral double-stranded ribonucleic acid for analysis by Vet Pract (Jan):17–18.
polycrylamide gel electrophoresis. J Clin Microbiol Winiarzyk, S., and Paul, P. S. 1997. In preparation.
14:273–280. Woode, G. N. 1986. Porcine rotavirus infection. In Diseases
Theil, K. W.; Saif, L. J.; Moorhead, P. D.; and Whitemoyer, of Swine, 6th ed. Ed. A. D. Leman, B. Straw, R. D. Glock,
R. E. 1985. Porcine rotavirus-like virus (group B ro- W. L. Mengeling, R. H. C. Penny, and E. Scholl. Ames:
tavirus): Characterization and pathogenicity for gnoto- Iowa State Univ Press, pp. 368–382.
biotic pigs. J Clin Microbiol 21:340–345. Woode, G. N., and Bridger, J. C. 1975. Viral enteritis of
Thouless, M. E.; Bryden, A. S.; Flewett, T. H.; Woode, G. N.; calves. Vet Rec 96:85–88.
CHAPTER 21 ROTAVIRUS AND REOVIRUS Paul, Stevenson 275

Woode, G. N., and Crouch, C. F. 1978. Naturally occurring Prostaglandins in the plasma and stool of children with
and experimentally induced rotaviral infections of do- rotavirus gastroenteritis. J Pediatr Gastroenterol Nutr
mestic and laboratory animals. J Am Vet Med Assoc 9:322–327.
173:522–526. Zaberezhny, A. D.; Lyoo, Y. S.; and Paul, P. S. 1994. Preva-
Woode, G. N.; Bridger, J.; Hall, G. A.; Jones, J. M.; and Jack- lence of P types among porcine rotaviruses using subge-
son, G. 1976. The isolation of reovirus-like agents (ro- nomic VP4 gene probes. Vet Microbiol 39:97–110.
taviruses) from acute gastroenteritis of piglets. Med Mi- Zindel, F.; Bommeli, W.; and Kihm, U. 1980. A survey of
crobiol 9:203–209. neutralizing antibodies against some porcine viruses in
Yamashiro, Y.; Shimizu, T.; Oguchi, S.; and Sato, M. 1989. Swiss pig herds. Proc Int Congr Pig Vet Soc 6:65.
Swine Influenza
22 B. C. Easterday and K. Van Reeth

Swine Influenza (SI) is an acute, infectious, respiratory adaptation of the virus to laboratory hosts, antigenic re-
disease of swine caused by type A influenza viruses. The lationships with other influenza viruses, and mainte-
disease is characterized by a sudden onset, coughing, nance of the disease in nature, including his controver-
dyspnea, fever, and prostration, followed by a rapid re- sial lungworm/earthworm hypothesis for the
covery. Lesions generally develop rapidly in the respira- interepizootic survival of the virus. From all accounts,
tory tract and regress quickly, but in a few cases severe vi- the infection and disease in swine have not changed sig-
ral pneumonia may be followed by death. The course, nificantly in the north-central United States since SI was
nature, and severity of the disease are likely to vary with first observed in 1918. However, as will be described lat-
the strain of the virus, the age and immune status of the er, the infection and disease may be caused by type A in-
pig, and the presence of intercurrent infections. fluenza viruses with different antigenic and/or biologic
SI has received considerable attention since the dis- characteristics.
ease was first described in 1918. In the late summer of It is clear, based on clinical observations and serolog-
that year an epizootic disease, having many clinical and ic and virologic surveillance studies, that the infection
pathologic similarities to influenza in humans, appeared and disease are found throughout the United States.
among swine in the north-central United States. The ap- About 25–33% of all pigs 6–7 months old slaughtered in
pearance of the disease in swine coincided with the 1918 the United States have antibody against SIV. The rate is
influenza pandemic that was responsible for the death of higher (about 45%) among older swine (2 or more years)
an estimated 20 million people throughout the world. that are slaughtered (i.e., those animals that have been
The exact date and locality of the initial occurrence of SI used for breeding purposes) (Woods 1975; Pirtle et al.
are unknown, but observers stated that cases were seen 1976; Hinshaw et al. 1978; Chambers et al. 1991). Those
in August 1918 on farms in western Illinois (Shope studies indicated no significant seasonal variation in the
1964). Although the infection and disease may have ex- rate of infection.
isted among swine populations before that time, it is un- Prior to 1975, there were few reports of SI in places
likely that a disease with such distinctive characteristics other than the United States. However, since that time,
would have gone unnoticed and unreported. clinical SI and various type A viruses and their antibod-
According to Dorset et al. (1922), Dr. J. S. Koen, an ies have been observed and reported in most parts of the
inspector in the division of hog cholera control of the world where swine are found. In Europe, SI viruses of the
U.S. Department of Agriculture, Bureau of Animal In- H1N1 subtype were reported occasionally in Czechoslo-
dustry, was the first to recognize the disease as being dif- vakia (Harnach et al. 1950), the United Kingdom (Blake-
ferent from any previously encountered. Koen was im- more and Gledhill 1941), and West Germany (Kaplan
pressed by the coincidental prevalence of epidemic and Payne 1959) between 1940 and 1950. Thereafter, the
human influenza and the similarity of signs and symp- virus/disease was not reported until 1976, when clinical
toms seen in humans and in swine. He was convinced SI reemerged on farms in northern Italy. The viruses re-
that the two diseases were the same and he was the first sponsible for these first outbreaks were closely related to
to apply the name “flu” to this new disease of hogs. His classic H1N1 viruses and probably had been introduced
opinion that the condition represented a new epizootic to Italy with a shipment of pigs from the United States
disease of swine and that swine had been infected from (Nardelli et al. 1978).
humans was shared by veterinarians and farmers in the From 1979, H1N1 influenza virus epizootics occurred
area. in France, Belgium, the Netherlands, Germany, and oth-
Various aspects of the disease, such as signs, lesions, er countries of continental Europe (Vandeputte et al.
and course, were described by Dorset et al. (1922), 1980; Ottis et al. 1981; Soerensen et al. 1981; Gourreau et
McBryde (1927), and McBryde et al. (1928) during the al. 1983; Masurel et al. 1983; Sinnecker et al. 1983; Mar-
decade following the first report. However,it was not un- tinsson et al. 1983). The viruses isolated in Europe after
til 1930 that the SI virus (SIV) was isolated and identified 1979 were related to, but clearly distinguishable from,
by Shope (1931b). For the next 25 years Shope expanded the classic H1N1 viruses. Their hemagglutinins were
his work to include studies of immunity, transmission, more closely related to the avian H1 and they were most

277
278 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

likely transmitted to swine from ducks (Pensaert et al. (Guo et al. 1992). Another 72 H1N1 viruses collected at
1981). These avian viruses generally are more invasive random in southern China from 1976 to 1982 were all
and pathogenic than the SI viruses in the United States, shown to be characteristic of the classic H1N1 viruses. In
resulting in economically devastating outbreaks and Japan, influenza viruses isolated during outbreaks of in-
widespread occurrence. Classic H1N1 viruses have been fluenza-like disease in swine, or recognized indirectly by
reported in only a few European countries and have serologic studies, include swine H1N1, human H1N1,
largely been replaced by the avianlike variants (Abusug- human H3N2 (Goto et al. 1992), and an H1N2 which is a
ra et al. 1987; Barigazzi et al. 1996). reassortant with the H1 from a recent-lineage swine
In Great Britain, however, the situation has not been H1N1 and the N2 from an early H3N2 (Ouchi et al.
the same as on the European continent. Beginning in 1996). Antibodies against H3N2 virus were demonstrat-
1986, classic SIV similar to the North American SIV cir- ed in 10% of 140 swine tested on Andaman and Nicobar
culated at a moderately high prevalence, but it was of mi- Islands (Joshi et al. 1993). Yamaoka et al. (1991) report-
nor clinical importance (Roberts et al. 1987). In 1992, a ed that 19% of 240 swine sera tested had antibody
large increase in outbreaks of respiratory disease was as- against type C influenza virus.
sociated with the isolation of an H1N1 virus antigenical-
ly distinguishable from the previous Great Britain strains ETIOLOGY
(Brown et al. 1993b). That virus represented the first
avianlike H1N1 virus transmitted from pigs in continen- SI is caused by influenza A viruses, in the family
tal Europe, where it had been circulating for more than Orthomyxoviridae. An enormous amount of information
10 years. Currently, classic and avian H1N1 viruses are is available on antigenic, genetic, structural, and biologic
cocirculating in the British swine population, along with characteristics of influenza A viruses (for review, see
H3N2 viruses (Brown et al. 1995). The latter represent Murphy and Webster 1990 and Lamb and Krug 1996). SI
human viruses that emerged from Asia in 1968 and be- viruses are pleomorphic, medium-sized, enveloped viri-
came established in pigs throughout Europe during the ons with glycoproteins, commonly called “spikes,” ex-
1970s (Kundin 1970; Tumova et al. 1976; Ottis et al. tending from their surface. These glycoproteins are the
1982). Evidence of clinical disease in swine caused by major surface antigens and are of two distinct kinds:
H3N2 viruses, however, was not reported until 1984 in hemagglutinin (H) and neuraminidase (N). Hemagglu-
continental Europe (Haesebrouck et al. 1985) and a few tinin is responsible for attachment of viruses to cells and
years later in Great Britain (Wibberley et al. 1988). causes agglutination of erythrocytes. Neuraminidase is
In recent years, H1N2 and H3N2 viruses have been responsible for the enzymatic elution of virus from ery-
associated with severe disease of swine in Canada. Cana- throcytes, and it may play a role in the release of virus
dian investigators have isolated several different influen- from infected cells. Antibodies to the hemagglutinin are
za viruses from pigs with influenza-like disease. These of the most importance for preventing infection with an
viruses have included H1N1 similar to the early (1930) influenza virus containing the same hemagglutinin,
H1N1 and an H1N1 more similar to the more recent whereas antibodies against the neuraminidase restrict
North American H1N1 viruses (Bikour et al. 1995a) and the spread of virus from infected cells. The H and N gly-
H3N2 similar to the early human H3N2 viruses (Bikour coprotein “spikes” are embedded in a lipid envelope that
et al. 1995b). Interestingly, the prevalence of infection surrounds the core of the virus particle. Matrix (M) pro-
with the H3N2 subtype viruses in the United States has tein molecules line the underside of the envelope and
been very low. surround the core, inside which is a helical complex of
Since the late 1970s, there have been several reports molecules consisting of ribonucleic acid (RNA) in associ-
of SI in various places in Asia. Most of those outbreaks ation with viral nucleoprotein (NP) and polymerases (en-
have been due to swine H1N1 or H3N2 viruses. Out- zymes that initiate replication). Influenza viruses are
breaks due to H1N1 viruses similar to the classic H1N1 classified as type A, B, or C, based on the antigenic relat-
were reported in Taiwan in 1975 (Hsu et al. 1976), on a edness of the NP and M proteins. The viral genome con-
large farm in Sarawak after breeding stock was intro- sists of 8 single-stranded RNA segments that encode 10
duced from the United States (Teik and Lan 1985), and in viral proteins.
pigs with influenza-like disease in Thailand (Kupradinun The antigenic characteristics of the two types of sur-
et al. 1991). Swine H1N1 virus was isolated. In China, face glycoproteins serve as the basis for dividing the
there are reports of the isolation of H3N2 viruses from viruses into subtypes. So far, 15 hemagglutinins and 9
pigs in which the H3 is similar to the early human H3 neuraminidases have been identified among all influenza
antigen (Kida et al. 1988), and other H3N2 viruses from A viruses. The current system of nomenclature of in-
pigs are similar to the more recent human H3 viruses. Be- fluenza viruses, introduced in 1980, designates the type,
tween October 1991 and January 1992, 20 H1N1 viruses host, place, strain number (if any), year of isolation, and
isolated from asymptomatic pigs at a Beijing abattoir antigenic subtype (WHO 1980). For example, a SIV iso-
were shown to be of swine, not avian or human, origin lated in Wisconsin in 1984 would be designated
CHAPTER 22 SWINE INFLUENZA Easterday, Van Reeth 279

A/Swine/Wis/1/84(H1N1). The first character, A, iden- incubation ranges from 33˚C to 37˚C. The infected em-
tifies the type and is followed by the host of origin (ex- bryos usually do not die, and the presence of the virus is
cept human), geographic origin, strain number, the year demonstrated after 48–72 hours of incubation by
of isolation, and the antigenic subtype of the H and N in hemagglutination tests on the allantoic and/or amniotic
parentheses. fluids. With the diversity of influenza viruses that have
The three most common subtypes of influenza that been recovered from swine, it is reasonable to expect cul-
circulate among swine worldwide are the “classic” H1N1, tural differences, for example, temperature of incuba-
the avianlike H1N1, and the humanlike H3N2. Immuni- tion for optimal growth, time of incubation, and embryo
ty against an H1N1 influenza virus does not provide pro- death. Although the embryonated chicken egg is the cul-
tection against the viruses of the H3N2 subtype. Anti- ture system most commonly used, various cell culture
genic characterizations of classic H1N1 viruses (Sherrar systems have been used for the growth and assay of SIV.
et al. 1989) and avian H1N1 variants (Scholtissek et al. These include calf kidney cells, fetal pig lung cells, canine
1983) indicate that these viruses have remained remark- kidney cells, pig kidney cells, chicken embryo fibro-
ably conserved since their introduction into swine popu- blasts, human diploid cells, and Chang conjunctival cells
lations. An H1N1 virus was isolated in the United States (Easterday 1975). Other cell culture systems include a pig
with slight antigenic variations in its hemagglutinin oviduct cell line (Bouillant et al. 1975) and a swine testi-
(Olsen et al. 1993), but such events are uncommon. cle cell line (Potgeiter et al. 1977). Fetal pig trachea, lung,
H3N2 viruses are somewhat less stable, and recent iso- and nasal epithelial organ cultures and tracheal organ
lates may show minor antigenic variations when com- cultures from chickens, horses, and ferrets also support
pared to the older prototype viruses (Haesebrouck and the growth of SIV (Nakamura and Easterday 1970;
Pensaert 1988; Kaiser et al. 1991). However, Bikour et Schmidt et al. 1974).
al.(1995a) showed that one of viruses responsible for SI
in Quebec was similar to early human H3N2 viruses. In EPIDEMIOLOGY
contrast to the influenza viruses that circulate in swine,
the human H1N1 and H3N2 viruses continue to have Major considerations in any discussion of the epidemi-
marked antigenic changes. The reason for this is not en- ology of SI include the epizootic characteristics in swine
tirely clear, but it seems likely that there is little immune and interspecies transmission and public health aspects.
pressure on swine viruses because pigs are short-lived
and their viruses are continually transmitted to nonim- Acute Swine Influenza
mune, susceptible pigs. The first appearance of SI in a swine population is com-
Because the viral RNA is segmented, genetic ex- monly associated with the movement of animals, for ex-
change, or reassortment, between different influenza A ample, the introduction of breeding stock, introduction
viruses can (and does) occur during mixed infections. of feeder pigs, or return of show stock to the farm. Many
Genetic reassortment between influenza viruses of hu- outbreaks are clearly related to the movement of animals
man and nonhuman origin is considered a likely mecha- from infected to susceptible herds. It is commonly re-
nism for the origin of new human pandemic strains. In ported that outbreaks are explosive, with all of the pigs
support of this, human and avian virus reassortants in the herd becoming ill at the same time. However, own-
(H1N1) have been isolated from pigs reared and kept un- ers who observe their herds closely are usually aware of
der commercial conditions and from children associated one to a few pigs with signs of the disease 2–5 days be-
with infected pigs (see below). In addition, other reassor- fore the whole herd is involved.
tant viruses have also been isolated from swine. Viruses The primary route of transmission is presumed to be
of the H1N2 subtype were isolated from swine in Japan direct pig-to-pig transmission by the nasopharyngeal
in 1978 (Sugimura et al. 1980), in France in 1987 and route. Nasal secretions are laden with virus during the
1988 (Gourreau et al. 1994), and in Great Britain in 1994 acute febrile stages of infection, providing an abundant
(Brown et al. 1995). H3N1 (Brown et al. 1993b) and source of infectious materials to infect susceptible ani-
H1N7 (Brown et al. 1994) virus subtypes were isolated in mals. Swine are readily infected under experimental pro-
Great Britain in the early 1990s. Except for the H1N2 cedures by instillation of virus suspensions into the nos-
virus in Great Britain, these reassortants did not spread trils or by exposure to small-particle aerosols. Contact
in the swine populations. Antibodies against type B and transmission is easily demonstrated under experimental
type C influenza viurses have been reported in swine in conditions. In densely swine-populated regions, air-
Great Britain (Brown et al. 1995). borne spread may contribute to explosive epidemics over
large geographic areas, particularly in an immunologi-
Laboratory Cultivation cally naive population. This was the case in the late 1970s
SIV grows readily when inoculated into embryonated and early 1980s in Belgium and France (Vandeputte et al.
chicken eggs. It may be inoculated by either the intraal- 1980; Gourreau et al. 1980). In Brittany (France), the first
lantoic or the intraamniotic routes. The temperature of outbreaks occurred in December 1981, and 70% of the
280 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

herds (4 million swine) were infected by September 1982 strains cocirculate continuously at a high frequency and
(Gourreau et al. 1980). have become enzootic in many regions. Serologic exami-
Once the infection has appeared in a swine breeding nation of finishing pigs has shown the prevalence of the
operation or any situation where there is no complete de- H1N1 and H3N2 strains to be 92% and 57%, respective-
population, the possibility of continuous virus circula- ly, in Belgium (Maes et al. 1996), 60% and 30% in the
tion exists (Madec et al. 1985). Under these conditions, Netherlands (Elbers et al. 1990), 73% and 62% in Spain
pigs may become infected at a very young age in the pres- (Yus et al. 1992), and 55% and 51% in Germany
ence of waning maternal immunity. In most instances, (Groschup et al. 1993). In some countries the distribu-
however, influenza viruses disappear from a herd after an tion is not uniform throughout the country, a situation
outbreak. Depending on their prevalence in a particular which is most likely a function of the differences in swine
region, the viruses may be introduced at some later time density and management-related risk factors.
(months or years), causing infections of the seronegative In the enzootic regions, it is unlikely that many farms
breeding and fattening stock. The prevalence and distri- escape repeated exposure to the viruses. Consequently,
bution of influenza viruses and the prevailing subtypes the majority of the litters on a farm are born from im-
are different in different parts of the world, and there mune sows and acquire serum antibodies through
may be regional differences within a country or conti- colostrum. The colostral antibody may provide protec-
nent. tion against disease but may not provide protection
Episodes of acute SI in the north-central United against infection. In herds with continuous virus circula-
States have historically been reported from the late sum- tion, young pigs may become infected in the presence of
mer into the winter, although it is clear that the viruses maternal antibody. A typical infection pattern has been
are circulating throughout the year. These outbreaks of- observed in commercial finishing herds operating on an
ten coincide with the onset of marked fluctuations in all-in/all-out basis. In this situation, pigs usually enter
outdoor temperatures from moderate daytime tempera- such units at 9–12 weeks of age, and they are likely to
tures to below freezing at night and with cold autumn originate from multiple breeding herds. The chance is
rains. As more swine are held in confinement housing high that some of those animals will be infected when
and not subjected to those environmental fluctuations, they arrive in the finishing unit. A serologic study in
the disease may become less seasonal in nature. During commercial fattening herds in Belgium showed that 17 of
one period of 14 months, Hinshaw et al. (1978) conduct- 17 groups became infected with H1N1 virus within the
ed extensive virologic and serologic surveillance on first few weeks after entry and that 9 of 17 groups had
slaughter swine in the north-central and south-central been infected with the H3N2 virus during the same peri-
United States. They recovered 478 influenza viruses from od (Van Reeth and Pensaert 1994a). Some of the groups
approximately 9400 swine tested (about 5% infectivity that were negative for H3N2 became infected later dur-
rate). Antibodies against SIV (H1N1) were found in 21% ing the fattening period. Serologic data from this and
of those swine. Studies in both areas revealed the pres- other studies (Madec et al. 1987; Elbers et al. 1990; Laval
ence of viruses in the pigs throughout the year, but the et al. 1991; Houben et al. 1995) document that intercur-
frequency of virus isolation was much higher in the rent infections with H1N1 and H3N2 influenza viruses,
northern states. The north-central states showed peak the porcine respiratory coronavirus (PRCV), and the
virus activity from October to December, whereas the porcine reproductive and respiratory syndrome virus
southern states did not have a marked peak other than a (PRRSV) are common in intensive production systems in
slight increase in the spring months. These results are in Europe.
agreement with those of Nakamura et al. (1972) and Pir-
tle et al. (1976), who reported a similar prevalence and INTERSPECIES TRANSMISSION
presence throughout the year. A serologic survey (Cham- Influenza A viruses infect many different species in na-
bers et al. 1991) on U.S. pigs in 1988–89 indicated that ture, including humans, lower mammals (including ma-
51% of pigs in the north-central United States had anti- rine mammals), and birds. It is clear that swine are in-
bodies to H1N1 viruses, emphasizing the continued cir- volved in the natural exchange of these viruses (Hinshaw
culation of this virus at high frequency. et al. 1984). Swine H1N1 viruses can be introduced into
Over the years there has been speculation about a car- avian populations and have been responsible for disease
rier state that would provide for the interepizootic sur- in turkeys (Mohan et al. 1981; Hinshaw et al. 1983; An-
vival of SIV. The widespread occurrence of SI in the Unit- dral et al. 1985; Ludwig et al. 1994).
ed States and other parts of the world at all times of the Infection of swine with human H3N2 viruses has
year supports the probability that the virus is constantly been conclusively demonstrated. An H3N2 virus, like the
circulating. There are no clear data to support or reject A/Hong Kong/68 virus, was isolated from pigs in Taiwan
the existence of a long-term true carrier state of influen- soon after it appeared in the human population (Kundin
za viruses in swine. 1970). Subsequently, several of the later variants of the
In Europe, avianlike H1N1 and H3N2 influenza virus human H3N2 viruses were found to have been transmit-
CHAPTER 22 SWINE INFLUENZA Easterday, Van Reeth 281

ted to pigs (Romvary and Tanyi 1975; Tumova et al. vestigation did not reveal a probable animal source of
1976). These viruses continue to be isolated from pigs the virus. Other cases include the recovery of a swine
many years after their disappearance as epidemic strains H1N1 virus from a 32-year-old pregnant woman who
from the human population (Haesebrouck et al. 1985; died of primary viral pneumonia after she attended a
Mancini et al. 1985; Pritchard et al. 1987; Castro et al. livestock show where sick pigs were present (Kaufman et
1988; Bikour et al. 1995b; W. Loeffen, pers. comm.— al. 1988; Wells et al. 1991); the recovery of a swine H1N1
1996) and cause significant disease outside the United virus from a 27-year-old male animal caretaker who died
States. with acute respiratory distress syndrome (Wentworth et
al. 1994); and the recovery of H1N1 viruses from two
Public Health Aspects people in Switzerland and one person in the Nether-
A major milestone in the chronology of zoonotic in- lands, two of whom had been in contact with diseased
fluenza came in January 1976 with the isolation of an in- pigs. The Swiss patients had mild symptoms and the
fluenza virus, A/NJ/8/76(H1N1), closely related to SI Dutch patient had severe pneumonia (de Jong et al.
viruses, from sick U.S. military recruits at Fort Dix, N.J. 1988). Infection and mild disease occurred in two indi-
(Goldfield et al. 1977; Kendal et al. 1977). There were viduals who became infected with a swine H1N1 virus
many cases of acute respiratory disease, and serologic in- while doing experimental infections in pigs (Wentworth
vestigations revealed that several hundred recruits had et al. 1997). In 1993, two H3N2 viruses, probably of pig
been infected. Extensive epidemiologic investigations origin, were isolated from young children in the Nether-
could not identify infected or sick pigs that might have lands (Claas et al. 1994).
been the source of the virus for that outbreak. There was These documented cases of transmission of SIV re-
speculation and concern that the virus would be the new sulting in acute, fatal respiratory disease in humans in
epidemic strain of influenza for humans—perhaps a re- contact with swine constitute a clear signal of the
turn of the virus that was responsible for the 1918 pan- zoonotic potential of influenza viruses that infect swine
demic. Consequently, a national program was initiated and the potential role of pigs in the transmission of new
to vaccinate humans against A/NJ/8/76(H1N1) virus, pandemic strains to humans. Since there are thousands
and a surveillance program was also initiated to investi- of human-swine interactions daily in occupational and
gate pigs and their human contacts. casual situations, the zoonotic potential of SI viruses
Speculation about the zoonotic nature of SIV came to must be recognized and respected.
an end in November 1976 when SIV (H1N1) was isolat-
ed from pigs and their caretaker on a farm in southern CLINICAL SIGNS
Wisconsin (Easterday et al. 1976; Hinshaw et al. 1978).
Pigs had been sick for 2 or 3 days with classic SI clinical Classic SI is a herd disease. The signs of the disease ob-
signs when one of the caretakers also became ill. He had served now are essentially as they were described in the
moderate to severe symptoms of influenza requiring bed 1920s (Dorset et al. 1922; McBryde 1927; Shope 1931a).
rest for 2 days. Nasal swabs from the pigs and throat The onset is sudden, after an incubation period of 1–3
washings from the man were collected on the same day. days. Most of the animals in the herd show signs at the
The virus was recovered from six of eight pigs sampled same time. There is anorexia, inactivity, prostration,
and from three throat washings collected from the young huddling, and piling. One can walk among the inactive
man over a period of approximately 18 hours. There was animals and they will not move, even with vigorous prod-
no evidence that the virus spread from the caretaker to ding. There is also open-mouthed, labored, jerking, and
any of his family or other human contacts. The charac- abdominal breathing, especially when the animals are
terization of the viruses from the man and the pigs indi- forced to move. Furthermore, moving may be accompa-
cated they were identical (Palese and Ritchey 1977; Hin- nied by severe paroxysms of coughing that may sound
shaw et al. 1978). Two weeks later, a similar case like a herd of barking dogs. Fever is usually in the range
occurred in a 14-year-old boy on a farm approximately of 40.5–41.7˚C. One may also observe conjunctivitis,
100 km away. In that case the virus was isolated from one rhinitis, nasal discharge, and sneezing. There is obvious
of five pigs and from one throat washing taken from the loss of weight and weakness related to the anorexia and
boy. Further testing indicated that the virus had spread inactivity. Morbidity is high (near 100%) but mortality is
from that boy to at least one and probably three of his low (usually less than 1%) unless there are intercurrent
close schoolmates. infections and/or the pigs are very young. Generally, re-
Dasco et al. (1980) reported the isolation of an H1N1 covery begins 5–7 days after onset and is as sudden and
virus from a young man who had worked at a large swine remarkable as the onset. Acute outbreaks of clinically
show. Patriarca et al. (1984) recovered an H1N1 virus typical SI, as described here, are generally limited to ful-
similar to contemporary strains isolated from swine in ly susceptible, seronegative pigs.
the United States following a fatal infection in an im- On the European continent, the sudden reintroduc-
munocompromised child. Extensive epidemiologic in- tion of influenza viruses into temporarily negative
282 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

breeding-fattening farms is frequently associated with ratory disease problems, fever, and decreased feed in-
acute disease in fattening swine of 50 kg and more. A take. Nevertheless, financial losses result from high costs
1996 longitudinal study in breeding-fattening herds in for medication, increased mortality, and generally poor
the Netherlands demonstrated that the infection and performance.
characteristic disease predominantly occur around the The hypothesis that influenza viruses may precipitate
age of 18 weeks (W. Loeffen, pers. comm.—1996). Most a more severe disease in combination with other respira-
breeding animals that have acquired active immunity as tory viruses has been supported by experimental dual-in-
a result of previous infections and their maternally im- fection studies (Van Reeth and Pensaert 1994a; Van
mune offspring remain unaffected. Infections with Reeth et al. 1996a; Van Reeth et al. 1996b). In these stud-
H1N1 and H3N2 subtype viruses are clinically alike, and ies, fever, respiratory disease, and growth retardation
both viruses have been correlated with acute respiratory were significantly more severe and lasting in dual infec-
episodes in Belgium, France (Laval et al. 1991; Madec et tions than in single-virus-infected pigs. Nevertheless,
al. 1987), Spain (Castro et al. 1988), Italy (Barigazzi et al. several unknown factors seem to be critical to the devel-
1996), and the Netherlands (W. Loeffen, pers. comm.— opment of clinical effects following dual virus infections.
1996). To illustrate, the effect of experimental PRRSV-SIV in-
Typical outbreaks of SI can be reproduced experi- fections is not reproducible to the same level in different
mentally only under very specific conditions. Inocula- experimental groups. Furthermore, other researchers
tion of fattening swine (±100 kg) with high virus doses did not observe an enhancement of clinical signs follow-
(107–107.5 egg infective doses 50 [EID50] per pig) of either ing simultaneous inoculation of pigs with PRCV and ei-
of the two influenza viruses H1N1 or H3N2 directly into ther H1N1 or H3N2 viruses (Lanza et al. 1992).
the trachea results in high fever, complete anorexia, and A growing body of evidence suggests that the “avian
dyspnea within 24 hours after inoculation (Maes et al. variant,” or avianlike, H1N1 strains circulating in Europe
1984). Mean growth arrest periods of 5–8 days and have acquired a more pathogenic character than the
mean weight losses of 5–6 kg have been recorded. Nev- “classic” H1N1 strains that circulate in the United States
ertheless, recovery was very rapid, with clinical signs as well as in other countries. Observations and reports
lasting only 2 days. Conversely, inoculation of the same suggest that SI now occurs more frequently and has a
amount of virus by the less invasive oronasal route leads greater economic significance in Europe than in the Unit-
to mild clinical signs or a subclinical infection. So far, ed States.
there are no indications of differences in the site of repli- Producers and veterinarians occasionally report re-
cation in the lungs with different influenza virus strains. productive, farrowing, and neonatal problems such as
In addition to the clinically apparent disease, subclin- abortions, stillbirths, infertility, and small weak litters
ical infections frequently occur, as indicated by the high associated with outbreaks of SI. There are insufficient
seroprevalence of both subtypes in finishing pigs in the data to conclude that SI viruses have a specific and sig-
absence of significant respiratory disease during the fat- nificant adverse association with reproductive, farrow-
tening period. Multiple factors, including immune sta- ing, and perinatal problems.
tus, age, infection pressure, intercurrent infections, cli-
matic conditions, and housing, may determine the PATHOGENESIS
clinical outcome of an infection with influenza virus. Al-
though infection occurs throughout the year, the clinical Infection with influenza virus is generally limited to the
disease is observed mainly in the colder seasons. respiratory tract, and viremia has only rarely been de-
Intercurrent infections are among the most impor- tected. Brown et al. (1993a) reported virus isolation from
tant complicating factors of an influenza virus infection. the serum of experimentally infected pigs 1 and 3 days
It has been known for years that secondary infections by postinoculation. However, the virus could be isolated
respiratory bacteria such as Actinobacillus pleuropneumo- during only one day, and virus titers in the serum sam-
niae, Pasteurella multocida, Haemophilus parasuis, and ples were low. Attempts to demonstrate virus replication
Streptococcus suis type 2 complicate the severity and in extrarespiratory sites have been largely unsuccessful.
course of an infection with influenza viruses. More re- Virus replication has been demonstrated in nasal mu-
cent observations under natural conditions suggest that cosa, tonsils, trachea, tracheobronchial lymph nodes,
respiratory viruses, as well, act as complicating factors. It and lungs (Lanza et al. 1992; Nayak et al. 1965). The
has been shown that the prevalence of dual or triple in- lungs seem to be the major target organ.
fections with influenza virus(es) and either PRCV or Following intratracheal inoculation of pigs, virus
PRRSV is extremely high in intensive fattening units in titers in the lungs may be greater than 108 EID50 /gram of
Europe (Madec et al. 1987; Van Reeth and Pensaert tissue (Haesebrouck and Pensaert 1986). The amount of
1994a; Houben et al. 1995). The associated clinical pic- virus that reaches the deeper airways and the resulting
ture is less characteristic than that of acute SI outbreaks, production of infectious virus in the lungs seem to de-
and usually only 20–50% of pigs in the herd show respi- termine the severity of illness. Experimental inoculation
CHAPTER 22 SWINE INFLUENZA Easterday, Van Reeth 283

of high virus quantities (107–107.5 EID50) via the nasal masked by intercurrent, especially bacterial, infections.
route in fattening swine (±100 kg) resulted in subclinical Morin et al. (1990) described a severe proliferative
infections, whereas intratracheal inoculation at the same and necrotizing pneumonia (PNP) in pigs as a newly rec-
virus dose produced typical clinical signs within 24 ognized disease in the province of Quebec, Canada. They
hours (Maes et al. 1984). Immunofluorescence studies of reported confluent consolidation of the cranial and mid-
lung tissue demonstrate the rapidity of virus replication dle lobes and the lower half of the caudal lobes. The
and the highly specific tropism for bronchiolar epitheli- lobes were not collapsed; they were red or gray, “moist
um (Nayak et al. 1965; Haesebrouck and Pensaert 1986; and meaty,” and interlobular edema was common. Type
Brown et al. 1993a). So far, there are no indications of A influenza virus was isolated from the lungs of affected
differences in the site of replication in the lungs with dif- animals. Subsequently, Girard et al. (1992) reproduced
ferent influenza virus strains. the disease with the virus isolated by Morin et al. (1990).
In the studies by Nayak et al. (1965), bronchial ep- Dea et al. (1992) reported similar lesions, especially the
ithelial cells stained positive within 2 hours postinfec- consolidated lungs. They concluded that the virus was a
tion. By 16 hours, there were large fluorescent areas of swine H1N1 distantly related to the contemporary H1N1
bronchial epithelium. Staining was intense through 72 viruses circulating in North America. Rekik et al. (1994)
hours postinfection and then diminished. Antigen was concurred that a new antigenic variant of H1N1 swine
also detected in the alveolar septa within 4 hours after in- influenza virus was associated with the PNP. However,
fection, and at 24 hours there were numerous fluorescent Bikour et al. (1994a) described a virus isolated from cas-
cells in the alveoli and alveolar ducts. Fluorescent stain- es of PNP in Quebec as a “new strain isolated for the first
ing in both bronchioli and alveoli disappeared by day 9. time in the porcine population of Canada [that] is relat-
Immunofluorescence of bronchi may involve nearly ed to A/Sw/Hong Kong/76 H3N2 swine influenza
100% of the epithelial cells (Haesebrouck and Pensaert virus.” Thus, it is not clear what factor(s) are responsible
1986). Typically, bronchi and bronchioli contain exudate for this unique pathological lesion. Winkler and Cheville
with degenerated and detached fluorescent mucosal cells (1986) reported that following inoculation of virus di-
and neutrophils. rectly into the trachea of anesthetized pigs, they ob-
Little information is available about influenza virus served dark areas of consolidation over all lung lobes.
pathogenesis at the cellular level. Recent studies in These areas were small and covered a small portion of
colostrum-deprived pigs suggest that bronchoalveolar the lung 24 hours after infection, but by 48–96 hours,
production of the cytokines tumor necrosis factor and confluent areas of consolidation completely covered the
interleukin-1 contributes to the typical constitutional ef- lung lobes, and the lung tissue was consistently firm.
fects and lung inflammatory changes following virus in- Differences in lesion severity have been observed
fection (Van Reeth and Pensaert 1997). In most experi- with the different influenza virus variants circulating in
mental studies, virus clearance was extremely rapid. SIV Great Britain. Classical H1N1 and H3N2 strains circulat-
could not be isolated from lungs or other respiratory ing since 1986 produced minimal gross lesions and a
tract tissues on or after day 7 (Brown et al. 1993a; Van very mild interstitial pneumonitis, both in natural cases
Reeth and Pensaert 1994b). Using highly sensitive ELISA and experimentally (Done et al. 1994). The more recent
techniques, influenza-specific antibodies could be de- H1N1 variant, on the other hand, produced marked
tected in serum on day 3 and nasal swabs on day 4 (Lee gross lesions and more severe histopathological changes
et al. 1995). involving severe necrosis of bronchial epithelium, alveo-
lar exudation, and neutrophil infiltration (Brown et al.
LESIONS 1993a). These findings have been confirmed experimen-
tally. The pathological data provide further evidence of
Gross Lesions an increased pathogenicity of the avian H1N1 strains in
The gross lesions found in uncomplicated SI are mainly Europe compared to the classic H1N1 strains circulating
those of a viral pneumonia. The changes are most often in the United States.
limited to the apical and cardiac lobes of the lungs, al-
though in severe cases more than half of the lung may be Microscopic Lesions
affected. Generally, there is a sharp line of demarcation As with the gross lesions, the microscopic lesions in un-
between the affected and normal lung tissue. The in- complicated cases of SI are consistent with viral pneu-
volved areas will be purple and firm. Some interlobular monia. Bachmann (1989) provided a concise summary
edema may be evident. The airways may be filled with of the microscopic lesions of SI: “Histologically, a wide-
blood-tinged, fibrinous exudate, and the associated spread degeneration and necrosis of the epithelium in
bronchial and mediastinal lymph nodes are usually en- bronchi and bronchioli can be observed. The lumen of
larged. In severe cases there may be fibrinous pleuritis bronchi, bronchioli, and alveoli are filled with exudate
(Shope 1931a; Nayak et al. 1965; Nakamura 1967). The containing desquamated cells and neutrophils, later
lesions in naturally occurring SI are often complicated or mostly monocytes. Furthermore, variable hyperemia
284 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

with dilatation of the capillaries and infiltration of the bated at 35˚C. Since SIV usually does not kill the chick
alveolar septae with lymphocytes, histiocytes, and plas- embryo, allantoic fluid is collected after 72 hours incuba-
ma cells occurs. Widespread alveolar atelectasis, intersti- tion and tested for its ability to agglutinate chicken red
tial pneumonia, and emphysema accompany these le- blood cells, which is presumptive evidence for the pres-
sions. There is also peribronchial and perivascular ence of an influenza virus. The hemagglutinin subtype is
cellular infiltration (Shope 1931a; Urman et al. 1958; determined by the hemagglutination-inhibition (HI)
Witte et al. 1981).” The PNP histopathologic lesions are test, and the neuraminidase subtype is determined with
described extensively by Morin et al. (1990), Girard et al. the neuraminidase-inhibition (NI) test (Centers for Dis-
(1992), and Dea et al. (1992). Lesions described include ease Control 1982). Cell culture systems are more com-
exudative lesions characterized by alveoli filled with pro- monly used for the isolation of influenza viruses from
tein-rich edema and large macrophages; necrotizing human specimens.
bronchiolitis affecting mainly terminal bronchioles; co- Serologic diagnosis of SI requires the use of paired
agulates of necrotic cells in alveolar ducts and alveoli; serum samples, one obtained during the acute phase of
and proliferative lesions marked by proliferation of type the disease and the second 3–4 weeks later, to demon-
II pneumocytes responsible for alveolar epithelialization strate an increase in the amount of antibody when tested
and hyaline membrane formation in the lumen of the against appropriate antigens. The most common test for
terminal alveolar ducts. antibody is the HI test. The diagnostician must be aware
of the possibility of the presence of nonspecific in-
DIAGNOSIS hibitors and nonspecific agglutinins found in swine
serum that may interfere with the test.
Virus isolation and/or the detection of specific antibody Other methods for detecting virus, viral antigen, or
are necessary for a specific diagnosis of SI. SI may be sus- specific antibody are direct immunofluorescence tech-
pected when there is an outbreak of acute respiratory niques applied to lung tissue (Barigazzi et al. 1993), indi-
disease involving most or all of the pigs in a herd, espe- rect immunofluorescence applied to nasal epithelial cells
cially in the colder seasons of fall and early winter. SI (Onno et al. 1990), immunofluorescence microscopy ap-
must be differentiated from a variety of respiratory dis- plied to bronchioalveolar lavage contents (Runge et al.
eases of swine. 1996), immunohistochemical detection in fixed tissue
The best sample for virus isolation from a live animal (Haines et al. 1993), enzyme-linked immunosorbent as-
is nasal mucus obtained by swabbing the nasal passages, say (ELISA) (Lee et al. 1993a, b, 1995), polymerase chain
or in the case of very small pigs where it is difficult to reaction (PCR) (Schorr et al. 1994), rapid cell culture as-
swab the nasal passages, pharyngeal mucus may be ob- say using immunoperoxidase staining for typing and
tained by swabbing. The virus is more likely to be found subtyping (Ziegler et al. 1995), and an enzyme im-
in nasal and pharyngeal secretions during the febrile pe- munoassay membrane test (Directigen FLU-A) which
riod than after the fever has subsided. The swab is sus- rapidly detects influenza A antigen in clinical specimens
pended in a suitable transport medium (such as glycerol (Ryan-Poirier et al. 1992). Because of the large number of
saline). It is very important to keep the sample moist and reagents necessary for the specific identification of in-
cold (refrigerator temperature, 4˚C) during transport to fluenza viruses, the definitive determination of the anti-
the laboratory. If the samples for virus isolation can be genic character of the virus(es) involved is generally
tested within 48 hours after collection, they should be done by a designated reference center.
kept at 4˚C. If the samples must be held longer, storage Positive diagnosis of the disease by serologic and vi-
at −70˚C is recommended. Storage in sealed containers rologic means in suckling or weanling pigs from dams
in dry ice or liquid nitrogen is appropriate, but the virus- with antibody to the virus may be complicated. Maternal
es are not stable at −20˚C. Filtration of the samples antibody persists for 2–4 months, depending on the ini-
should be avoided to conserve small amounts of virus tial level. It has been shown that weanling pigs with ma-
that might be in the transport medium. Adventitious ternal antibody may be infected and may shed virus. The
bacterial and fungal agents may be controlled with the rate of virus recovery and severity of signs of disease are
addition of appropriate antimicrobial agents to the inversely related to level of maternal antibody (Easter-
transport medium. The virus may also be isolated from day 1971, 1972; Renshaw 1975). Depending on the level
lung tissue of pigs that die or are killed during the acute of antibody, it may be very difficult to isolate virus. In ad-
stage of the disease. The lung tissue should be held un- dition, the level of antibody in the convalescent-phase
der the same conditions as swab material until ready for serum will be lower than in the acute-phase serum be-
culturing. At that time the tissue is ground (minced) and cause of the inhibition of active antibody production by
suspended in saline. maternal antibody (Mensik 1960, 1963, 1966; Easterday
Chicken eggs, embryonated for 10 days, remain a re- 1971; Renshaw 1975). After maternal antibody is deplet-
liable and economical culture medium for isolating in- ed, pigs may be infected again, shed virus, have signs of
fluenza viruses. The inoculated eggs are generally incu- disease, and have a typical primary antibody response.
CHAPTER 22 SWINE INFLUENZA Easterday, Van Reeth 285

TREATMENT 1994). It is an oil-in-water vaccine containing an unspec-


ified H1N1 virus that was developed for the protection of
There is no specific therapy for SI. Careful nursing is im- weaned pigs and breeding animals against SI. The au-
portant, with the provision of comfortable, draft-free thors conclude that it “is a safe and efficacious vaccine for
shelter and clean, dry, dust-free bedding. To avoid addi- the prevention of clinical disease caused by SIV.” Bikour
tional stress, pigs should not be moved or transported et al. (1994b) have shown that a combination of Quil A
during the acute stages of the disease. Fresh, clean water and Alhydrogel is a valuable immunopotentiating adju-
should be accessible at all times, because most animals vant for immunizing swine against influenza virus.
will be febrile. There is a marked loss of appetite during In Europe, vaccination against SIV is a common prac-
the course of the disease, but it returns quickly with clin- tice. Inactivated whole-virus vaccines and split-product
ical improvement. Expectorants are commonly used as a vaccines prepared from detergent-treated virus are com-
herd treatment and are administered in the drinking wa- mercially available. These vaccines contain H1N1 and
ter. Antibiotics and other antimicrobial treatments have H3N2 influenza virus strains of human origin. The
been used on a herd basis to control concurrent or sec- H1N1 component, the prototype A/New Jersey/1/76
ondary bacterial infections. Individual animals may re- strain, confers a high degree of protection against the
quire additional supportive and antibacterial treatment “avian” H1N1 strains currently circulating in the Euro-
in severe cases. pean swine populations. As for the H3N2 component,
Amantadine (1-adamantanamine) has been shown to there should be careful consideration of the strain to be
be effective in reducing the febrile response and shed- included in vaccine to provide the best protection, be-
ding of virus in experimentally infected pigs (McGregor cause several H3N2 strains circulate in the swine popula-
and Easterday 1979). However, there are no antiviral tions (Haesebrouck et al. 1985).
treatments licensed for use against influenza in swine. The SIV vaccines are administered intramuscularly,
and protection is based on the presence of hemaggluti-
PREVENTION AND CONTROL nation-inhibiting antibodies in serum. Two injections of
vaccine 3 weeks apart are preferred. Under experimental
Standard biosecurity measures to prevent susceptible conditions, one vaccination of fattening swine with
animals from contacting infected animals are appropri- monovalent vaccine based on the A/New Jersey/1/76
ate to prevent the introduction of SI into a herd. Because (H1N1) strain or the A/Port Chalmers/1/73(H3N2)
of the known occurrences of interspecies transfer of in- strain resulted in clinical protection and reduced virus
fluenza viruses, biosecurity measures should include replication in the lungs at challenge (Vandeputte et al.
measures to prevent contact with other species, especial- 1980; Haesebrouck and Pensaert 1986). Two doses of the
ly avian species. Also important, humans suspected of H1N1 vaccine a month apart induced complete protec-
having influenza virus infection should be excluded from tion against infection (Haesebrouck and Pensaert 1986).
contact with swine. Animals recovered from SI develop There are contradictory reports about the economic
hemagglutination-inhibiting, virus-neutralizing, com- benefit of SI vaccination in the field. In commercial fat-
plement-fixing, and neuraminidase-inhibiting antibody. tening herds, SIV infections occur very soon after entry
The degree and duration of resistance to subsequent in- of feeder pigs, so that immunity induced by vaccination
fection in swine recovered from influenza have not been with inactivated vaccines comes too late. On closed
precisely determined. Substantial levels of antibody may breeding-fattening farms in Belgium, vaccination of
be found for at least 6 months after infection. As with feeder pigs is considered cost-efficient, particularly dur-
many other infections, the relationship of the amount of ing winter when infections are more harmful. Care
antibody in the serum or respiratory secretions to the de- should be taken not to vaccinate before the age of 10
gree of resistance has not been defined, and there is con- weeks to avoid interference by maternal immunity. One
siderable variation in the antibody response of individ- vaccine for intradermal vaccination (by PIGJET) of feed-
ual pigs following exposure. er pigs has recently been licensed in Europe. Though vac-
Inactivated-virus vaccines have been developed cination by this method is highly laborsaving, the effica-
and/or tested by several investigators (Woods and cy in the field remains to be proven. In view of the
Mansfield 1976, 1978, 1980; Pirtle 1977). There is con- extensive circulation of H1N1 virus in pigs and the anti-
siderable variation in antibody response and protection genic conservation of these viruses, vaccination should
from infection following vaccination. In a study using a be a valuable component of SI control programs.
temperature-sensitive strain of H1N1, it was shown that
nonvaccinated, experimentally infected pigs required 2 REFERENCES
weeks longer to reach market weight than their vaccinat-
ed exposed cohorts (Easterday et al. 1977). There is one Abusugra, I. A.; Linne, T.; Klingeborn, B. 1987. The prove-
licensed vaccine, MaxiVac®-FLU, available in the United nance of the Swedish swine influenza H1N1 virus of
States for the prevention of SI (Brown and McMillen 1983. J Vet Med B 34:566–572.
286 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Andral, B.; Toquin, D.; Madec, F.; Aymard, M.; Gourreau, J. and Manvell, R. J. 1994. Isolation of an influenza A virus
M.; Kaiser, C.; Fontaine, M.; and Metz, M. H. 1985. Dis- of unusual subtype (H1N7) from pigs in England, and
ease in turkeys associated with H1N1 influenza virus fol- the subsequent experimental transmission from pig to
lowing an outbreak of the disease in pigs. Vet Rec pig. Vet Microbiol 39:125–134.
116:617–618. Brown, I. H.; Harris, P. A.; and Alexander, D. J. 1995. Sero-
Bachmann, P. A. 1989. Swine influenza virus. In Virus Infec- logical studies of influenza viruses in pigs in Great Brit-
tions of Porcines. Ed. M. B. Pensaert. Amsterdam: Else- ian, 1991–2. Epidemiol Infect 114:511–520.
vier Science, pp. 193–207. Castro, J. M.; del Pozo, M.; and Simarro, I. 1988. Identifica-
Barigazzi, G.; Bonardi, S.; Fibu, E.; Candotti, P.; Ferrari, B.; tion of H3N2 influenza virus isolated from pigs with res-
and Bedogni, I. 1993. Use of the direct immunofluores- piratory problems in Spain. Vet Rec 122:418–419.
cence technique to diagnose swine influenza: A compar- Centers for Disease Control. 1982. Concepts and procedures
ison with virus isolation. In Atti del XIX Meet Annu Soc for laboratory-based influenza surveillance. Washing-
Ital Patol e Allev d Suini, 1992, pp. 309–318. ton, D.C.: Centers for Disease Control, U.S. Dept Health
Barigazzi, G.; Foni, E.; Candotti, P.; and Cataldi, M. 1996. and Human Serv.
Antigenic analysis of swine influenza viruses isolated in Chambers, T. M.; Hinshaw, V. S.; Kawaoka, Y.; Easterday, B.
Italy from 1976 to 1995. Proc Int Congr Pig Vet Soc C.; and Webster, R. G. 1991. Influenza viral infection of
14:100. swine in the United States, 1988–89. Arch Virol
Bikour, M. H.; Cornaglia, E.; Weber, J. M.; and Elazhary, Y. 116(1–4):261–265.
1994a. Antigenic characterization of an H3N2 swine in- Claas, E. C. J.; Kawaoka, Y.; de Jong, J. C.; Masurel, N.; and
fluenza virus isolated from pigs with proliferative and Webster, R. G. 1994. Infection of children with avian-
necrotizing pneumonia in Quebec. Can J Vet Res human reassortant influenza virus from pigs in Europe.
58(4):287–290. Virology 204:453–457.
Bikour, M. H.; Cornaglia, E.; and Elazhary, Y. 1994b. Com- Dasco, C. C.; Couch, R. B.; and Quarles, J. M. 1980. Sporadic
parative study of the immunostimulatory properties of occurrence of swine influenza in man. Abstr Annu Meet
different adjuvants administered with an inactivated in- Am Soc Microbiol 80:288.
fluenza virus vaccine and evaluation of passive immuni- Dea, S.; Bilodeau, R.; Sauvageau, R.; Montpetit, C.; and
ty in pigs. Imm and Inf Dis 4(3):166–172. Martineau, G. P. 1992. Antigenic variant of swine in-
Bikour, M. H.; Frost, E. H.; Deslandes, S.; Talbot, B.; and fluenza virus causing proliferative and necrotizing pneu-
Elazhary, Y. 1995a. Persistence of a 1930 swine influen- monia in pigs. J Vet Diag Invest 4(4):380–392.
za A (H1N1) virus in Quebec. J Gen Virol de Jong, J. C.; Paccaud, M. F.; de Ronde-Verloop, F. M.; Huf-
76(10):2539–2547. fels, N. H.; Verwei, C.; Weijers, T. F.; Bangma, P. J.; van
Bikour, M. H.; Frost, E. H.; Deslandes, S.; Talbot, B.; Weber, Kregten, E.; Kerckhaert, J. A.; and Wicki, F. 1988. Isola-
J. M.; and Elazhary, Y. 1995b. Recent H3N2 swine in- tion of swine-like influenza A (H1N1) viruses from man
fluenza virus with haemagglutinin and nucleoprotein in Switzerland and the Netherlands. Ann Inst Past
genes similar to 1975 strains. J Gen Virol 76(3):697–703. 139(4):429–437.
Blakemore, F., and Gledhill, A. W. 1941. Some observations Done, S. H.; Spencer, Y. I.; Brown, I. H.; Higgins, R.; and
on an outbreak of swine influenza in England. Vet Rec Hannam, D. A. 1994. Natural swine influenza virus
53:227–230. (A/swine/Eng/195852/92) infections in pigs in the UK:
Bouillant, A. M.; Dulac, G. C.; Willis, N.; Girard, A.; Grieg, Morphology, immunocytochemistry and aging of le-
A. S.; and Boulanger, P. 1975. Viral susceptibility of a sions. Proc Int Congr Pig Vet Soc 13:103.
cell line derived from the pig oviduct. Can J Comp Med Dorset, M.; McBryde, C. N.; and Niles, W. B. 1922. Remarks
39:450–456. on “hog flu.” J Am Vet Med Assoc 62:162–171.
Brown, G. B., and McMillen, J. K. 1994. MaxiVacTM-FLU: Easterday, B. C. 1971. Influenza virus infection of the suck-
Evaluation of the safety and efficacy of a swine influen- ling pig. Acta Vet (Suppl)2:33–42.
za vaccine. In Proc Am Assoc Swine Pract, 25th Annu ———. 1972. Immunologic considerations in swine in-
Meet, pp. 37–39. fluenza. J Am Vet Med Assoc 160:645–648.
Brown, I. H.; Done, S. H.; Spencer, Y. I.; Cooley, W. A.; Har- ———. 1975. Swine influenza. In Diseases of Swine, 4th ed.
ris, P. A.; and Alexander, D. J. 1993a. Pathogenicity of a Ed. H. W. Dunne and A. D. Leman. Ames: Iowa State
swine influenza H1N1 virus antigenically distinguish- Univ Press, pp. 141–167.
able from classical and European strains. Vet Rec Easterday, B. C.; Pawlisch, R.; Gunderson, J. A.; and Craven,
132:598–602. R. 1976. Unpublished data.
Brown, I. H.; Manvell, R. J.; Alexander, D. J.; Chakraverty, P.; Easterday, B. C.; Murphy, B. R.; and McGregor, S. 1977. In-
Hinshaw, V. S.; and Webster, R. G. 1993b. Swine in- fection and vaccination of pigs with influenza A/New
fluenza outbreaks in England due to a new H1N1 virus. Jersey/8/76 (Hsw1N1) virus. J Infect Dis 136(Sup-
Vet Rec 132:461–462. pl):699–702.
Brown, I. H.; Alexander, D. J.; Chakraverty, P.; Harris, P. A.; Elbers, A. R. W.; Tielen, M. J. M.; Cromwijk, W. A. J.; and
CHAPTER 22 SWINE INFLUENZA Easterday, Van Reeth 287

Hunneman, W. A. 1990. Sero–epidemiological screen- Vet Res 57(1):33–36.


ing of pig sera collected at the slaughterhouse to detect Harnach, R.; Hubik, R.; and Chvatal, O. 1950. Isolation of
herds infected with Aujeszky’s disease virus, porcine in- the virus of swine influenza in Czechoslovakia. Cas
fluenza virus and Actinobacillus (Haemophilus) pleuro- Ceskoslov Vet 5:289
pneumoniae in the framework of an integrated quality Hinshaw, V. S.; Bean, W. J., Jr.; Webster, R. G.; and Easter-
control (IQC) system. Vet Q 12:221–230. day, B. C. 1978. The prevalence of influenza viruses in
Girard, C.; Morin, M.; and Elazhary, Y. 1992. Experimental- swine and the antigenic and genetic relatedness of in-
ly induced porcine proliferative and necrotising pneu- fluenza viruses from man and swine. Virology 84:51–62.
monia with an influenza A virus. Vet Rec Hinshaw, V. S.; Webster, R. G.; Bean, W. J.; Downie, J.; and
130(10):206–207. Senne, D. A. 1983. Swine influenza–like viruses in
Goldfield, M.; Noble, G.; and Dowdle, W. R. 1977. Identifi- turkeys: Potential source of virus in humans? Science
cation and preliminary antigenic analysis of swine in- 220:206–208.
fluenza–like viruses isolated during an influenza out- Hinshaw, V. S.; Alexander, D. J.; Aymard, M.; Bachmann, P.
break at Fort Dix, New Jersey. J Infect Dis A.; Easterday, B. C.; Hannoun, C.; Kida, H.; Lipkind, M.;
136(Suppl):381–385. Mackenzie, J. S.; Nerome, K.; Schild, G. C.; Scholtissek,
Goto, H.; Takai, M.; Iguchi, H.; Benkele, W.; Ohta, C.; Ya- C.; Senne, D. A.; Shortridge, K. F.; Skehel, J. J.; and Web-
mamoto, Y.; Kida, H.; Noro, S.; and Sakurada, N. 1992. ster, R. G. 1984. Antigenic comparisons of swine-in-
Antibody responses of swine to type A influenza viruses fluenzalike H1N1 isolates from pigs, birds and humans:
in the most recent several years. J Vet Med Sci An international collaborative study. Bull WHO
54(2):235–241. 62:871–878.
Gourreau, J. M.; Kaiser, C.; Hannoun, C., Vaissaire, J.; and Houben, S.; Van Reeth, K.; and Pensaert, M. B. 1995. Pat-
Gayot, G. 1980. Premier isolement en France du virus de tern of infection with the porcine reproductive and res-
l’influenza du porc (Hsw1N1) dans un environnement piratory syndrome virus on swine farms in Belgium. J
pathologique plurimicrobien. Bull Acad Vet 53:181–188. Vet Med B 42:209–215.
Gourreau, J. M.; Kaiser, C.; Fontaine, M.; Madec, F.; Ay- Hsu, F. S.; Joseph, R. L.; Chang, C. R; Chen, W. F.; and Chou,
mard, M.; Vigouroux, A.; Salingardes, F.; and Labie, J. N. Y. 1976. An epizootic of swine in Taiwan. Proc Int
1983. Evolution de la grippe porcine en France. In La Congr Pig Vet Soc 4:16.
grippe, grippe humaine, grippes animals, les vaccins. Joshi, M. V.; Kadam, S. S.; and Rao, B. L. 1993. A note on
Collection Fondation Marcel Merieux 1983, pp. 65–67. prevalence of antibodies to influenza viruses in pigs
Gourreau, J. M.; Kaiser, C.; Valette, M.; Douglas, A. R.; La- from Andaman and Nicobar Islands. J Bombay Vet Coll
bie, J.; and Aymard, M. 1994. Isolation of two H1N2 in- 4(1–2):49–50.
fluenza viruses from swine in France. Arch Virol Kaiser, C.; Valette, M.; Million–Jolly, J.; et al. 1991. Mise en
135:365–382. evidence de variations des antigenes de surface du virus
Groschup, M. H.; Brun, A.; and Haas, B. 1993. Serological grippal H3N2 chez le parc en France a l’aide d’anticorps
studies on the potential synergism of porcine reproduc- monoclonux. Epidemiol et Santé Anim 19:75–84.
tive and respiratory syndrome virus and influenza–, Kaplan, M. M., and Payne, A. M. M. 1959. Serological sur-
corona– and paramyxoviruses in the induction of respi- vey in animals for type A influenza in relation to the
ratory symptoms in swine. J Vet Med B 40:681–689. 1957 pandemic. Bull WHO 20:465–488.
Guo, Yuanji; Webster, R. G.; Zuge, Yahai; Wang, Min; Kaufman, J.; Hassan, M.; Rytel, M.; Chayer, R.; Volkert, P.;
Zhang, Shalin; Zheng, Hongyong; and Liu, Lingyan. McKinney, P.; Michaels, J.; Farmer, C.; Sedmak, G.; Hin-
1992. Virus Info Exch Newsl for SE Asia and W Pacific shaw, V. S.; Berg, J.; Hopfensperger, D.; Procter, M.;
9(2):62. Davis, J. P.; and Smith, T. F. 1988. Human infection with
Haesebrouck, F., and Pensaert, M. 1986. Effect of intratra- swine influenza virus—Wisconsin. JAMA 260(21):3116.
cheal challenge of fattening pigs previously immunized Kendal, A. P.; Goldfield, M.; Noble, G. R.; and Dowdle, W.
with an inactivated influenza H1N1 vaccine. Vet Micro- R. 1977. Identification and preliminary antigenic analy-
biol 11:239–249. sis of swine influenzalike viruses isolated during an in-
———. 1988. Influenza in swine in Belgium (1969–1986): fluenza outbreak at Fort Dix, New Jersey. J Inf Dis
Epizootiological aspects. Comp Immun Microbiol Infect 136(Suppl):381–385.
Dis 11:215–222. Kida, H.; Shortridge, K. F.; and Webster, R. G. 1988. Origin
Haesebrouck, F.; Biront, P.; Pensaert, M. B.; and Leunen, J. of the hemagglutinin gene of H3N2 influenza viruses
1985. Epizootics of respiratory tract disease in swine in from pigs in China. Virol 162(1):160–166.
Belgium due to H3N2 influenza virus and experimental Kundin, W. D. 1970. Hong Kong A2 influenza virus infection
reproduction of disease. Am J Vet Res 46(9):1926–1928. among swine during a human epidemic in Taiwan. Na-
Haines, D. M.; Waters, E. H.; and Clark, E. G. 1993. Im- ture 228:857.
munohistochemical detection of swine influenza A virus Kupradinun, S.; Peanpijit, P.; Bhodhikosoom, C.; Yoshioka,
in formalin-fixed and paraffine-embedded tissues. Can J Y.; Endo, A.; and Nerome, K. 1991. The first isolation of
288 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

swine H1N1 influenza viruses from pigs in Thailand. McBryde, C. N. 1927. Some observations on “hog flu” and
Arch Virol 118(3–4):289–297. its seasonal prevalence in Iowa. J Am Vet Med Assoc
Lamb, R. A., and Krug, R. M. 1996. In Fields Virology, vol. 71:368–377.
1, 3d ed. Ed. B. N. Fields, D. M. Knipe, and P. M. How- McBryde, C. N.; Niles, W. B.; and Moskey, H. E. 1928. In-
ley. Philadelphia: Lippincott-Raven, pp. 1353–1445. vestigations on the transmission and etiology of hog flu.
Lanza, I.; Brown, I. H.; and Paton, D. J. 1992. Pathogenicity J Am Vet Med Assoc 73:331–346.
of concurrent infection of pigs with porcine respiratory McGregor, S., and Easterday, B. C. 1979. Unpublished data.
coronavirus and swine influenza virus. Res Vet Sci Mensik, J. 1960. Production and behavior of swine influen-
53:309–314. za antibodies. I. Influence of colostral antibodies on im-
Laval, A.; Le Foll, P.; Gestin, G.; and Reynaud, G. 1991. munity in piglets during early life. Sb Cesk Akad Zemed
Grippes et coronavirus respiratoire porcin: Étude Ved 5:599–619. Vet Bull 31, Abstr 127.
serologique dans dix élevages bretons. Rec Med Vet ———. 1963. Formation of antibodies in swine influenza.
167:521–528. III. Colostrol immunity and inhibition of antibody for-
Lee, B. W.; Bey, R. F.; Baarsch, M. J.; Morrison, R. B.; and mation. Ved Pr Ustav Vet 3:141–149.
Freese, W. 1993a. Determination of hemagglutination- ———. 1966. The formation and dynamism of antibodies
inhibition titers to influenza A virus in porcine sera by in swine influenza. V. A long-term depression of the an-
the use of an enzyme-linked immunosorbent assay. Am tibody formation in the progeny of hyperimmune moth-
J Vet Res 54(8):1270. ers. Vet Med (Prague) 11:589–595.
Lee, B. W.; Bey, R. F.; Baarsch, M. J.; and Simonson, R. R. Mohan, R.; Saif, Y. M.; Erickson, G. A.; Gustafson, G. A.;
1993b. ELISA method for detection of influenza A infec- and Easterday, B. C. 1981. Serologic and epidemiologic
tion in swine. J Vet Diag Invest 4(4):510–515. evidence of infection in turkeys with an agent related to
Lee, B. W.; Bey, R. F.; Baarsch, M. J.; and Larson, M. E. 1995. the swine influenza virus. Avian Dis 25:11–16.
Class specific antibody response to influenza A H1N1 in- Morin, M.; Girard, C.; Elazhary, Y.; Fajardo, R.; Drolet, R.;
fection in swine. Vet Microbiol 43:241–250. and Lagace, A. 1990. Severe proliferative and necrotiz-
Ludwig, S.; Haustein, A.; Kaleta, E. F.; and Scholtissek, C. ing pneumonia in pigs: A newly recognized disease. Can
1994. Recent influenza A (H1N1) infections in pigs and Vet J 31:12.
turkeys in northern Europe. Virol 202(1):281–286. Murphy, B. R., and Webster, R. G. 1990. Influenza viruses.
Madec, F.; Gourreau, J. M.; Kaiser, C.; Le Dantec, J.; Vannier, In Virology. Ed. B. Fields. New York: Raven Press, pp.
P.; and Aymard M. 1985. Étude de la persistance d’une 1091–1154.
activité du virus grippal H1N1 (Swine) dans les élevages Nakamura, R. M. 1967. In Vivo and in Vitro Studies of
porcins en dehors des phases épidemiques. Comp Im- Swine Influenza: A Hypothesis on the Interepizootic
mun Microbiol Infect Dis 8:247–258. Survival of Virus. Ph.D. diss., Univ Wisconsin—Madi-
Madec, F.; Kaiser, C.; Jestin, A.; Gourreau, J.-M.; Vannier, P.; son.
Kobisch, M.; Paboeuf, F.; and Aymard, M. 1987. Les syn- Nakamura, R. M., and Easterday, B. C. 1970. Studies on
dromes grippaux en porcherie d’engraissement: En- swine influenza. III. Propagation of swine influenza
quête “flash” réalisée en Bretagne. Courte communica- virus in explants of respiratory tract tissues from fetal
tion. Le Point Vet 19:654–659. pigs. Cornell Vet 60:28–34.
Maes, D.; Deluyker, H.; Verdonck, M.; Castryck, F.; Miry, C.; Nakamura, R. M.; Easterday, B. C.; Pawlisch, R.; and Walker
and de Kruif, A. 1996. Epidemiological investigations on G. L. 1972. Swine influenza: Epizootiological and sero-
respiratory diseases in pigs in Belgium. Proc Int Congr logical studies. Bull WHO 47:481–487.
Pig Vet Soc 14:405. Nardelli, L.; Pascucci, S.; Gualandi, G. R.; and Loda, P. 1978.
Maes, L.; Haesebrouck, F.; and Pensaert, M. 1984. Experi- Outbreaks of classical swine influenza in Italy in 1976.
mental reproduction of clinical disease by intratracheal Zentralbl Veterinärmed (B) 25:853–857.
inoculation of fattening pigs with swine influenza virus Nayak, D. P.; Twiehaus, M. T.; Kelley, G. W.; and Underdahl,
isolates. Proc Int Congr Pig Vet Soc 8:60. N. R. 1965. Immunocytologic and histopathologic de-
Mancini, G.; Donatelli, I.; Rozera, C.; Arangio Ruiz, G.; and velopment of experimental swine influenza infection in
Butto, S. 1985. Antigenic and biochemical analysis of in- pigs. Am J Vet Res 26:1271–1282.
fluenza “A” H3N2 viruses isolated from pigs. Arch Virol Olsen, C. W.; McGregor, M. W.; Colley, A. J.; Schantz, B.;
83:157–167. Hotze, R.; and Hinshaw, V. S. 1993. Antigenic and ge-
Martinsson, K.; Klingeborn, B.; and Rockborn, G. 1983. Ut- netic analysis of a recently isolated H1N1 swine influen-
brott av influenza suisI Sverige. Sven Veterinartidn za virus. Am J Vet Res 54:1630–1636.
35:37. Onno, M.; Jestin, A.; Nannier, P.; and Kaiser, C. 1990. Diag-
Masurel, N.; De Boer, G. F.; Anker, W. J. J.; and Huffels, A. nosis of swine influenza with an immunofluorescence
D. N. H. J. 1983. Prevalence of influenza viruses A-H1N1 technique using monoclonal antibodies. Vet Quart
and A-H3N2 in swine in the Netherlands. Comp Immun 12(4):251–254.
Microbiol Infect Dis 6:141–149. Ottis, K.; Bollwahn, P. A.; and Heinritzi, K. 1981. Ausbruch
CHAPTER 22 SWINE INFLUENZA Easterday, Van Reeth 289

von Schweineninfluenza in der Bundesrepublik Runge, M.; Ganter, M.; Delbeck, F.; Hartwick, W.; Ruffer,
Deutschland: Klinik, Nachweis und Differenzierung. A.; Franz, B.; and Amtsberg, G. 1996. Demonstration of
Tierärztl Umschau 36:608–612. pneumonia in swine as a constant problem: Culture and
Ottis, K.; Sidoli, L.; Bachmann, P. A.; Webster, R. G.; and immunofluorescence microscopic studies of bron-
Kaplan, M. M. 1982. Human influenza A viruses in pigs: chioalveolar lavage (BAL) and serological findings (in
Isolation of a H3N2 strain antigenically related to German). Berl Münch Tierärzt Woch 109:101–107.
A/England/42/72 and evidence for continuous circula- Ryan–Poirier, K. A.; Katz, J. M.; Webster, R. G.; and Kawao-
tion of human viruses in the pig population. Arch Virol ka, Y. 1992. Application of Directigen FLU-A for the de-
73:103–108. tection of influenza A virus in human and nonhuman
Ouchi, A.; Nerome, K.; Kanegae, Y.; Ishida, M.; Nerome, R.; specimens. J Clin Microb 30:1072–1075.
Hayashi, K.; Hashimoto, T.; Kaji, M.; Kaji, Y.; and Inaba, Schmidt, R. C.; Maasab, H. F.; and Davenport, F. M. 1974.
Y. 1996. Large outbreak of swine influenza in southern Infection by influenza A viruses of tracheal organ cul-
Japan caused by reassortant (H1N1) influenza viruses: tures derived from homologous and heterologous hosts.
Its epizootic background and characterization of the J Infect Dis 129:28–36.
causative viruses. J Gen Virol 77:1751–1759. Scholtissek, C.; Burger, H.; Bachmann, P. A.; and Hannoun,
Palese, P., and Ritchey, M. B. 1977. Polyacrylamide gel elec- C. 1983. Genetic relatedness of hemagglutinins of the
trophoresis of the RNAs of new influenza virus strains: H1 subtype of influenza A strains. Virology 102:13–20.
An epidemiological tool. Dev Biol Stand 39:411–415. Schorr, E.; Wentworth, D.; and Hinshaw, V. S. 1994. Use of
Patriarca, R. A.; Kendal, A. R.; Zakowski, R. C.; Cox, N. J.; polymerase chain reaction to detect swine influenza
Trautman, M. S.; Cherry, J. D.; Auervach, D. M.; Mc- virus in nasal swab specimens. Am J Vet Res 55:952–956.
Cusker, J.; Belliveau, R. R.; and Kappus, K. D. 1984. Lack Sherrar, M. G.; Easterday, B. C.; and Hinshaw, V. S. 1989.
of significant person-to-person spread of swine influen- Antigenic conservation of H1N1 swine influenza virus-
za-like virus following fatal infection in an immunocom- es. J Gen Virol 70:3297–3303.
promised child. Am J Epidemiol 119:152–158. Shope, R. E. 1931a. Swine influenza. I. Experimental trans-
Pensaert, M.; Ottis, K.; Vandeputte, J.; Kaplan, M. M.; and mission and pathology. J Exp Med 54:349–359.
Bachmann, P. A. 1981. Evidence for the natural trans- ———. 1931b. Swine influenza. III. Filtration experiments
mission of influenza A virus from wild ducks to swine and etiology. J Exp Med 54:373–385.
and its potential importance for man. Bull WHO ———. 1964. Swine influenza. In Diseases of Swine, 2d ed.
59:75–78. Ed. H. W. Dunne. Ames: Iowa State Univ Press, pp.
Pirtle, E. C. 1977. Evaluation of A/New Jersey/76 influenza 109–126.
whole-virus vaccine in hysterectomy-derived pigs. J Inf Sinnecker, H.; Sinnecker, R.; Zilske, E.; Strey, A.; and
Dis (Suppl) 136:703–705. Leopoldt, D. 1983. Influenzavirus A/swine-Ausbruche
Pirtle, E. C.; Hill, H. R.; Swanson, M. R.; and Van Deusen, R. bei Hausscheinen und Antikorperbefunde in Hu-
A. 1976. Hemagglutination-inhibiting antibodies manseren. Zentralbl Bakteriol Parasitenkd infektionstr
against swine influenza and Hong Kong influenza virus- Hyg Abt 1: Orig Reihe A 255:209–213.
es in swine sera in the U.S.A. Bull WHO 53:7–11. Soerensen, K. J.; Hoye, P. D.; Roensholt, L.; Hage, L.; West-
Potgeiter, L. N. D.; Stair, E. L.; Morton, R. J.; and Whitenack, ergaard, J. M.; and Pedersen, K. B. 1981. Influenza hos
D. L. 1977. Isolation of swine influenza virus in Okla- svin i Danmark. Dan Veterinaertidsskr 64:826–829.
homa. J Am Vet Med Assoc 171:758–760. Sugimura, T.; Yonemochi, H.; Ogawa, T.; Tanaka, Y.; and
Pritchard, G. C.; Dick, I. G. C.; Roberts, D. H.; and Wibber- Kumagai, T. 1980. Isolation of a recombinant influenza
ley, G. 1987. Porcine influenza outbreak in East Anglia virus (Hsw1N2) from swine in Japan. Arch Virol
due to influenza A virus (H3N2). Vet Rec 121:548. 66:271–274.
Rekik, M. R.; Arora, D. J.; and Dea, S. 1994. Genetic varia- Teik, L. K., and Lan, L. S. 1985. A preliminary report on the
tion in swine influenza virus A isolate associated with isolation and identification of influenza A virus from an
proliferative and necrotizing pneumonia in pigs. J Clin outbreak in Sarawak. Kaijian-Veterinar 17:137–140.
Microb 32:515–518. Tumova, B.; Mensik, J.; Stumpa, A.; Fedova, D.; and Pospisil,
Renshaw, H. W. 1975. Influence of antibody-mediated im- Z. 1976. Serological evidence and isolation of a virus
mune suppression on clinical, viral, and immune re- closely related to the human A/Hong Kong/68 (H3N2)
sponses to swine influenza infection. Am J Vet Res strain in swine populations in Czechoslovakia in
36:5–13. 1969–1972. Zentralbl Veterinärmed (B) 23:590–603.
Roberts, D. H.; Cartwright, S. F.; and Wibberley, G. 1987. Urman, H. K.; Underdahl, N. R.; and Young, G. A. 1958.
Outbreaks of classical swine influenza in pigs in England Comparative histopathology of experimental swine in-
in 1986. Vet Rec 121:53–55. fluenza and virus pneumonia of pigs in disease-free, an-
Romvary, J., and Tanyi, J. 1975. Hong Kong influenza virus tibody-devoid pigs. Am J Vet Res 19:913–917.
infections in animals in Hungary. In Proc 20th World Vandeputte, J.; Pensaert, M.; and Castryck, F. 1980. Serolo-
Vet Congr, pp. 1455–1456. gische diagnose en onderzoek naar verspreiding van het
290 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

varkensinfluenzavirus in Belgie. Vlaams Diergeneeskd Winkler, G. C., and Cheville, N. F. 1986. Ultrastructural
Tijdschr 49:1–7. morphometric investigation of early lesions in the pul-
Van Reeth, K., and Pensaert, M. 1994a. Prevalence of infec- monary alveolar region of pigs during experimental
tions with enzootic respiratory and enteric viruses in swine influenza infection. Am J Path 122:541–552.
feeder pigs entering fattening herds. Vet Rec Witte, K. H.; Nienhoff, H.; Ernst, H.; Schmidt, U.; and
135:594–597. Prager, D. 1981. First outbreak of swine influenza in pig
———. 1994b. Porcine respiratory coronavirus-mediated herds in the Federal Republic of Germany. Tierärztl Um-
interference against influenza virus replication in the sch 36:591–606.
respiratory tract of feeder pigs. Am J Vet Res Woods, G. T. 1975. Prevalence of hemagglutination-inhibi-
55:1275–1281. tion (HI) antibodies to influenza viruses A/swine/IV/63
———. 1997. Unpublished data. and A/swine/Taiwan/7310/70 (H3N2) in Illinois swine
Van Reeth, K.; Nauwynck, H.; and Pensaert, M. 1996a. Dual herds, 1971–1973. Res Commun Chem Pathol Pharma-
infections of feeder pigs with porcine reproductive and col 10:573–575.
respiratory syndrome virus followed by porcine respira- Woods, G. T., and Mansfield, M. E. 1976. Antigenicity of in-
tory coronavirus or swine influenza virus: A clinical and activated swine influenza virus concentrated by centrifu-
virological study. Vet Mic 48:325–335. gation. Res Commun Chem Pathol Pharmacol
Van Reeth, K.; Van Hoof, D.; Willems, L.; and Pensaert, M. 13:129–132.
1996b. A clinical study of dual infections with porcine ———. 1978. Serologic response of gilts of breeding age to
reproductive-respiratory syndrome virus and swine in- human influenza vaccines containing A/New Jersey/76
fluenza virus administered with different time intervals. (Hsw1N1) antigen. Res Commun Chem Pathol Pharma-
Proc Int Congr Pig Vet Soc 14:59. col 19:177–180.
Wells, D. L.; Hopfensperber, D. L.; Arden, N. H.; Harmon, ———. 1980. Experimental challenge of pregnant gilts af-
M. W.; Davis, J. P.; Tipple, M. A.; and Schonberger, L. B. ter vaccination with vaccine prepared for the national
1991. Swine influenza virus infections: Transmission vaccination program in the United States. Bovine Pract
from ill pigs to humans at a Wisconsin agricultural fair 1:41–44.
and subsequent probable person-to-person transmis- World Health Organization (WHO). 1980. Memorandum:
sion. JAMA 265:478–481. A revision of the system of nomenclature for influenza
Wentworth, D. E.; Thompson, B. L.; Xu, X.; Regnery, H. L.; virus. Bull WHO 58:585–591.
Cooley, A. J.; McGregor, M. W.; Cox, N. J.; and Hinshaw, Yamaoka, M.; Holta, H.; Itoh, M.; and Homma, M. 1991.
V. S. 1994. An influenza A (H1N1) virus closely related Prevalence of antibody to influenza C virus among pigs
to swine influenza virus, responsible for a fatal case of in Hyoga Prefecture, Japan. J Gen Virol 72:711–714.
human influenza. J Virol 68:2051–2058. Yus, E.; Sanjuan, M. L.; Garcia, F.; Castro, J. M.; and Simar-
Wentworth, D. E.; McGregor, M. W.; Macklin, M. D.; Neu- ro, I. 1992. Influenza A viruses, epidemiologic study in
mann, V.; and Hinshaw, V. S. 1997. Transmission of fatteners in Spain (1987–89). J Vet Med B 39:113–118.
swine influenza virus to humans after exposure to ex- Ziegler, T.; Hall, H.; Sanchez-Fauquier, A.; Gamble, W. C.;
perimentally infected pigs. J Inf Dis 175:7–15. and Cox, N. J. 1995. Type and subtype-specific detection
Wibberley, G.; Swallow, C.; and Roberts, D. H. 1988. Char- of influenza viruses in clinical specimens by rapid cul-
acterization of an influenza A (H3N2) virus isolated ture assay. J Clin Microb 33:318–321.
from pigs in England in 1987. Br Vet J 144:196–201.
Swine Pox
23 J. A. House and C. A. House

Swine pox (SwP) was first reported in 1842 by Spinola in distributed on the backs and sides of affected pigs
Europe, and in 1929 by McNutt et al. in North America. (Schwarte and Biester 1941).
Worldwide in distribution, SwP is usually associated Occasionally, horizontal transmission may occur in
with swine operations that have poor sanitation and/or the absence of insects, since SwPV is shed in nasal and
intensive breeding with open-herd management. SwP oral secretions and from lesions. The SwPV is present in
causes little economic loss and can usually be differenti- infected epithelium and in dry scabs produced in the lat-
ated from other diseases by its clinical signs and epi- er stages of infection. Abraded skin can serve as the
demiology. route of entry for SwPV. The virus may persist for up to
The clinical signs of infection may be caused by ei- a year in the desiccated form, likely accounting for the
ther vaccinia virus (VV) or SwP virus (SwPV) (Man- persistence of SwP in affected herds.
ninger et al. 1940; Shope 1940). Since the eradication of The morbidity of SwP may approach 100% in young
smallpox (variola) and the subsequent cessation of vac- stock up to 4 months of age where poor hygiene occurs.
cinia vaccination and VV infection of swine, the occur- The disease may have a seasonal incidence related to the
rence of SwP has decreased (Meyer and Conroy 1972). prevalence of insects. Mortality is usually less than 5%.

ETIOLOGY CLINICAL SIGNS

SwPV is classified in the family Poxviridae and is the only SwP is a typical pox disease, with the development of the
member of the genus Suipoxvirus (Francki et al. 1991). lesions progressing through the classic stages of macule
The large virion (300–450 × 176–260 nm) contains dou- (reddening), papule (reddening with edema), vesicle (flu-
ble-stranded DNA in a characteristic poxvirus core or id exuding from the pox lesion), and pustule or crust for-
nucleoid bordered by lateral bodies and surrounded by mation (drying of the lesion with scab formation) (Fig.
an outer coat of numerous proteins. Because SwPV is en- 23.1). The vesicular stage is not readily observed in SwP.
veloped, it is ether sensitive (Cheville 1966a). Typical of The time from macule formation to scabbing and resolu-
poxviruses, it shares antigenic epitopes with members of tion of lesions is 3–4 weeks. A lymphadenitis may occur.
other poxvirus genera (Ouchi et al. 1992). The large Complication with bacterial infection prolongs the reso-
genome has room for many insertions, and the SwPV has lution of the lesion (Miller and Olson 1978, 1980).
been considered a vector for the delivery of immunogens Young animals are more severely affected than adults.
to swine (Foley et al. 1991). Suckling piglets may have lesions in the perioral epitheli-
The virus, once adapted to grow in cell culture, read- um or a generalized disease with lesions all over the
ily reaches high titers and maintains its pathogenicity for body. Lesions are more prominent on the hairless parts
swine (Kasza et al. 1960; Kasza and Griesemer 1962). of the skin. Mechanical transmission by pig lice results
in lesions on the lower parts of the body, including the
EPIDEMIOLOGY udder and vulva. Transmission by flies and mosquitoes is
less common and may result in lesions on the dorsal
SwP is a disease of pigs only; SwPV does not infect cattle, parts of the body, including the snout and ears. Sporadic
horses, sheep, dogs, cats, domestic fowl, rabbits, guinea congenital infection has been reported (Borst et al. 1990;
pigs, rats, mice, or humans (Shope 1940; Schwarte and Paton et al. 1990).
Biester 1941; Datt 1964). The incubation period following experimental intra-
The pig louse (Haematopinus suis) serves as a me- dermal inoculation is 2–5 days. The incubation period
chanical vector and is considered the primary means of following intravenous inoculation is 10–14 days (Shope
transmission of SwPV (Shope 1940). The ventral distrib- 1940) but can be as short as 5 days (Kasza and Griesemer
ution of pox lesions can be correlated to the habitat of H. 1962), the range likely being related to the inoculation
suis. Flies and mosquitoes have been implicated as me- dosage. Incubation under field conditions may be up to
chanical vectors, and in this situation, pox lesions are 14 days.

291
292 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

23.1. Swine pox lesions on


the belly of a pig. Pa = papule;
Pu = pustule; N = nipple.

PATHOGENESIS which is also observed with capripoxvirus infection of


sheep, goats, and cattle, occurs in infected epithelial cells
SwP is usually initiated by SwPV entering a break in the containing ICIBs and is a result of margination of chro-
skin such as a louse bite or an abrasion. The SwPV repli- matin with nuclear filamentous matrix formation. There
cates in the cytoplasm of the cells of the stratum spi- may be intercellular edema, which could account for the
nosum. Lesions in local lymph nodes are not extensive, transient vesicular stage. Acantholysis, the separation of
and the virus is not readily isolated from them. Sec- individual cells of the stratum spinosum with loss of
ondary lesions are believed to result exclusively from the desmosomes and detachment into the vesicular fluid,
spread of the virus from an existing lesion, not by a cell- was not found with SwPV infection (Meyer and Conroy
associated viremia as in most other poxvirus infections. 1972). Necrosis of epithelial cells and the presence of
The virus has not been recovered from the blood (Kasza neutrophils and macrophages in the dermis and epider-
and Griesemer 1962; Shope 1940), but this may be due mis are common in later stages of the pox lesion (Fig.
to the fact that the SwPV is difficult to isolate, especially 23.2). The pustules involve the full thickness of epitheli-
when in low concentrations. Viremia must occur, as evi- um and may heal, leaving a light scar. Poxvirus particles
denced by congenital infection in piglets. can be observed in the cytoplasm of infected cells by
Following intravenous inoculation, gnotobiotic electron microscopy (Fig. 23.3).
piglets developed skin lesions uniformly over their entire
bodies but not in internal organs (Meyer and Conroy DIAGNOSIS
1972), confirming the propensity of SwPV to infect the
integument. In gnotobiotic pigs, the macular stage was SwP is typified by the presence of pox lesions on the skin
not observed; therefore, the early reddening may be the of the belly, ears, snout, vulva, and back. Secondary bac-
combined effect of reaction to louse bites, opportunistic terial infections may cause more extensive lesions and lo-
bacteria, and SwPV. cal abscess formation. Diseases to consider in the differ-
ential diagnosis of SwP include classic vesicular diseases,
LESIONS bites from needle teeth, pityriasis rosea, parakeratosis,
parasitic skin disorders, allergic skin reactions, early
The gross lesions of SwP are described in the section on stages of ringworm, staphylococcal or streptococcal epi-
clinical signs. dermitis, and cutaneous erysipelas.
The histopathological changes caused by SwPV are The SwPV infection may be confirmed by detection
similar to those of other poxviruses (Kasza and Griese- of viral antigens using immunofluorescence and electron
mer 1962; Cheville 1966a, b; Conroy and Meyer 1971; microscopy. The presence of ICIBs along with central nu-
Meyer and Conroy 1972; De Boer 1975). The virus repli- clear clearing in affected epithelial cells is pathognomon-
cates in the stratum spinosum, causing hydropic degen- ic for SwPV, as VV infection does not cause the central
eration. Sections stained with hematoxylin and eosin re- nuclear clearing (Teppema and De Boer 1975). The virus
veal one to three eosinophilic-staining intracytoplasmic may be isolated by multiple passages (up to five) in swine
inclusion bodies (ICIBs) in some cells during all stages of cell cultures and definitively identified by neutralization
the infection. Characteristic central nuclear clearing, with specific-reference antiserum.
23.2. Swine pox lesions in the epidermis: ballooning
degeneration of cytoplasm (B), intracytoplasmic inclusion
bodies (I), and central nuclear clearing (N).

23.3. Electron micrograph of an epidermal cell with cytoplasmic degeneration and numerous
poxvirus particles (V); a second cell has marginated chromatin (MC) and a filamentous matrix
(F) in the nucleus, which corresponds to the central nuclear clearing seen histologically. Insert
shows typical poxvirus particles. (Courtesy of D. A. Gregg.)
294 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Serologic diagnosis utilizes the agar gel immunodif- De Boer, G. F. 1975. Swinepox: Virus isolation, experimental
fusion test or the more sensitive counterelectrophoresis infections and the differentiation from vaccinia virus in-
test (De Boer 1975). Swine do not develop high levels of fections. Arch Virol 49:141–150.
neutralizing antibody (Shope 1940; Kasza et al. 1960), Foley, P. L.; Paul, P. S.; Levings, R. L.; Hanson, S. K.; and
and negative results on the virus neutralization test Middle, L. A. 1991. Swinepox virus as a vector for the de-
should not be interpreted as the absence of SwPV infec- livery of immunogens. Ann NY Acad Sci 646:220–222.
tion. Francki, R. B.; Faquot, C. M.; Knudsen, D. L.; and Brown, F.
1991. Classification and Nomenclature of Viruses. Fifth
TREATMENT Report of the International Committee on Taxonomy of
Virus. Arch Virol, Suppl 2. New York: Springer Verlag,
There is no known treatment for SwP, although antibi- pp. 320–326.
otics may diminish the complications caused by infec- Kasza, L., and Griesemer, R. A. 1962. Experimental swine
tion with secondary bacteria. Affected animals should be pox. Am J Vet Res 23:443–450.
isolated. Ectoparasites should be controlled by the use of Kasza, L.; Bohl, E. H.; and Jones, D. O. 1960. Isolation and
licensed insecticides, and the premises should be aggres- cultivation of swinepox virus in primary cell cultures of
sively cleaned and disinfected. swine origin. Am J Vet Res 21:269–273.
Manninger, R.; Csontos, J.; and Salyi, J. 1940. Über die Ati-
PREVENTION ologie des pockenartigen Ausschlags der Ferkel. Arch
Tierheilkd 75:159.
Vaccines for SwP have not been developed due to the rel- McNutt, S. H.; Murray, C.; and Purwin, P. 1929. Swinepox. J
atively low economic importance of this disease. Recov- Am Vet Med Assoc 74:752.
ered animals are specifically immune to SwP (Shope Meyer, R. C., and Conroy, J. D. 1972. Experimental swine
1940) even in the absence of demonstrable neutralizing pox in gnotobiotic piglets. Res Vet Sci 13:334–338.
antibody, implying the importance of cell-mediated im- Miller, R. B., and Olson, L. D. 1978. Epizootic of concurrent
munity. cutaneous streptococcal abscesses and swinepox in a
Animals introduced into herds should be carefully in- herd of swine. Am J Vet Med Assoc 172:676–680.
spected for skin lesions and ectoparasites. Good hygien- ———. 1980. Experimental induction of cutaneous strep-
ic practices are essential for the prevention and control of tococcal abscesses as a sequela to swinepox. Am J Vet Res
SwP. 41:341–347.
Ouchi, M.; Fujiwara, M.; Hatano, Y.; Yamada, M.; and Shi-
REFERENCES ro, N. H. 1992. Analysis of swinepox virus antigens us-
ing monoclonal antibodies. J Vet Med Sci 54:731–737.
Borst, G. H. A.; Kimman, T. G.; Gielicens, A. L. J.; and Van Paton, D. K.; Brown, I. H.; Fitton, J.; and Wrathall, A. E.
der Kamp, J. S. 1990. Four sporadic cases of congenital 1990. Congenital pig pox: A case report. Vet Rec
swinepox. Vet Rec 127:61–63. 127:204.
Cheville, N. F. 1966a. The cytopathology of swinepox in the Schwarte, L. H., and Biester, H. E. 1941. Pox in swine. Am J
skin of swine. Am J Pathol 49:339–352. Vet Res 2:136–140.
———. 1966b. Immunofluorescent and morphologic stud- Shope, R. E. 1940. Swine pox. Arch Gesamte Virusforsch
ies on swinepox. Pathol Vet 3:556–564. 1:457–467.
Conroy, J. D. And Meyer, R. C. 1971. Electron microscopy of Spinola, M. 1842. Krankheiten der Schweine. Ed. A.
swinepox virus in germfree pigs and in cell culture. Am J Hieschwald. Berlin, p. 204.
Vet Res 32:2021–2032. Teppema, J. S., and De Boer, G. F. 1975. Ultrastructural as-
Datt, N. S. 1964. Comparative studies of pigpox and vac- pects of experimental swinepox with special reference to
cinia viruses. I. Host range pathogenicity. J Comp Pathol inclusion bodies. Arch Virol 49:151–163.
74:62–80.
24 Transmissible Gastroenteritis and
Porcine Respiratory Coronavirus
L. J. Saif and R. D. Wesley

Transmissible gastroenteritis (TGE) is a highly conta- early spring; and (4) the commercial vaccines available
gious, enteric viral disease of swine characterized by are of limited effectiveness (Bohl et al. 1975; Moxley and
vomiting, severe diarrhea, and a high mortality (often Olson 1989a; Saif and Theil 1990).
100%) in piglets under 2 weeks of age. Although swine of
all ages are susceptible to this viral infection, the mortal- ETIOLOGY
ity in swine over 5 weeks of age is low.
The disease was first reported by Doyle and Hutch- The viral etiology of TGE was suggested by the initial re-
ings (1946) as occurring in the United States in 1945, al- port of Doyle and Hutchings (1946) when they described
though it undoubtedly had existed prior to this time. the filterable nature of the infectious agent. TGEV is a
Subsequent to its recognition in the United States, TGE species belonging to the genus Coronavirus of the family
was reported in Japan in 1956 (Sasahara et al. 1958) and Coronaviridae (Cavanagh et al. 1995).
England in 1957 (Goodwin and Jennings 1958). Since
then it has been reported in most countries worldwide. Size, Morphology, and Morphogenesis
The disease is most frequently diagnosed and causes TGEV is enveloped and pleomorphic, with an overall di-
the most loss when it occurs in herds at farrowing time. ameter of 60–160 nm as viewed by negative-staining
In contrast, it often goes undiagnosed when it occurs in electron microscopy (EM) (Fig. 24.1) (Okaniwa et al.
growing, finishing, or adult swine because of the mild 1968; Phillip et al. 1971; Saif et al. 1977; Granzow et al.
clinical signs, which usually consist only of inappetence 1981). It has a single layer of club-shaped surface projec-
and diarrhea of a few days’ duration. Because of these tions that are 12–25 nm in length and widely spaced.
undiagnosed infections, serologic surveys are a more ac- The morphogenesis of coronaviruses has been re-
curate indication of the prevalence of TGE virus (TGEV). viewed (Siddell et al. 1983). TGEV antigen can be
Such surveys indicated that 19–54% of sampled swine demonstrated in the cytoplasm by immunofluorescence
herds in Europe and North America were seropositive for (IF) as early as 4–5 hours postinfection (Pensaert et al.
TGEV antibodies (Gagnon et al. 1974; Toma et al. 1978; 1970a). Maturation of virus occurs in the cytoplasm by
Egan et al. 1982), and a more recent survey confirmed a
similar TGEV seroprevalence (36%) in the United States
(USDA APHIS VS 1992). Currently in many European
countries, nearly 100% of swine herds are seropositive
for TGEV antibodies. This situation is due to a TGEV res-
piratory variant, porcine respiratory coronavirus
(PRCV), that appeared in 1984 and rapidly spread in Eu-
ropean swine (Brown and Cartwright 1986; Pensaert et
al. 1986; Pensaert 1989; Pensaert and Cox 1989; Laude et
al. 1993; Pensaert et al. 1993). Economic losses from TGE
can be severe, as reviewed in earlier studies (Pritchard
1982, 1987; Mousing et al. 1988). In North America, the
economic impact is still serious, but in Europe it has de-
creased following the establishment of enzootic PRCV
(Pensaert and Cox 1989; Laude et al. 1993).
In the densely swine-populated areas of North Amer-
ica, TGE remains a major cause of sickness and death in
piglets. Swine producers are especially apprehensive
about this disease because (1) mortality is high in new-
born pigs; (2) there is no effective, practical treatment; 24.1. Electron micrograph of a TGEV particle show-
(3) strict biosecurity measures are needed to prevent en- ing typical coronavirus morphology. Arrow points to the
try of the virus into a herd, especially in the winter and virus peplomers, or spikes. Bar = 100 nm.

295
296 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

budding through endoplasmic reticulum, and viral parti-


cles (65–90 nm in diameter) are often observed within
cytoplasmic vacuoles (Fig. 24.2A) (Thake 1968; Pensaert
et al. 1970b; Wagner et al. 1973). The virus is frequently
seen lining the host cell membranes after exit from in-
fected cells (Fig. 24.2B). TGEV glycoproteins have been
identified on the surface of infected swine testicular (ST)
cells (Laviada et al. 1990).

Biological Properties
TGEV is very stable when stored frozen but somewhat la-
bile at room temperature or above. No detectable drop in
titer occurred when virus of pig intestine origin was
stored at −20˚C for 6-18 months (Young et al. 1955; Hael-
terman and Hutchings 1956). In contrast, virus of pig in-
testine origin, when allowed to dry and putrefy at 21˚C,
was rather labile; after 3 days only two of four inoculated
pigs became sick; and after 10 days no viable virus was
detected by pig inoculation (Bay et al. 1952). When held
at 37˚C, there was a log 10 reduction in infectivity titer
every 24 hours (Young et al. 1955). Storage of cell culture
virus at −20˚C, −40˚C, or −80˚C for 365 days did not re-
sult in any significant drop in titer, whereas storage at
37˚C for 4 days resulted in total loss of infectivity (Hara-
da et al. 1968). Infectious virus persisted in a liquid ma-
nure slurry for more than 8 weeks at 5˚C, 2 weeks at
20˚C, and 24 hours at 35˚C (Haas et al. 1995).
The virus is highly photosensitive. Haelterman
(1963) reported that fecal material containing 105 pig-in-
fectious doses (PID) was inactivated within 6 hours when
exposed to sunlight. Cartwright et al. (1965) reported the
photosensitivity of a cytopathic strain when it was ex-
posed to ultraviolet light on a laboratory bench.
In terms of chemical stability, TGEV is inactivated by
exposure to 0.03% formalin, 1% Lysovet (phenol and
aldehyde), 0.01% beta propiolactone, 1 mM binary ethyl-
enamine, sodium hypochlorite, NaOH, iodines, quater-
nary ammonium compounds, ether, and chloroform
(Harada et al. 1968; Nakao et al. 1978; Brown 1981; Van
Cott et al. 1993).
As reported for other enteric viruses, TGEV field
strains are trypsin resistant, relatively stable in pig bile,
and stable at pH 3 (Harada et al. 1968; Moscari 1980a;
Laude et al. 1981). These properties allow the virus to
survive in the stomach and small intestine. However, at-
tenuated strains, as well as field strains, of TGEV vary in
these properties, and most studies have failed to show a
correlation between susceptibility to these various treat-
ments and cell culture passage level or degree of viru-
lence (Furuuchi et al. 1975; Hess and Bachmann 1976; 24.2. (A) TGEV in vesicles of the endoplasmic
Moscari 1980a; Laude et al. 1981). reticulum of a pig kidney cell (36 hours postinfection).
Bar = 100 nm. (B) TGEV lining the cell membrane of a
pig kidney cell (36 hours postinfection). N = nucleus;
Molecular Properties bar = 200 nm.
TGEV and the deletion mutant, PRCV, are enveloped
viruses containing one large, polyadenylated, single-
stranded, genomic RNA of positive-sense polarity. The
CHAPTER 24 TRANSMISSIBLE GASTROENTERITIS Saif, Wesley 297

genome organization, replication strategy, and expres- nating activity is located at the N-terminal region of the
sion of viral proteins are similar to those of other coron- TGEV S protein, a region that is missing from the PRCV
aviruses (Laude et al. 1990; 1993; Enjuanes and Van der S protein; thus, determining the presence or absence of
Zeijst 1995). The genomic RNA of TGEV is infectious hemagglutinating activity (Schultze et al. 1996) could be
(Brian et al. 1980), and the complete sequence of the a method to differentiate PRCV and TGEV strains.
Purdue-115 strain of TGEV is 28,579 nucleotides long MABs have been produced against attenuated
(Kapke and Brian 1986; Rasschaert and Laude 1987; (Jimenez et al. 1986; Laude et al. 1986; Correa et al. 1990;
Rasschaert et al. 1987; Eleouet et al. 1995). Delmas et al. 1990) and virulent (Welch and Saif 1988;
TGEV has a buoyant density in sucrose of 1.18–1.20 Zhu et al. 1990; Simkins et al. 1992, 1993) strains of
g/mL (Brian et al. 1980; Jimenez et al. 1986). The phos- TGEV and used to characterize TGEV proteins and map
pholipids and glycolipids incorporated into the virus en- epitopes which elicit antibodies. The major neutraliza-
velope are derived from the host cell, and thus, the enve- tion determinants on the S protein were highly con-
lope composition is host cell dependent (Pike and served for TGEV and PRCV strains (Jimenez et al. 1986;
Garwes 1977). Laude et al. 1986; Garwes et al. 1987; Welch and Saif
Intact TGEVs contain four structural proteins: a 1988; Sanchez et al. 1990; Simkins et al. 1992, 1993). Use
large surface glycoprotein (spike or S); a small mem- of neutralizing MABs to map epitopes on the S glyco-
brane protein (sM); an integral membrane glycoprotein protein revealed four distinct antigenic sites (A, B, C, D),
(M); and a nucleocapsid protein (N) (Garwes and Pocock with site A-B as the highly conserved immunodominant
1975; Spaan et al. 1988; Godet et al. 1992). The N protein epitope recognized by strongly neutralizing MABs (Gar-
(47 kDa) interacts with viral RNA to form a helical ri- wes et al. 1987; Simkins et al. 1992, 1993; Correa et al.
bonucleoprotein complex. This structure in association 1990; Delmas et al. 1990; Gebauer et al. 1991), although
with M protein forms a spherical subviral internal core other sites (D, C) also can induce neutralizing antibodies
(Risco et al. 1996). The 29–36 kDa M glycoprotein, firm- (Laude et al. 1986; Posthumus et al. 1990). The locations
ly embedded in the viral envelope by three or four mem- of each of the four major antigenic sites were mapped on
brane-spanning regions, is present in two topological the primary structure of the S glycoprotein. Starting
forms (Risco et al. 1995). In one conformation, the N ter- from the amino terminal end, these sites were designat-
minus of the M protein is external to the virus envelope ed C, B, D, and A by the Madrid group (Correa et al.
and the C terminus is internal, whereas, in the second 1990) and D, C, and A-B by the Paris group (Delmas et al.
conformation, both ends of the molecule are on the out- 1990). Paris sites D, C, and A-B (the designation used in
side of the virion. In both conformations, the hy- this chapter unless specified otherwise) overlap Madrid
drophilic N terminus with a single accessible glycosyla- sites B, D, and A, respectively. Three subsites (Aa, Ab,
tion site is responsible for interferon induction (Charley Ac) for site A were recognized using TGEV-MAB-resis-
and Laude 1988). Epitopes on protuding N-terminal and tant mutants (Correa et al. 1990).
C-terminal ends of M protein molecules bind comple-
ment-dependent neutralizing monoclonal antibodies Antigenic and Genomic Relationships
(MABs) (Woods et al. 1988; Laude et al. 1992; Risco et al. Only one serotype of TGEV is known (Kemeny 1976).
1995). S glycoproteins, probably as trimer complexes PRCV strains are not distinguished from enteropatho-
(Delmas and Laude 1990), are visible in electron micro- genic strains of TGEV by the virus neutralization (VN)
graphs as the virus “corona” (Fig. 24.1). Functions attrib- test (Pensaert 1989; Pensaert and Cox 1989) but can be
uted to the S glycoprotein (170–220 kDa) include virus distinguished by a blocking enzyme-linked immunosor-
neutralization (complement-independent), virus-cell at- bent assay (ELISA) test (see the section on diagnosis be-
tachment, membrane fusion, as well as hemagglutina- low) or by a large deletion in the S gene of PRCV result-
tion. Garwes et al. (1978/79) have shown that purified S ing in a smaller S protein (170–190 kDa) than the S
glycoprotein elicited the production of TGEV-neutraliz- protein of TGEV (220 kDa).
ing antibodies, which neutralize at multiple steps in the Seven coronaviruses are related antigenically or by
virus replication cycle (Nguyen et al. 1986; Sune et al. their genomic sequence (Enjuanes and Van der Zeijst
1990). 1995; Siddell 1995). These are TGEV, PRCV, canine cor-
Aminopeptidase N and a second 200 kDa surface navirus (CCV), feline infectious peritonitis virus (FIPV),
protein on ST cells and on porcine intestinal enterocytes feline enteric coronavirus (FECV), porcine epidemic di-
have been identified as virus receptors (Delmas et al. arrhea virus (PEDV), and human coronavirus (HCV
1992; Weingartl and Derbyshire 1994). The receptor 229E). Within this group, only TGEV, PRCV, CCV, FIPV,
binding site for aminopeptidase N and the major neu- and FECV form an antigenically related subset based on
tralizing site (site A) on the S protein are located within cross-reactivity in VN and IF tests and with MABs to the
the same domain (Godet et al. 1994). Treatment of S, N, or M protein (Pedersen et al. 1978; Reynolds et al.
TGEV with sialidase enhanced hemagglutinating activity 1980; Woods 1982; Enjuanes and Van der Zeijst 1995).
(Noda et al. 1987; Schultze et al. 1996). This hemaggluti- Moreover, all members of this antigenic subset share the
298 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

antigenic subsite Ac on the S protein (Enjuanes and Van Molecular probes and MABs are also used to detect
de Zeijst 1995). Additional cross-reactivity between the and differentiate among these antigenically related coro-
S, M, and N proteins of TGEV, CCV, and FIPV was naviruses. In terms of the S glycoprotein gene that con-
shown by radioimmunoprecipitation, immunoblotting, fers host range specificity, the 300 amino acid residues at
and ELISA, and it was suggested that these viruses may the N terminus are the most variable. In this domain
actually represent host range mutants of an ancestral CCV and FIPV are more similar to each other than to
virus strain (Horzinek et al. 1982). Similar cross-reactivi- TGEV (Jacobs et al. 1987; Wesseling et al. 1994). Thus,
ty among the S, M, and N structural proteins of TGEV cDNA probes developed from this domain of the TGEV
and PRCV was observed using polyclonal antisera in im- S gene reacted with TGEV RNA but failed to recognize
munoblotting assays (Callebaut et al. 1988). CCV or FIPV RNA under conditions of high stringency.
PEDV is another coronavirus of pigs that causes a Either cDNA probes or RT-PCR (reverse transcriptase-
disease similar to TGEV; this disease has been docu- polymerase chain reaction) techniques are used to differ-
mented only in swine in Europe and Asia. Antibodies to entiate U.S. strains of PRCV from prototype strains of
PEDV have not been detected in a limited survey of adult TGEV (Bae et al. 1990; Vaughn et al. 1994; Jackwood et
swine sera from the United States (DeBouck et al. 1982; al. 1995). Similar differentiation of the TGEV-related
L. J. Saif and M. B. Pensaert—unpublished data, 1990). coronaviruses was possible using specific MABs to the S
Some one-way, immunoblotting cross-reactivity with the glycoprotein of TGEV that recognized TGEV strains but
N protein has been reported for PEDV, FIPV, CCV, TGEV, failed to react with PRCV, FIPV, or CCV (Laude et al.
and a putative mink coronavirus (Yaling et al. 1988; Have 1988; Callebaut et al. 1989; Sanchez et al. 1990; Simkins
et al. 1992). However, no antigenic cross-reactivity be- et al. 1992).
tween PEDV and TGEV-related coronaviruses has been PRCV strains, isolated during the mid-1980s and
detected using polyclonal antisera or MABs in other 1990s, have been characterized and partially sequenced
serologic assays (Pensaert and DeBouck 1978; Callebaut (Rasschaert et al. 1990; Wesley et al. 1991b; Britton et al.
et al. 1988; Enjuanes and Van der Zeijst 1995). Another 1991; Vaughn et al. 1995). An overall nucleotide and
coronavirus of pigs, hemagglutinating en- amino acid sequence homology of 96% between TGEV
cephalomyelitis virus, is antigenically unrelated to the and PRCV suggests that PRCV evolved from TGEV and
coronaviruses in group I (Enjuanes and Van der Zeijst that this occurred on a number of independent occa-
1995; Siddell 1995). sions. Two striking features characterize the PRCV
Although TGEV, CCV, and FIPV are antigenically genome: (1) a large deletion (621–681 nucleotides) at a
closely related, Reynolds et al. (1980) suggested that consistent site near the N terminus of the S gene that
TGEV and CCV could be distinguished serologically gives rise to a smaller S protein; and (2) a variable region
(with sera from naturally infected animals) by employing with deletions that often eliminate the open reading
two-way cross-neutralization tests. Additional in vitro bi- frame (ORF) immediately downstream of the S gene.
ological differences also have been detected; whereas These genetic changes may account for the altered tissue
both TGEV and CCV will grow in either canine kidney tropism of PRCV. From sequence data, significant ho-
cells (Welter 1965; Reynolds et al. 1980) or an estab- mologies occur in the three genes encoding the three
lished feline cell line (Woods 1982), neither CCV nor main structural proteins within TGEV-related viruses
FIPV will grow in ST or porcine thyroid cells, both of (TGEV, PRCV, CCV, FECV, FIPV) and between TGE-re-
which support the growth of TGEV isolates (Reynolds et lated viruses and HCV 229E and PEDV (Enjuanes and
al. 1980). Van der Zeijst 1995). Thus, sequencing data show a ge-
In vivo biological differences also exist among TGEV, nomic relationship between these latter two viruses and
CCV, and FIPV in their pathogenicity for neonatal pigs TGEV-related viruses that is not demonstrated by sero-
(Binn et al. 1974; Woods and Pedersen 1979). Whereas logic analysis (Enjuanes and Van der Zeijst 1995; Siddell
virulent FIPV caused diarrhea and intestinal lesions sim- 1995).
ilar to those of virulent TGEV, CCV caused no clinical An altered phenotype was reported for a small plaque
signs and only slight villous atrophy. IF, using a porcine (SP) variant of TGEV (Woods 1978; Woods et al. 1981).
anti-TGEV serum conjugate, occurred in villous entero- Although the S gene of the SP variant and the S gene of
cytes of TGEV- and FIPV-infected pigs but predominated wild-type TGEV are similar, a large deletion (462 nu-
in crypt enterocytes of CCV-infected pigs. CCV shed by cleotides) is present in the SP viral genome just down-
acutely infected dogs was shown to infect baby pigs and stream from the S gene (Wesley et al. 1990a). This dele-
induced serum neutralizing antibodies for both CCV and tion eliminated the ORF of one potential viral encoded
TGEV (Woods and Wesley 1992). However, baby pigs protein and eliminated the N-terminal portion of a sec-
and pregnant gilts experimentally infected with FIPV did ond potential viral protein. These two ORFs are also
not produce TGEV-neutralizing antibodies but did devel- missing from the high cell culture–passaged Nouzilly
op some immunity to a TGEV challenge (Woods and strain, a second attenuated TGEV with an SP phenotype
Pedersen 1979). (Britton et al. 1994). Presumably, the altered pathogenic-
CHAPTER 24 TRANSMISSIBLE GASTROENTERITIS Saif, Wesley 299

ity of SP virus, the Nouzilly virus, and the PRCVs results immunity of the sow. Mortality is usually less than
from these genetic deletions. 10–20%, being determined by the age when infected and
by the variable degree of immunity obtained from im-
EPIDEMIOLOGY mune sows. Enzootic TGE in suckling or recently weaned
pigs can be difficult to diagnose and must be differenti-
The epizootiology of TGE has been reviewed (Haelter- ated from other types of enzootic diarrheal problems
man 1962; Ferris 1973; Toma et al. 1978; Wood 1979; commonly occurring in young pigs, such as rotaviral di-
Pritchard 1982, 1987). On a herd basis, two epizootio- arrhea and colibacillosis. Enzootic TGE will persist in the
logic forms of TGE can be described: epizootic and en- herd as long as susceptible or partially immune swine are
zootic. In addition, infections with the TGEV variant exposed to TGEV.
PRCV present a different pattern and greatly complicate The recrudescence or reintroduction of TGEV may
seroprevalence studies of the epizootiology of TGEV occur in herds that contain immune sows, resulting in
(Pensaert 1989; Pensaert and Cox 1989). discrete episodes of disease (Pritchard 1987). This situa-
tion commonly occurs in herds in the concentrated
Epizootic TGE swine-rearing areas of the United States or other coun-
Epizootic TGE refers to the occurrence of TGE in a herd tries. Each winter, herds often become reinfected, and
where most, if not all, of the animals are susceptible. the disease is especially seen in growing and finishing
When TGEV is introduced in such a herd, the disease swine. Such animals are susceptible, since the herd infec-
usually spreads rapidly to swine of all ages, especially tion from the previous winter has usually terminated
during the winter. Some degree of inappetence, vomi- during the intervening summer and autumn. If the dis-
tion, or diarrhea will occur in most animals. ease enters the farrowing house, the disease in suckling
Suckling pigs become very sick, dehydrating rapidly; or weaned pigs will resemble that described above since
mortality is very high in pigs under 2–3 weeks of age but the sows will usually be immune. It is unclear whether
gradually decreases in older pigs. Lactating sows often the source of virus in these circumstances comes from re-
become sick, developing anorexia and agalactia, which activation of virus shedding in carrier swine or reintro-
further contribute to piglet mortality. The history and se- duction of virus into the herd from an external source.
vere clinical signs aid in diagnosis of epizootic TGE in
the United States, since other similar diseases have not Porcine Respiratory Coronavirus
been reported. However, in Europe, PED has similar clin- Evidence that led to the discovery of PRCV was obtained
ical signs (Pensaert and DeBouck 1978), and the pres- from serologic surveys of slaughterhouse sows or swine
ence of PRCV appears to have greatly reduced the inci- tested for international trade. In 1984, this led to the first
dence and severity of epizootic TGE (Pensaert and Cox isolation of PRCV, a nonenteropathogenic coronavirus,
1989; Brown and Paton 1991; Laude et al. 1993; Pensaert in Belgium (Pensaert et al. 1986). In 1989, two herds in
et al. 1993). the United States unexpectedly were seropositive for
TGEV antibodies although these herds had not been vac-
Enzootic TGE cinated for TGE nor had they experienced clinical disease
Enzootic TGE refers to a persistence of the virus and dis- (Hill et al. 1990; Wesley et al. 1990b). PRCV is a TGEV
ease in a herd, occurring as a result of a continual or fre- variant that infects epithelial cells of the respiratory tract
quent influx of susceptible swine that, when infected, and alveolar macrophages (Pensaert 1989; Pensaert and
tend to perpetuate the disease. Enzootic TGE is limited Cox 1989; Paul et al. 1994). After experimental challenge,
to seropositive herds that have frequent farrowings PRCV infects only a few unidentified cells in the small in-
(Stepanek et al. 1979) or have frequent additions or com- testine, and thus, virus shedding in feces is limited or not
mingling of susceptible swine and represents a common detected (O’Toole et al. 1989; Cox et al. 1990a, b; Van
sequel to a primary outbreak in large breeding herds. In Cott et al. 1993, 1994; Brim et al. 1994, 1995). PRCV-in-
these situations, TGEV spreads slowly among adult fected pigs produce antibodies that neutralize TGEV.
swine, particularly herd replacements (Morin et al. 1978, Swine density, distances between farms, and season
1983; Pritchard 1987). Females saved for breeding will be of the year influence the epizootiology of PRCV. PRCV
immune and will transfer via their colostrum and milk a infects pigs of all ages by airborne transmission or by
variable degree of passive immunity to their progeny contact, and in areas with high swine density, the virus
during the suckling period. Sows usually are not sick. In can spread to pigs on neighboring farms that are several
these herds, rather mild TGE viral diarrhea will be seen kilometers away (Pensaert and Cox 1989). In addition,
primarily in pigs from the age of about 6 days until about the risk of a farm becoming infected increases as the size
2 weeks after weaning. The pig is clinically affected when of a neighboring herd increases (Henningsen et al.
viral exposure exceeds the pig’s passive immunity, and 1989). PRCV infections are subclinical and the virus has
the age when this occurs is related to, or reflects, the spread rapidly and extensively in pigs of Western Euro-
management system used in the herd and the degree of pean countries (Brown and Cartwright 1986; Jestin et al.
300 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

1987; Lange et al. 1988; Henningsen et al. 1989; Pensaert rather labile when exposed to warm temperature or to
1989; Van Nieuwstadt and Pol 1989; Laude et al. 1993; sunlight. This would allow a greater opportunity for the
Martin et al. 1994) and even into European countries virus to be transmitted in a viable state between herds in
that were previously free of TGEV (Pensaert 1989). A winter, particularly on inanimate objects, such as during
similar rapid and extensive spread of PRCV has not been transport of feed or animals. The observation has been
detected in the United States (USDA APHIS VS 1992); made that reduced or fluctuating ambient temperatures
however, a limited serologic survey in 1995 suggested markedly predispose feeder pigs to clinical manifesta-
that many asymptomatic swine herds in Iowa were tions of TGE (Shimizu et al. 1978; Shimizu and Shimizu
seropositive for PRCV (Wesley et al. 1997). 1979a), thus contributing to transmission in winter be-
PRCV has become enzootic in many European swine cause of the diarrheic state.
herds (Pensaert 1989; Lanza et al. 1993; Laude et al. What constitutes the reservoir of TGEV between sea-
1993; Pensaert et al. 1993). The virus circulates in the sonal epizootics? Haelterman (1962) proposed at least
herd infecting pigs before the age of 20–26 weeks, after three reservoirs: (1) associated pig farms in which the
passively acquired maternal antibodies have declined virus spreads subclinically; (2) hosts other than swine;
(Pensaert et al. 1993). Introduction of pigs into fattening and (3) carrier pigs. The most probable explanation for
units and commingling of PRCV-negative and PRCV- maintenance of the disease is that it exists in the enzoot-
positive pigs from diverse sources resulted in serocon- ic form in feeder-pig operations (Morin et al. 1978) or in
version to PRCV in pigs shortly after introduction into herds that are on a continuous farrowing program. These
most units (Van Reeth and Pensaert 1994a). Susceptible situations could constitute foci for maintenance of the
pigs experimentally infected with PRCV were shown to disease during the warmer months and for dissemina-
shed virus from nasal secretions for a period of less than tion during the winter months. This hypothesis is sup-
2 weeks (Onno et al. 1989; Wesley et al. 1990b; Bourgueil ported by the finding that TGEV infection can spread
et al. 1992; Van Cott et al. 1993; Brim et al. 1994). There rather slowly through a group of growing swine under
is no evidence for the fecal-oral transmission of PRCV. certain conditions, such as during the summer months
Pensaert et al. (1993) have shown that pigs on closed (Maes and Haelterman 1979).
breeding farms become infected shortly after weaning There is also evidence for the existence of TGEV in
even in the presence of maternal antibodies, and thus, nonporcine hosts. Cats, dogs, and foxes have been sug-
PRCV persists in the herd by regularly infecting newly gested as possible carriers of TGEV from one herd to an-
weaned pigs. PRCV can persist in the herd throughout other since they can shed virus in their feces for variable
the entire year or it can disappear temporarily from a periods (Haelterman 1962; McClurkin et al. 1970). Al-
herd mainly in the spring and summer months and reap- though cats and dogs fed TGEV showed no clinical signs
pear in the nursery and fattening units during the colder (except after repeated passage in dogs; Klemm and Ristic
months of the year (Pensaert et al. 1993). Thus, waves of 1976) and seroconverted to TGEV, virus excreted by dogs
infection, without clinical disease, coincide with the fog- was shown to be infectious for pigs (Haelterman 1962;
gy and rainy season in Europe (Laude et al. 1993). Reynolds and Garwes 1979).
The seroprevalence of TGEV infections among Euro- Massive concentrations of starlings (Sturnus vulgaris)
pean swine was examined by blocking ELISA following in winter in feeding areas of swine may provide a method
the widespread dissemination of PRCV infections by which TGEV can be mechanically carried from one
(Brown and Paton 1991; Lanza et al. 1993; Pensaert et al. farm to another. Pilchard (1965) reported that TGEV was
1993). The investigators reported no or low (0.6%) detected in the droppings of starlings for as long as 32
TGEV-positive serum samples from breeding sows in hours after they were fed TGEV. Houseflies (Musca do-
Spain (Lanza et al. 1993) and from pigs in Great Britain mestica) have also been proposed as possible mechanical
(Brown and Paton 1991), respectively. By comparison, vectors for TGEV. TGEV antigen was detected in flies
7.6% of sera (of 160 tested) from slaughterhouse sows in within a swine herd in which TGE was enzootic, and ex-
Belgium were TGEV positive (Pensaert et al. 1993). Thus, perimentally inoculated flies excreted TGEV for 3 days
the seroprevalence of TGEV infections in Europe has de- (Gough and Jorgenson 1983).
creased coincident with the spread of PRCV. A third possibility relating to the transmission of
TGE is the length of time infected swine eliminate viable
Transmission and Reservoirs TGEV and the role of the carrier pig. Only one report
One of the significant epizootiologic features of TGE is (Lee et al. 1954) has indicated that fecal shedding under
its seasonal appearance, that is, during the winter natural conditions lasted more than the commonly re-
months, usually from the middle of November to about ported 2-week period (Pensaert et al. 1970a). As for res-
the middle of April. Several explanations have been giv- piratory shedding of TGEV, the virus has been detected
en for this seasonal occurrence. Haelterman (1962) has in nasal swabs for postexposure periods up to 11 days
suggested that this is probably due to the characteristics (Kemeny et al. 1975). However, from lung homogenates,
of the virus, since the virus is very stable when frozen but the virus was detected for postexposure periods up to
CHAPTER 24 TRANSMISSIBLE GASTROENTERITIS Saif, Wesley 301

104 days (Underdahl et al. 1975). TGEV has also been re- close contact with their affected piglets or to hormonal
covered from milk of infected sows during the acute factors that may influence susceptibility. In contrast,
phase of the disease (Kemeny et al. 1975; Kemeny and sows in the field having no contact with young infected
Woods 1977) and following intramammary infusion or pigs usually have rather mild clinical signs or none.
injection of lactating sows with live TGEV (Saif and Bohl The incubation period is short, usually 18 hours to 3
1983). In the latter study, demonstration of TGEV anti- days. Infection generally spreads rapidly through the en-
gen in mammary gland tissue indicates the virus may tire group of swine so that in 2–3 days most animals are
replicate in the mammary glands of lactating sows, affected, but this is more likely to occur in winter than
which may contribute to agalactia in sows. Transmission summer (Maes and Haelterman 1979).
of the virus via milk to suckling piglets, as suggested in
the same study, may account, in part, for the rapid Enzootic TGE
spread of the infection among a litter. Enzootic TGE is most likely to occur in large herds that
Although TGEV has been detected in the intestinal have frequent farrowings, as discussed in the section on
and respiratory tracts for periods of up to 104 days epidemiology. The clinical signs shown by infected pigs
postinfection, it is unknown whether such virus can be will usually be similar but less severe than those seen in
eliminated from the body in a viable state that will result susceptible pigs of the same age. Death loss is usually
in new infections. Addition of sentinel pigs to a herd at low, especially if pigs can be kept warm. The clinical
3, 4, and 5 months after a previous TGE outbreak result- signs in suckling pigs can resemble those of “white
ed in no infections in the introduced pigs, as determined scours,” which is most commonly caused by rotavirus
by serologic tests (Derbyshire et al. 1969). (Bohl et al. 1978). In some herds, depending on manage-
Nasal shedding of PRCV in experimentally infected ment, enzootic TGE is manifested mainly in weaned pigs
pigs occurs through postexposure day 10 (Onno et al. and may be confused with Escherichia coli, coccidia, or ro-
1989; Wesley et al. 1990b). However, it is unknown for tavirus infections (Morin et al. 1983; Pritchard 1987).
how long pigs recovered from PRCV infections in the
field remain infectious. The possible role of the long- Porcine Respiratory Coronavirus
term carrier hog in transmitting TGEV or PRCV is un- The severity of clinical signs and the degree of patho-
known. genicity appear to be PRCV strain dependent, and it is
speculated that these differences might correlate with
CLINICAL SIGNS slightly different genetic deletions. In addition, infection
of swine with PRCV and other respiratory viruses, espe-
Epizootic TGE cially in association with porcine reproductive and respi-
The typical clinical signs of TGE in piglets are transient ratory syndrome virus (PRRSV), can alter the severity of
vomiting, accompanied or rapidly followed by a watery disease and clinical signs. Ultimately, the manifestation
and usually yellowish diarrhea, rapid loss of weight, de- of respiratory disease is dependent on a number of vari-
hydration, and high morbidity and mortality in pigs un- ables, including environmental and seasonal factors,
der 2 weeks of age. Diarrhea in young pigs is usually pro- management practices, virus load or dose, and presence
fuse, and feces will often contain small curds of of other bacterial or viral pathogens in the herd (Van
undigested milk. The odor of the feces is very offensive. Reeth and Pensaert 1994a, b).
Severity of the clinical signs, duration of the disease, and Many European and American PRCV strains cause
mortality are inversely related to the age of the pig. Most subclinical infections, and by histological examination, a
pigs under 7 days of age will die in 2–7 days after first mild interstitial pneumonia may be present (Van Reeth
showing clinical signs. Most suckling pigs over 3 weeks and Pensaert 1992; Laude et al. 1993; Enjuanes and Van
of age will survive but are likely to remain unthrifty for a der Zeijst 1995). Experimentally infected feeder pigs had
time. only a transient weight loss, whereas 4- and 6-day-old
Clinical signs of TGE in growing and finishing swine pigs infected with PRCV showed only a reduced rate of
and in sows are usually limited to inappetence and diar- weight gain in comparison with control pigs (Vannier
rhea for 1 or a few days, with vomiting observed in an oc- 1990; Lanza et al. 1992; Van Reeth et al. 1996; Wesley
casional animal. The very few deaths observed are prob- and Woods 1996). In one study, when American PRCV
ably due to complicating factors such as stress or strains were compared in pigs under the same condi-
concurrent infections, which frequently occur after tions, differences were observed in pathogenicity among
weaning. However, Bachmann et al. (1972) reported a strains (Halbur et al. 1994). Two PRCV isolates, AR310
mortality between 25% and 30% in a group of 2- to 6- and LEPP with the same S gene deletion, induced signs of
month-old swine in Germany. Some lactating sows be- moderate respiratory disease in pigs from 4 to 10 days
come very sick, with an elevated temperature, agalactia, postinfection, while a third American strain (1894) pro-
vomiting, inappetence, and diarrhea. These severe signs duced only mild respiratory disease in pigs. However, at
may be due to a high degree of exposure to the virus from a lower dose, even the AR310 strain was asymptomatic in
302 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

gnotobiotic pigs (Halbur et al. 1993). A Canadian PRCV pig. Furthermore, they suggested that the presence of
strain (1Q90) isolated in Quebec caused severe pneumo- undigested lactose exerts an osmotic force which causes
nia and 60% mortality in 1-week-old pigs given 108.5 a retention of fluid in the lumen of the intestine and
TCID50 (median tissue culture infectious doses) of virus. even a withdrawal of fluid from the tissues of the body
It is noteworthy, however, that littermates of these pigs and thus contributes to diarrhea and dehydration.
exposed to PRCV by contact, presumbly at a lower dose, Additional mechanisms contributing to diarrhea in
exhibited only rapid breathing and fever (Jabrane et al. TGEV-infected pigs include altered sodium transport in
1994). Thus, clinical signs seen after experimental PRCV the jejunum, resulting in accumulation of electrolytes
exposure may be highly dose related and also influenced and water in the intestinal lumen (Butler et al. 1974), and
by the age of the pigs and inoculation techniques. The re- loss of extravascular protein (Prochazka et al. 1975). The
sults of experimental studies were also influenced by the ultimate cause of death, as suggested by Cornelius et al.
health status of the pigs and their subsequent treatment (1968), is probably dehydration and metabolic acidosis
(Vannier 1990). coupled with abnormal cardiac function resulting from
Besides a single-virus infection with PRCV, pigs can hyperkalemia.
be exposed concurrently to other respiratory viruses and A marked shortening or atrophy of the villi occurs in
agents. Often this occurs when pigs from multiple the jejunum (Fig. 24.3) and to a lesser extent in the ileum,
sources are grouped together in nurseries or grower-fin- but it is often absent in the proximal portion of the duo-
isher units (Van Reeth and Pensaert 1994a). The effects denum (Hooper and Haelterman 1966). Both virus pro-
of dual-virus inoculations have been investigated and, duction and villous atrophy were greater in newborn
generally, found to be more severe than for a single-virus pigs than in 3-week-old pigs (Moon et al. 1973; Norman
infection. In particular, respiratory signs were enhanced et al. 1973), suggesting higher susceptibility of neonates
in PRCV-infected pigs when the pigs were inoculated 2 to TGEV infection. Several mechanisms have been pro-
days later with either swine influenza virus or pseudora- posed to account for this age-dependent resistance to
bies virus (Van Reeth and Pensaert 1994b, 1996). Com- clinical disease. First, the rapidity with which infected
bined infections with PRRSV followed by PRCV also re- villous epithelial cells can be replaced by migration of ep-
sulted in a prolonged fever with respiratory disease and a ithelial cells from crypts of Lieberkühn may partially ac-
reduced rate of weight gain (Van Reeth et al. 1996). Be- count for the lower fatality rate in older than in newborn
cause many respiratory viruses and bacteria are common pigs. Moon (1978) reported that 3-week-old pigs nor-
in swine populations, these combined infections with mally replace villous enterocytes in the small intestine
PRCV may increase the occurrence and severity of respi- about three times more rapidly than newborn pigs.
ratory disease. This subject requires additional research. These newly replaced villous enterocytes are reportedly
resistant to TGEV infection (Pensaert et al. 1970b; Shep-
PATHOGENESIS herd et al. 1979), possibly due to onset of the immune re-
sponse, presence of intestinal interferon (LaBonnardiere
The pathogenesis of TGE has been reviewed (Hooper and Laude 1981), or inability of these regenerating cells
and Haelterman 1966; Moon 1978; Shepherd et al. to support virus growth. Second, TGEV accumulates and
1979). The early events have been described as follows: replicates in the apical tubulovascular system of villous
TGEV is ingested, infects the mucosa of the small intes- absorptive cells in newborn pigs; this system is lacking in
tine, and causes villous atrophy because of a rapid and pigs older than 3 weeks (Wagner et al. 1973). Third, virus
extensive loss of functional epithelial cells. dose may play a major role in infections. Witte and
Walther (1976) demonstrated that the infectious dose of
Intestinal Replication TGEV needed to infect a market hog (about 6 months
Whether by the oral or by the nasal route, the virus is old) was 104 times greater than that needed to infect a 2-
swallowed and, being able to resist the effects of a low day-old piglet.
pH and proteolytic enzymes, remains viable until it However, the severity of clinical signs due to TGE is
comes in contact with the highly susceptible villous ep- also increased when pigs are (1) fed a zinc-deficient ra-
ithelial cells of the small intestine. Infection and rapid tion (Whitenack et al. 1978); (2) anemic (Ackerman et al.
destruction or alteration in function of a high propor- 1972); (3) exposed to a low temperature or fluctuation in
tion of these cells result in a marked reduction in enzy- temperature (Shimizu et al. 1978); or (4) injected with a
matic activity in the small intestine, which disrupts di- synthetic corticosteroid, dexamethazone (Shimizu and
gestion and cellular transport of nutrients and Shimizu 1979a). In regard to the last two items, the
electrolytes, causing an acute malabsorption syndrome mechanism is thought to be due to an interference with
(Moon 1978). Hooper and Haelterman (1966) suggested the early initiation of a local cell-mediated immune re-
that the inability of infected pigs to hydrolyze lactose, sponse (Shimizu and Shimizu 1979a).
and possibly other nutrients, results in the marked dep- The failure of cell-cultured attenuated strains of
rivation of nutrients that can be so critical to the young TGEV to infect the epithelial cells located in the cranial
CHAPTER 24 TRANSMISSIBLE GASTROENTERITIS Saif, Wesley 303

A B
24.3. Villi of the jejunum from a normal pig (A) and from a TGEV-infected pig (B), as viewed
through a dissecting microscope (approximately ×10).

portion of the small intestines of pigs probably explains fected piglets (Kemeny et al. 1975).
why such strains do not cause diarrhea as severe as that In addition, studies have shown that TGEV is capable
observed with virulent strains (Frederick et al. 1976; of replicating in mammary tissue of lactating sows (Saif
Hess et al. 1977; Furuuchi et al. 1979; Pensaert 1979). and Bohl 1983) and that infected sows shed virus in milk
Furthermore, there was a reverse correlation between the (Kemeny and Woods 1977). The significance of possible
level of cell culture attenuation of TGEV and the extent mammary gland infection with TGEV under field condi-
of intestinal infection (Hess et al. 1977). tions is unclear. Whether it plays a role in the agalactia,
Cell-cultured strains of TGEV of reduced virulence in often seen in TGEV-infected sows, or rapid spread of in-
combination with a mildly virulent E. coli have been fection among piglets warrants investigation.
shown to cause a more severe disease in germfree pigs Although natural infection of the porcine fetus with
than when either organism was given alone (Underdahl TGEV has not been reported, intrafetal inoculation re-
et al. 1972). Concurrent infections with TGEV and E. coli sults in the production of villous atrophy and serocon-
or porcine rotavirus have been reported (Hornich et al. version to TGEV (Redman et al. 1978).
1977; Theil et al. 1979).
PRCV. PRCV has a tropism for cells of the respiratory
Extraintestinal Replication Sites tract. It replicates to high titer in porcine lungs and also
TGEV. Although ingestion is undoubtedly the most infects epithelial cells of the nares, trachea, bronchi,
common portal of entry for the virus, the nasal route, or bronchioles, and alveoli and alveolar macrophages (Pen-
airborne infection, may be important, but the efficiency saert et al. 1986; O’Toole et al. 1989). Viremia occurs fol-
of this route of transmission for the spread of TGEV ap- lowing primary infection, and virus spreads to parenchy-
pears much less significant than for PRCV. Macroscopic mal organs and lymph nodes.
lung lesions were observed in gnotobiotic pigs inoculat- Only a few scattered cells containing PRCV antigen
ed oronasally with TGEV, but no clinical pneumonia re- are found in the small intestine even when the virus is di-
sulted (Underdahl et al. 1975). A preliminary report rectly inoculated into the intestinal lumen. These infect-
also indicated that TGEV was present in alveolar ed cells are located in or underneath the epithelial layer
macrophages of infected neonatal pigs, suggesting a pos- of the intestinal villi and crypts, and the virus does not
sible role for these cells in lung infection. However, only spread to adjacent cells (Cox et al. 1990a, b). This limited
cell culture–adapted, but not virulent, TGEV replicated intestinal replication of PRCV explains why infectious
in cultures of alveolar macrophages in vitro (Laude et al. virus is seldom detected in feces of PRCV-infected swine
1984). Highly attenuated strains of TGEV have also been (Van Cott et al. 1993, 1994).
reported that replicate in the upper respiratory tract and Researchers have attempted to use MAB neutraliza-
lung but not in the intestine of newborn pigs, resem- tion–resistant mutants and recombinant TGEV strains
bling PRCV infections (Furuuchi et al. 1979). Moreover, to explore the molecular basis for the difference in path-
nasal shedding of TGEV was detected in infected piglets ogenicity and tissue tropism between TGEV and PRCV
(Van Cott et al. 1993) and in lactating sows exposed to in- strains (Bernard and Laude 1995; Ballesteros et al. 1997).
304 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Bernard and Laude (1995) reported that most TGEV these dimensions average about 795 µm and 110 µm, re-
(Purdue-115 strain) MAB-resistant mutants selected spectively, giving a villi-crypt ratio of about 7:1; in in-
with MABs to S protein site D exhibited a reduced en- fected piglets the corresponding figures are about 180
teropathogenicity in pigs, which was correlated with a µm and 157 µm, giving a ratio of about 1:1 (Hooper and
point mutation or small deletion in the S protein gene Haelterman 1969). Other lesions reported in experimen-
encoding the N-terminal subregion of the S protein (sim- tally challenged 8-week-old pigs include microulceration
ilar region deleted in PRCV strains). Using recombinants of the dome epithelium over Peyer’s patches, especially
generated between enteric/respiratory (PUR46) and res- in the cranial portion of the small intestine (Chu et al.
piratory (PTV) strains of TGEV, Ballesteros et al. (1997) 1982a).
concluded that a substitution in amino acid 219 of the S Scanning EM has been used to reveal the develop-
protein of the PUR46 recombinants was responsible for ment of intestinal lesions of TGE and correlates well
the loss of enteric tropism observed after inoculation of with lesions observed by light microscopy (Waxler 1972;
pigs with the PUR46 recombinants. The authors specu- Moxley and Olson 1989b). Using scanning EM, Moxley
lated that two different domains of the S protein (around and Olson (1989b) showed that the level of passive im-
amino acid 219) are required to infect intestinal epithe- munity in TGEV-infected pigs influenced not only the de-
lial cells: one domain binds to the cellular receptor, gree of villous atrophy but also its segmental distribu-
aminopeptidase N, and the other domain may be a bind- tion. Villous atrophy was minimal in pigs nursing sows
ing site for an undefined intestinal coreceptor. previously infected with virulent TGEV, compared to
pigs nursing seronegative sows or sows given live attenu-
LESIONS ated vaccines. In partially protected pigs, villous atrophy
was seen primarily in the ileum instead of the jejunum.
Gross Lesions Similar observations were noted in pigs from herds with
Gross lesions with TGE are usually confined to the gas- enzootic TGE.
trointestinal tract, with the exception of dehydration. Transmission EM of TGEV-infected epithelial cells of
The stomach is often distended with curdled milk. Con- the small intestine has revealed alterations in the mi-
gestion of the mucosa is a variable sign. Hooper and crovilli, mitochondria, endoplasmic reticulum, and oth-
Haelterman (1969) reported that about 50% of the pigs er cytoplasmic components. Virus particles, primarily in
euthanized during the first 3 days of infection had a cytoplasmic vacuoles, were observed in villous entero-
small area of hemorrhage on the diaphragmatic side of cytes and in M cells, lymphocytes, and macrophages in
the stomach at the border of the diverticulum ventricu- the dome regions of Peyer’s patches (Thake 1968; Wagn-
lus. er et al. 1973; Chu et al. 1982a).
The small intestine is distended with yellow and fre-
quently foamy fluid and usually contains flecks of cur- PRCV Lesions
dled undigested milk. The wall is thin and almost trans- In the case of PRCV, as noted earlier, villous atrophy is
parent, probably due to atrophy of the villi. not observed. However, microscopic examination of
Although lung lesions have been observed in experi- lungs from asymptomatic pigs reveals that PRCV causes
mentally infected gnotobiotic pigs (Underdahl et al. a diffuse interstitial pneumonia in a high percentage of
1975), they have not been reported in pigs naturally in- inoculated animals (O’Toole et al. 1989; Van Nieuwstadt
fected with TGEV. and Pol 1989; Cox et al. 1990a). Others reported more se-
vere respiratory lesions dependent upon the strain and
Subgross Lesions dose of PRCV used to experimentally infect pigs (Halbur
A highly significant lesion of TGE is the markedly short- et al. 1993, 1994; Jabrane et al. 1994; Paul et al. 1994).
ened villi of the jejunum and ileum, which Hooper and
Haelterman (1969) referred to as villous atrophy (Fig. DIAGNOSIS
24.3). However, this is also seen in rotavirus diarrhea but
is not usually as severe or extensive as in TGE (Bohl et al. This subject has been reviewed by Bohl (1981). Collec-
1978). Infections with some strains of E. coli and coccidia tion and preservation of appropriate clinical specimens
have also been reported to produce this lesion (Hornich are necessary for reliable diagnosis. Although villous at-
et al. 1977). However, the pathologic findings and extent rophy is a consistent lesion in severely affected pigs, it
of villous atrophy were highly variable in pigs from en- frequently occurs in other enteric infections as well (ro-
zootically infected herds (Pritchard 1987). tavirus, PED, coccidia, and sometimes E. coli). Laborato-
ry diagnosis of TGE may be accomplished by one or
Microscopic Lesions more of the following procedures: detection of viral anti-
The degree of villous atrophy can be judged in histologic gen, detection of viral nucleic acids, microscopic detec-
sections by comparing the length of the jejunal villi with tion of virus, isolation and identification of virus, or de-
the depth of the crypts of Lieberkühn. In normal piglets tection of a significant antibody response.
CHAPTER 24 TRANSMISSIBLE GASTROENTERITIS Saif, Wesley 305

Unfortunately, the serologic assays are complicated Recently an immunoperoxidase technique using
by the failure of polyclonal antibodies to differentiate be- MAB to the highly conserved N protein of TGEV has
tween PRCV and TGEV (discussed under differential been applied to the detection of TGEV (intestinal tis-
blocking ELISA). However, evaluation of clinical signs, sues) or PRCV (lungs) using formalin-fixed paraffin-em-
histologic lesions, and tissue distribution of viral antigen bedded tissues (Shoup et al. 1996). This permits the di-
may provide a presumptive diagnosis, because PRCV agnosis of TGEV/PRCV on the same tissues as used for
does not cause diarrhea or villous atrophy and replicates histopathology and allows retrospective screening of
almost exclusively in respiratory tissues (Pensaert 1989; fixed tissues for TGEV/PRCV. Although polyclonal anti-
Pensaert and Cox 1989). Thus PRCV is suspected if there bodies will not differentiate between TGEV and PRCV,
is antigen in lung tissues, seroconversion to TGEV/PRCV certain MABs have been produced that react with TGEV
by VN test, and no signs of enteric disease. but fail to recognize PRCV. These differentiating MABs
have been used in IF and immunoperoxidase tests (Gar-
Detection of Viral Antigen wes et al. 1988; Van Nieuwstadt and Pol 1989). PRCV has
The detection of TGE viral antigen in epithelial cells of been detected in respiratory tissues and nasal epithelial
the small intestine is probably the most common cells by IF and immunoperoxidase tests, but use of dif-
method for diagnosing TGE in young pigs. Either IF ferentiating MABs is necessary for confirmation, since
(Pensaert et al. 1970a) or immunoperoxidase (Becker et enteric strains of TGEV may also replicate in these tis-
al. 1974; Chu et al. 1982b; Shoup et al. 1996) techniques sues.
may be used, but the former is more common. For best A double antibody sandwich ELISA using monoclon-
results, pigs in the early stages of diarrhea are killed. Mu- al or polyclonal antibodies to TGEV as capture or sec-
cosal scrapings (Black 1971) or frozen sections from the ondary antibodies is becoming more widely used to de-
jejunum and ileum are prepared and stained by either tect TGEV antigens in cell culture, feces, and intestinal
the direct (Fig. 24.4) or indirect IF method. Mucosal contents (Bernard et al. 1986; Van Nieustadt et al. 1988;
scrapings generally yield a greater sampling of the in- Lu et al. 1991; Cornaglia et al. 1994; Lanza et al. 1995) or
testinal mucosa. Problems that may be encountered in IF PRCV antigen in nasal secretions or lung homogenates
tests include (1) lack of sensitivity or specificity of (Cornaglia et al. 1994). The ELISA tests are rapid, applic-
reagents (primary or secondary reagents used must be able for use on samples from live pigs, and suitable for
free of antibodies to other enteric organisms, particular- testing large numbers of samples.
ly rotavirus); (2) failure to obtain specimens early after
onset of diarrhea before the loss of infected cells (piglets Detection of Viral Nucleic Acids
must be euthanized to obtain specimens); and (3) cross- Recently, nucleic acid hybridization probes have been de-
reactions with FIPV, CCV, and PRCV. However, replica- veloped to detect TGEV genome sequences in fecal sam-
tion of PRCV in villous enterocytes is uncommon, and IF ples, infected tissues, or infected cell cultures (Shockley
or immunoperoxidase staining of villous enterocytes in et al. 1987; Benfield et al. 1991). Moreover, nucleic acid
conjunction with diarrhea almost certainly indicates probes derived from the 5′ end of the TGEV peplomer
TGEV. gene can distinguish between PRCV and TGEV. In a hy-
bridization assay, these probes selectively differentiated
enteric TGEV isolates from the United States, Japan, and
England, including live attenuated TGEV vaccine strains
from U.S. isolates of PRCV, FIPV, and CCV (Bae et al.
1990; Wesley et al. 1991a). RT-PCR and nonradioactive
cDNA probes have also been used to differentiate TGEV
and PRCV isolates (Vaughn et al. 1994, 1996).

Electron Microscopy
TGEV has been demonstrated in the intestinal contents
and feces of infected pigs by negative-contrast transmis-
sion EM (Fig. 24.5) (Saif et al. 1977). In laboratories hav-
ing access to an electron microscope and personnel
trained in virus recognition by EM, this procedure has
had increasing use. Furthermore, immune EM (IEM) has
advantages over conventional EM techniques in that it is
24.4. Immunofluorescing cells from a TGEV-infected more sensitive for detecting TGEV and can provide sero-
pig. A compression smear was made from a mucosal logic identification of the virus from either clinical spec-
scraping of the jejunum and stained by the direct imens or cell culture harvests. In addition, use of IEM en-
fluorescent antibody test (×350). ables one to more readily differentiate TGEV from
306 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

common enveloped membranous debris and to concur- upon primary isolation of field strains; additional pas-
rently detect the presence of other enteric viruses (Saif et sages may be required before CPE is evident. A charac-
al. 1977). IEM is at least as sensitive as IF for detection of teristic type of CPE usually seen in ST and porcine thy-
TGEV. IEM is also applicable for detection of PRCV roid cells consists of greatly enlarged, rounded, or
shedding in nasal secretions (L. J. Saif, unpublished— elongated cells that have a balloonlike appearance (Ke-
1990). However, this method cannot distinguish be- meny 1978). The ST cell line has been used for detecting
tween TGEV and PRCV unless MABs are used, although field strains of TGEV by CPE, plaques, or IF (Kemeny
shedding of large numbers of PRCV in feces would not 1978; Bohl 1979). For detecting viral CPE or plaques, the
be expected (Van Cott et al. 1993, 1994). sensitivity of ST cells can be further enhanced by adding
pancreatin or trypsin to cell culture media (Bohl 1979;
Isolation and Identification of Virus Woods 1982) and using older cells (Stark et al. 1975).
Oral exposure of young pigs is probably the most sensi- Pocock and Garwes (1975) reported that TGEV replicat-
tive method for isolating or detecting TGEV (Dulac et al. ed at highest titer in a slightly acidic medium in sec-
1977). However, this procedure is very expensive; conse- ondary pig thyroid cells.
quently, cell cultures are more frequently used. Pig kidney and particularly ST cells have been the
Primary and secondary pig kidney cells (Bohl and Ku- cells of choice for isolation of PRCV, but PRCV also
magai 1965) or pig kidney cell lines (Laude et al. 1981), grows in a continuous cat fetus cell line (Laude et al.
primary porcine salivary gland cells (Stepanek et al. 1993). Nasal swab fluids or lung tissue homogenates
1971), porcine thyroid cells (Witte 1971), and the Mc- from affected swine are used for isolation of PRCV. The
Clurkin swine testicle (ST) cell line (McClurkin and Nor- CPE resembles that produced by TGEV strains, with syn-
man 1966) have been successfully used for isolating cytia formation frequently observed for PRCV (Pensaert
TGEV from feces or gut contents of infected pigs. The 1989; Pensaert and Cox 1989).
virus also replicates in organ cultures from pig esopha- Identification of cell culture virus can be done by VN,
gus, ileum, cecum, and colon (Rubenstein et al. 1970). IF, or IEM using specific TGEV antiserum. However,
However, parvovirus contamination of some batches of MABs specific for TGEV are required to confirm TGEV
cells prepared from porcine thyroid glands may be a dis- and exclude PRCV (Garwes et al. 1988; Laude et al.
advantage to the use of these cells (Dulac et al. 1977). 1988), or isolates can be differentiated using RT-PCR or
Distinct cytopathogenic effect (CPE) may be negligible specific cDNA probes (Laude et al. 1993; Enjuanes and
Van der Zeijst 1995). Confusion with cross-reacting CCV
and FIPV should not occur, since these viruses do not
replicate in ST or secondary pig thyroid cells (Reynolds
et al. 1980).

Serologic Diagnosis
The detection of TGEV antibodies can assist in diagnosis
and control in several different ways. However, the sero-
logic diagnosis of TGEV is complicated by the finding
that both TGEV and PRCV induce neutralizing antibod-
ies that are qualitatively and quantitatively similar (Pen-
saert 1989; Pensaert and Cox 1989). A blocking ELISA
test (described later) is necessary to differentiate these
antibodies. A rise in antibody titer between acute and
convalescent serum samples provides retrospective evi-
dence for epizootic TGE or infection with PRCV. Howev-
er, the history of the herd with respect to disease and
serologic status is needed to help interpret serologic find-
ings. To determine if enzootic TGE or PRCV is a problem
in a herd, serum samples from 2- to 6-month-old swine
can be tested for antibodies. At this age, passively ac-
quired antibodies should be absent (Derbyshire et al.
1969); thus, positive results suggest enzootic TGEV or
PRCV. Serologic tests can also be used to monitor the
TGEV or PRCV infection status of a herd. The entrance
24.5. Typical virus-antibody aggregates observed by of only serologically negative swine will also help main-
IEM of TGEV and gnotobiotic pig anti-TGEV serum. tain a herd free of TGEV and PRCV. Neutralizing anti-
Bar = 100 nm. bodies to TGEV can be detected in serum as early as 7–8
CHAPTER 24 TRANSMISSIBLE GASTROENTERITIS Saif, Wesley 307

days after infection and may persist for at least 18 formation required for TGEV seropositive animals. Cur-
months: little is known about the persistence of neutral- rently, the reliability of commercial ELISAs for differen-
izing antibodies to PRCV within a herd (Cartwright tiating U.S. strains of PRCV and TGEV is unclear.
1968; Vannier et al. 1982).
TGEV antibodies have been detected by several dif- TREATMENT
ferent serologic tests. The VN test has been the most
widely used, using cell culture–adapted viruses in cell Antiviral agents have not yet been developed for the spe-
culture systems by a variety of procedures: inhibition of cific treatment of TGE. Some inhibition of TGEV repli-
CPE in microtiter plates (Toma and Benet 1976) and cation in cell culture has been reported for the antiviral
plaque reduction assays (Bohl and Kumagai 1965; compounds amantadine (Dimitrov 1982) and isathia-
Thomas and Dulac 1976) are the most common. An IF zone (Potopalsky et al. 1983). Although high levels of
test was developed (Benfield et al. 1978) but was less sen- type 1 interferon were detected in the intestine in pigs in
sitive and reliable than the VN test (Hortig et al. 1980). the early phase of TGEV infection, the role of this inter-
Very sensitive passive hemagglutination tests (Labadie et feron in the recovery or pathogenesis of TGE was unde-
al. 1977; Shimizu and Shimizu 1977) and ELISA tests termined (LaBonnardiere and Laude 1981). Recent stud-
(Nelson and Kelling 1984; Paul et al. 1986; Hohdatsu et ies suggest that interferon may activate natural killer
al. 1987; Garwes et al. 1988; Bernard et al. 1989; Calle- cells in newborn pigs, thereby contributing a degree of
baut et al. 1989; Van Nieuwstadt et al. 1989; Berthon et resistance to challenge with TGEV (Lesnick and Der-
al. 1990) have been described, but both require concen- byshire 1988; Loewen and Derbyshire 1988). In addi-
trated purified virus for sensitizing red blood cells or tion, during a field outbreak of TGE, 1- to 12-day-old
coating ELISA plates, respectively. Other serologic tests piglets treated orally for 4 days with 1–20 IU of human
include an indirect immunoperoxidase test adapted to interferon-α had significantly greater survival rates than
detect immunoglobulin (Ig) class-specific antibodies placebo-treated piglets (Cummins et al. 1995). However,
(Kodama et al. 1981) and radioimmunoprecipitation no increased survival was seen in piglets given human in-
tests (Kodama et al. 1980). Complement-fixing antibod- terferon-α shortly after birth. Whether such therapy
ies could not be demonstrated in convalescing swine could be cost-effective for treatment of TGEV-infected
(Dulac et al. 1977). piglets was not assessed.
The only treatment for TGE presently available is to
Blocking ELISA Test for Serologic alleviate starvation, dehydration, and acidosis. Parenter-
Differentiation of PRCV and TGEV al treatment with fluids, electrolytes, and nutrients
Studies using MABs to TGEV have shown that certain would be effective in treating young pigs, but not practi-
antigenic sites on TGEV are not present on the spike pro- cal under farm conditions. Oral therapy with balanced
tein of PRCV (Laude et al. 1988; Callebaut et al. 1989; electrolyte and glucose solutions is contraindicated in
Sanchez et al. 1990; Simkins et al. 1992, 1993). Thus, young pigs (Moon 1978). The following measures are
some antigenic determinants on the spike protein of suggested: provide a warm (preferably above 32˚C),
TGEV are absent because of the deletion in the S protein draft-free, and dry environment and make water or nu-
of PRCV. Differences in the capacity of TGEV MABs to trient solution freely accessible to the thirsty TGEV-in-
bind to PRCV serve as the basis of serologic tests to de- fected pigs. Such measures will tend to reduce mortality
termine if a swine herd is infected with TGEV or PRCV in pigs that are infected at more than 3–4 days of age.
(Garwes et al. 1988; Bernard et al. 1989; Callebaut et al. Antibacterial therapy might be beneficial in 2- to 5-week-
1989; Van Nieuwstadt and Boonstra 1992; Simkins et al. old pigs, especially if there is a concurrent infection with
1993). In the blocking ELISA, TGEV antigen is reacted pathogenic strains of E. coli. Cross-suckling or putting in-
with either TGEV or PRCV antiserum followed by the fected or susceptible litters onto TGE-immune sows was
distinguishing MAB. TGEV antiserum contains compet- found useful in various field outbreaks (Stepanek et al.
ing antibody that blocks the binding of the MAB, where- 1979; Pritchard 1982).
as the PRCV antiserum does not block and allows the
MAB to bind. Thus, a negative (no blocking) reaction in PREVENTION
ELISA and a positive result in the VN test are evidence of
a PRCV infection. The test should only be evaluated on a Management
herd basis because some pigs with low TGEV or PRCV PREVENTING ENTRANCE OF TGEV INTO A HERD.
antibody titers, as occurs early in the infection process Swine in the incubative stage of the disease or those in
(7–14 days) or after infection with some TGEV strains, the viral shedding or carrier state can provide a source of
may go undiagnosed (Callebaut et al. 1989; Van Nieuw- entrance of TGEV into a herd. Some precautions to help
stadt and Boonstra 1992; Simkins et al. 1993). Presently, prevent these possibilities are to introduce swine that
to export TGE-free swine from countries where PRCV in- originate from herds known to be free of TGE, are sero-
fections occur, only this test provides the differential in- logically negative, and/or have been placed in isolation
308 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

on the farm for 2–4 weeks before being added to the herd in attempting to control or terminate an enzootic TGE
proper. A frequent question is, How soon after a TGE herd problem. First, pregnant seropositive sows can be
outbreak can pigs be moved to another herd without vaccinated intramuscularly (IM) or intramammarily late
spreading the disease? A practical answer to this ques- in gestation or shortly after farrowing with live attenuat-
tion is that a period of 4 weeks should elapse from the ed TGEV to boost immunity. Although only limited in-
last sign of disease before introducing such animals into formation is available, this procedure should boost milk
a “clean” herd. antibody levels (Saif and Bohl 1983), providing longer
Starlings have been incriminated as a means by passive immunity to suckling pigs (Leopoldt and Meyer
which the disease is spread between herds in winter 1978; Stepanek et al. 1979; Lutter et al. 1982). Although
months, owing largely to their tendencies to gather in this procedure may only delay the onset of TGE in ex-
large flocks and feed around swine. Cats, dogs, or foxes posed pigs, it can be beneficial in reducing mortality.
might play a role in spreading TGE between herds under Second, alterations in management can be made to
certain situations (see the section on epidemiology). break the cycle of infection by eliminating reservoirs of
Feces from TGEV-infected swine can be carried on susceptible pigs in a unit: prevent the continual influx of
boots, shoes, clothing, truck beds, feeds, etc., and can be susceptible animals into the herd (e.g., by temporarily al-
a source of infection to other herds. Especially in winter tering the farrowing schedule); temporarily utilize other
this is probably an important means by which TGEV is facilities; and have smaller farrowing and nursing units,
transmitted during the transport of livestock and feed. to more nearly achieve an all-in/all-out management sys-
Consequently, precautions should be taken to minimize tem.
such occurrences. If visitors, especially animal and feed
truckers, have direct contact with swine during the win- Immunoprophylaxis
ter months, it is well to provide them with clean ACTIVE IMMUNITY. The duration of active immuni-
footwear. ty in swine after oral infection with virulent TGEV has
not been well characterized. Intestinal infection of
AFTER ONSET OF TGE. When TGE has occurred on breeding-age swine results in detectable serum antibod-
a farm and pregnant animals have not yet been exposed, ies that persist for at least 6 months to possibly several
two procedures may minimize losses of newborn pigs. years (Stepanek et al. 1979). However, the serum anti-
(1) If the animals are due to farrow more than 2 weeks body titer, although providing serologic evidence of
hence, purposely expose them to virulent virus, such as TGE, provides little indication of the degree of active im-
the minced intestines of infected pigs, so they will be im- munity. Swine that have recovered from TGE are im-
mune at farrowing time. (2) If the animals will farrow in mune to subsequent short-term challenge, presumably
less than 2 weeks, attempt to provide facilities and man- due to local immunity within the intestinal mucosa (Van
agement procedures so they will not be exposed to TGEV Cott et al. 1994; Saif et al. 1994; Brim et al. 1995). The
until at least 3 weeks postfarrowing. To minimize deaths, age and immune status of the animal at initial infection
provide young pigs with a warm, dry, draft-free environ- and the severity of the challenge may greatly influence
ment and access to water, nutrient solution, or milk re- the completeness and duration of this active immunity.
placer (see the section on treatment). The mechanism of active immunity in the gut proba-
Some success has been achieved in elimination of bly relates to stimulation of the secretory IgA (sIgA) im-
TGEV from epizootically infected closed breeder herds mune system with production of intestinal sIgA anti-
without depopulation by the following procedures (Har- bodies by lymphoid cells within the lamina propria (Van
ris et al. 1987): (1) Bring in all breeding stock replace- Cott et al. 1993, 1994; Saif et al. 1994). IgA TGEV anti-
ments for the next 4–6 months. (2) In the face of an out- bodies and antibody-secreting cells (ASCs) have been de-
break, feed TGEV-infected minced intestines tected in the intestine and serum of pigs after oral, but
simultaneously to all pigs in the herd (including replace- not parenteral, inoculation with TGEV (Kodama et al.
ment stock) to eliminate susceptible hosts, thereby 1980; Sprino and Ristic 1982; Van Cott et al. 1993, 1994;
shortening the time the disease progresses through the Saif et al. 1994). Kodama et al. (1980) proposed that de-
herd and ensuring more uniform exposure levels in all tection of IgA antibody in the serum, presumably in-
pigs. (3) Maintain strict all-in/all-out production in far- testinally derived, might serve as an indicator of active
rowing and nursery units. (4) Add sentinel seronegative immunity to TGE. In another study, oral inoculation of
pigs about 2 months after clinical signs of TGE disappear gnotobiotic pigs with TGEV resulted in development of
and monitor these pigs for seroconversion to TGEV. Po- both serum and intestinal TGEV-neutralizing antibodies
tential hazards associated with feed-back control of TGE detectable from 5 to at least 35 days postexposure (DPE)
include possible spread of other pathogens to pregnant (Saif 1976). IgM (5–15 DPE) and IgA immunocytes
sows and throughout the herd. (which remained predominant from 7 to 35 DPE) were
detected in the intestinal lamina propria of the TGEV-in-
ENZOOTIC TGE. Two approaches can be considered fected gnotobiotic pigs. More recently, an enzyme-linked
CHAPTER 24 TRANSMISSIBLE GASTROENTERITIS Saif, Wesley 309

immunospot (ELISPOT) technique was used to investi- nant TGEV vaccines.


gate the kinetics of IgA and IgG TGEV antibody produc- Because lymphocyte cytotoxicity was absent in new-
tion by the pig’s systemic and local gut associated lym- born piglets and decreased in parturient sows, it was pro-
phoid tissues (GALT). High numbers of IgA ASCs were posed that a lack of K and NK cell activity against TGEV-
induced in GALT only by virulent TGEV: live attenuated infected cells may correlate with the increased
(vaccine) TGEV or PRCV strains induced significantly susceptibility of newborn piglets and parturient sows to
fewer IgA ASCs (Van Cott et al. 1993, 1994; Saif et al. TGEV infection (Cepica and Derbyshire 1984). Thus,
1994; Berthon et al. 1990). These and other studies CMI may play a role in either recovery from TGEV infec-
(Stone et al. 1982) indicate that the pig is immunocom- tion or resistance to reinfection via the rapid elimination
petent at birth in regard to humoral and mucosal anti- of TGEV-infected epithelial cells by any one or all of a
body production, but in the intestine, additional matu- combination of SCMC, ADCMC, or sensitized T lym-
rational time may be required for antibody responses to phocyte-mediated cytotoxicity.
reach adult levels.
Besides local antibody-mediated immunity, cell-me- VACCINATION OF NEONATAL OR WEANED PIGS.
diated immune responses may also be important in ac- TGEV Vaccines. Neonatal pigs have been orally vac-
tive immunity against TGEV infections. A number of cinated with attenuated TGEV in an attempt to induce
tests have been used to demonstrate cell-mediated im- rapid protection via either interference or local immuni-
munity (CMI) to TGEV, including macrophage migra- ty. No early interference has been demonstrated, and
tion inhibition (Frederick and Bohl 1976), leukocyte mi- generally more than 5 days were required before protec-
gration inhibition (Woods 1977; Liou 1982), direct tion due to active immunity could be induced (Pensaert
lymphocyte cytotoxicity (Shimizu and Shimizu 1979b), 1979). One study reported a slightly earlier onset of pro-
lymphocyte proliferation (Shimizu and Shimizu 1979c; tection, by 3–4 DPI, after maintaining vaccinated pigs at
Welch et al. 1988; Brim et al. 1994, 1995; Anton et al. a lowered temperature (18–20˚C) to enhance replication
1995, 1996), spontaneous cell-mediated cytotoxicity of the attenuated virus (Furuuchi et al. 1976). Failure to
(SCMC), and antibody-dependent cell-mediated cytotox- induce an early interference phenomenon and the delay
icity (ADCMC) (Cepica and Derbyshire 1983). Only indi- required for development of active immunity make
rect evidence exists concerning the role of CMI in resis- neonatal vaccination an unlikely method of providing
tance to TGEV infection. CMI was demonstrated with immediate protection against TGEV within the critical
lymphocytes obtained from GALT of swine orally infect- first few days of life.
ed with virulent TGEV (Frederick and Bohl 1976; Active immunization of suckling or feeder pigs could
Shimizu and Shimizu 1979c; Welch et al. 1988; Brim et be important for control of enzootic infections, especial-
al. 1994, 1995); swine parenterally or oronasally inocu- ly in newly weaned pigs, in which TGEV infections may
lated with attenuated TGEV or PRCV developed CMI result in increased mortality. Live attenuated and inacti-
mainly in systemic sites (spleen or peripheral blood lym- vated TGEV vaccines have been federally licensed for oral
phocytes). Lymphoproliferative responses to TGEV per- or intraperitoneal administration, respectively, shortly
sisted within GALT, but not systemic lymphocytes, for at after birth. One limited preliminary study reported
least 110 days after oral infection of 6-month-old swine greater protection in vaccinated, compared to control,
(Shimizu and Shimizu 1979c) but for only about 14–21 seropositive suckling pigs even though serum antibody
days after infection of younger (7- to 11-day-old) pigs levels were not enhanced but were comparable in the two
(Welch et al. 1988; Brim et al. 1994). Recently, it was con- groups (Graham 1980). However, challenge in older
firmed that CD4 T-helper cells are involved in lympho- piglets usually is much more difficult to standardize, due
proliferative responses to TGEV (Anton et al. 1996). to age resistance to infection. Further studies reported
Although T-cell epitopes were identified by lymphopro- that the presence of maternal antibodies in vaccinated
liferation studies for each of the three major proteins of pigs decreased (Hess et al. 1982; Lanza et al. 1995; Sestak
TGEV, a dominant functional T-helper epitope was de- et al. 1996) or completely suppressed (Furuuchi et al.
fined on the N protein (N321) (Anton et al. 1995). The N321 1978) active antibody production following oral admin-
peptide-induced T cells collaborated in the in vitro syn- istration of live TGEV. In the latter study, conducted in
thesis of TGEV-neutralizing antibodies specific for the S suckling piglets nursing naturally infected sows (Furu-
protein. These investigators further reported that pro- uchi et al. 1978), high levels of both passive circulating
duction of high levels of TGEV-specific antibodies in vit- and intestinal antibodies in the suckling piglets probably
ro (from in vivo TGEV-primed mesenteric lymph node accounted for the complete interference with active im-
cells) required stimulation by at least two TGEV struc- munization by an attenuated strain of TGEV.
tural proteins, with maximal responses induced by na- Other approaches have been used in attempts to ac-
tive S protein rosettes in combination with recombinant tively immunize young pigs against TGEV. Woods and
N protein. Such findings have important implications for Pedersen (1979) noted 33% mortality in challenged pigs
the optimal design of TGEV subunit or other recombi- (3 of 9) vaccinated orally/intraperitoneally with two dos-
310 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

es of the antigenically related live virulent FIPV. In com- against intestinal infection (Hooper and Haelterman
parison, 100% of the challenged pigs (3 of 3) died that 1966). The concentration of IgG decreases about 30-fold
were orally vaccinated once with an attenuated SP vari- during the first week of lactation, whereas sIgA concen-
ant of TGEV. Gough et al. (1983a) reported that 10 trations decline only about 3-fold, becoming the pre-
young weaned pigs inoculated IM with two or three dos- dominant Ig in milk (Porter and Allen 1972). Cells seed-
es of an adjuvanted, soluble, undefined TGEV subunit ed from the intestine produce sIgA locally in the
(23 kDa) vaccine were protected against virus challenge. mammary tissue (Roux et al. 1977). The piglet does not
However, parenteral administration of three doses of a absorb sIgA milk antibodies, but they play an important
baculovirus-expressed recombinant S protein (contain- role in passive intestinal immunity.
ing the four major antigenic sites, A–D) in adjuvant to Mechanisms of passive immunity to TGEV infections
suckling piglets elicited neutralizing antibodies to TGEV have been reviewed (Pensaert 1979; Saif and Bohl 1979a,
in serum but failed to protect pigs against TGEV chal- 1981a; Saif 1985; Saif and Theil 1990). Swine that have
lenge (Shoup et al. 1997). recovered from TGE can transmit passive immunity to
their suckling pigs (Bay et al. 1953). Suckling pigs are
PRCV. The observation that epizootic outbreaks of protected as a result of the frequent ingestion of
TGE in Europe have declined dramatically following the colostrum or milk that contains TGEV-neutralizing anti-
widespread dissemination of PRCV among European bodies. Such antibodies in the lumen of the intestine
swine prompted researchers to examine if a respiratory neutralize any ingested TGEV and thus protect the sus-
PRCV infection could induce active intestinal immunity ceptible epithelial cells of the small intestine. Haelter-
and protection against TGEV. The consensus from sever- man (1963, 1965) referred to this immunogenic mecha-
al studies was that prior infection of nursing or weaned nism as lactogenic immunity. This is accomplished
pigs with PRCV provides partial immunity against TGEV naturally when immune sows allow their piglets to suck-
challenge, evident by a reduced duration and level of le about every 2 hours. Passive protection can also be ac-
virus shedding and diarrhea in most (Cox et al. 1993; Van complished artificially by continuous feeding of anti-
Cott et al. 1994; Brim et al. 1995; Wesley and Woods serum to piglets (Haelterman 1963; Noble 1964).
1996), but not all (Van Nieuwstadt et al. 1989), pigs stud- Although presently impractical because of the expense
ied. The mechanism of this partial immunity presum- of the antiserum and the necessity of frequent adminis-
ably is related to the rapid increase in TGEV-neutralizing tration, these problems might eventually be overcome by
antibody titers (Cox et al. 1993; Wesley and Woods 1996) use of MABs in some type of slow-release delivery sys-
and numbers of IgG and IgA ASCs observed in the in- tem. A question of major importance in possible appli-
testines of PRCV-exposed pigs after TGEV challenge cation of MABs is whether they could be used in the face
(Van Cott et al. 1994; Saif et al. 1994). The latter re- of an outbreak to reduce morbidity and mortality.
searchers speculated that the migration of PRCV-specific TGEV antibodies in colostrum and milk of sows are
IgG and IgA ASCs from the bronchus-associated lym- primarily associated with IgA or IgG (Abou-Youssef and
phoid tissues (BALT) to the gut of the PRCV-exposed Ristic 1972; Bohl et al. 1972; Saif et al. 1972). TGEV milk
pigs after TGEV challenge might explain the rapid antibodies of the IgA class provide the most effective
anamnestic response and the partial protection induced. protection, but IgG antibodies were also protective if
However, after PRCV exposure of neonatal pigs, at least high titers could be maintained in milk (Bohl and Saif
6–8 days were required to develop partial immunity to 1975) or by artificial feeding of colostral IgG (Stone et al.
TGEV challenge (Wesley and Woods 1996), suggesting 1977). The greater efficacy of IgA TGEV antibodies is
that induction of active immunity might be too late to probably because (1) they occur in higher levels in milk
protect seronegative newborn piglets from epizootic (Porter and Allen 1972), (2) they are more resistant to
TGE. proteolytic enzymes (Underdown and Dorrington 1974),
and (3) they selectively bind to gut enterocytes (Nagura
PASSIVE IMMUNITY. Passive immunity is of prima- et al. 1978). TGEV antibodies in milk of the IgG class are
ry importance in providing newborn piglets with imme- produced as a result of parenteral or systemic immu-
diate protection against TGEV infection. Swine are born nization. IgA TGEV antibodies occur in the milk follow-
devoid of immunoglobulins (Igs), which they acquire af- ing intestinal infection. To explain their occurrence it
ter birth via colostrum. Colostral Igs, which consist pri- was proposed that after antigenic sensitization in the
marily of IgG, represent a serum transudate that is trans- gut, IgA immunocytes migrate to the mammary gland,
ferred from the dam across the piglet’s intestinal where they localize and secrete IgA antibodies into the
epithelium to its circulation, thus providing the neonate colostrum and milk (Saif and Bohl 1979a, 1981a; Saif
with the same complement of serum antibodies as in the 1985; Saif and Theil 1990; Saif et al. 1994). This “gut-
dam (Porter and Allen 1972; Bourne 1973). These pas- mammary” immunologic axis, first proposed in refer-
sively acquired humoral antibodies function mainly in ence to TGEV infections in swine (Bohl et al. 1972; Saif et
protection against systemic infection but do not protect al. 1972), is an important concept in designing optimal
CHAPTER 24 TRANSMISSIBLE GASTROENTERITIS Saif, Wesley 311

vaccination procedures to provide effective lactogenic TGEV antibodies were evident, and mortality among
immunity. challenged pigs from vaccinated dams ranged from 25%
to 100% (Saif and Bohl 1979a, b; Voets et al. 1980; Mox-
TGEV VACCINATION OF THE SERONEGATIVE ley and Olson 1989a; Saif and Theil 1990).
PREGNANT DAM. A variety of viral vaccines (viru- Concerns that attenuated strains of TGEV might not
lent, attenuated, inactivated, and subunit) and routes of survive passage through the acidic environment of the
administration (oral, intranasal, intramuscular, subcuta- stomach prompted studies using lyophilized attenuated
neous, and intramammary) (Bohl et al. 1975; Kaji and virus in enteric-coated gelatin capsules (Hess et al. 1978;
Shimizu 1978; Pensaert 1979; Saif and Bohl 1979a; Voets Voets et al. 1980). Hess et al. (1978), using the high-
et al. 1980; Moxley and Olson 1989a; Saif and Theil titered B1 strain of TGEV (300 cell culture passages), re-
1990; Saif et al. 1994) have been tested for induction of ported high levels of IgA TGEV antibodies in milk and
lactogenic immunity. Oral administration of live viru- only 10% piglet mortality. Voets et al. (1980) used the
lent virus to pregnant sows generally gave the highest lev- high-passaged Purdue strain and found that six of nine
el of immunity, resulting in protective immunity for the sows failed to seroconvert after oral vaccination; even the
sow and consistently producing high titers of persisting three sows that seroconverted had a 44% piglet mortality
IgA TGEV antibodies in milk associated with protective rate. Fichtner et al. (1982) noted 30% mortality in chal-
lactogenic immunity for suckling pigs. lenged piglets after feeding attenuated Riems virus to
There are presently several federally licensed TGEV their dams for 10 days during gestation. In further efforts
vaccines. All contain inactivated or live attenuated TGEV to ensure that vaccine virus reached the small intestine,
and are approved for use in pregnant or neonatal swine. two studies used direct inoculation of attenuated viruses
These vaccines and their efficacy will be considered in into the intestinal lumen. Again, protection was poor
the following sections according to their respective (62% mortality) (Voets et al. 1980) in challenged piglets
routes of administration. Many variables complicate the from dams given a single intralumenal inoculation of at-
evaluation of both experimental and commercial TGEV tenuated Purdue TGEV during gestation, whereas
vaccines, often resulting in conflicting data. These in- greater protection (10% mortality) was noted in chal-
clude the challenge dose and strain of TGEV; the age of lenged piglets after their dams received intralumenal in-
the pig at challenge; environmental conditions, especial- oculation of attenuated Riems TGEV for several days
ly temperature; the milking efficiency of the vaccinated during gestation (Fichtner et al. 1982).
sow; and the immune status of the dam at the time of Other researchers selected variants of high- and low-
vaccination. passaged TGEV strains resistant to low pH and prote-
olytic enzymes in vitro and used these strains as vaccines
Oral and/or Intranasal Vaccination. Based on the for passive protection studies (Aynaud et al. 1985; Chen
observation that sows infected with TGEV during gesta- 1985; Shirai et al. 1988; Bernard et al. 1989). They re-
tion could transmit immunity to their piglets, “planned” ported inconsistent results, with mortality varying from
infection of pregnant swine with virulent TGEV has been 0% to 73% among litters challenged with virulent TGEV.
used to mimic this natural immunity. This procedure is In the latter two studies, data interpretation was con-
usually accomplished by feeding virulent autogenous founded somewhat by variations in the ages of the pigs
virus to pregnant swine at least 2 weeks before farrowing. at challenge, a factor shown in other studies to dramati-
The virus may consist of minced guts from young pigs cally influence piglet survivability (Moxley and Olson
acutely infected with TGEV and is administered to sows 1989a).
with food or in frozen gelatin capsules given orally by A live attenuated SP variant TGEV grown in a persis-
means of a balling gun. tently infected porcine leukocyte cell line has been used
Oral vaccination of pregnant swine with attenuated to vaccinate pregnant swine by the oral/IN and/or intra-
TGEV would appear to be the logical route of vaccina- mammary routes (Woods 1978, 1984). Challenge of the
tion for stimulating milk IgA TGEV antibodies so as to suckling pigs resulted in mortality of 14–34%. In the lat-
duplicate the natural route of infection and induction of ter study the author reported generally high TGEV anti-
immunity. The intranasal (IN) route has been used alone body titers in both IgA and IgG fractions of 3–4 days
or together with the oral route, because attenuated postfarrowing (DPF) milk. However, three of eight sows
strains of TGEV are known to replicate in the respirato- vaccinated with SP TGEV became mildly sick after chal-
ry tract (Furuuchi et al. 1979) and upon being swallowed lenge exposure of their nursing pigs. Although diarrhea
might seed additional virus to the gut. However, results was observed in pigs nursing SP-TGEV vaccinates (48%
using attenuated strains orally and/or IN have generally morbidity), it was reportedly mild and delayed in onset
been disappointing (Saif and Bohl 1979a, 1981a; Voets et (3 DPF). The SP TGEV has been reported to be avirulent
al. 1980; Henning and Thomas 1981; Moxley and Olson for newborn pigs, replicating within the intestinal lami-
1989a). In previous studies using the high-passaged Pur- na propria but not epithelial cells (Woods et al. 1981).
due strain of TGEV orally, or orally and IM, few IgA Molecular analysis of several attenuated TGEV strains
312 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

revealed changes in mRNA 2 and 3, affecting the S pro- lactating sows resulted in IgA and IgM TGEV antibodies.
tein and nonstructural protein 3 (Register and Wesley Specific antibody activity was found not only in milk
1994). from injected glands but also in milk from noninjected
Moscari (1980b) reported induction of IgA TGEV an- glands (Bohl and Saif 1975; Saif and Bohl 1983). Protec-
tibodies in milk after oral administration of the CKp at- tion was good (14–26% mortality) in litters of intramam-
tenuated strain of TGEV; however, protection studies marily vaccinated pregnant swine, presumably because
were not described. Inconsistent results were noted in exceptionally high levels of IgG antibodies persisted in
vaccination studies using a commercial vaccine (Ambico, the milk at the time of challenge, 3 DPF (Shibley et al.
Inc.) administered twice orally (in feed) and once IM. 1973; Bohl and Saif 1975). A similar, greatly enhanced,
Whereas Welter (1980) reported 8% mortality among predominantly IgG milk antibody titer was noted in two
challenged pigs, others reported higher mortality, simi- sows vaccinated IM/IN with the high-titered (108–109.3
lar to that in piglets suckling unvaccinated sows (Saif TCID50) attenuated TO163 strain of TGEV. No mortality
and Bohl 1981b; Bohl et al. 1982; Moxley and Olson occurred in either of these litters, confirming the protec-
1989a). tive ability of IgG TGEV antibodies when present in high
The generally poor results obtained with oral or IN titers in milk (Kaji and Shimizu 1978).
vaccination of sows using attenuated TGEV strains may
be attributed to the superficial or limited replication of Heterologous Vaccines. The antigenic relationship
most attenuated strains in the sow’s intestine (Frederick between TGEV and FIPV was the basis for studies of the
et al. 1976; Hess et al. 1977). Consequently, this results in possible efficacy of FIPV as a heterologous coronavirus
little antigenic stimulation of underlying intestinal IgA vaccine in swine. Preliminary studies indicated that
plasma cells and correspondingly little IgA antibody se- some immunity (25% mortality) against TGE was con-
cretion in milk. Attempts to use a low cell culture–pas- ferred in pigs nursing two sows vaccinated during gesta-
saged TGEV to induce passive immunity led to erratic re- tion orally/IN and intramammarily with live virulent
sults in terms of both seroconversion in orally/IN FIPV. However, this FIPV was also pathogenic in new-
exposed sows and protection in piglets (Saif and Bohl born pigs (Woods and Pedersen 1979). Subsequent stud-
1979b). The dilemma remains of how to commercially ies using cell culture–adapted attenuated FIPV in sows
develop a TGEV vaccine capable of stimulating an IgA by the same routes of inoculation resulted in higher litter
response in the gut of sows but being sufficiently attenu- mortality (52%) and low TGEV antibody titers of the IgG
ated or noninfectious so as not to produce disease in class in milk (Woods 1984).
newborn pigs.
VACCINATION OF TGEV-INFECTED SWINE. Vac-
Parenteral Vaccination. Various experimental and cines have been used on two populations of pregnant
two commercial vaccines were comprised of live attenu- swine: those that have and those that have not previous-
ated virus administered IM about 6 and 2 weeks prefar- ly been naturally infected with TGEV. There are signifi-
rowing. Experimental evaluations of this vaccination cant differences in the immune responses and, conse-
regime have generally indicated reduced piglet mortality quently, piglet protection in these two groups of animals
(38–56% in vaccinates compared with 71–92% in con- following vaccination. These differences may account for
trols) but not morbidity (Bohl et al. 1975; Voets et al. some of the discrepant results seen in vaccine challenge
1980; Moxley and Olson 1989a). However, vaccination studies if previously infected swine were unknowingly
results were poor when compared with almost complete used. This possibility can only be eliminated by using a
protection (0–9% mortality) in litters of naturally infect- very sensitive test (such as plaque reduction VN) to mea-
ed sows. Henning and Thomas (1981) and Matisheck et sure TGEV antibodies and by knowing the herd history
al. (1982) reported more favorable vaccination results of test animals in terms of previous TGE outbreaks. Oc-
with mortality of 10% and 18% using two commercial currence of PRCV in herds may further complicate fu-
vaccines. ture TGEV vaccine studies.
The IM vaccination procedure has two major disad- Limited laboratory research has indicated that par-
vantages. (1) Vaccinated swine develop little or no gut enteral inoculation of previously infected swine during
immunity; they usually get sick when exposed to TGEV. gestation using attenuated TGEV resulted in a boost in
If this occurs during lactation, their suckling pigs will be TGEV milk antibodies in both the IgA and IgG classes
deprived of adequate milk. (2) The TGEV antibodies (Saif and Bohl 1981a, b, 1983; Saif 1985). Others have al-
found in the milk of these vaccinated sows are of the IgG so reported greatly increased milk TGEV antibody titers
class and of low titer and fail to provide optimal passive after intramammary inoculation of previously infected
protection to suckling pigs. swine with inactivated TGEV (Thorsen and Djurickovic
Intramammary injection of seronegative pregnant 1971). These titers were about four- to sevenfold greater
swine with TGEV resulted in high titers of primarily IgG than in seronegative intramammarily vaccinated gilts or
TGEV antibodies in milk, whereas similar injections in nonvaccinated infected sows. Currently available par-
CHAPTER 24 TRANSMISSIBLE GASTROENTERITIS Saif, Wesley 313

enterally administered TGEV vaccines may be more ef- that only the recombinant S protein fragments contain-
fective in boosting immunity in previously infected preg- ing antigenic site A induced neutralizing antibodies in
nant swine than in initiating immunity in previously un- serum which delayed the onset of clinical signs and virus
infected seronegative pregnant swine. These vaccines shedding when artificially fed to TGEV-challenged
may be especially useful in herds in which enzootic TGE piglets. Similarly, Shoup et al. (1997) reported that only
is a problem (Leopoldt and Meyer 1978; Stepanek et al. full or partial (789 residues) length baculovirus recombi-
1979; Lutter et al. 1982). nant S glycoprotein constructs containing the four major
antigenic sites (A–D) induced moderate to high neutral-
NEW EXPERIMENTAL VACCINE APPROACHES izing antibody titers to TGEV in serum and primed pigs
Native Subunit Vaccines. In two studies, virus sub- for secondary antibody responses after challenge. A re-
units purified from virulent TGEV and administered combinant S protein containing only antigenic sites C
with adjuvants by parenteral routes were evaluated as and D (397 residues) induced low neutralizing antibody
vaccines (Garwes et al. 1978/79; Gough et al. 1983b). titers in serum but also primed pigs for a secondary anti-
Garwes et al. (1978/79) injected pregnant sows intra- body response after TGEV challenge. Parenteral inocula-
mammarily with virus surface projections (S glycopro- tion of pigs or sows with the recombinant glycoproteins
tein) or subunit particles: neutralizing antibodies were failed to induce a high level of active or passive immuni-
present only in serum and colostrum of sows injected ty, respectively, against TGEV challenge. The parenteral-
with S glycoproteins. Colostral antibodies were associat- ly inoculated sows had IgG, but not IgA, antibodies to
ed with IgG, except in one sow that had both IgG and IgA TGEV in colostrum and low or no TGEV antibody titers
antibodies. However, protection of challenged piglets in milk, confirming previous reports that the presence of
was poor (100% mortality in four of six litters). serum antibodies and low titers of colostral antibodies
Gough et al. (1983b) used a low molecular weight un- (of the IgG isotype) are not associated with passive pro-
defined soluble subunit (about 23 kDa) purified from tection against TGEV (Haelterman 1965; Saif and Bohl
TGEV by sonication, isopycnic centrifugation, and gel fil- 1979a, b, 1981a, 1983).
tration to IM inject pregnant gilts. These investigators
detected neutralizing antibodies in serum and milk and Live Recombinant Vector Vaccines. Because stim-
observed only 4% mortality among challenged piglets. ulation of intestinal immunity is important to induce
The Ig isotype of TGEV antibodies in the milk of vacci- protection against TGEV, investigators have attempted
nates was not determined. These results have not been to develop genetically engineered vaccines using live re-
reproduced further or confirmed. combinant vectors that replicate in the intestinal tract.
Studies of a live-vector vaccine for TGEV initially focused
rDNA Subunit Vaccines. Limited data exist con- on the use of a prokaryotic expression system such as at-
cerning the immunogenicity of rDNA-produced viral tenuated strains of Salmonella typhimurium, which pos-
subunits or peptides of TGEV. The S protein fragments sess a tropism for intestinal Peyer’s patches (Smerdou et
expressed in E. coli were not glycosylated, were difficult al. 1996). The TGEV S protein site D (similar to Paris site
to isolate due to aggregation and insolubility, and did not C) was expressed as a fusion protein with E. coli labile
induce neutralizing antibodies to TGEV in mice (Hu et toxin B in a recombinant S. typhimurium. Oral inocula-
al. 1985). Posthumus et al. (1990) noted that a peptide tion of rabbits with the recombinant bacteria induced
from the S protein (residues 379–386) raised neutraliz- TGEV antibodies in serum and intestinal secretions as
ing antibodies to TGEV in rabbits. Higher titers were in- measured by a radioimmunoassay but only low titers of
duced when this peptide was coupled to a second S pro- neutralizing antibodies in the intestinal secretions. How-
tein peptide (residues 1160–1180), but the protective ever, no pig protection studies have been reported.
efficacy of these peptides was not tested in swine. Studies are in progress using a live eukaryotic vector
Eukaryotic vectors have been used to express TGEV S with a respiratory and enteric tropism (human adeno-
protein genes encoding determinants dependent on gly- virus 5 and porcine adenovirus) to express the TGEV S
cosylation (site A-B). The S glycoprotein of TGEV ex- glycoprotein (Torres et al. 1996). A human adenovirus 5
pressed in vaccinia induced low titers of neutralizing an- recombinant expressing the TGEV S protein (containing
tibodies but no protection (Hu et al. 1985). The S, M, and sites A–D) or the amino terminal 26% of the S protein
N proteins of TGEV were also expressed in insect cells (Madrid sites C and B; Paris site D) induced neutralizing
using a baculovirus expression system (Pulford et al. antibodies to TGEV in the serum of swine inoculated
1990; Godet et al. 1991; Tuboly et al. 1994, 1995; Shoup oronasally and intraperitoneally. Newborn piglets were
et al. 1997). The baculovirus-expressed S glycoproteins orally challenged with a TGEV-immune serum mixture
elicited neutralizing antibodies to TGEV in the serum of and then artificially fed immune serum (3 times per day)
rats (Godet et al. 1991) and pigs (Tuboly et al. 1994, from the immunized swine. Only in the group fed serum
1995; Shoup et al. 1997). Using TGEV S gene constructs from swine inoculated with the adenovirus–TGEV S pro-
of various lengths, Tuboly et al. (1994, 1995) showed tein recombinant (containing sites A–D) were newborn
314 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

piglets passively protected against diarrheal mortality, spring after TGEV challenge. Besides levels of IgA anti-
but they were not protected against TGEV infection. bodies in milk, a hallmark of protection in these and an-
However, no results were reported for direct TGEV chal- other study (Wesley and Woods 1993) of passive immu-
lenge of recombinant TGEV-adenovirus-inoculated pigs. nity to TGEV induced by PRCV appeared to be induction
Others (Callebaut et al. 1996) developed a recombi- of active immunity to TGEV in the sow, preventing ill-
nant human adenovirus 5 expressing the S protein of ness and/or agalactia. Diarrhea mortality was consis-
PRCV. Piglets inoculated oronasally with the recombi- tently lower in litters of PRCV-exposed sows that did not
nant adenovirus developed low PRCV-neutralizing anti- become ill or develop agalactia after TGEV challenge of
bodies in serum and shed virus nasally after PRCV chal- their litters.
lenge at similar levels as control pigs. However, the The mechanism for the induction of IgA antibodies
recombinant-inoculated pigs had a shorter duration of observed inconsistently in the milk of PRCV-exposed
virus shedding coinciding with a rapid anamnestic neu- sows is unclear, as is the correlation of such antibodies
tralizing antibody response after PRCV challenge. with passive protection to TGEV challenge (Pensaert and
A recent novel strategy for generation of recombi- Cox 1989; Callebaut et al. 1990). After a primary expo-
nant vaccines is based on the use of synthetic sure to PRCV, IgA antibodies occurred in the milk of on-
minigenomes from TGEV defective interfering RNAs ly 30% of sows; this percentage increased to 84% in sows
and helper rescue virus to express heterologous proteins. reinfected with PRCV. These results concur with a report
For example, a recombinant minigenome expressing the by Sestak et al. (1996) that the titers of IgA (and IgG) an-
S protein gene of TGEV used in combination with a res- tibodies to TGEV were significantly increased in the milk
piratory TGEV strain as a helper virus has been used to of sows multiply exposed to PRCV during two sequential
create chimeric viruses expressing the S protein from pregnancies. The variable IgA antibody responses ob-
TGEV and the respiratory strain (Izeta et al. 1997). If suf- served in the milk of sows after primary exposure to
ficiently attenuated, such chimeric viruses might prove PRCV could relate to the low numbers of virus-specific
useful as new candidate vaccines for TGEV. IgA ASCs observed in BALT, GALT, and mesenteric
lymph nodes of seronegative pigs exposed to PRCV (Van
PRCV Vaccines to Prevent TGEV. Since PRCV be- Cott et al. 1993, 1994). Reinfections with PRCV may be
came widespread in the European swine population, the necessary to boost numbers of IgA ASCs in BALT and to
incidence and severity of TGE in countries with PRCV increase the efficiency of a possible sIgA immunologic
have declined (Pensaert and Cox 1989; Laude et al. link between BALT and the mammary gland. Mainly
1993). This suggests that previous exposure of swine to virus-specific IgG ASCs were observed in BALT after
PRCV imparts a degree of immunity to a subsequent oronasal inoculation of seronegative pigs with PRCV
TGEV infection (Pensaert 1989; Pensaert and Cox 1989). (Van Cott et al. 1993). Following TGEV challenge, in-
A number of researchers have examined the relation- creased numbers of IgG ASCs (and IgA ASCs) appeared
ship between PRCV infection of sows and passive immu- in GALT of the PRCV-exposed pigs (Van Cott et al. 1994).
nity to TGEV in piglets. Prior natural exposure of sows to Because IgG antibodies are also prevalent in the milk of
PRCV induced a variable degree (44–53% mortality) of sows exposed to viruses that replicate in the respiratory
passive protection against experimental TGEV challenge tract, for example, attenuated TGEV strains and
of suckling pigs (Bernard et al. 1989; Paton and Brown pseudorabies virus (Saif and Bohl 1977), it is conceivable
1990). Variable protection in the field during TGE out- that an IgG immunologic link may also exist between
breaks was also noted among litters of PRCV-exposed BALT and the mammary gland in sows, or alternatively,
sows (Pensaert and Cox 1989; Callebaut et al. 1990). BALT stimulation contributes serum IgG antibodies that
Similar variable levels of protection (average of are subsequently transudated into the milk.
30–67% mortality) were reported after TGEV challenge Besides quantitative differences in the levels of IgA
of piglets suckling sows that had been experimentally in- antibodies induced in milk after exposure to TGEV or
fected or reinfected with PRCV during pregnancy (Calle- PRCV, researchers have investigated potential differ-
baut et al. 1990; DeDiego et al. 1992; Wesley and Woods ences in virus epitopes recognized by the IgA milk anti-
1993; Lanza et al. 1995; Sestak et al. 1996). Noteworthy bodies induced after TGEV versus PRCV infection of
in the latter two studies, litter mortality was lowest sows (DeDiego et al. 1992, 1994). In TGEV-infected sows,
(range = 0–27%, average = 14%) and IgA and IgG milk an- antigenic subsite A (Aa, Ab, Ac), followed by antigenic
tibody titers were highest in sows multiply exposed to subsite D (Madrid), was the best inducer of IgA antibod-
PRCV during two subsequent pregnancies. These experi- ies, whereas after PRCV infection, antigenic site D and
mental findings agree with other reports (Callebaut et al. subsite Ab were immunodominant. These authors con-
1990) that naturally PRCV-exposed sows that are rein- cluded that only IgA recognizing at least antigenic sites A
fected with PRCV during pregnancy secreted IgA TGEV and D conferred good protection in vivo, whereas any Ig
antibodies in milk and provided a high degree of protec- class recognizing only one antigenic site neutralized
tion (0–12.5% mortality in three of six litters) to their off- virus in cell culture.
CHAPTER 24 TRANSMISSIBLE GASTROENTERITIS Saif, Wesley 315

Clearly, additional studies are necessary to clarify the munoperoxidase method for detection of viral and
levels and mechanisms of active and passive immunity chlamydial antigens. III. Demonstration of TGE antigen
to TGEV established in swine by previous exposure to in pig thyroid cell cultures. Zentralbl Veterinärmed (B)
PRCV. In particular, it is important to elucidate the 21:59–65.
mechanism by which IgA antibodies are induced after in- Benfield, D. A.; Haelterman, E. O.; and Burnstein, T. 1978.
fection with PRCV, if a BALT-mammary gland immuno- An indirect fluorescent antibody test for antibodies to
logic link exists but is less efficient than the gut-mamma- transmissible gastroenteritis of swine. Can J Comp Med
ry link for induction of IgA in milk, why IgA antibodies 42:478–482.
occur only in some PRCV-exposed sows, and the effec- Benfield, D. A.; Jackwood, D. J.; Bae, I.; Saif, L. J.; and Wes-
tiveness of these IgA antibodies in protecting suckling ley, R. D. 1991. Detection of transmissible gastroenteri-
pigs against intestinal TGEV infections. tis virus using cDNA probes. Arch Virol 116:91–106.
Bernard, S., and Laude H. 1995. Site-specific alteration of
REFERENCES transmissible gastroenteritis virus spike protein results
in markedly reduced pathogenicity. J Gen Virol
Abou-Youssef, M. F., and Ristic, M. 1972. Distribution of 76:2235–2241.
antibodies to transmissible gastroenteritis virus in Bernard, S.; Lantier, I.; Laude H.; and Aynaud, J. M. 1986.
serum and milk of sows: Isolation and identification of Detection of transmissible gastroenteritis coronavirus
the immunoglobulin classes of swine serum and milk. antigens by a sandwich enzyme-linked immunosorbent
Am J Vet Res 33:975–979. assay technique. Am J Vet Res 47:2441–2444.
Ackerman, L. J.; Morehouse, L. G.; and Olson, L. D. 1972. Bernard, S.; Bottreau, E.; Aynaud, J. M.; Have, P.; and Szy-
Transmissible gastroenteritis in three-week-old pigs: mansky, J. 1989. Natural infection with the porcine res-
Study of anemia and iron absorption. Am J Vet Res piratory coronavirus induces protective lactogenic im-
33:115–120. munity against transmissible gastroenteritis. Vet
Anton, I. M.; Sune, C.; Meloen, R. H.; Borras-Cuesta, F.; and Microbiol 21:1–8.
Enjuanes, L. 1995. A transmissible gastroenteritis coro- Berthon, P.; Bernard, S.; Salmon, H.; and Binns, R. 1990. Ki-
navirus nucleoprotein epitope elicits T-helper cells that netics of the in vitro antibody response to transmissible
collaborate in the in vitro antibody synthesis to the three gastroenteritis virus from pig mesenteric lymph node
major structural viral proteins. Virol 212:746–751. cells using ELISASPOT and ELISA tests. J Immunol
Anton, I. M.; Gonzalez, S.; Bullido, M. J.; Corsin, M.; Risco, Methods 131:173–182.
C.; Langeveld, J. P. M.; and Enjuanes, L. 1996. Coopera- Binn, L. N.; Lazar, E. C.; Keenan, K. P.; Huxsoll, D. L.;
tion between transmissible gastroenteritis coronavirus Marchwicki, R. H.; and Strano, A. J. 1974. Recovery and
(TGEV) structural proteins in the in vitro induction of characterization of a coronavirus from military dogs
virus-specific antibodies. Virus Res 46:111–124. with diarrhea. Proc US Anim Health Assoc 78:359–366.
Aynaud, J. M.; Nguyen, T. D.; Bottreau, E.; Brun, A.; and Black, J. W. 1971. Diagnosis of TGE by FA: Evaluation of ac-
Vannier, P. 1985. Transmissible gastroenteritis (TGE) of curacy on field specimens. Proc US Anim Health Assoc
swine: Survivor selection of TGE virus mutants in stom- 75:492–498.
ach juice of adult pigs. J Gen Virol 66:1911–1917. Bohl, E. H. 1979. Diagnosis of diarrhea in pigs due to trans-
Bachmann, P. A.; Haniche, T.; Danner, K.; and Bibrack, B. missible gastroenteritis virus or rotavirus. In Viral En-
1972. Epidemiology of TGE in the pig. Zentralbl Veter- teritis in Humans and Animals. Ed. F. Bricout and R.
inärmed (B) 19:166–174. Scherrer. INSERM (Paris) 90:341–343.
Bae, I.; Jackwood, D. J.; Benfield, D. A.; Saif, L. J.; Wesley, R. ———. 1981. Coronaviruses: Diagnosis of infections. In
D.; and Hill, H. 1990. Differentiation of transmissible Comparative Diagnosis of Viral Diseases, vol. 4. Ed. E.
gastroenteritis virus from porcine respiratory coron- Kurstak and C. Kurstak. New York: Academic Press, pp.
avirus and other antigenically related coronaviruses by 301–328.
using cDNA probes specific for the 5′ region of the S gly- Bohl, E. H., and Kumagai, T. 1965. The use of cell cultures
coprotein gene. J Clin Microbiol 29:215–218. for the study of swine. Proc US Livest Sanit Assoc
Ballesteros, M. L.; Sanchez, C. M.; and Enjuanes, L. 1997. 69:343–350.
Two amino acid changes at the N-terminus of transmis- Bohl, E. H., and Saif, L. J. 1975. Passive immunity in trans-
sible gastroenteritis coronavirus spike protein result in missible gastroenteritis of swine: Immunoglobulin char-
the loss of enteric tropism. Virol 227:378–388. acteristics of antibodies in milk after inoculating virus
Bay, W. W.; Doyle, L. P.; and Hutchings, L. M. 1952. Some by different routes. Infect Immun 11:23–32.
properties of the causative agent of transmissible gas- Bohl, E. H.; Gupta, R. K. P.; Olquin, M. V. F.; and Saif, L. J.
troenteritis in swine. Am J Vet Res 13:318–321. 1972. Antibody responses in serum, colostrum and milk
———. 1953. Transmissible gastroenteritis in swine: A of swine after infection or vaccination with transmissi-
study of immunity. J Am Vet Med Assoc 122:200–202. ble gastroenteritis virus. Infect Immun 6:289–301.
Becker, W.; Teufel, P.; and Mields, W. 1974. The im- Bohl, E. H.; Frederick, G. T.; and Saif, L. J. 1975. Passive im-
316 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

munity in transmissible gastroenteritis of swine: Intra- porcine respiratory coronavirus. J Gen Virol
muscular injection of pregnant swine with a modified 69:1725–1730.
live-virus vaccine. Am J Vet Res 36:267–271. Callebaut, P.; Pensaert, M. B.; and Hooyberghs, J. 1989. A
Bohl, E. H.; Kohler, E. M.; Saif, L. J.; Cross, R. F.; Agnes, A. competitive inhibition ELISA for the differentiation of
G.; and Theil, K. W. 1978. Rotavirus as a cause of diar- serum antibodies from pigs infected with transmissible
rhea in pigs. J Am Vet Med Assoc 172:458–463. gastroenteritis virus (TGEV) or with the TGEV-related
Bohl, E. H.; Saif, L. J.; and Jones, J. E. 1982. Observations on porcine respiratory coronavirus. Vet Microbiol 20:9–19.
the occurrence of transmissible gastroenteritis (TGE) in Callebaut, P.; Cox, E.; Pensaert, M.; and Van Deun, K. 1990.
a vaccinated herd. In Ohio Swine Res Ind Rep, Anim Sci Induction of milk IgA antibodies by porcine respiratory
Ser 82-1, Ohio State Univ, pp. 66–69. coronavirus infection. Adv Exp Med Biol 276:421–428.
Bourgueil, E.; Hutet, E.; Cariolet, R.; and Vannier, P. 1992. Callebaut, P.; Enjuanes, L.; and Pensaert M. 1996. An adeno-
Experimental infection of pigs with the porcine respira- virus recombinant expressing the spike glycoprotein of
tory coronavirus (PRCV): Measure of viral excretion. Vet porcine respiratory coronavirus is immunogenic in
Microbiol 31:11–18. swine. J Gen Virol 77:309–313.
Bourne, F. J. 1973. Symposium on nutrition of the young Cartwright, S. F. 1968. Transmissible gastroenteritis of
farm animal: The immunoglobulin system of the suck- swine (TGE). Br Vet J 124:410–413.
ling pig. Proc Nutr Soc 32:205–214. Cartwright, S. F.; Harris, H. M.; Blandford, T. B.; Fingham,
Brian, A. B.; Dennis, D. E.; and Grey, J. S. 1980. Genome of I.; and Gitter, M. 1965. A cytopathic virus causing a
porcine transmissible gastroenteritis virus. J Virol transmissible gastroenteritis in swine. I. Isolation and
34:410–415. properties. J Comp Pathol 75:386–395.
Brim, T. A.; Van Cott, J. L.; Lunney, J. K.; and Saif, L. J. 1994. Cavanagh, D.; Brian, D. A.; Brinton, M. A.; Enjuanes, L.;
Lymphocyte proliferative responses of pigs inoculated Holmes, K. V.; Horzinek, M. C.; Lai, M. M. C.; Laude, H.;
with transmissible gastroenteritis virus or porcine respi- Plagemann, P. W.; Siddell, S. G.; Spaan, W. J. M.;
ratory coronavirus. Am J Vet Res 55:494–501. Taguchi, F.; and Talbot, P. J. 1995. Coronaviridae. In
Brim, T. A.; Van Cott, J. L.; Lunney, J. K.; and Saif, L. J. 1995. Virus Taxonomy: The Classification and Nomenclature
Cellular immune responses of pigs after primary inocu- of Viruses. 6th Rep of ICTV. Ed. F. A. Murphy et al. Vi-
lation with porcine respiratory coronavirus or transmis- enna and New York: Springer-Verlag, pp. 407–411.
sible gastroenteritis virus and challenge with transmissi- Cepica, A., and Derbyshire, J. B. 1983. Antibody-dependent
ble gastroenteritis virus. Vet Immunol Immunopath cell-mediated cytotoxicity and spontaneous cell-mediat-
48:35–54. ed cytotoxicity against cells infected with porcine trans-
Britton, P.; Mawditt, K. L.; and Page, K. W. 1991. The missible gastroenteritis virus. Can J Comp Med
cloning and sequencing of the virion protein genes from 47:298–303.
a British isolate of porcine respiratory coronavirus: ———. 1984. Antibody-dependent and spontaneous cell-
Comparison with transmissible gastroenteritis virus mediated cytotoxicity against transmissible gastroen-
genes. Virus Res 21:181–198. teritis virus-infected cells by lymphocytes from sows, fe-
Britton, P.; Kottier, S.; Chen, C.-M.; Pocock, D. H.; Salmon, tuses and neonatal piglets. Can J Comp Med
H.; and Aynaud, J. M. 1994. The use of PCR genome 48:258–261.
mapping for the characterization of TGEV strains. Adv Charley, B., and Laude, H. 1988. Induction of alpha inter-
Exp Med Biol 342:29–34. feron by transmissible gastroenteritis coronavirus: Role
Brown, I., and Cartwright, S. 1986. New porcine coron- of transmembrane glycoprotein E1. J Virol 62:8–11.
avirus? Vet Rec 119:282–283. Chen, K. S. 1985. Enzymatic and acidic sensitivity profiles of
Brown, I. H., and Paton, D. J. 1991. Serological studies of selected virulent and attenuated transmissible gastroen-
transmissible gastroenteritis in Great Britain, using a teritis viruses of swine. Am J Vet Res 46:632–636.
competitive ELISA. Vet Record 128:500–503. Chu, R. M.; Glock, R. D.; and Ross, R. F. 1982a. Changes in
Brown, T. T. 1981. Laboratory evaluation of selected disin- gut-associated lymphoid tissues of the small intestine of
fectants as viricidal agents against porcine parvovirus, eight-week-old pigs infected with transmissible gas-
pseudorabies virus, and transmissible gastroenteritis troenteritis virus. Am J Vet Res 43:67–76.
virus. Am J Vet Res 42:1033–1036. Chu, R. M.; Li, N. J.; Glock, R. D.; and Ross, R. F. 1982b. Ap-
Butler, D. G.; Gall, D. G.; Kelly, M. H.; and Hamilton, J. R. plication of peroxidase staining technique for detection
1974. Transmissible gastroenteritis: Mechanisms re- of transmissible gastroenteritis virus in pigs. Am J Vet
sponsible for diarrhea in an acute enteritis in piglets. J Res 43:77–81.
Clin Invest 53:1335–1342. Cornaglia, E.; Chretien, M.; Charara, S.; and Elazhary, Y.
Callebaut, P.; Correa, I.; Pensaert, M.; Jimenez, G.; and En- 1994. Detection of porcine respiratory coronavirus and
juanes, L. 1988. Antigenic differentiation between trans- transmissible gastroenteritis virus by an enzyme-linked
missible gastroenteritis virus of swine and a related immunosorbent assay. Vet Microbiol 42:349–359.
CHAPTER 24 TRANSMISSIBLE GASTROENTERITIS Saif, Wesley 317

Cornelius, L. M.; Hooper, B. E.; and Haelterman, E. O. 1968. Dimitrov, P. 1982. Effect of amantadine on different stages
Changes in fluid and electrolyte balance in baby pigs of replication of porcine transmissible gastroenteritis
with transmissible gastroenteritis. Am J Clin Pathol virus. Vet Med Nauki 19:90–96. In Bulgarian with Eng-
2:105–113. lish abstract.
Correa, I.; Gebauer, F.; Bullido, M. J.; Sune, C.; Baay, M. F. Doyle, L. P., and Hutchings, L. M. 1946. A transmissible gas-
D.; Zwaagstra, K. A.; Posthumus, W. P. A.; Lenstra, J. A.; troenteritis in pigs. J Am Vet Med Assoc 108:257–259.
and Enjuanes, L. 1990. Localization of antigenic sites of Dulac, G. C.; Ruckerbauer, G. M.; and Boulanger, P. 1977.
the E2 glycoprotein of transmissible gastroenteritis Transmissible gastroenteritis: Demonstration of the
virus. J Gen Virol 71:271–279. virus from field specimens by means of cell culture and
Cox, E.; Hooyberghs, J.; and Pensaert, M. B. 1990a. Sites of pig inoculation. Can J Comp Med 41:357–363.
replication of a porcine respiratory coronavirus related Egan, I. T.; Harris, D. L.; and Hill, H. T. 1982. Prevalence of
to transmissible gastroenteritis virus. Res Vet Sci swine dysentery, transmissible gastroenteritis, and
48:165–169. pseudorabies in Iowa, Illinois and Missouri swine. In
Cox, E.; Pensaert, M. B.; Callebaut, P.; and Van Deun, K. Proc 86th Annu Meet US Anim Health Assoc, pp.
1990b. Intestinal replication of a porcine coronavirus 497–502.
closely related antigenically to the enteric transmissible Eleouet, J.-F.; Rasschaert, D.; Lambert, P.; Levy, L.; Vende, P.;
gastroenteritis virus. Vet Microbiol 23:237–243. and Laude, H. 1995. Complete sequence (20 kilobases)
Cox, E.; Pensaert, M. B.; and Callebaut, P. 1993. Intestinal of the polyprotein-encoding gene 1 of transmissible gas-
protection against challenge with transmissible gas- troenteritis virus. Virology 206:817–822.
troenteritis virus of pigs immune after infection with the Enjuanes, L., and Van der Zeijst, B. A. M. 1995. Molecular
porcine respiratory coronavirus. Vaccine 11:267–272. basis of transmissible gastroenteritis virus epidemiolo-
Cummins, J. M.; Mock, R. E.; Shive, B. W.; Krakowka, S.; gy. In The Coronaviridae. Ed. S. G. Siddell. New York:
Richards, A. B.; and Hutcheson, D. P. 1995. Oral treat- Plenum Press, pp. 337–376.
ment of transmissible gastroenteritis with natural hu- Ferris, D. H. 1973. Epizootiology of porcine transmissible
man interferon alpha: A field study. Vet Immunol Im- gastroenteritis (TGE). In Advances in Veterinary Science
munopathol 45:355–360. and Comparative Medicine, vol. 17. New York and Lon-
DeBouck, P.; Callebaut, P.; and Pensaert, M. 1982. Preva- don: Academic Press, pp. 57–86.
lence of the porcine epidemic diarrhea (PED) virus in Fichtner, D.; Leopoldt, D.; and Meyer, U. 1982. Unter-
the pig population of different countries. Proc Int Congr suchungen zur Ermittlung der minimalen Antigen-
Pig Vet Soc 7:45. menge bei der oralen Muttertierimmunisierung gegen
DeDiego, M.; Laviada, M. D.; Enjuanes, L.; and Escribano, J. die Transmissible Gastroenteritis der Schweine mit
M. 1992. Epitope specificity of protective lactogenic im- Riemser TGE-Vakzine. Arch Exp Veterinärmed
munity against swine transmissible gastroenteritis 36:577–585.
virus. J Virol 66:6502–6508. Frederick, G. T., and Bohl, E. H. 1976. Local and systemic
DeDiego, M.; Rodriguez, F.; Alcaraz, C.; Gomez, N.; Alonso, cell-mediated immunity against transmissible gastroen-
C.; and Escribano, J. M. 1994. Characterization of the teritis, an intestinal viral infection of swine. J Immunol
IgA and subclass IgG responses to neutralizing epitopes 116:1000–1004.
after infection of pregnant sows with the transmissible Frederick, G. T.; Bohl, E. H.; and Cross, R. F. 1976. Patho-
gastroenteritis virus or the antigenically related porcine genicity of an attenuated strain of transmissible gas-
respiratory coronavirus. J Gen Virol 75:2585–2593. troenteritis virus for newborn pigs. Am J Vet Res
Delmas, B., and Laude, H. 1990. Assembly of coronavirus 37:165–169.
spike protein into trimers and its role in epitope expres- Furuuchi, S.; Shimizu, Y.; and Kumagai, T. 1975. Compari-
sion. J Virol 64:5367–5375. son of properties between virulent and attenuated
Delmas, B.; Rasschaert, D.; Godet, M.; Gelfi, J.; and Laude, strains of TGE. Natl Inst Anim Health Q (Tokyo)
H. 1990. Four major antigenic sites of the coronavirus 15:159–164.
transmissible gastroenteritis virus are located on the ———. 1976. Vaccination of newborn pigs with an attenu-
amino-terminal half of spike glycoprotein S. J Gen Virol ated strain of transmissible gastroenteritis virus. Am J
71:1313–1324. Vet Res 37:1401–1404.
Delmas, B.; Gelfi, J.; L’Haridon, R.; Vogel, L. K.; Sjestrem, H.; Furuuchi, S.; Shimizu, M.; and Shimizu, Y. 1978. Field trials
Noren, O.; and Laude, H. 1992. Aminopeptidase N is a on transmissible gastroenteritis live virus vaccine in
major receptor for the enteropathogenic coronavirus newborn piglets. Natl Inst Anim Health Q (Tokyo)
TGEV. Nature 357:417–420. 18:135–142.
Derbyshire, J. B.; Jessett, D. M.; and Newman, G. 1969. An Furuuchi, S.; Shimizu, Y.; and Kumagai, T. 1979. Multiplica-
experimental epidemiological study of porcine trans- tion of low and high cell culture passaged strains of
missible gastroenteritis. J Comp Pathol 79:445–452. transmissible gastroenteritis virus in organs of newborn
318 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

piglets. Vet Microbiol 3:169–178. ble Gastroenteritis des Schweines im Negativekon-


Gagnon, A. N.; Dulack, G. C.; Marsolais, G.; Lussier, G.; and trastverfahren. Arch Exp Veterinärmed 35:177–186.
Marois, P. 1974. Maladies porcines a etiologie virale Haas, B.; Ahl, R.; Bohm, R.; and Strauch, D. 1995. Inactiva-
dans la Province de Quebec. II. Gastro–enterite trans- tion of viruses in liquid manure. Rev Sci Tech Off Int
missible. Can Vet J 15:316–318. Epiz 14:435–445.
Garwes, D. J., and Pocock, D. H. 1975. The polypeptide Haelterman, E. O. 1962. Epidemiological studies of trans-
structure of transmissible gastroenteritis virus. J Gen Vi- missible gastroenteritis of swine. Proc US Livest Sanit
rol 29:25–34. Assoc 66:305–315.
Garwes, D. J.; Lucas, M. H.; Higgins, D. A.; Pike, B. V.; and ———. 1963. Transmissible gastroenteritis of swine. Proc
Cartwright, S. F. 1978/79. Antigenicity of structural 17th World Vet Congr, Hannover, 1:615–618.
components from porcine transmissible gastroenteritis ———. 1965. Lactogenic immunity to transmissible gas-
virus. Vet Microbiol 3:179–190. troenteritis of swine. J Am Vet Med Assoc 147:1661.
Garwes, D. J.; Stewart, F.; and Elleman, C. J. 1987. Identifi- Haelterman, E. O., and Hutchings, L. M. 1956. Epidemic di-
cation of epitopes of immunological importance on the arrheal diseases of viral origin in newborn swine. Ann
peplomer protein of porcine transmissible gastroenteri- NY Acad Sci 66:186–190.
tis virus. Adv Exp Med Biol 218:509–515. Halbur, P. G.; Paul, P. S.; Vaughn, E. M.; and Andrews, J. J.
Garwes, D. J.; Stewart, F.; Cartwright, S. F.; and Brown, I. 1993. Experimental reproduction of pneumonia in gno-
1988. Differentiation of porcine coronavirus from trans- tobiotic pigs with porcine respiratory coronavirus iso-
missible gastroenteritis virus. Vet Rec 122:86–87. late AR310. J Vet Diagn Invest 5:184–188.
Gebauer, F.; Posthumus, W. P. A.; Correa, I.; Sune, C.; Smer- Halbur, P.; Paul, P.; and Vaughn, E. 1994. Virulent porcine
dou, C.; Sanchez, C. M.; Lenstra, J. A.; Meloen, R. H.; respiratory coronavirus isolates exist in the United
and Enjuanes, L. 1991. Residues involved in the anti- States. Proc Int Congr Pig Vet Soc 13:70.
genic sites of transmissible gastroenteritis coronavirus S Harada, K.; Kaji, T.; Kumagai, T.; and Sasahara, J. 1968.
glycoprotein. Virology 183:225–238. Studies on transmissible gastroenteritis in pigs. IV.
Godet, M.; Rasschaert D.; and Laude, H. 1991. Processing Physicochemical and biological properties of TGE virus.
and antigenicity of entire and anchor-free spike glyco- Natl Inst Anim Health Q (Tokyo) 8:140–147.
protein S of coronavirus TGEV expressed by recombi- Harris, D. L.; Bevier, G. W.; and Wiseman, B. S. 1987. Erad-
nant baculovirus. Virology 185:732–740. ication of transmissible gastroenteritis virus without de-
Godet, M.; L’Haridon, R.; Vautherot, J.-F.; and Laude, H. population. In Proc Am Assoc Swine Pract, Des Moines,
1992. TGEV corona virus ORF4 encodes a membrane Iowa, p. 555.
protein that is incorporated into virions. Virology Have, P.; Moving, V.; Svansson, V.; Uttenthal, A.; and Bloch,
188:666–675. B. 1992. Coronavirus infection in mink (Mustela vison):
Godet, M.; Grosclaude, J.; Delmas, B.; and Laude, H. 1994. Serological evidence of infection with a coronavirus re-
Major receptor-binding and neutralization determi- lated to transmissible gastroenteritis virus and porcine
nants are located within the same domain of the trans- epidemic diarrhea virus. Vet Microbiol 31:1–10.
missible gastroenteritis virus (coronavirus) spike pro- Henning, E. R., and Thomas, P. C. 1981. Comparison of in-
tein. J Virol 68:8008–8016. tramuscular and oral modified live virus TGE vaccines.
Goodwin, R. F. W., and Jennings, A. R. 1958. A highly infec- Vet Med Small Anim Clin 76:1789–1792.
tious gastroenteritis of pigs. Vet Rec 70:271–272. Henningsen, D.; Mousing, J.; and Aalund, O. 1989. Porcint
Gough, P. M., and Jorgenson, R. D. 1983. Identification of corona virus (PCV) i Danmark: En epidemiologisk traer-
porcine transmissible gastroenteritis virus in house flies snitsanalyse baseret pa screening-omrade sporgeskema
(Musca domestica Linneaus). Am J Vet Res data. Dansk Vet Tidsskr 71:1168–1177.
44:2078–2082. Hess, R. G., and Bachmann, P. A. 1976. In vitro differentia-
Gough, P. M.; Ellis, C. H.; Frank, C. J.; and Johnson, C. J. tion and pH sensitivity of field and cell culture attenuat-
1983a. A viral subunit immunogen for porcine trans- ed strains of transmissible gastroenteritis. Infect Immun
missible gastroenteritis. Antiviral Res 3:211–221. 13:1642–1646.
Gough, P. M.; Frank, C. J.; Moore, D. G.; Sagona, M. A.; and Hess, R. G.; Bachmann, P. A.; and Hanichen, T. 1977. At-
Johnson, C. J. 1983b. Lactogenic immunity to transmis- tempts to establish an immunoprophylaxis for trans-
sible gastroenteritis virus induced by a subunit im- missible gastroenteritis virus infection of pigs. I. Patho-
munogen. Vaccine 1:37–41. genicity of the B1 strain after serial passages. Zentralbl
Graham, J. A. 1980. Induction of active immunity to TGE in Veterinärmed (B) 24:753–763.
neonatal pigs nursing seropositive dams. Vet Med Small Hess, R. G.; Bachmann, P. A.; and Mayr, A. 1978. Attempts
Anim Clin 75:1618–1619. to develop an immunoprophylaxis against transmissible
Granzow, H.; Meyer, U.; Solisch, P.; Lange, E.; and Fichtner, gastroenteritis (TGE) in pigs. III. Passive immune trans-
D. 1981. Die Morphologie der Koronaviren–elektronen- fer after oral vaccination with attenuated TGE virus
mikroskopische Darstellung des Virus der Transmissi- strain B1. Zentralbl Veterinärmed (B) 25:308–318.
CHAPTER 24 TRANSMISSIBLE GASTROENTERITIS Saif, Wesley 319

Hess, R. G.; Chen, Y. S.; and Bachmann, P. A. 1982. Active peplomer gene of porcine transmissible gastroenteritis
immunization of feeder pigs against transmissible gas- (TGEV): Comparison with the sequence of the peplom-
troenteritis (TGE): Influence of maternal antibodies. er protein of feline infectious peritonitis virus (FIPV).
Proc Int Congr Pig Vet Soc 7:1. Virus Res 8:363–371.
Hill, H.; Biwer, J.; Wood, R.; and Wesley, R. 1990. Porcine Jestin, A.; Le Forban, Y.; Vannier, P.; Madec, F.; and Gour-
respiratory coronavirus isolated from two U.S. swine reau, J. M. 1987. Un nouveau coronavirus porcin: Études
herds. In Proc Am Assoc Swine Pract, Des Moines, Iowa, sero-epidemiologiques retrospectives dans les élevages
p. 333. de Bretagne. Rec Med Vet 163:567–571.
Hohdatsu, T.; Eiguchi, Y.; Ide, S.; Baba, K.; Yamagishi, H.; Jimenez, G.; Correa, I.; Melgosa, M. P.; Buillido, M. J.; and
Kume, T.; and Matumoto, M. 1987. Evaluation of an en- Enjuanes, L. 1986. Critical epitopes in transmissible gas-
zyme-linked immunosorbent assay for the detection of troenteritis virus neutralization. J Virol 60:131–139.
transmissible gastroenteritis virus antibodies. Vet Mi- Kaji, T., and Shimizu, Y. 1978. Passive immunization
crobiol 13:93–97. against transmissible gastroenteritis virus in piglets of
Hooper, B. E., and Haelterman, E. O. 1966. Concepts of ingestion of milk of sows inoculated with attenuated
pathogenesis and passive immunity in transmissible virus. Natl Inst Anim Health Q (Tokyo) 18:43–52.
gastroenteritis of swine. J Am Vet Med Assoc Kapke, P. A., and Brian, D. A. 1986. Sequence analysis of the
149:1580–1586. porcine transmissible gastroenteritis coronavirus nucle-
———. 1969. Lesions of the gastrointestinal tract of pigs ocapsid protein gene. Virology 151:41–49.
infected with transmissible gastroenteritis. Can J Comp Kemeny, L. J. 1976. Antibody response in pigs inoculated
Med 33:29–36. with transmissible gastroenteritis virus and cross reac-
Hornich, M.; Salajka, E.; and Stepanek, J. 1977. Malabsorp- tions among ten isolates. Can J Comp Med 40:209–214.
tion in newborn piglets with diarrhoeic Escherichia coli ———. 1978. Isolation of transmissible gastroenteritis
infection and transmissible gastroenteritis. Zentralbl virus from pharyngeal swabs obtained from sows at
Veterinärmed (B) 24:75–86. slaughter. Am J Vet Res 39:703–705.
Hortig, H.; Roder, B.; and Bruske, R. 1980. Vergleichende Kemeny, L. J., and Woods, R. D. 1977. Quantitative trans-
Untersuchungen zur Verwendbarkeit des Microneutrali- missible gastroenteritis virus shedding patterns in lac-
sationstests, des indirekten Immunofluoreszenztests tating sows. Am J Vet Res 38:307–310.
und des Virus-Inhibitionstests zum Nachweis von An- Kemeny, L. J.; Wiltsey, V. L.; and Riley, J. L. 1975. Upper res-
tikorpen gegen das Virus der Transmissible Gastroen- piratory infection of lactating sows with transmissible
teritis (TGE) des Schweines. DTW 87:192–196. gastroenteritis virus following contact exposure to in-
Horzinek, M. C.; Lutz, H.; and Pedersen, N. C. 1982. Anti- fected piglets. Cornell Vet 65:352–362.
genic relationships among homologous structural Klemm, R. C., and Ristic, M. 1976. The effect of propaga-
polypeptides of porcine, feline, and canine coronavirus- tion of transmissible gastroenteritis (TGE) virus in pups
es. Infect Immun 37:1148–1155. and the lungs of baby pigs on the immunologic proper-
Hu, S.; Bruszewski, J.; Smalling, R.; and Browne, J. K. 1985. ties of the virus. Proc Int Congr Pig Vet Soc 4:K11.
Studies of TGEV spike protein gp 195 expressed in E. Kodama, Y.; Ogata, M.; and Shimizu, Y. 1980. Characteris-
coli and by a TGE-vaccinia virus recombinant. In Im- tics of immunoglobulin A antibody in serum of swine
munobiology of Proteins and Peptides—III. Ed. M. inoculated with transmissible gastroenteritis virus. Am J
Zouhair–Atassi and H. L. Bachrach. New York: Plenum Vet Res 40:740–745.
Press, p. 63. ———. 1981. Serum immunoglobulin A antibody response
Izeta, A.; Smerdou, C.; Sanchez, C. M.; Alonso, S.; Penzes, in swine infected with transmissible gastroenteritis
Z.; and Enjuanes, L. 1997. Replication, packaging, and virus, as determined by indirect immunoperoxidase an-
expression of foreign genes using synthetic tibody test. Am J Vet Res 42:437–442.
minigenomes derived from transmissible gastroenteritis Labadie, J. P.; Aynaud, J. M.; Vaissaire, J.; and Renault, L.
coronavirus defective interfering RNAs. Abst 7th Int 1977. Porcine transmissible gastroenteritis. Antibody
Symp on Coronavirus and Arteriviruses. May 10–15, detection by passive haemagglutination test: Applica-
1997, Segovia, Spain. tions to diagnosis and epidemiology. Rec Med Vet
Jabrane, A.; Girard, C.; and Elazhary, Y. 1994. Pathogenicity 153:931–936.
of porcine respiratory coronavirus isolated in Quebec. LaBonnardiere, C., and Laude, H. 1981. High interferon
Can Vet J 35:86–92. titer in newborn pig intestine during experimentally in-
Jackwood, D. J.; Kwon, H. M.; and Saif, L. J. 1995. Molecular duced viral enteritis. Infect Immun 32:28–31.
differentiation of transmissible gastroenteritis virus and Lange, E.; Schirrmeier, H.; Granzow, H.; Fichtner, D. P.; and
porcine respiratory coronavirus strains. Adv Exp Med Leopoldt, D. 1988. Isolierung und erste Charakter-
Biol 380:35–41. isierung eines coronavirus aus Lungen und Tonsillen
Jacobs, L.; deGroot, R.; Van der Zeijst, B. A.; Horzinek, M. klinisch gesunder Schweine. Monatsh Veterinärmed
C.; and Spaan, W. 1987. The nucleotide sequence of the 43:273–274.
320 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Lanza, I.; Brown, I. H.; and Paton, D. J. 1992. Pathogenicity 8:135–141.


of concurrent infection of pigs with porcine respiratory Loewen, K. G., and Derbyshire, J. B. 1988. The effect of in-
coronavirus and swine influenza virus. Res Vet Sci terferon induction in parturient sows and newborn
53:309–314. piglets on resistance to transmissible gastroenteritis.
Lanza, I.; Rubio, P.; Fernandez, M.; Munoz, M.; and Can Vet Res 52:149–153.
Carmenes, P. 1993. Seroprevalence of porcine respirato- Lu, W.; Osorio, F. A.; Rhodes, M. B.; and Moxley, R. A. 1991.
ry coronavirus infection in Spanish breeding sows. Prev A capture-enzyme immunoassay for rapid diagnosis of
Vet Med 17:263–269. transmissible gastroenteritis virus. J Vet Diagn Invest
Lanza, I.; Shoup, D. I.; and Saif, L. J. 1995. Lactogenic im- 3:119–123.
munity and milk antibody isotypes to transmissible gas- Lutter, K.; Klahn, J.; and Kokles, R. 1982. Erfahrungen bei
troenteritis virus in sows exposed to porcine respiratory der Sanierung der Transmissible Gastroenteritis nach
coronavirus during pregnancy. Am J Vet Res dem Selektionsverfahren in einer Schweinezuchtanlage
56:739–748. mit 1200 produktiven Sauen. Monatsh Veterinärmed
Laude, H.; Gelfi, J.; and Aynaud, J. M. 1981. In vitro proper- 37:121–126.
ties of low- and high-passaged strains of transmissible Maes, R. K., and Haelterman, E. O. 1979. A seroepizootio-
gastroenteritis coronavirus of swine. Am J Vet Res logical study of five viruses in a swine-evaluation sta-
42:447–449. tion. Am J Vet Res 40:1642–1645.
Laude, H.; Charley, B.; and Gelfi, J. 1984. Replication of Martin, M.; Casal, J.; Lanza, I.; Rubio, P.; and Carmenes, P.
transmissible gastroenteritis coronavirus (TGE) in swine 1994. Porcine respiratory coronavirus spread in
alveolar macrophages. J Gen Virol 65:327–332. Catalunya, Spain, a previously infection-free area. Prev
Laude, H.; Chapsal, J. M.; Gelfi, J.; Labiau, S.; and Vet Med 21:65–74.
Grosclaude, J. 1986. Antigenic structure of transmissi- Matisheck, P.; Emerson, W.; and Searl, R. C. 1982. Results of
ble gastroenteritis virus. I. Properties of monoclonal an- laboratory and field tests of TGE vaccine. Vet Med Small
tibodies directed against virion proteins. J Gen Virol Anim Clin 77:262–264.
67:119–130. McClurkin, A. W., and Norman, J. O. 1966. Studies on trans-
Laude, H.; Gelfi, J.; Rasschaert, D.; and Delmas, B. 1988. missible gastroenteritis of swine. II. Selected character-
Caracterisation antigenique du coronavirus respiratoire istics of a cytopathogenic virus common to five isolates
porcin à l’aide d’anticorps monoclonaux diriges contre from transmissible gastroenteritis. Can J Comp Med
le virus de la gastro-enterite transmissible. J Rech 30:190–198.
Porcine Fr 20:89–94. McClurkin, A. W.; Stark, S. L.; and Norman, J. O. 1970.
Laude, H.; Rasschaert, D.; Delams, B.; Godet, M.; Gelfi, J.; Transmissible gastroenteritis (TGE) of swine: The possi-
and Charley, B. 1990. Molecular biology of transmissi- ble role of dogs in the epizootiology of TGE. Can J Comp
ble gastroenteritis. Vet Microbiol 23:147–154. Med 34:347–349.
Laude, H.; Gelfi, J.; Lavenant, L.; and Charley, B. 1992. Sin- Moon, H. W. 1978. Mechanisms in the pathogenesis of di-
gle amino acid changes in the viral glycoprotein M affect arrhea: A review. J Am Vet Med Assoc 172:443–448.
induction of alpha interferon by the coronavirus trans- Moon, H. W.; Norman, J. O.; and Lambert, G. 1973. Age de-
missible gastroenteritis virus. J Virol 66:743–749. pendent resistance to TGE of swine. I. Clinical signs and
Laude, H.; Van Reeth, K.; and Pensaert, M. 1993. Porcine some mucosal dimensions in the small intestine. Can J
respiratory coronavirus: Molecular features and virus- Comp Med 37:157–166.
host interactions. Vet Res 24:125–150. Morin, M.; Solorzano, R. F.; Morehouse, L. G.; and Olson, L.
Laviada, M. D.; Videgain, S. P.; Moreno, L.; Alonso, F.; En- D. 1978. The postulated role of feeder swine in the per-
juanes, L.; and Escribano, J. M. 1990. Expression of petuation of the transmissible gastroenteritis virus. Can
swine transmissible gastroenteritis virus envelope anti- J Comp Med 42:379–384.
gens on the surface of infected cells: Epitopes externally Morin, M.; Turgeon, D.; Jolette, J.; Robinson, Y.; Phaneuf, J.
exposed. Virus Res 16:247–254. B., Sauvageau, R.; Beauregard, M.; Teuscher, E.; Higgins,
Lee, K. M.; Moro, M.; and Baker, J. A. 1954. Transmissible R.; and Larivere, S. 1983. Neonatal diarrhea of pigs in
gastroenteritis in pigs. Am J Vet Res 15:364–372. Quebec: Infectious causes of significant outbreaks. Can
Leopoldt, D., and Meyer, U. 1978. Transmissible gastro-en- J Comp Med 47:11–17.
teritis of swine: A model of infectious diarrhoea. Arch Moscari, E. 1980a. Physicochemical properties of field and
Exp Veterinärmed 32:417–425. cell culture attenuated strains of swine transmissible
Lesnick, C. E., and Derbyshire, J. B. 1988. Activation of nat- gastroenteritis (TGE) coronavirus. Acta Vet Acad Sci
ural killer cells in newborn piglets by interferon induc- Hung 28:341–350.
tion. Vet Immunol Immunopathol 18:109–117. ———. 1980b. Vaccination experiments against transmis-
Liou, P. P. 1982. Cellular immunity in transmissible gas- sible gastroenteritis (TGE) of swine. VII. Immunoglobu-
troenteritis virus–infected pigs: Influence of viral anti- lin characteristics of antibodies in milk of sows vacci-
gens in leukocyte migration. J Chin Soc Vet Sci nated with the “CKp” strain of TGE virus. Acta Vet Acad
CHAPTER 24 TRANSMISSIBLE GASTROENTERITIS Saif, Wesley 321

Sci Hung 28:131–145. porcine immunoglobulins. Proc Int Congr Pig Vet Soc
Mousing, J.; Vagsholm, I.; Carpenter, T. E.; Gardner, I. A.; 9:190.
and Herd, D. W. 1988. Financial impact of transmissible Paul, P. S.; Halbur, P. G.; and Vaughn, E. M. 1994. Signifi-
gastroenteritis in pigs. J Am Vet Med Assoc cance of porcine respiratory coronavirus infection.
192:756–759. Compendium Cont Educ Pract Vet 16:1223–1234.
Moxley, R. A., and Olson, L. D. 1989a. Clinical evaluation of Pedersen, N. C.; Ward, J.; and Mengeling, W. L. 1978. Anti-
transmissible gastroenteritis virus vaccination proce- genic relationship of the feline infectious peritonitis
dures for inducing lactogenic immunity in sows. Am J virus to coronaviruses of other species. Arch Virol
Vet Res 50:111–118. 58:45–53.
———. 1989b. Lesions of transmissible gastroenteritis Pensaert, M. B. 1979. Immunity in TGE of swine after infec-
virus infection in experimentally inoculated pigs suck- tion and vaccination. In Viral Enteritis in Humans and
ling immunized sows. Am J Vet Res 50:708–716. Animals. Ed. F. Bricout and R. Scherrer. INSERM (Paris)
Nagura, H.; Nakane, D. K.; and Brown, W. R. 1978. Breast 90:281–293.
milk IgA binds to jejunal epithelia in suckling rats. J Im- ———. 1989. Transmissible gastroenteritis virus (respira-
munol 120:1330–1339. tory variant). In Virus Infections of Porcines. Ed. M. B.
Nakao, J.; Hess, R. G.; Bachmann, P. A.; and Mahnel, H. Pensaert. Amsterdam: Elsevier Science, pp. 154–165.
1978. Tenacity and inactivation of transmissible gas- Pensaert, M. B., and Cox, E. 1989. Porcine respiratory coro-
troenteritis (TGE) virus of pigs. Berl Münch Tierärztl navirus related to transmissible gastroenteritis virus.
Wochenschr 91:353–357. Agri-Pract 10:17–21.
Nelson, L. D., and Kelling, C. L. 1984. Enzyme-linked im- Pensaert, M. B., and DeBouck, P. 1978. A new coronavirus-
munosorbent assay for detection of transmissible gas- like particle associated with diarrhea in swine. Arch Vi-
troenteritis virus antibody in swine sera. Am J Vet Res rol 58:243–247.
45:1654–1657. Pensaert, M. B.; Haelterman, E. O.; and Burnstein, T. 1970a.
Nguyen, T. D.; Bottreau, E.; Bernard, S.; Lantier, I.; and Ay- Transmissible gastroenteritis of swine: Virus-intestinal
naud, J. M. 1986. Neutralizing secretory IgA and IgG do cell interactions. I. Immunofluorescence, histopatholo-
not inhibit attachment of transmissible gastroenteritis gy and virus production in the small intestine through
virus. J Gen Virol 67:939–943. the course of infection. Arch Gesamte Virusforsch
Noble, W. A. 1964. Methods used to combat transmissible 31:321–334.
gastroenteritis. Vet Rec 76:51. Pensaert, M. B.; Haelterman, E. O.; and Hinsman, E. J.
Noda, M.; Yamashita, H.; Koide, F.; Kadoi, K.; Omori, T.; 1970b. Transmissible gastroenteritis of swine: Virus-in-
Asagi, M.; and Inaba, Y. 1987. Hemagglutination with testinal cell interactions. II. Electron microscopy of the
transmissible gastroenteritis virus. Arch Virol epithelium in isolated jejunal loops. Arch Gesamte
96:109–115. Virusforsch 31:335–351.
Norman, J. O.; Lambert, G.; Moon, H. W.; and Stark, S. L. Pensaert, M.; Callebaut, P.; and Vergote, J. 1986. Isolation of
1973. Age dependent resistance to transmissible gas- a porcine respiratory, non-enteric coronavirus related to
troenteritis (TGE). II. Coronavirus titer in tissues of pigs transmissible gastroenteritis. Vet Q 8:257–261.
after exposure. Can J Comp Med 37:167–170. Pensaert, M.; Cox, E.; Van Deun, K.; and Callebaut, P. 1993.
Okaniwa, A.; Harada, K.; and Park, D. K. 1968. Structure of A sero-epizootiological study of porcine respiratory
swine transmissible gastroenteritis virus examined by coronavirus in Belgian swine. Vet Q 15:16–20.
negative staining. Natl Inst Anim Health Q (Tokyo) Phillip, J. I. H.; Cartwright, S. F.; and Scott, A. C. 1971. The
8:175–181. size and morphology of T.G.E. and vomiting and wast-
Onno, M.; Jestin, A.; Cariolet, R.; and Vannier, P. 1989. ing disease of pigs. Vet Rec 88:311–312.
Rapid diagnosis of TGEV-like coronavirus in fattened Pike, B. V., and Garwes, D. J. 1977. Lipids of transmissible
pigs by indirect immunofluorescence labelling in nasal gastroenteritis virus and their relation to those of two
cells. J Vet Med (B) 36:629–634. different host cells. J Gen Virol 34:531–535.
O’Toole, D.; Brown, I.; Bridges, A.; and Cartwright, S. F. Pilchard, E. I. 1965. Experimental transmission of transmis-
1989. Pathogenicity of experimental infection with sible gastroenteritis virus by starlings. Am J Vet Res
“pneumotropic” porcine coronavirus. Res Vet Sci 26:1177–1179.
47:23–29. Pocock, D. H., and Garwes, D. J. 1975. The influence of pH
Paton, D. J., and Brown, I. H. 1990. Sows infected in preg- on the growth and stability of transmissible gastroen-
nancy with porcine respiratory coronavirus show no ev- teritis virus in vitro. Arch Virol 49:239–247.
idence of protecting their suckling piglets against trans- Porter, P., and Allen, W. D. 1972. Classes of immunoglobu-
missible gastroenteritis. Vet Res Commun 14:329–337. lins related to immunity in the pig: A review. J Am Vet
Paul, P. S.; Mengeling, W. L.; Saif, L. J.; and Van Deresen, R. Med Assoc 160:511–518.
A. 1986. Detection of classes of antibodies to TGE and Posthumus, W. P.; Lenstra, J. A.; Schaaper, W. M.; Van
rotavirus of swine using monoclonal antibodies to Nieuwstadt, A. P., Enjuanes, L.; and Meloen, R. H. 1990.
322 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Analysis and simulation of a neutralizing epitope of teins. J Virol 70:4773–4777.


transmissible gastroenteritis virus. J Virol Roux, M. E.; McWilliams, M.; Phillips-Quagliata, J. M.;
64:3304–3309. Weisz-Carrington, P.; and Lamm, M. E. 1977. Origin of
Potopalsky, A. I.; Spivak, N. Y.; Shved, A. D.; Melnichenko, IgA-secreting cells in the mammary gland. J Exp Med
V. S.; and Krasnova, E. F. 1983. Activity of certain 146:1311.
chemopreparations with respect to the transmissible Rubinstein, D.; Tyrrell, A. J.; Derbyshire, J. B.; and Collins,
gastroenteritis virus and pig enterovirus. Mikrobiol Zh A. P. 1970. Growth of porcine transmissible gastroen-
45:75–78. In Russian with English abstract. teritis virus in organ cultures of pig tissue. Nature
Pritchard, G. C. 1982. Observations on clinical aspects of 227:1348–1349.
transmissible gastroenteritis of pigs in Norfolk and Suf- Saif, L. J. 1976. The Immune Response of Swine to Trans-
folk, 1980–81. Vet Rec 110:465–469. missible Gastroenteritis Virus. Ph.D. thesis, Ohio State
———. 1987. Transmissible gastroenteritis in endemically Univ.
infected breeding herds of pigs in East Anglia, ———. 1985. Passive immunity to coronavirus and ro-
1981–1985. Vet Rec 120:226–230. tavirus infections in swine and cattle: Enhancement by
Prochazka, Z.; Hampl, J.; Sedlacek, M.; Maasek, J.; and maternal vaccination. In Infectious Diarrhoea in the
Stepanek, J. 1975. Protein loss in piglets infected with Young. Ed. S. Tzipori. Amsterdam: Elsevier Science, p.
transmissible gastroenteritis virus. Zentralbl Veter- 456.
inärmed (B) 22:138–146. Saif, L. J., and Bohl, E. H. 1977. The immunoglobulin class-
Pulford, D. J.; Britton, P.; Page, K. W.; and Garwes, D. J. es of antibodies in milk of swine after intranasal expo-
1990. Expression of transmissible gastroenteritis virus sure to pseudorabies virus or transmissible gastroenteri-
structural genes by virus vectors. Adv Exp Med Biol tis virus. Infect Immun 16:961–966.
276:223. ———. 1979a. Role of SIgA in passive immunity of swine
Rasschaert, D., and Laude, H. 1987. The predicted primary to enteric viral infections. In Immunology of Breast
structure of the peplomer protein E2 of the porcine Milk. Ed. P. L. Ogra and D. H. Dayton. New York: Raven.
coronavirus transmissible gastroenteritis virus. J Gen Vi- ———. 1979b. Passive immunity in transmissible gas-
rol 68:1883–1890. troenteritis of swine: Immunoglobulin classes of milk
Rasschaert, D.; Gelfi, J.; and Laude, H. 1987. Enteric coron- antibodies after oral-intranasal inoculations of sows
avirus TGEV: Partial sequence of the genomic RNA, its with a live low cell culture–passaged virus. Am J Vet Res
organization and expression. Biochim 69:591–600. 40:115–117.
Rasschaert, D.; Duarte, M.; and Laude, H. 1990. Porcine res- ———. 1981a. Keynote address: Passive immunity against
piratory coronavirus differs from transmissible gas- enteric viral infections. Proc 3d Int Symp Neonatal Diar-
troenteritis virus by a few genomic deletions. J Gen Virol rhea. VIDO, Saskatoon, Canada.
71:2599–2607. ———. 1981b. Experimental studies using TGE vaccines. In
Redman, D. R.; Bohl, E. H.; and Cross, R. F. 1978. Intrafetal Ohio Swine Res Ind Rep, Anim Sci Ser 81-2, Ohio State
inoculation of swine with transmissible gastroenteritis Univ, pp. 58–59.
virus. Am J Vet Res 39:907–911. ———. 1983. Passive immunity to transmissible gastroen-
Register, K. B., and Wesley, R. D. 1994. Molecular charac- teritis virus: Intramammary viral inoculation of sows.
terization of attenuated vaccine strains of transmissible Ann NY Acad Sci 409:708–723.
gastroenteritis virus. J Vet Diagn Invest 6:16. Saif, L. J., and Theil, K. W., eds. 1990. Viral Diarrheas of
Reynolds, D. J., and Garwes, D. J. 1979. Virus isolation and Man and Animals. Boca Raton, Fla.: CRC Press.
serum antibody responses after infection of cats with Saif, L. J.; Bohl, E. H.; and Gupta, R. K. P. 1972. Isolation of
transmissible gastroenteritis virus. Arch Virol porcine immunoglobulins and determination of the im-
60:161–166. munoglobulin classes of transmissible gastroenteritis vi-
Reynolds, D. J.; Garwes, D. J.; and Lucey, S. 1980. Differen- ral antibodies. Infect Immun 6:600–609.
tiation of canine coronavirus and porcine transmissible Saif, L. J.; Bohl, E. H.; Kohler, E. M.; and Hughes, J. H. 1977.
gastroenteritis virus by neutralization with canine, Immune electron microscopy of transmissible gastroen-
porcine and feline sera. Vet Microbiol 5:283–290. teritis virus and rotavirus (reovirus-like agent) of swine.
Risco, C.; Antón, I. M.; Sune, C.; Pedregosa, A. M.; Martin- Am J Vet Res 38:13–20.
Alonso, J. M.; Parra, F.; Carrascosa, J. L.; and Enjuanes, Saif, L. J.; Van Cott, J. L.; and Brim, T. A. 1994. Immunity to
L. 1995. Membrane protein molecules of transmissible transmissible gastroenteritis virus and porcine respira-
gastroenteritis coronavirus also expose the carboxy-ter- tory coronavirus infections in swine. Vet Immunol Im-
minal region on the external surface of the virion. J Virol munopathol 43:89–97.
69:5269–5277. Sanchez, C. M.; Jimenez, G.; Laviada, M. D.; Correa, I.;
Risco, C.; Antón, I. M.; Enjuanes, L.; and Carrascosa, J. L. Sune, C.; Bullido, M. J.; Gebauer, F.; Smerdou, C.; Calle-
1996. The transmissible gastroenteritis coronavirus con- baut, P.; Escribano, J. M.; and Enjuanes, L. 1990. Anti-
tains a spherical core shell consisting of M and N pro- genic homology among coronaviruses related to trans-
CHAPTER 24 TRANSMISSIBLE GASTROENTERITIS Saif, Wesley 323

missible gastroenteritis virus. Virology 174:410–417. nant baculovirus-expressed TGEV spike (S) glycoprotein
Sasahara, J.; Harada, K.; Hayashi, S.; and Watanabe, M. vaccines. Am J Vet Res (in press).
1958. Studies on transmissible gastroenteritis in pigs in Siddell, S. G. 1995. The Coronaviridae: An introduction. In
Japan. Jpn J Vet Sci 20:1–6. The Coronaviridae. Ed. S. G. Siddell. New York: Plenum
Schultze, B.; Krempl, C.; Ballesteros, M. L.; Shaw, L.; Press, pp. 1–10.
Schauer, R.; Enjuanes, L.; and Herrler, G. 1996. Trans- Siddell, S. G.; Wege, H.; and Ter Meulen, V. 1983. The biol-
missible gastroenteritis coronavirus, but not the related ogy of coronaviruses. J Gen Virol 64: 761–776.
porcine respiratory coronavirus, has a sialic acid (N-gly- Simkins, R. A.; Weilnau, P. A.; Bias, J.; and Saif, L. J. 1992.
colylneuraminic acid) binding activity. J Virol Antigenic variation among transmissible gastroenteritis
70:5634–5637. virus (TGEV) and porcine respiratory coronavirus
Sestak, K.; Lanza, I.; Park, S. K.; Weilnau, P. A.; and Saif, L. strains detected with monoclonal antibodies to the S
J. 1996. Contribution of passive immunity to porcine protein of TGEV. Am J Vet Res 53:1253–1258.
respiratory coronavirus to protection against transmis- Simkins, R. A.; Weilnau, P. A.; Van Cott, J.; Brim, T.; and
sible gastroenteritis virus challenge exposure in suckling Saif, L. J. 1993. Competition ELISA, using monoclonal
pigs. Am J Vet Res 57:664–671. antibodies to the TGEV S protein, for serologic differen-
Shepherd, R. W.; Gall, D. G.; Butler, D. G.; and Hamilton, J. tiation of pigs infected with TGEV or porcine respirato-
R. 1979. Determinates of diarrhea in viral enteritis: The ry coronavirus. Am J Vet Res 54:254–259.
role of ion transport and epithelial changes in the ileum Smerdou, C.; Anton, I. M.; Plana, J.; Curtiss, R.; and En-
in transmissible gastroenteritis in piglets. Gastroen- juanes, L. 1996. A continuous epitope from transmissi-
terology 76:20–24. ble gastroenteritis coronavirus S protein fused to E. coli
Shibley, G. P.; Salsbury, D. L.; Djurickovic, S. M.; and John- heat-labile toxin B subunit expressed by attenuated Sal-
son, G. 1973. Application of an intramammary route of monella induces serum and secretory immunity. Virus
vaccination against transmissible gastroenteritis in Res 41:1–9.
swine. Vet Med Small Anim Clin 68:59–61. Spaan, W.; Cavanagh, D.; and Horzinek, M. C. 1988. Coron-
Shimizu, M., and Shimizu, Y. 1977. Micro-indirect hemag- aviruses: Structure and genomic expression. J Gen Virol
glutination test for detection of antibodies against 69:2939–2952.
transmissible gastroenteritis virus of pigs. J Clin Micro- Sprino, P. J., and Ristic, M. 1982. Intestinal, pulmonary, and
biol 6:91–95. serum antibody responses of feeder pigs exposed to
———. 1979a. Effects of ambient temperatures on clinical transmissible gastroenteritis virus by oral and oral-in-
and immune responses of pigs infected with transmissi- tranasal routes of inoculation. Am J Vet Res 43:255–261.
ble gastroenteritis virus. Vet Microbiol 4:109–116. Stark, S. L.; Fernelius, A. L.; Booth, G. D.; and Lambert, G.
———. 1979b. Demonstration of cytotoxic lymphocytes of 1975. Transmissible gastroenteritis (TGE) of swine: Ef-
virus-infected target cells in pigs inoculated with trans- fect of age of swine testes cell culture monolayers on
missible gastroenteritis virus. Am J Vet Res 40:208–213. plaque assays of TGE virus. Can J Comp Med
———. 1979c. Lymphocyte proliferative response to viral 39:466–468.
antigen in feeder pigs infected with transmissible gas- Stepanek, J.; Pospisil, Z.; and Mesaros, E. 1971. Growth ac-
troenteritis. Infect Immun 23:239–243. tivity of transmissible gastroenteritis (TGE) virus in pri-
Shimizu, M.; Shimizu, Y.; and Kodama, Y. 1978. Effects of mary cultures of pig kidney cells and pig salivary gland
ambient temperatures on induction of transmissible cells. Acta Vet Brno 40:235–240.
gastroenteritis in feeder pigs. Infect Immun 21:747–752. Stepanek, J.; Mensik, J.; Franz, J.; and Hornich, M. 1979.
Shirai, J.; Lantier, I.; Bottreau, E.; Aynaud, J. M.; and Epizootiology, diagnosis and prevention of viral diar-
Bernard, S. 1988. Lactogenic immunity to transmissible rhoea in piglets under intensive husbandry conditions.
gastroenteritis of swine induced by attenuated Nouzilly Proc 21st World Vet Congr, Moscow, 6:43.
strain of TGE virus: Neutralizing antibody classes and Stone, S. S.; Kemeny, L. J.; Woods, R. D.; and Jensen, M. T.
protection. Ann Rech Vet 19:267–272. 1977. Efficacy of isolated colostral IgG, IgA and IgM(A)
Shockley, L. J.; Kapke, P. A.; Lapps, W.; Brian, D. A.; Potgi- to protect neonatal pigs against the coronavirus of
eter, L. D.; and Woods, R. 1987. Diagnosis of porcine transmissible gastroenteritis. Am J Vet Res
and bovine enteric coronavirus infections using cloned 38:1285–1288.
cDNA probes. J Clin Microbiol 25:1591–1596. Stone, S. S.; Kemeny, L. J.; and Jensen, M. T. 1982. Serum
Shoup, D. I.; Swayne, D. E.; Jackwood, D. J.; and Saif, L. J. antibody responses of neonatal and young adult pigs to
1996. Immunocytochemistry of transmissible gastroen- transmissible gastroenteritis coronavirus. Vet Immunol
teritis virus antigens in fixed paraffin-embedded tissues. Immunopathol 3:529–533.
J Vet Diag Invest 8:161–167. Sune, C.; Jimenez, G.; Correa, I.; Bullido, M. J.; Gebauer, F.;
Shoup, D. I.; Jackwood, D. J.; and Saif, L. J. 1997. Active and Smerdou, C.; and Enjuanes, L. 1990. Mechanisms of
passive immune responses to transmissible gastroen- transmissible gastroenteritis coronavirus neutralization.
teritis virus (TGEV) in swine inoculated with recombi- Virology 177:559–569.
324 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Thake, D. C. 1968. Jejunal epithelium in transmissible gas- ry or enteric strains of coronavirus to immunity against
troenteritis of swine (an electron microscopic and histo- enteric coronavirus challenge. J Immunol
chemical study). Am J Pathol 53:149–168. 152:3980–3990.
Theil, K. W.; Saif, L. J.; Bohl, E. H.; Agnes, A. G.; and Kohler, Vannier, P. 1990. Disorders induced by the experimental in-
E. M. 1979. Concurrent porcine rotaviral and transmis- fection of pigs with the porcine respiratory coronavirus
sible gastroenteritis viral infections in a three-day-old (P.R.C.V.). J Vet Med (B) 37:177–180.
conventional pig. Am J Vet Res 40:719–721. Vannier, P.; Toma, B.; Madec, F.; and Aynaud, J. M. 1982.
Thomas, F. C., and Dulac, G. C. 1976. Transmissible gas- Valuation of duration of TGE virus spread among sows
troenteritis virus: Plaques and a plaque neutralization of 2 infected herds by means of a serological survey of
test. Can J Comp Med 40:171–174. antibodies persistence. Proc Int Congr Pig Vet Soc 7:3.
Thorsen, J., and Djurickovic, S. 1971. Experimental immu- Van Nieuwstadt, A. P., and Boonstra, J. 1992. Comparison of
nization of sows with inactivated TGE virus. Can J Comp the antibody response to transmissible gastroenteritis
Med 35:99–102. virus and porcine respiratory coronavirus, using mono-
Toma, B., and Benet, J. J. 1976. A technique of research on clonal antibodies to antigenic sites A and X of the S gly-
microplates of the antibodies neutralizing transmissible coprotein. Am J Vet Res 53:184–190.
gastroenteritis virus of swine. Rec Med Vet Van Nieuwstadt, A. P., and Pol, J. M. A. 1989. Isolation of a
152:565–568. TGE virus–related respiratory coronavirus causing fatal
Toma, B.; Vannier, P.; and Aynaud, J. M. 1978. Epidemio- pneumonia in pigs. Vet Rec 124:43–44.
logical studies on porcine transmissible gastroenteritis Van Nieuwstadt, A. P.; Cornelissen, J. B.; and Zetstra, T.
in France. Rec Med Vet 154:853–858. 1988. Comparison of two methods for detection of
Torres, J. M.; Alonso, C.; Ortega, A.; Mittal, S.; Graham, F.; transmissible gastroenteritis virus in feces of pigs with
and Enjuanes, L. 1996. Tropism of human adenovirus experimentally induced infection. Am J Vet Res
type 5–based vectors in swine and their ability to protect 49:1836–1843.
against transmissible gastroenteritis coronavirus. J Virol Van Nieuwstadt, A. P.; Zetstra, T.; and Boonstra, J. 1989. In-
70:3770–3780. fection with porcine respiratory coronavirus does not
Tuboly, T.; Nagy, E.; Dennis, J. R.; and Derbyshire, J. B. fully protect pigs against intestinal transmissible gas-
1994. Immunogenicity of the S protein of transmissible troenteritis virus. Vet Rec 125:58–60.
gastroenteritis virus expressed in baculovirus. Arch Vi- Van Reeth, K., and Pensaert, M. 1992. Experimental infec-
rol 137:55–67. tions with different porcine respiratory coronavirus field
Tuboly, T.; Nagy, E.; and Derbyshire, J. B. 1995. Passive pro- isolates: Clinical and virological aspects. Proc Int Congr
tection of piglets by recombinant baculovirus induced Pig Vet Soc 12:152.
transmissible gastroenteritis virus specific antibodies. ———. 1994a. Prevalence of infections with enzootic res-
Can J Vet Res 59:70–72. piratory and enteric viruses in feeder pigs entering fat-
Underdahl, N. R.; Mebus, C. A.; Stair, E. L.; and Twiehaus, tening herds. Vet Rec 135:594–597.
M. J. 1972. The effect of cytopathogenic transmissible ———. 1994b. Porcine respiratory coronavirus–mediated
gastroenteritis–like viruses and/or Escherichia coli on interference against influenza virus replication in the
germfree pigs. Can Vet J 13:9–16. respiratory tract of feeder pigs. Am J Vet Res
Underdahl, N. R.; Mebus, C. A.; and Torres-Medina, A. 55:1275–1281.
1975. Recovery of transmissible gastroenteritis virus ———. 1996. A clinical and virological study in pigs infect-
from chronically infected experimental pigs. Am J Vet ed with Aujeszky’s disease virus shortly after infection
Res 36:1473–1476. with porcine respiratory coronavirus. Proc Int Congr Pig
Underdown, B. J., and Dorrington, K. J. 1974. Studies on the Vet Soc 14:137.
structural and conformational basis for the relative re- Van Reeth, K.; Nauwynck, H.; and Pensaert, M. 1996. Dual
sistance of serum and secretory immunoglobulin A to infections of feeder pigs with porcine reproductive and
proteolysis. J Immunol 112:949–959. respiratory syndrome virus followed by porcine respira-
USDA APHIS VS. 1992. National Animal Health Monitor- tory coronavirus or swine influenza virus: A clinical and
ing System Survey (Data Collection: 1989–1990). Tech virological study. Vet Microbiol 48:325–335.
Rep, USDA APHIS VS. Vaughn, E. M.; Halbur, P. G.; and Paul, P. S. 1994. Three new
Van Cott, J. L.; Brim, T. A.; Simkins, R. A.; and Saif, L. J. isolates of porcine respiratory coronavirus with various
1993. Antibody-secreting cells to transmissible gas- pathogenicities and spike (S) gene deletions. J Clin Mi-
troenteritis virus and porcine respiratory coronavirus in crobiol 32:1809–1812.
gut- and bronchus-associated lymphoid tissues of ———. 1995. Sequence comparison of porcine respiratory
neonatal pigs. J Immunol 150:3990–4000. coronavirus isolates reveals heterogeneity in the S, 3,
Van Cott, J. L.; Brim, T. A.; Lunney, J. K.; and Saif, L. J. 1994. and 3-1 genes. J Virol 69:3176–3184.
Contribution of antibody secreting cells induced in mu- ———. 1996. Use of nonradioactive cDNA probes to dif-
cosal lymphoid tissues of pigs inoculated with respirato- ferentiate porcine respiratory coronavirus and transmis-
CHAPTER 24 TRANSMISSIBLE GASTROENTERITIS Saif, Wesley 325

sible gastroenteritis virus isolates. J Vet Diagn Invest in Iowa swine. Can J Vet Res 61:305–308.
8:241–244. Wesseling, J. G.; Vennema, H.; Godeke, G.-J.; Horzinek, M.
Voets, M. T.; Pensaert, M.; and Rondhuis, P. R. 1980. Vacci- C.; and Rottier, J. M. 1994. Nucleotide sequence and ex-
nation of pregnant sows against transmissible gastroen- pression of the spike (S) gene of canine coronavirus and
teritis with two attenuated virus strains and different in- comparison with the S proteins of feline and porcine
oculation routes. Vet Q 2:211–219. coronaviruses. J Gen Virol 75:1789–1794.
Wagner, J. E.; Beamer, P. D.; and Ristic, M. 1973. Electron Whitenack, D. L.; Whitehair, C. K.; and Miller, E. R. 1978.
microscopy of intestinal epithelial cells of piglets infect- Influence of enteric infection on zinc utilization and
ed with a transmissible gastroenteritis virus. Can J Comp clinical signs and lesions of zinc deficiency in young
Med 37:177–188. swine. Am J Vet Res 39:1447–1454.
Waxler, G. L. 1972. Lesions of transmissible gastroenteritis Witte, K. H. 1971. Isolation of the virus of transmissible
in the pig as determined by scanning electron mi- gastroenteritis (TGE) from naturally infected piglets in
croscopy. Am J Vet Res 33:1323–1328. cell cultures. Zentralbl Veterinärmed (B) 18:770–778.
Weingartl, H. M., and Derbyshire, J. B. 1994. Evidence for a Witte, K. H., and Walther, C. 1976. Age-dependent suscep-
putative second receptor for porcine transmissible gas- tibility of pigs to infection with the virus of transmissi-
troenteritis virus on the villous enterocytes of newborn ble gastroenteritis. Proc Int Congr Pig Vet Soc 4:K3.
pigs. J Virol 68:7253–7259. Wood, E. N. 1979. Transmissible gastroenteritis epidemic
Welch, S.-K. W., and Saif, L. J. 1988. Monoclonal antibodies diarrhoea of pigs. Br Vet J 135:305–314.
to a virulent strain of transmissible gastroenteritis virus: Woods, R. D. 1977. Leukocyte migration-inhibition proce-
Comparison of reactivity against the attenuated and vir- dure for transmissible gastroenteritis viral antigens. Am
ulent virus strain. Arch Virol 101:221–235. J Vet Res 38:1267–1269.
Welch, S.-K. W.; Saif, L. J.; and Ram, S. 1988. Cell-mediated ———. 1978. Small plaque variant transmissible gastroen-
immune responses of nursing pigs inoculated with at- teritis virus. J Am Vet Med Assoc 173:643–647.
tenuated or virulent transmissible gastroenteritis virus. ———. 1982. Studies of enteric coronaviruses in a feline
Am J Vet Res 49:1228–1234. cell line. Vet Microbiol 7:427–435.
Welter, C. J. 1965. TGE of swine. I. Propagation of virus in ———. 1984. Efficacy of vaccination of sows with serolog-
cell cultures and development of a vaccine. Vet Med ically related coronaviruses for control of transmissible
Small Anim Clin 60:1054–1058. gastroenteritis in nursing pigs. Am J Vet Res
———. 1980. Experimental and field evaluation of a new 45:1726–1729.
oral vaccine for TGE. Vet Med Small Anim Clin Woods, R. D., and Pedersen, N. C. 1979. Cross-protection
75:1757–1759. studies between feline infectious peritonitis and porcine
Wesley, R. D., and Woods, R. D. 1993. Immunization of transmissible gastroenteritis viruses. Vet Microbiol
pregnant gilts with PRCV induces lactogenic immunity 4:11–16.
for protection of nursing piglets from challenge with Woods, R. D., and Wesley, R. D. 1992. Seroconversion of
TGEV. Vet Microbiol 38:40. pigs in contact with dogs exposed to canine coronavirus.
———. 1996. Induction of protective immunity against Can J Vet Res 56:78–80.
transmissible gastroenteritis virus after exposure of Woods, R. D.; Cheville, N. F.; and Gallagher, J. E. 1981. Le-
neonatal pigs to porcine respiratory coronavirus. Am J sions in the small intestine of newborn pigs inoculated
Vet Res 57:157–162. with porcine, feline, and canine coronaviruses. Am J Vet
Wesley, R. D.; Woods, R. D.; and Cheung, A. K. 1990a. Ge- Res 42:1163–1169.
netic basis for the pathogenesis of transmissible gas- Woods, R. D.; Wesley, R. D.; and Kapke, P. A. 1988. Neu-
troenteritis virus. J Virol 64:4761–4766. tralization of transmissible gastroenteritis virus by com-
Wesley, R. D.; Woods, R. D.; Hill, H. T.; and Biwer, J. D. plement dependent monoclonal antibodies. Am J Vet
1990b. Evidence for a porcine respiratory coronavirus, Res 49:300–304.
antigenically similar to transmissible gastroenteritis Yaling, Z.; Ederveen, J.; Egberink, H.; Pensaert, M.; and
virus, in the United States. J Vet Diagn Invest 2:312–317. Horzinek, M. C. 1988. Porcine epidemic diarrhea virus
Wesley, R. D.; Wesley, I. V.; and Woods, R. D. 1991a. Differ- (CV777) and feline infectious peritonitis virus (FIPV)
entiation between transmissible gastroenteritis virus are antigenically related. Arch Virol 102:63–71.
and porcine respiratory coronavirus using a cDNA Young, G. A.; Hinz, R. W.; and Underdahl, N. R. 1955. Some
probe. J Vet Diagn Invest 3:29–32. characteristics of transmissible gastroenteritis in dis-
Wesley, R. D.; Woods, R. D.; and Cheung, A. K. 1991b. Ge- ease-free antibody-devoid pigs. Am J Vet Res
netic analysis of porcine respiratory coronavirus, an at- 16:529–535.
tenuated variant of transmissible gastroenteritis virus. J Zhu, X. L.; Paul, P. S.; Vaughn, E.; and Morales, A. 1990.
Virol 65:3369–3373. Characterization and reactivity of monoclonal antibod-
Wesley, R. D.; Woods, R. D.; McKean, J. D.; Senn, M. K.; and ies to the Miller strain of transmissible gastroenteritis
Elazhary, Y. 1997. Prevalence of coronavirus antibodies virus of swine. Am J Vet Res 51:232–238.
Vesicular Diseases
25 J. A. House and C. A. House

Vesicular diseases in swine are identical clinically. They World War I (Cotton 1927), but infection in swine was
can be caused by infection with the viruses of foot-and- not reported until 1943 (Schoening 1943). The first re-
mouth disease (FMD), vesicular stomatitis (VS), vesicu- port of VES was in 1932 in California (Traum 1934), al-
lar exanthema of swine (VES), and swine vesicular dis- though it was initially thought to be FMD. A new vesicu-
ease (SVD). FMD, VES, and SVD are exotic to the United lar disease, SVD, emerged in Italy in 1966 (Nardelli et al.
States, while VS is enzootic. The importance of vesicular 1968). San Miguel sea lion viruses (SMSVs), isolated
diseases in today’s world-trade environment remains from marine mammals, were shown to cause vesicular
high. Because FMD is extremely contagious and has such disease in inoculated swine (Smith et al. 1973; Berry et al.
a dramatic economic impact, vesicular diseases must be 1990). The history and distribution of these viruses are
properly diagnosed, reported, and controlled. given in Table 25.1.
In Italy in 1546, Fracastorius made what is probably
the first report of a vesicular disease (Bulloch 1927). ETIOLOGY
FMD was the first animal disease shown to be caused by
a filterable agent (Loeffler and Frosch 1898). VS was rec- The characteristics of the viruses that cause vesicular dis-
ognized in horses and cattle in the United States during eases are shown in Table 25.2.

Table 25.1. Chronological and geographical distribution of vesicular diseases


Disease First Agent
Virus Reported Identified Distribution Areas Free
Foot-and-mouth disease 1546 1898 Sporadic: Europe North and Central America,
Enzootic: South America, Australia, New Zealand,
Asia, Africa Japan
Swine vesicular disease 1966 1968 Europe, Japan, Hong Kong, Taiwan North, Central, and South
America; Africa; United
Kingdom
Vesicular exanthema of swine 1932 1934 Last North American case in 1956 World
San Miguel sea lion None 1973 Pacific coast of North America Remainder of world
Vesicular stomatitis 1916 1927 North, Central, and South America Remainder of world

Table 25.2. Classification of the etiologic agents of vesicular diseases


Disease Agents Classification Nucleic Acid Size and Stability Proteins/Antigens Serotypes
a
Foot-and-mouth Picornaviridae SS RNA 22–30 nm; labile 4 structural 7: numerous subtypes
disease virus Aphthovirus (positive below pH 7.0; proteins, plus a show varied degrees of
sense) ether-resistant group of reactive cross-protection within
nonstructural each serotype
proteins
Swine vesicular Picornaviridae SS RNA 22–30 nm; acid- 4 structural 1: related to Coxsackie
disease virus Enterovirus (positive stable; ether- proteins B-5 virus
sense) resistant
Vesicular Caliciviridae SS RNA 35–39 nm; labile 1 polypeptide in 26 or more (13 known for
exanthema of Calicivirus (positive below pH 3; ether- coat; plus group VESVs; at least 13 for
swine/San Miguel sense) resistant reactive antigens SMSVs)
sea lion viruses
Vesicular stomatitis Rhabdoviridae SS RNA 70 × 170 nm; bullet- 1 immunogenic 2 of importance to swine:
virus Vesiculovirus (negative shaped; ether-labile; glycoprotein New Jersey and
sense) stable at pH 5–10 Indiana 1
a
SS RNA = single-stranded RNA.
327
328 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Foot-and-Mouth Disease closest relative is the human enterovirus Coxsackie B-5


FMD viruses (FMDVs) are in the family Picornaviridae (Graves 1973).
and the genus Aphthovirus (Francki et al. 1991c). Aphtha
is the Greek word for “vesicles in the mouth.” These Vesicular Exanthema of Swine and San
viruses are small (22–30 nm), nonenveloped, and labile Miguel Sea Lion Viruses
below pH 7.0, and they have single-stranded positive- The VESVs and SMSVs, members of the Caliciviridae
sense RNA with approximately 8450 nucleotides that family, are closely related caliciviruses (Smith et al.
serve as messenger RNA. The high rate of nucleotide 1973), named from the Latin calices (cups) for the cup-
substitutions in the single-stranded RNA viruses ac- shaped depressions on the virion. The viruses are 35–39
counts for their wide variation (Domingo et al. 1993; nm in diameter, nonenveloped, moderately acid-stable,
Vosloo et al. 1996). Perhaps one of the most important and contain single-stranded positive-sense RNA (Francki
sites for substitutions is the GH loop (codons 133–158) et al. 1991b). The capsomers are composed of one major
of viral protein 1 (VP1) (Acharya et al. 1989), which is polypeptide.
adjacent to the common aphthovirus receptor site RGD There are 13 known serotypes of VESVs, which are
(Fox et al. 1989). There are four structural viral proteins, sequentially designated (except for strains 1934B and
of which VP1 has been shown to elicit protective immu- 101) by a letter followed by the year of isolation (e.g.,
nity in cattle (Bachrach 1977). Viral RNA polymerase, a VESV A48). There are at least 13 SMSV serotypes, which
nonstructural protein, has been termed “virus infection are numbered consecutively (with no SMSV 3, which
associated antigen” (VIAA) for diagnostic purposes but does not exist) in order of discovery.
is more accurately termed “FMDV nonstructural protein
3D.” One copy of 3D occurs in each viral particle (New- Vesicular Stomatitis
man et al. 1994). The FMDV nonstructural protein 2C is The bullet-shaped VSVs belong to the family Rhabdoviri-
present in clarified FMD vaccines at levels insufficient to dae, genus Vesiculovirus. The viral particles are 70 × 170
elicit an immune response (Lubroth et al. 1996). nm, enveloped, ether-labile, and stable at pH 5–10, and
There are seven serotypes of FMDV: A, O, C, Asia 1, they contain a single-stranded, negative-sense RNA.
and South African Territories (SAT) 1, 2, and 3. There is There are five structural viral proteins: L, G, N, NS, and
no cross-protection between the serotypes (Brooksby M. The glycosylated G protein confers the type specifici-
1982). All serotypes have numerous subtypes, which ty and elicits neutralizing and hemagglutinating anti-
may arise during acute or persistent infection (Gebauer bodies (Francki et al. 1991a). The viral nucleoprotein N
et al. 1988; Vosloo et al. 1996). The serologic relatedness and the matrix protein M account for serologic-group
of subtypes is determined by “r-values,” the ratio of an- cross-reactions. There are at least 11 vesiculovirus
tibody titers between new and reference viruses (Pereira serotypes (Tesh et al. 1983; Travassos da Rosa et al.
1977). 1984), of which serotypes New Jersey (NJ), Indiana 1, In-
diana 2, and Indiana 3 are of importance to livestock.
Swine Vesicular Disease
SVD virus (SVDV) is in the family Picornaviridae, genus EPIDEMIOLOGY
Enterovirus (Francki et al. 1991c). The virions are small
(22–30 nm), nonenveloped, and acid-stable. Each has a The epidemiological features of the vesicular diseases are
single-stranded positive-sense RNA and four structural summarized in Table 25.3.
viral proteins. There is one serotype of SVDV, whose

Table 25.3. Epidemiological features of vesicular diseases


Main Species Contamination of
Disease Agent Affected Transmission Morbidity Carrier State Meat and By-Products
Foot-and-mouth Swine, cattle, sheep, Aerosol, contact, High Cattle, sheep, goats, Frozen meat, lymph
disease virus goats, African fomites African buffaloes nodes, bone marrow,
buffaloes; some milk products
strains are host
restricted
Swine vesicular Swine, humans Contact, fomites Moderate No Virus persists in cured meat
disease virus and gland products
Vesicular Swine, various Contact, fomites Moderate No Meat
exanthema of marine animals
swine/San Miguel
sea lion viruses
Vesicular stomatitis Horses, cattle, Biting insects; some Moderate to No Not documented
virus swine, humans by contact low
CHAPTER 25 VESICULAR DISEASES House, House 329

Foot-and-Mouth Disease has occurred nine times in the United States, with the
FMD is considered the most contagious disease of live- last outbreak in 1929. Serotypes A, O, and C occur in
stock. Essentially all cloven-footed species are suscepti- South America, and six serotypes (A, O, and C, SAT-1,
ble to FMDV. In some texts, the possibility of human in- SAT-2, and SAT-3) occur in Africa. The SAT serotypes are
fection is stated, but this has yet to be documented. generally restricted to sub-Saharan Africa. Serotypes A,
Infected animals shed large quantities of virus into their O, C, and Asia 1 occur in Asia and the Middle East.
surroundings for at least 7 days. Aerosol exposure is the Japan, Australia, New Zealand, and certain Pacific is-
major means of spread, particularly in temperate cli- lands are free of FMD. A consistent vaccination program
mates, where high humidity and moderate temperatures in Western Europe and most of Eastern Europe greatly
can favor long-distance airborne spread (Sellers and diminished the prevalence of FMD. The European Union
Parker 1969; Thomson 1994). Tremendous aerosols are stopped vaccinating for FMD in January 1992. Sporadic
generated by swine, which are often called an “amplifier local outbreaks occurred in Italy in 1993 (Maragon et al.
host” (Sellers and Parker 1969; Donaldson and Ferris 1994), in the Balkans in 1995, and in sheep in Greece in
1980; Donaldson et al. 1987). Swine may produce up to 1996. These outbreaks were controlled either by stamp-
108 infectious doses contained in aerosols per day during ing out or by stamping out and vaccination.
peak viremia (Sellers et al. 1971), which is up to 1500 Stringent importation regulations (Section 306A of
times greater than that produced by cattle (Donaldson the Tariff Act of 1930 and acts of 1890 and 1903) on an-
and Ferris 1980). In hot, arid regions, the disease is main- imals and animal products have effectively protected the
ly spread by direct contact, as aerosols cannot travel long FMD-free status of the United States. The U.S. Depart-
distances due to low humidity and high temperatures. ment of Agriculture (USDA) conducts research and diag-
Other means of spread include contaminated semen, nostic studies with FMDV at its high-containment facili-
meat and milk products, and fomites. Semen from in- ties (greater than biosafety level 3) at the Plum Island
fected cattle contained and transmitted FMDV by artifi- Animal Disease Center (PIADC).
cial insemination (AI) (Cottral et al. 1968), whereas se- FMD results in direct economic losses through the
men from infected swine contained FMDV but did not loss of meat, milk, draft power, and genetic stocks, and
transmit the disease by AI (McVicar et al. 1978). indirect losses through embargoes and loss of trade. The
The most dramatic proof of carrier buffaloes infect- direct loss from FMD during the first year of an outbreak
ing contact animals was shown by Vosloo et al. (1996). in the United States was estimated at $4 billion, with in-
Sporadically over two years, both contact cattle and buf- direct costs up to 10 times more (Blackwell 1984).
faloes became infected by a cloned SAT-2 virus. The nu-
cleotide sequence varied significantly from the original Swine Vesicular Disease
clone (1.649% nucleotide substitution/year). An inadver- SVD is a moderately contagious disease of swine. Mice
tent introduction of a SAT-1 strain infected some buf- may be infected experimentally. Persons working with or
faloes, and ultimately these carrier animals infected an- around infected pigs can harbor SVDV in their nasal pas-
other buffalo. The nucleotide substitution rate for the sages (Sellers and Herniman 1974), and human infection
SAT-1 strain was 1.549% per year. African buffaloes have has been observed (Brown et al. 1976). Swine vesicular
clearly transmitted FMDV to cattle (Dawe et al. 1994; disease virus is spread primarily by contact with infected
Hedger and Condy 1985), as well as under experimental swine or by garbage feeding of SVDV-contaminated
conditions (Dawe et al. 1994). The major transmission of meat products. Fecal transmission is not common (Chu
FMDV from African buffaloes to cattle occurs when year- et al. 1979; Mann and Hutchings 1980), which is unusu-
ling buffalo calves, whose maternal antibody has waned, al for an enterovirus. The SVDV can survive up to 38
become infected with virus from carrier buffaloes. It is days in a pH range of 3.9–9.1 at refrigerator tempera-
hypothesized that buffalo calves then shed large quanti- tures (Herniman et al. 1973). Earthworms from soil
ties of virus and pass the virus to susceptible cattle dur- above pits used to bury infected pigs yielded SVDV when
ing close contact or by sharing water holes (Thomson their gut tracts and surfaces were sampled, emphasizing
1995). the ability of SVDV to persist in the environment
Some strains of FMDV may be host restricted under (Coombs 1973). SVDV persisted for 560 days in the
field conditions, as exemplified by the porcinophilic na- lymph nodes of hams prepared from infected swine
ture of the O1 strain that infected virtually the entire (Mebus et al. 1993b). The feeding of infected meat rep-
swine population of Taiwan in the spring of 1997. No in- resents a disease threat even though the oral route of in-
fections were reported in the 300,000 Taiwanese cattle. fection may require at least 300,000 infectious units
Experimentally, O1 Taiwan did not readily infect cattle (Mann et al. 1975; Mann and Hutchings 1980). Abraded
even when administered in massive doses (Dunn and mucosa is more readily infected, requiring only about
Donaldson 1997). 100 infectious particles.
On the North American continent, FMD last oc- There is no known carrier state for SVDV. Outbreaks
curred in Mexico in 1946–54 and in Canada in 1952. It of SVD in Hong Kong (1970) and in England (1972) once
330 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

defined its eastern and western limits. Recently, SVD has clared an exotic disease in 1959 in the United States, and
been reported in Italy (1995 and 1996) and in Portugal all countries worldwide remain free of VES.
(1995).
Vesicular Stomatitis
Vesicular Exanthema of Swine and San VS is considered to be a disease of moderate to low con-
Miguel Sea Lion Viruses tagious potential, as it is primarily an insect-borne dis-
VES is a moderately contagious disease. Under certain ease. The VSVs can cause clinical disease or subclinical
management situations—namely, the feeding of infect- infections in a wide range of animals, including swine,
ed seal meat or contaminated garbage—swine were the cattle, horses, and humans.
only livestock to show clinical VES. Marine animals (fish, The origin of outbreaks and means of dissemination
sea lions, fur seals, elephant seals, etc.) along the Pacific of VSV is not thoroughly understood. The ecological dis-
coast from California to Alaska and likely along the coast tribution of VS is river basins and wooded areas, point-
of northeastern Russia are apparently the natural hosts ing to insect transmission. Serotypes Indiana 1 and NJ
of the numerous serotypes of VESVs and SMSVs. A cali- have been isolated from sand flies (Lutzomyia shannoni),
civirus was isolated from a California sea lion (Zalophus and transovarial transmission was demonstrated (Tesh
californianus) from San Miguel Island and designated et al. 1971; Comer et al. 1990). Sand flies infected with
San Miguel sea lion virus (Smith et al. 1973). Numerous VSV NJ transmitted the virus to suckling mice (Comer et
serotypes of SMSVs were isolated from fish and marine al. 1990). Studies on Ossabaw Island, Georgia, have
mammals (Smith et al. 1977, 1979, 1980, 1983b) and shown that feral swine become seropositive to VSV NJ
from frozen meat of fur seals fed to mink (Sawyer et al. during late spring months when insect activity resumes
1978). Bovine calicivirus (BCV, Tillamook virus), a cali- (Stallknecht et al. 1985). Black flies were shown to bio-
civirus serotype isolated from three dairy calves in Ore- logically transmit VSV NJ in their saliva for up to 10 days
gon (Smith et al. 1983a), may be of marine origin, as an- after feeding on viral fluids (Cupp et al. 1992). Fomites
tibodies to BCV were found in various marine mammals may play a role in the spread of VS in swine. Contact
(Barlough et al. 1987). Many SMSVs have been shown to transmission may occur (Schoening 1943) and apparent-
produce vesicular disease in experimentally inoculated ly requires abrasions in the epithelium (Patterson et al.
swine (Smith et al. 1973; Berry et al. 1990). Low levels of 1955). A carrier state for VSV was not detected by virus
antibodies to VESVs and SMSVs have been found in ter- isolation or nucleic acid hybridization studies (Redelman
restrial mammals (feral swine, foxes, buffaloes, donkeys, et al. 1989).
and cattle) on the west coast of the United States and on VS has been recognized in the United States since at
Santa Barbara Channel Islands (Smith and Latham least 1916 (Cotton 1927). Vesicular disease outbreaks in
1978). The relationship of antibodies to natural disease cattle and horses in 1925 and 1926 were caused by infec-
is not clear. The infection of humans with SMSVs is in- tion with two distinct serotypes of VSV (Cotton 1927),
ferred but not corroborated. later called Indiana 1 and New Jersey (NJ). VS was first
The VESVs were shown to be present in the meat of described in U.S. swine that were used for the production
infected swine (Mott et al. 1953) and are spread mainly of hog cholera hyperimmune serum in Missouri in 1943
by feeding of infected meats, contact, or fomites (Schoening 1943). In the United States, epizootics, most-
(Bankowski et al. 1955). Long-term carriers have not ly serotype NJ, occur about every 10–13 years, starting in
been shown for VESV. early summer and ending with the onset of freezing
VES was confined to California from its first descrip- weather. These epizootics generally start in the south-
tion in 1932 until 1951, when pork trimmings from an western states and then spread northward into the Rocky
interstate California passenger train were fed to swine in Mountain states. The disease primarily affects horses
Wyoming. The ensuing epizootic involved 42 states plus and cattle, with an occasional spillover to swine. The
the District of Columbia. The last foci of VES were three 1982–83 VS NJ epizootic persisted into winter, causing
large garbage-feeding operations in Secaucus, New Jer- speculation that VS may be altering its epidemiological
sey. The epizootic was controlled in 1956, costing $33 pattern. Movement of infected animals extended the
million in direct costs and indemnities (Mulhern 1953). scope of this outbreak to Idaho and California. The 1995
Two cases of VES occurred outside the United States. outbreak of VS NJ was typical, being confined to western
Butcher hogs aboard ships en route to Hawaii in 1946 states and disappearing with the onset of cold weather.
and 1947 were intercepted and were slaughtered. The VS NJ is endemic in the coastal regions of Georgia (Stall-
second occurrence was in swine fed uncooked garbage knecht et al. 1985).
from a U.S. military base in Iceland in 1955, which was In Central and South America and in Mexico,
also promptly controlled (Bankowski 1965). After enact- serotypes NJ and Indiana 1 have been associated with VS
ment of laws prohibiting feeding of raw garbage to in cattle, horses, and swine annually. Subtypes Indiana 2
swine and requiring the slaughter of infected animals, and Indiana 3 were isolated from mites from rice rats in
VES was eradicated in the United States. VES was de- Trinidad (Jonkers et al. 1964) and in outbreaks of VS in
CHAPTER 25 VESICULAR DISEASES House, House 331

Argentina and Brazil (Garcia Perazzi et al. 1963), respec- epithelium become blanched, followed by the formation
tively. Indiana 2 and Indiana 3 (Federer et al. 1967) occur of vesicles and erosions after loss of the epithelium. Vesi-
in Central and South America, respectively, but have not cles up to 3 cm in diameter may be found on the snout,
been reported to produce clinical disease in swine under with vesicular lesions extending into the nares, on the
field conditions. There are numerous other serotypes of lips, tongue, hard and soft palate, coronary band, on the
vesiculoviruses (House and Wilks 1983; Tesh et al. 1983; soft tissues of the feet (interdigital clefts and bulbs), and
Travassos da Rosa et al. 1984) but none has been report- soft tissues around the dewclaws (see Figs. 25.1–25.4).
ed to cause vesicular disease in swine under natural con- Vesicular lesions may occur in other areas of the skin, es-
ditions. pecially where there is mechanical pressure or abrasion.
Nursing sows may have vesicles on the teats. Slobbering
CLINICAL SIGNS and chomping are common. Pregnant sows may abort
due to fever. The viruses of FMD, SVD, VES, and VS are
The clinical signs of FMD, VS, VES, SVD, and SMSV not reported to infect the fetus.
cannot be distinguished from each other in the field. Vesicles yield a serous fluid if they rupture (usually
Generally, within 1–5 days postexposure, the body tem- 6–24 hours after formation), and the epithelial layers
perature rises sharply to 40.5˚C or higher. Areas of the above the stratum germinativum may detach. Eroded,

25.1. Lesions on a pig’s foot, the


second day of clinical signs of
FMD. Note blanching (arrow A),
cracking of the epithelium (arrow
B), and an erosion left where
epithelium has been lost (arrow C).

25.2. Lesions on a pig’s foot,


the third day of clinical signs of
FMD. Note the blanched coronary
band of the dewclaw (arrow A)
and debris adhering to vesicular
tissue on the coronary band of the
foot (arrow B).
332 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

25.3. Lesions on a pig’s snout,


the first day of clinical signs of
FMD. Note the vesicles (arrows).

25.4. Lesions on a pig’s snout,


the third day of clinical signs of
FMD. Note the crusty areas (arrow
A) and darker areas where vesicles
have ruptured and fluid has leaked
through the epithelium (arrow B).

hyperemic, hemorrhagic lesions are present. The hoof diseases. In young animals infected with FMDV or
may become detached if the vesicles have coalesced SVDV, myocardial necrosis may occur and mortality may
around the coronary band. Severe laminitis may be fol- reach 50–100%. There have been occasional reports of
lowed by a chronically deformed foot. After the initial specific FMD isolates causing high mortality in mature
stage of infection, the animal’s temperature may drop to animals. Cattle affected with a strain of C4 in Argentina,
40˚C. The lesions heal within 7–14 days if no secondary sheep in northern Africa affected with a strain of O1, and
bacterial infection occurs. mountain gazelles in Israel affected with another strain
FMD infects many or all of the exposed animals with of O1 (Shimshony et al. 1986) represent some examples
no immunity in temperate climates or confined quarters. of 50% or greater mortality in adult animals.
In partially immune populations or in tropical climates,
morbidity may be decreased. VES is moderately conta- PATHOGENESIS
gious, with highly pathogenic strains affecting greater
numbers of animals. With SVD, the entire herd may not Foot-and-Mouth Disease
be affected, and subclinical cases may occur. VS may The major route of transmission for FMDV is aerosol
spread horizontally in swine in confined quarters. (Sellers and Parker 1969; Donaldson et al. 1970; Thom-
Low mortality (<5%) is usually seen with vesicular son 1995). Relatively large aerosol particles (3–6 microns
CHAPTER 25 VESICULAR DISEASES House, House 333

or greater) adhere to the upper respiratory tract, while epithelium via an abrasion, the virus multiplies in the
smaller particles (3 microns or less) reach the lower res- stratum germinativum of the epidermis. Hydropic de-
piratory tract. Viral nucleic acid, as detected by in situ generation occurs, followed by necrosis of infected cells.
hybridization, was widespread in epithelial tissues of Intracellular edema (hydropic degeneration) and inter-
cattle only 6 hours after aerosol exposures (Brown et al. cellular edema account for vesicle formation (Madin and
1992). In swine following intradermal inoculation in the Traum 1955). Virus spreads from cell to cell as the infec-
snout, widespread FMDV nucleic acid was detected by in tion progresses. Local lymph nodes may become in-
situ hybridization on the first sampling 24 hours after in- volved, with extensive lymphocyte destruction and con-
fection (Brown et al. 1995). Langerhans cells may be the gestion; a low-level viremia occurs and may account for
primary means of this rapid distribution of virus. The some secondary lesions.
pattern of distribution is “segmentally diffuse.” Follow-
ing seeding of secondary targets (epithelium, mucosa, Vesicular Stomatitis
and myocardium), the virus replicates, and viremia gen- VSV apparently requires introduction into the epitheli-
erally persists for 3–5 days (Sutmoller and McVicar um through insect bites or via fomites into minor abra-
1976). Within 2–3 days, vesicles develop where stress sions of the oral or nasal mucosa, teats, or feet (Patterson
and mechanical abrasions occur. In swine this includes et al. 1955). Virus replicates in the stratum germina-
the snout, feet, dewclaws, skin over the joints and pres- tivum. A vesicle and an exudate form, usually 2–3 days
sure points of the extremities, mouth, tongue, and teats postinoculation. Without secondary infection, which
of nursing sows. may involve the subcutaneous tissue, the lesion heals in
In addition, FMDV replication in the epithelial cells 1–2 weeks.
of the mammary gland is well documented. The FMDV The presence of a viremia is controversial in VSV in-
is generally shed in the milk of cattle for up to 10 days fection. Early literature indicates that there may be a
postexposure, corresponding to the development of viremia. However, this has not been confirmed by more
virus-neutralizing antibodies (Blackwell et al. 1982, recent reports. In pigs, virus was isolated from local
1983). In some cattle, however, the virus may be shed in lymph nodes but not blood following intradermal inocu-
the milk for up to 7 weeks, even in the presence of neu- lation in the snout to approximate the natural route of
tralizing antibodies in the serum (Burrows et al. 1971). infection (Redelman et al. 1989).
Similar patterns of virus shedding in milk probably oc-
cur in swine. The virus is found in large quantities in oral LESIONS
and respiratory secretions 2–7 days postexposure. In
young animals severe myocardial necrosis may occur. The principal lesion, the vesicle, of FMD, SVD, VES, and
VS is identical by gross pathology (Jubb et al. 1985). Vesi-
Swine Vesicular Disease cles occur in the epithelium of the oral and stratified
Lesions are evident 3–11 days after eating contaminated squamous nasal mucosa, feet, teats, pressure points on
food (McKercher and Graves 1981) and as early as 2–4 the extremities, interdigital spaces, eyelids, and around
days after experimental inoculation (Burrows et al. 1973; the coronary bands.
Lai et al. 1979). The first lesions generally appear on the A vesicular lesion begins as a small blanched area and
coronary band. A fever, which persists for up to 5 days, progresses to a blanched, slightly raised area that ex-
essentially coincides with the development of a cell-free pands as vesiculation progresses. The epithelium may
viremia and vesicular lesions. The peak viremia occurs separate from the stratum basale, leaving a red lesion
2–4 days postexposure and persists for about 7 days. The with shreds of torn epithelium. Vesicular fluid may leak
virus persists for at least 7 days in the tissues of the through the stratum corneum without the formation of a
snout, tongue, coronary band, tonsil, cardiac muscle, true vesicle (Seibold and Sharp 1960). Since vesicles are
and central nervous system. The stratum spinosum is the most often seen at the site of mechanical stress, they usu-
primary site of viral replication in the epithelium. A non- ally rupture quickly, leaving a red erosion. Often, lesions
suppurative meningoencephalitis most frequently oc- are contaminated with dirt and debris (Fig. 25.2), result-
curs in the cerebrum, thalamus, brain stem, and olfacto- ing in infection with opportunistic bacteria. The foot le-
ry lobes. Necrosis and an inflammatory reaction occur in sions, especially of VES, may be associated with a celluli-
the endocardium and myocardium (Lai et al. 1979). tis with persistent swelling, resulting in lameness and
local lymph node involvement. In young animals infect-
Vesicular Exanthema of Swine ed with FMDV, myocardial necrosis may produce pale
VES is primarily introduced by feeding contaminated necrotic areas referred to as “tiger heart.”
raw garbage. However, oral infection with VESV is esti-
mated to require 100–1000 times the amount of virus Histopathology
needed to produce a lesion by intradermal inoculation The histopathology of FMD, SVD, and VES is considered
into the snout (Mott et al. 1953). Following entry into the quite similar. The lesions begin in the stratum spinosum,
334 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

where vesicles form because of hydropic degeneration DIAGNOSIS


and intercellular edema. Keratinocytes die, become
spherical, and float as single or clustered cells in the The diagnosis of FMD, SVD, VES, and VS must be con-
vesicular fluid (spongiosa) (Fig. 25.5A). The stratum ducted in the laboratory, for the diseases are clinically in-
basale essentially remains intact but may be somewhat distinguishable from each other. In countries where
disrupted by VESV infection (Mebus 1977). vesicular diseases are endemic, national animal disease
The histopathology of VS in cattle has been carefully diagnostic laboratories are generally available. Samples
studied by Seibold and Sharp (1960) and described by may be shipped to the World Reference Laboratory for
Jubb et al. (1985) and is probably equivalent for swine. Foot-and-Mouth Disease at Pirbright, Great Britain, or to
The lesions develop in the stratum spinosum with inter- regional vesicular disease diagnostic laboratories includ-
cellular edema that stretches the intercellular bridges ing the Foreign Animal Disease Diagnostic Laboratory
(desmosomes). Cells remain attached at the ends parallel (FADDL) at the PIADC in Plum Island, New York, Unit-
to the stratum basale but are separated lengthwise by in- ed States, or the Pan American Foot-and-Mouth Disease
tercellular edema, giving a reticular or “Japanese Laboratory in Rio de Janeiro, Brazil. The director of the
lantern” appearance (Fig. 25.5B). Cells become necrotic laboratory should be contacted before shipment. Per-
in the later stages of the lesion. The epithelial layers mits may be needed for shipment to some laboratories,
above the stratum basale separate in about 30% of the and ever-changing international shipping regulations for
cases. Loss of vesicular fluid through the stratum diagnostic samples must be followed.
corneum occurs frequently, and vesicles may not be ob- Samples from acutely ill swine should include vesicu-
served. lar tissue and, if possible, vesicular fluid from gently

A B
25.5. (A) A multilocular microvesicle of FMD with necrosis of the stratum spinosum (acanthol-
ysis) and keratinocytes floating in the vesicular fluid (spongiosa). (B) Severe intercellular edema
with separation of the cells of the entire stratum spinosum, giving a reticulated pattern typical of
VS; the vesicle forms when the full thickness of epithelium separates from the basal layer (arrow).
(Courtesy of D. A. Gregg.)
CHAPTER 25 VESICULAR DISEASES House, House 335

washed, active lesions on the tongue, lips, gums, nares, spectrum of different oligonucleotides, has been used to
feet, dewclaws, extremities, or teats. Heparinized and study the relationship of FMDVs (Frisby et al. 1976).
clotted blood should be collected for virus isolation and However, nucleotide sequencing, which provides the or-
serology, respectively. Clotted blood for serum should be der of the nucleotides in the native RNA, is a more pre-
collected from recovered animals. Necropsy samples of cise tool to study the variable areas of the genome of
tissues for virus isolation should include vesicular lesions FMDV (Beck and Strohmaier 1987; Westbury et al. 1988;
and, particularly from newborn or young animals, skele- Marquardt and Adam 1990). The polymerase chain reac-
tal muscle and myocardium. These samples should be tion (Meyer et al. 1991; Donn et al. 1994; Marquardt et
shipped on wet ice if they will arrive at a diagnostic lab- al. 1995), combined with nucleotide sequencing, pro-
oratory within 24 hours. If a longer shipping time is an- vides definitive epidemiological information on the ori-
ticipated, freezing the tissue in airtight vials and ship- gin of viral strains.
ping on dry ice is recommended. For histopathology, a
complete set of tissues representing all organ systems, TREATMENT
but with emphasis on organs with lesions, should be col-
lected and preserved in buffered 10% formalin. No specific treatments are known for the vesicular dis-
In swine, the differential diagnosis of FMD, SVD, eases. Palliative measures include feeding soft feeds, re-
VES, and VS should include swine pox, hog cholera, moving animals from hard-surfaced or concrete floors,
pseudorabies, porcine parvovirus infection (Kresse et al. and providing access to clean water. Providing hygienic
1985), parsnip or celery contact dermatitis (Mont- conditions, using antiseptic solutions on affected epithe-
gomery et al. 1987), chemical burns, trauma, and my- lium, and administering antibiotics may help prevent
cotic stomatitis. and control secondary infections.
Epithelial and vesicular fluid samples are tested by
complement fixation (CF) or an antigen capture ELISA PREVENTION
for the presence of viral antigens of FMD, SVD, and VS.
ELISA can detect and identify serotypes of FMD viral Exotic Vesicular Diseases
antigens at lower concentrations than the CF test under It is imperative to prevent the infection of swine with
experimental (Roeder and LeBlanc Smith 1987) and field FMDV since they can produce tremendous infectious
conditions (Westbury et al. 1988). The results of these aerosols. Prevention of FMD and SVD is accomplished
tests can be available within 4 hours of receipt of the by strictly regulating movement of animals and animal
sample. Virus isolation in cell cultures (Snowdon 1966; products. Swine from FMD-endemic areas were first im-
House and House 1989) of suckling mice, electron mi- ported into the United States from China in 1989 using
croscopy (EM), and animal host inoculation studies may the methods developed for cattle importation (House et
be done. In nonenzootic areas, virus isolation should be al. 1983). Swine semen has also been imported using
performed. A positive FMDV isolation with serotype testing and quarantine procedures. The importation of
identification may be completed as early as 24–72 hours swine embryos may be complicated by the depth of
after receipt of the sample. A negative virus isolation sperm tracts and other undefined characteristics of
study may take up to 2 weeks, as multiple passages may porcine embryos that may cause adherence of microbial
be required. agents to the zona (Callis 1996).
Serum samples may be tested for precipitating anti- Regulations for the international movement of ani-
bodies to the FMD VIAA, for neutralizing antibodies to mal products other than germ plasm are based on virus
FMDVs, SVDV, VESVs, and VSVs, and for CF antibodies inactivation studies. Inactivation of FMDV and SVDV in
to VSVs, with results available in 1–3 days. Virus-neu- meat products has been reviewed (Blackwell 1984). Pork
tralizing antibodies are serotype-specific for FMDV, and products from swine infected with FMDV and SVDV
animals infected for the first time characteristically de- (McKercher et al. 1985, 1987) and from swine infected
velop the highest titer against the homologous subtype. with FMDV, SVDV, African swine fever virus, and hog
Antibodies to FMD VIAA were used historically to dif- cholera virus (classic swine fever virus) (Mebus et al.
ferentiate the immune response to active infection from 1993a, b) were free of virus when subjected to prolonged
that of vaccination. However, repeated vaccination with curing processes. Virus in the milk of FMDV-infected
killed FMD vaccine may elicit a transient antibody re- cattle was inactivated by pasteurization at ultrahigh tem-
sponse to VIAA (Pinto and Garland 1979). An ELISA em- peratures for a short time (148˚C for 2 seconds) (Cunliffe
ploying baculovirus-expressed FMD nonstructural pro- et al. 1979).
teins 3D and 2C can be used to differentiate vaccinated Regulations for cooking of garbage fed to swine re-
from convalescent animals (Meyer et al. 1997). sulted in the control and eradication of VES (Madin and
RNA fingerprinting, which uses ribonuclease T1 to Traum 1953). Swine fed seal meat or other marine-origin
specifically cleave the RNA at unique sites and produce a feed supplements could become infected with cali-
336 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

civiruses, but such practices are no longer in use and VES (1990) have synthesized empty capsids of FMDV in in-
has not recurred. sect cells using baculovirus-expression vectors, but there
are no reports of their efficacy.
Domestic Vesicular Diseases Inactivated VS vaccines were produced and provi-
Vaccination has not been used to prevent VS in swine in sionally licensed for use in cattle during the 1982–83 epi-
the United States (Buisch 1983). The segregation of zootic, and for horses and cattle in 1995. Efficacy data on
swine from infected cattle, horses, and wildlife, including these products are not available. There is no vaccine
feral swine, in epizootic regions is a practical suggestion. available for SVD, since eradication is the preferred
The role of insects in the spread of VS justifies an insect method of control.
control program during an epizootic.
REFERENCES
CONTROL
Acharya, R.; Fry, E.; Stuart, D.; Fox, D; Rowlands, D.; and
In case of an outbreak of exotic vesicular disease in Brown, F. 1989. The three-dimensional structure of foot-
swine, the USDA emergency disease guidelines call for and-mouth disease virus at 2.9A revolution. Nature
stamping out of the disease upon identification of the 337:709–716.
exotic agent. The plan calls for immediate quarantine of Bachrach, H. L. 1977. Foot-and-mouth disease virus: Proper-
the affected premises, restriction of animal movement, ties, molecular biology and immunogenicity. In
public-alert statements, eradication by slaughter, and Beltsville Symposium in Agricultural Research I. Virolo-
close communication and cooperation with industry. gy in Agriculture. Ed. J. A. Romberger. Montclair, N.J.:
Today, the loss of genetic material and animal pro- Allanheld, Osmum & Co., pp. 46–49.
tein, environmental regulations, and animal welfare con- Bankowski, R. A. 1965. Vesicular exanthema. Adv Vet Sci
cerns warrant the consideration of vaccination as a tool 10:23–64.
for controlling and eradicating FMD. Bankowski, R. A.; Perkins, A. G.; Stuart, E. E.; and Kummer,
The North American Foot-and-Mouth Disease Vac- M. 1955. Epizootiology of vesicular exanthema in Cali-
cine Bank, maintained by USDA in cooperation with fornia. In Proc 59th Annu Meet US Livestock Sanit As-
Canada and Mexico and other international FMD vac- soc, pp. 356–367.
cine banks, has reserves of inactivated, safety- and po- Barlough, J. E.; Berry, E. S.; Smith, A. W.; and Skilling, D. E.
tency-tested FMD vaccine antigens available to be for- 1987. Prevalence and distribution of serum neutralizing
mulated into FMD vaccines. Vaccinated animals could be antibodies to Tillamook (bovine) calicivirus in selected
permanently identified and their movement restricted. populations of marine mammals. J Wildl Dis 23:45–51.
Vaccines for FMD, used throughout the world in Beck, E., and Strohmaier, K. 1987. Subtyping of European
FMD-endemic areas, are inactivated, adjuvanted vac- foot-and-mouth disease virus strains by nucleotide se-
cines. These vaccines are produced in BHK (baby ham- quence determination. J Virol 61:1621–1629.
ster kidney) suspension cell cultures and inactivated with Berry, E. S.; Skilling, B. S.; Barlough, J. E.; Vedros, N. A.;
ethylenamine derivatives. Vaccination with the prevalent Gage, L. J.; and Smith, A. W. 1990. New marine cali-
serotypes 2–4 times a year is recommended. The tradi- civirus serotype infective for swine. Am J Vet Res
tional aluminum hydroxide–saponin adjuvanted FMD 51:1184–1187.
vaccines used for cattle are less efficacious in swine than Bittle, J. L.; Houghten, R. A.; Alexander, H.; Shinnick, T. M.;
are oil-adjuvanted FMD vaccines (McKercher and Gior- Sutcliffe, J. G.; Leaner, R. A.; Rowlands, D. J.; and Brown,
dano 1967). F. 1982. Protection against foot-and-mouth disease by
No genetically engineered vaccines for FMD are com- immunization with a chemically synthesized peptide
mercially available, and their practicality is uncertain. produced from viral nucleotide sequence. Nature
Experimentally, genetic sequences for the key immuno- 298:30.
genic FMD VP1 were cloned into Escherichia coli, and Blackwell, J. H. 1984. Foreign animal disease agent survival
VP1 was successfully expressed (Kleid et al. 1981). The in animal products: Recent developments. J Am Vet Med
cloned protein protected vaccinated cattle and swine Assoc 184:674–679.
against a virulent challenge inoculation. The system was Blackwell, J. H.; McKercher, P. D; Kosikowski, F. V;
successful with selected subtypes of serotypes A and C, Carmichael, L. E.; and Gorewit, R. C. 1982. Concentra-
but for serotype O, the cloned proteins did not provide tion of foot-and-mouth disease virus in milk of cows in-
consistent protection. The amino acid sequence of criti- fected under simulated field conditions. J Dairy Sci
cal polypeptides, which could induce neutralizing anti- 65:1624–1631.
bodies to serotype O, was deduced from the nucleotide ———. 1983. Histological and histochemical characteriza-
sequence. Synthetic polypeptides protected guinea pigs tion of mammary gland tissue of cows infected with
(Bittle et al. 1982) and cattle (DiMarchi et al. 1986) foot-and-mouth disease by contact exposure. Res Vet Sci
against a virulent challenge inoculation. Roosien et al. 35:106–113.
CHAPTER 25 VESICULAR DISEASES House, House 337

Brooksby, J. B. 1982. Portraits of viruses: Foot-and-mouth DiMarchi, R.; Brooke, G.; Gale, C.; Cracknell, V.; Dale, T.;
disease virus. Intervirology 18:1–23. and Mowat, N. 1986. Protection of cattle against foot-
Brown, C. C.; Meyer, R. F.; Olander, H. J.; House, C.; and and-mouth disease by a synthetic peptide. Science
Mebus, C. A. 1992. A pathogenesis study of foot-and- 232:639–641.
mouth disease in cattle, using in situ hybridization. Can Domingo, E.; Diez, J.; Martinez, M. A.; Hernandez, J.; Hol-
J Vet Res 56:189–193. guin, A.; Borrego, B.; and Mateu, M. G. 1993. New ob-
Brown, C. C.; Olander, H. J.; and Meyer, R. F. 1995. Patho- servations on antigenic diversification of RNA viruses:
genesis of foot-and-mouth disease in swine, studied by Antigenic variation is not dependent on immune selec-
in-situ hybridization. J Comp Path 113:51–58. tion. J Gen Virol 74:2039–2045.
Brown, F.; Goodridge, D.; and Burrows, R. 1976. Infection of Donaldson, A. I., and Ferris, N. P. 1980. Sites of release of
man with swine vesicular disease virus. J Comp Path airborne foot-and-mouth disease virus from infected
86:409–414. pigs. Res Vet Sci 29(3):315–319.
Buisch, W. W. 1983. Fiscal year 1982–1983 vesicular stom- Donaldson, A. I.; Herniman, K. A. J.; Parker, J.; and Sellers,
atitis outbreak. In Proc 87th Annu Meet US Anim R. F. 1970. Further investigations on the airborne excre-
Health Assoc, pp. 78–84. tion of foot-and-mouth disease virus. J Hyg (Camb)
Bulloch, W. 1927. De contagione et contagiosis morbis et cu- 68:557–564.
ratione. J Comp Path 40:75–76. Donaldson, A. I.; Gibson, C. F.; Oliver, R.; Hamblin, C.; and
Burrows, R.; Mann, J. A.; Greig, A.; Chapman, W. G.; and Kitching, R. P. 1987. Infection of cattle by airborne foot-
Goodridge, D. 1971. The growth and persistence of foot- and-mouth disease virus: Minimal doses with O1 and
and-mouth disease virus in the bovine mammary gland. SAT 2 strains. Res Vet Sci 43:339–346.
J Hyg (Camb) 69:307–321. Donn, A.; Martin, L. A.; and Donaldson, A. I. 1994. Im-
Burrows, R; Greig, A.; and Goodridge, D. 1973. Swine vesic- proved detection of persistent foot-and-mouth disease
ular disease. Res Vet Sci 15:141–144. infection in cattle by the polymerase chain reaction. J Vi-
Callis, J. J. 1996. Evaluation of the presence and risk of foot- rol Meth 49:179–186.
and-mouth disease virus by commodity in international Dunn, C. S., and Donaldson, A. I. 1997. Natural adaption to
trade. Res Sci Tech Off Int Epiz 15:1075–1085. pigs of a Taiwanese isolate of foot-and-mouth disease
Chu, R. M.; Moore, D. M.; and Conroy, J. D. 1979. Experi- virus. Vet Rec 141:174–175.
mental swine vesicular disease, pathology and immuno- Federer, K. E.; Burrows, R.; and Brooksby, J. B. 1967. Vesicu-
fluorescence studies. Can J Comp Med 43:29–30. lar stomatitis virus: The relationship between some
Comer, J. A.; Tesh, R. B.; Modi, G. B.; Coin, J. T.; and Nettles, strains of the Indiana serotypes. Res Vet Sci 8:103–117.
V. F. 1990. Vesicular stomatitis virus New Jersey Fox, G.; Parry, N. R.; Barnett, P. V.; McGinn, B.; Rowlands,
serotype: Replication in and transmission by Lutzomyia D. J.; and Brown, F. 1989. The cell attachment site on
shannoni (Diptera: Psychodidae). Am J Trop Med Hyg foot-and-mouth disease virus includes the amino acid
42:483–490. sequence RGD (argenine-glycine-aspartic acid). J Gen
Coombs, G. P. 1973. A study of swine vesicular disease in Virol 70:625–637.
England. In Proc 77th Annu Meet US Anim Health As- Francki, R. B.; Fauquet, C. M.; Knudsen, D. L.; and Brown, F.
soc, pp. 332–335. 1991a. Classification and Nomenclature of Viruses. Fifth
Cotton, W. E. 1927. Vesicular stomatitis. Vet Med Report of the International Committee on Taxonomy of
22:169–175. Virus. Arch Virol, Suppl 2. New York: Springer Verlag,
Cottral, G. E.; Gailiunas, P.; and Cox, B. F. 1968. Foot-and- pp. 250–254.
mouth disease virus in semen of bulls and its transmis- ———. 1991b. Classification and Nomenclature of Viruses.
sion by artificial insemination. Arch Gesamte Virus- Fifth Report of the International Committee on Taxono-
forsch 23:362–377. my of Virus. Arch Virol, Suppl 2. New York: Springer
Cunliffe, H. R.; Blackwell, J. H.; Dors, R.; and Walker, J. S. Verlag, pp. 300–302.
1979. Inactivation of milk-borne foot-and-mouth dis- ———. 1991c. Classification and Nomenclature of Viruses.
ease virus at ultrahigh temperatures. J Food Prot Fifth Report of the International Committee on Taxono-
42(2):135–137. my of Virus. Arch Virol, Suppl 2. New York: Springer
Cupp, E. W.; Mare, J.; Cupp, M. S.; and Ramberg, F. B. 1992. Verlag, pp. 320–326.
Biological transmission of vesicular stomatitis virus Frisby, D. P.; Newton, C.; Carey, N. H.; Fellner, P.; Newman,
(New Jersey) by Simulium vittatum (Diptera: Simulidae) J J. F. E.; Harris, T. J. R.; and Brown, F. 1976. Oligonu-
Med Ent 29:137–140. cleotide mapping of picornavirus RNAs by two-dimen-
Dawe, P. S.; Sorenson, K.; Ferris, N. P.; Barnett, I. T. R.; Arm- sional electrophoresis. Virol 71:379–388.
stong, R. M.; and Knowles, N. J. 1994. Transmission of Garcia Perazzi, A. J.; Caggiano, C. H.; and Fernandez, A. A.
foot-and-mouth disease from carrier African buffalo 1963. Publication Tecnica No 2. CADEFA, Ministry of
(Syncerus caffer) to cattle under experimental conditions Agriculture, Buenos Aires, Argentina.
in Zimbabwe. Vet Rec 134:211–215. Gebauer, F.; De La Torro, J. C.; Gomes, I.; Mateu, M. G.;
338 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

Barahona, H.; Tiraboschi, B.; Bergmann, I.; Demello, P.; ———. 1955. Vesicular exanthema of swine. Bacteriol Rev
and Domingo, E. 1988. Rapid selection of genetic and 19:6–19.
antigenic variants of foot-and-mouth disease virus dur- Mann, J. A., and Hutchings, G. H. 1980. Swine vesicular dis-
ing persistence in cattle. J Virol 62:2041–2049. ease: Pathways of infections. J Hyg 84:355–363.
Graves, J. H. 1973. Serological relationship of swine vesicu- Mann, J. A.; Burrows, R.; and Goodridge, D. 1975. Mild sub-
lar disease virus and Coxsackie B-5 virus. Nature clinical infections with swine vesicular disease virus. Bull
245:314. Off Int Epiz 83:117–122.
Hedger, R. S., and Condy, J. B. 1985. Transmission of foot Maragon, S.; Facchin, F.; Moutou, I.; Massirio, G.; Vincenzi,
and mouth disease from African buffalo virus carriers to G.; and Davies, G. 1994. The 1993 Italian foot-and-
bovines. Vet Rec 117:205. mouth disease epidemic: Epidemiological features of
Herniman, K. A. J.; Medhurst, P. M.; Wilson, J. N.; and Sell- the four outbreaks identified in Verona province (Ventro
ers, R. F. 1973. The action of heat, chemicals and disin- region). Vet Rec 135:53–57.
fectants on swine vesicular disease virus. Vet Rec Marquardt, O., and Adam, K. 1990. Foot-and-mouth disease
93:620–624. virus subtyping by sequencing VP1 genes. Vet Microbiol
House, C., and House, J. A. 1989. Evaluation of techniques 23:175–183.
to demonstrate foot-and-mouth disease virus in bovine Marquardt, O.; Straub, O. C.; Ahl, R.; and Haas, B. 1995. De-
tongue epithelium: Comparison of the sensitivity of cat- tection of foot-and-mouth disease virus in nasal swabs
tle, mice, primary cell cultures, cryopreserved cell cul- of asymptomatic cattle by RT-PCR within 24 hours. J Vi-
tures, and established cell lines. Vet Microbiol rol Meth 53:255–261.
20:99–109. McKercher, P. D., and Giordano, A. R. 1967. Foot-and-
House, J. A., and Wilks, C. R. 1983. Studies on exotic mouth disease in swine. I. The immune response of
vesiculoviruses. In Proc 87th Ann Meet US Animal swine to chemically treated and nontreated foot-and-
Health Assoc, pp. 276–288. mouth disease virus. Arch Gesamte Virusforsch
House, J. A.; Yedloutschnig, R. J.; Dardiri, A. H.; Herrick, D. 20:39–53.
E.; and Acree, J. A. 1983. Procedures used for the impor- McKercher, P. D., and Graves, J. H. 1981. Swine vesicular
tation of Brazilian Zebu cattle into the United States. disease. In CRC Handbook Series in Zoonoses, Sec B. Vi-
Proc 26th Annu Meet Am Assoc Vet Lab Diagn, pp. ral Zoonoses, vol. 2. Ed. J. H. Steele. Boca Raton, Fla.:
13–24. CRC Press, pp. 161–167.
Jonkers, A. H.; Shope, R. E.; Aitken, T. H. G.; and Spence, L. McKercher, P. D.; Blackwell, J. H.; Murphy, R.; Callis, J. J.;
1964. Cocal virus, a new agent in Trinidad related to Panina, G. F.; Civardi, A.; Bugnetti, M.; Desimone, F.;
vesicular stomatitis virus. Am J Vet Res 25:236–242. and Scatozza, F. 1985. Survival of swine vesicular dis-
Jubb, K. V. F.; Kennedy, P. C.; and Palmer, N. 1985. Patholo- ease virus in “Prosciutto di Parma” (Parma Ham). Can
gy of Domestic Animals, vol. 2. New York: Academic Inst Food Sci Technol J 18:163–167.
Press, pp. 90–110. McKercher, P. D.; Yedloutschnig, R. J.; Callis, J. J.; Murphy,
Kleid, D. G.; Yansura, D.; Small, B.; Dowbenko, D.; Moore, R.; Panina, G. F.; Civardi, A.; Bugnetti, M.; Foni, E.; Lad-
D. M.; Grubman, M. J.; McKercher, P. D.; Morgan, D. damada, A.; Scarano, C.; and Scatozza, F. 1987. Survival
O.; Robertson, B. H.; and Bachrach, H. L. 1981. Cloned of viruses in “Prosciutto di Parma” (Parma Ham). Can
viral protein vaccine for foot-and-mouth disease: Re- Inst Food Sci Technol J 20:267–272.
sponses in cattle and swine. Science 214:1125–1129. McVicar, J. W.; Eisner, R. J.; Johnson, L. A.; and Pursel, V. G.
Kresse, J. I.; Taylor, W. D.; Stewart, W. C.; and Eernissee, K. 1978. Foot-and-mouth disease and swine vesicular dis-
A. 1985. Parvovirus infection in pigs with necrotic and ease viruses in boar semen. In Proc 81st Annu Meet US
vesicle-like lesions. Vet Microbiol 10:525–531. Anim Health Assoc, pp. 221–230.
Lai, S. S; McKercher, P. D.; Moore, D. M.; and Gillespie, J. H. Mebus, C. A. 1977. Ulcerative diseases of animals with an
1979. Pathogenesis of swine vesicular disease in pigs. infectious etiology. J Oral Path 7:365–371.
Am J Vet Res 40:463–468. Mebus, C. A.; House, C.; Ruiz Gonzalvo, F.; Pineda, J. M.;
Loeffler, F., and Frosch, P. 1898. Zentralbl Bakt 1 Orig Tapiador, J.; Pire, J. J.; Bergada, J.; Yedloutschnig, R. J.;
28:371. Sahu, S.; Becerra, V.; and Sanchez-Vizcaino, J. M. 1993a.
Lubroth, J.; Grubman, M. J.; Burrage, T. G.; Newman, J. F. Survival of foot-and-mouth disease, African swine fever
E.; and Brown, F. 1996. Absence of protein 2C from clar- and hog cholera viruses in Spanish Serrano cured hams
ified foot-and-mouth disease virus vaccine provides the and Iberian cured hams, shoulders and loins. Food Mi-
basis for distinguishing convalescent from vaccinated crobiol 10:133–143.
animals. Vaccine 14:419–427. Mebus, C. A.; House, C.; Ruiz Gonzalvo, F.; Pineda, J. M.;
Madin, S. H., and Traum, J. 1953. Experimental studies with Tapiador, J.; Pire, J. J.; Bergada, J.; Yedloutschnig, R. J.;
vesicular exanthema of swine. II. Studies on stability. and Sanchez-Vizcaino, J. M. 1993b. Survival of swine
Vet Med 48:443–450. vesicular disease virus in Spanish Serrano cured hams
CHAPTER 25 VESICULAR DISEASES House, House 339

and Iberian cured hams, shoulders and loins. Food Mi- Roosien, J.; Belsham, G. J.; Ryan, M. D.; King, A. M. Q.; and
crobiol 10:263–268. Vlak, J. M. 1990. Synthesis of foot-and-mouth disease
Meyer, R. F.; Brown, C. C.; House, C.; House, J.; and Molitor virus capsid proteins in insect cells using baculovirus ex-
T. W. 1991. Rapid and sensitive detection of foot-and- pression vectors. J Gen Virol 71:1703–1711.
mouth disease virus in tissues by enzymatic RNA ampli- Sawyer, J. C.; Madin, S. H.; and Skilling, D. E. 1978. Isolation
fication of the polymerase gene. J Virol Methods of San Miguel sea lion virus from samples of an animal
34:161–172. food product produced from Northern fur seal (Callorhi-
Meyer, R. F.; Babcock, G. D.; Newman, J. F. E.; Burrage, T. nus ursinus) carcasses. Am J Vet Res 39:137–139.
G.; Toohey, K.; Lubroth, J.; and Brown, F. 1997. Bac- Schoening, H. W. 1943. Vesicular stomatitis in swine. In
ulovirus expressed 2C of foot-and-mouth disease virus Proc 47th Annu Meet US Livest Sanit Assoc, pp. 85–86.
has the potential for differentiating convalescent from Seibold, H. R., and Sharp, J. B. 1960. A revised concept of
vaccinated animals. J Virol Meth, in press. the pathological changes of the tongue in cattle with
Montgomery, J. F.; Oliver, R. E.; and Poole, W. S. H. 1987. A vesicular stomatitis. Am J Vet Res 21:35–51.
vesiculo-bullous disease in pigs resembling foot-and- Sellers, R. F., and Herniman, K. A. J. 1974. The airborne ex-
mouth disease. I. Field cases. NZ Vet J 35:21–26. cretion by pigs of swine vesicular disease virus. J Hyg
Mott, L. O.; Patterson, W. C.; Songer, J. R.; and Hopkins, S. (Camb) 72:61–65.
R. 1953. Experimental infections with vesicular exanthe- Sellers, R. F., and Parker, J. 1969. Airborne excretion of foot-
ma. II. Feeding of viral suspensions and infected tissues. and-mouth disease virus. J Hyg (Camb) 67:671–677.
In Proc 57th Annu Meet US Livest Sanit Assoc, pp. Sellers, R. F.; Herniman, K. A. J.; and Donaldson, A. I. 1971.
349–360. The effects of killing or removal of animals affected with
Mulhern, F. J. 1953. Present status of vesicular exanthema foot-and-mouth disease on the amounts of airborne
eradication program. In Proc 57th Annu Meet US Livest virus present in loose boxes. Brit Vet J 127:358–365.
Sanit Assoc, pp. 326–333. Shimshony, A.; Orgad, U.; Baharv, D.; Prudovsky, S.; Yakob-
Nardelli, L.; Lodetti, E.; Gualandi, G. L.; Burrows, R.; son, B.; Bar Moshe, B.; and Dagan, D. 1986. Malignant
Goodridge, D.; Brown, F.; and Cartwright, B. 1968. A foot-and-mouth disease in mountain gazelles. Vet Rec
foot-and-mouth disease syndrome in pigs caused by an 119:175–176.
enterovirus. Nature 219:1275–1276. Smith, A. W., and Latham, A. B. 1978. Prevalence of vesicu-
Newman, J. F. E.; Piatti, P. G.; Gorman, B. M.; Burrage, T. lar exanthema of swine antibodies among feral mam-
G.; Ryan, M. D.; Flint, M.; and Brown, F. 1994. Foot-and- mals associated with the southern California coastal
mouth disease virus particles contain replicase protein zones. Am J Vet Res 39:291–296.
3D. Proc Natl Acad Sci USA 91:733–737. Smith, A. W.; Akers, T. G.; Madin, S. H.; and Vedros, N. A.
Patterson, W. C.; Jenny, E. W; and Holbrook, A. A. 1955. Ex- 1973. San Miguel sea lion virus isolation, preliminary
perimental infections with vesicular stomatitis in swine. characterization, and relationships to vesicular exanthe-
I. Transmission by direct contact and feeding infected ma of swine virus. Nature 244:108–109.
meat scraps. In Proc 59th Annu Meet US Livest Sanit As- Smith, A. W.; Prato, C. M.; and Skilling, D. E. 1977. Charac-
soc, pp. 368–378. terization of two new serotypes of San Miguel sea lion
Pereira, H. G. 1977. Subtyping of foot-and-mouth disease virus. Intervirology 8:30–36.
virus. In Developments in Biological Standardization. Smith, A. W.; Akers, T. B.; Latham, A. B; Skilling, D. E.; and
Ed. C. Mackowiak and R. H. Regamey. Basel: S. Karger, Bray, H. L. 1979. A new calicivirus isolated from a ma-
vol. 35, pp. 167–174. rine mammal. Arch Virol 61:255–259.
Pinto, A. A., and Garland, A. J. M. 1979. Immune response Smith, A. W.; Skilling, D. E.; Dardiri, A. H.; and Latham, A.
to virus-infection-associated (VIA) antigen in cattle re- B. 1980. Calicivirus pathogenic for swine: A new
peatedly vaccinated with foot-and-mouth disease virus serotype isolated from opaleye Girella nigricans, an
inactivated by formalin or acetylethyleneimine. J Hyg ocean fish. Science 209:940–941.
(Camb) 82:41–50. Smith, A. W.; Mattson, D. E.; Shilling, D. E.; and Schmitz, J.
Redelman, D.; Nichol, S.; Kleeforth, R.; Van der Matten, M.; A. 1983a. Isolation and partial characterization of a cali-
and Whetstone, C. 1989. Experimental vesicular stom- civirus from calves. Am J Vet Res 44:851–855.
atitis virus infection of swine: Extent of infection and Smith, A. W.; Ritter, D. G.; Ray, G. C; Skilling, D. E.; and
immunological response. Vet Immunol Immunopathol Wartzok, D. 1983b. New calicivirus isolates from feces of
20:345–361. walrus (Odobenus rosmarus). J Wildl Dis 19:86–183.
Roeder, P. L., and Leblanc Smith, P. M. 1987. Detection and Snowdon, W. A. 1966. Growth of foot-and-mouth disease
typing of foot-and-mouth disease virus by enzyme- virus in monolayer cultures of calf thyroid cells. Nature
linked immunosorbent assay: A sensitive, rapid, and re- 210:1079–1080.
liable technique for primary diagnosis. Res Vet Sci Stallknecht, D. E.; Nettles, V. F.; Fletcher, W. D.; and Erick-
43:225–232. son, G. A. 1985. Enzootic vesicular stomatitis New Jersey
340 SECTION 2 VIRAL DISEASES W. L. Mengeling, Editor

type in an insular feral swine population. Am J Epidemi- Traum, J. 1934. Vesicular exanthema of swine. In Proc 12th
ol 122:876–883. Int Vet Congr, New York, vol. 2, pp. 5–9.
Sutmoller, P., and McVicar, J. W. 1976. Pathogenesis of foot- Travassos da Rosa, A. P. A.; Tesh, P. B.; Travassos da Rosa,
and-mouth disease: The lung as an additional portal of J. S.; Herve, J. P.; and Main, A. J. 1984. Carajas and Mara-
entry of the virus. J Hyg (Camb) 77:235–243. ba virus, two new vesiculoviruses isolated from phle-
Tesh, R. B.; Chaniotis, B. H.; and Johnson, K. M. 1971. Vesic- botomine sand-flies in Brazil. Am J Trop Med Hyg
ular stomatitis virus (Indiana serotype) transovarial 33:999–1006.
transmission by phlebotomine sandflies. Science Vosloo, W.; Bastos, A. D.; Kirkbride, E.; Esterhuysen, J. J.;
175:1477–1499. Janse Van Rensburg, D.; Bengis, R. G.; Keet, D. W.; and
Tesh, R. B.; Travassos da Rosa, A. P. A.; and Travassos da Thomson, G. R. 1996. Persistent infection of African
Rosa, J. S. 1983. Antigenic relationship among rhab- buffalo (Syncerus caffer) with SAT-type foot and mouth
doviruses infecting terrestrial vertebrates. J Gen Virol disease viruses: Rate of fixation of mutations, antigenic
64:169–176. change and interspecies transmission. J Gen Virol
Thomson, G. R. 1994. Foot-and-mouth disease. In Infec- 77:1457–1467.
tious Diseases of Livestock with Special Reference to Westbury, H. A.; Doughty, W. J.; Forman, A. J.; Suchinta
Southern Africa. Ed. J. A. W. Coetzer, G. R. Thomson, Tangchaitrong; and Kongthan, A. 1988. A comparison
and R. C. Tustin. vol. 2. New York: Oxford University of enzyme-linked immunosorbent assay, complement
Press, pp. 825–852. fixation, and virus isolation for foot-and-mouth disease
———. 1995. Overview of foot and mouth disease in south- diagnosis. Vet Microbiol 17:21–28.
ern Africa. Rev Sci Tech Off Int Epiz 14:503–520.

You might also like