Recent Advances in Biology and Immunobiology of Eimeria Species

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

CLINICAL MICROBIOLOGY REVIEWS, Jan. 2002, p. 58–65 Vol. 15, No.

1
0893-8512/02/$04.00⫹0 DOI: 10.1128/CMR.15.1.58–65.2002

Recent Advances in Biology and Immunobiology of Eimeria Species


and in Diagnosis and Control of Infection with These Coccidian
Parasites of Poultry
P. C. Allen* and R. H. Fetterer
Parasite Biology, Epidemiology, and Systematics Laboratory, Animal and Natural Resources Institute, Agricultural
Research Service, U.S. Department of Agriculture, Beltsville, Maryland 20705

INTRODUCTION .........................................................................................................................................................58
LIFE CYCLE, GENETICS, AND BIOCHEMISTRY...............................................................................................58
SPECIES DETERMINATION AND DIAGNOSIS...................................................................................................59
IMMUNOBIOLOGY ....................................................................................................................................................59
CONTROL OF COCCIDIOSIS ..................................................................................................................................60

Downloaded from cmr.asm.org at MAHIDOL UNIV FAC OF MED on June 25, 2007
Role of Poultry House Management ......................................................................................................................60
Prophylactic Control with Anticoccidials ..............................................................................................................60
Vaccination ................................................................................................................................................................61
Recombinant vaccines ..........................................................................................................................................61
Live vaccines..........................................................................................................................................................61
Use of cytokines as adjuvants .............................................................................................................................62
Alternative Controls Including Natural-Product Feed Additives.......................................................................62
DIRECTION OF FUTURE RESEARCH ..................................................................................................................63
REFERENCES ..............................................................................................................................................................63

INTRODUCTION development. Within the host cells, sporozoites undergo asex-


ual reproduction (schizogony or merogony) in which nuclear
Coccidiosis is recognized as the parasitic disease that has the division is followed by cytoplasmic differentiation, resulting in
greatest economic impact on poultry production. The annual merozoites that break free and penetrate other host cells.
worldwide cost is estimated at about $800 million (107), and These may carry out several more merogonic generations. Sex-
that for the American broiler industry about $450 million. ual reproduction or gametogeny follows the last merogonic
These estimates include the costs of prophylactic in-feed med- cycle. Merozoites enter host cells and develop into either male
ication for broilers and broiler-breeders, alternative treatments (microgamonts) or female (macrogamonts) forms. The mi-
(e.g., with amprolium) if the medications fail, and losses due to crogamonts give rise to many microgametes, that exit, seek,
mortality, morbidity, and poor feed conversions of birds that and penetrate (fertilize) the macrogamonts that then develop
survive outbreaks. into oocysts. Oocysts are shed with the feces. Prepatent periods
LIFE CYCLE, GENETICS, AND BIOCHEMISTRY may generally range from 4 to 5 days postinfection. Maximum
oocyst output ranges from 6 to 9 days postinfection.
Avian Eimeria spp. have homoxenous life cycles, which have The biochemical and genetic mechanisms that control the
been well described (38). A web site (http://www.iah.bbsrc development of Eimeria spp. within host cells are not known.
.ac.uk/eimeria/) that clearly details the biology of coccidia has However, through study of precocious and drug-resistant lines
also become available. In general, oocysts shed in feces un- of E. tenella (86, 87), two linkage groups associated with these
dergo sporogony (a meiotic process) in the external environ-
traits have been identified and mapped to parasite chromo-
ment, a process requiring oxygen, taking about 24 h. The
somes 1 and 2. This information may help identify other ge-
sporulated oocysts contain four sporocysts, each containing
netic loci involved in regulating the life cycle of E. tenella.
two sporozoites. Upon ingestion by a suitable host, oocysts
Genetic segregation data from this study are available through
excyst within the intestinal lumen. This process is aided by
the internet (http://www.genome.org). Other researchers (70)
trypsin, bile, and CO2. The released sporozoites penetrate the
have recently identified a gene (ets3a) whose expression is
villous epithelial cells. Sporozoites of some species (E. brunetti
developmentally regulated and which may be important in
and E. praecox) develop within cells at the site of penetration.
controlling the life cycle of E. tenella. More genetic informa-
Sporozoites of other species (E. acervulina, E. maxima, E.
tion on Eimeria and many other parasites can be found on the
necatrix, and E. tenella) are transported (1, 49, 95, 97) to other
sites, for example the crypt epithelium, where they undergo internet. Some of the websites include Eimeria (http://www
.iah.bbsrc.ac.uk/eimeria/genome.htm), GenBank (http://www
.ncbi.nlm.nih.gov), Parasitology Online (http://www.parasitology-
* Corresponding author. Mailing address: Parasite Biology, Epide- online.com/), and George Washington School of Medicine
miology, and Systematics Laboratory, Animal and Natural Resources
Institute, Agricultural Research Service, U.S. Department of Agricul-
(http://genome.wustl.edu/gsc/).
ture, Beltsville, MD 20705. Phone: (301) 504-8772. Fax: (301) 504- The metabolism of Eimeria spp. has come under new scru-
5306. E-mail: pallen@ANRI.barc.usda.gov. tiny with the increase in our efforts to define protective anti-

58
VOL. 15, 2002 BIOLOGY AND IMMUNOBIOLOGY OF EIMERIA 59

gens for use in vaccines and to target metabolic pathways for cally altered mice (88). In mice, TCR␥␦⫹ CD4⫹ major histo-
chemotherapy. For example, a mannitol cycle has been char- compatibility complex type II (MHC-II)-restricted T cells that
acterized in E. tenella (12, 80) that apparently provides man- produce gamma interferon (IFN-␥) are essential for antigen-
nitol as an energy source for sporulation. This pathway may be specific resistance to both primary and secondary infections.
widespread within Eimeria spp. (12). Newly characterized en- Additionally, B cells and interleukin-6 IL-6 are minor but nec-
zymes in E. tenella and E. maxima have been reported also, as essary components of resistance to primary infections (88).
well as a list of enzyme activities previously reported in avian Other, non-class II-restricted cells appear to be involved in
Eimeria (108). effecting good “memory” responses to challenge infections.
Lack of chickens with genetic modifications of the immune
SPECIES DETERMINATION AND DIAGNOSIS
effector systems (targeted gene disruptions), as well as the
There are seven valid species of chicken coccidia, E. acervu- paucity of monoclonal antibody reagents, has made it more
lina, E. brunetti, E. maxima, E. mitis, E. necatrix, E. praecox, and difficult to dissect the immune responses to avian coccidiosis.
E. tenella (83), each species developing in a particular location Nevertheless, the following facts are clear. There are genetic
within the chick digestive tract. It is common to find at least six components to resistance to primary infections (50, 114).
species (e.g., E. acervulina, E. maxima, E. tenella, E. brunetti, E. Chickens develop solid immunity to homologous secondary
mitis, and E. praecox) in litter samples from a single flock infections (74). Immunity does not prevent sporozoite invasion

Downloaded from cmr.asm.org at MAHIDOL UNIV FAC OF MED on June 25, 2007
during its first 6 weeks (106). Five of the species, E. acervulina, of cells but does prevent sporozoite development (14).
E. brunetti, E. maxima, E. necatrix, and E. tenella, are well The effectors of immune responses to primary and challenge
known and identifiable with relative ease, because they pro- coccidial infections are primarily T cells residing in the gut-
duce characteristic gross lesions. Their pathogenicities range associated lymphoid tissues (58, 59, 60). Humoral immune
from moderate to severe. On the other hand, E. praecox and E. responses also occur, but antibodies play a minor role in resis-
mitis do not kill chickens or produce pathognomic lesions and tance and immunity to coccidia (51, 74). Chicken intestinal
often have been considered to be benign. However, experimen- lymphocytes have been phenotyped using monoclonal antibod-
tal infections result in enteritis, diarrhea, and reduced feed ies, and found to exhibit markers homologous to those of
efficiencies (107), indicating that these two species certainly murine and human CD3, CD4, and CD8 T cells (24, 27, 53, 60,
can cause commercial losses and hence need to be controlled. 94). The CD3 polypeptide complex is found in association with
Generally, the five most pathogenic species listed above can the antigen-specific T-cell receptor (TCR) (21) characterized
be differentiated in the host on the basis of clinical signs, by ␣␤ or ␥␦ heterodimers (27). The CD4⫹ T (helper) cells
characteristic lesions at particular sites of infection in the recognize MHC-II antigens, and CD8⫹ T (cytotoxic/suppres-
chicken intestine, and consideration of the prepatent period, sor) cells recognize MHC-I antigens (60). In chickens,
size of oocysts, and morphology of intracellular stages. How- TCR␥␦⫹ T cells can be either CD4⫹ or CD8⫹ (60).
ever, the less pathogenic species such as E. mitis and E. praecox The roles of these T cells in response to avian Eimeria
might be overlooked. A very useful handbook is available (29) infections appear somewhat different from those in murine
which describes standard diagnostic and testing procedures for Eimeria infections. Studies in which these T-cell populations in
Eimeria species and includes colored photographs of gross chickens infected with E. acervulina or E. tenella have been
lesions caused by the most commonly found species. selectively depleted by treatment with monoclonal antibody
Shirley (82) was the first to use a molecular biological ap- (96) show a different contribution of CD4⫹ and CD8⫹ T cells
proach to differentiate species on the basis of isoenzyme pat- to resistance and immunity, which appears, as judged by oocyst
terns of oocysts by starch block electrophoresis. Ellis and Bum- output, to be dependent on the infecting species of coccidia. In
stead (37) were among the first to demonstrate that rRNA and chickens infected with E. acervulina, CD4⫹ T-cell depletion did
rDNA probes could be used to identify individual species not significantly increase oocyst production during either pri-
through characteristic restriction fragment patterns. Procunier mary or challenge infection (96). Additionally, fewer oocysts
et al. (72) used a randomly amplified polymorphic DNA assay were produced during challenge than during primary infection.
to differentiate E. acervulina and E. tenella and detect within- These results suggest that CD4⫹ T cells are not significant
strain differences. Recombinant DNA techniques have been effectors of resistance or immunity to E. acervulina. On the
used to discriminate different strains of E. tenella (84) and other hand, depletion of CD4⫹ cells in E. tenella infections
develop markers for precocious and drug-resistant strains (86), resulted in increased oocyst output during primary but not
and PCR amplification of internal transcribed spacer region 1 challenge infection, indicating that CD4⫹ cells are important
from genomic DNA has been used to detect and differentiate effectors of resistance to primary E. tenella infections.
six Eimeria species (81). Eight species (including E. hagani) are Depletion of CD8⫹ T cells significantly decreased oocyst
claimed to be differentiated using a two-step PCR procedure output during primary infections of both E. acervulina and E.
(98), and six Australian species have been characterized using tenella. It is theorized that this effect is due to a reduction in the
a PCR-linked restriction fragment length polymorphism ap- number of CD8⫹ cells that serve as transporters for sporozo-
proach (110). These PCR methods should prove very useful for ites (see below). However, CD8⫹ cell depletion results in a
epidemiological surveys of avian coccidiosis. larger oocyst output after a challenge infection. These results
indicate CD8⫹ cells may not be effectors of resistance in pri-
IMMUNOBIOLOGY
mary infection but are necessary effectors in immunity to chal-
Great progress has been made in our understanding of the lenge infections (59, 96). The importance of CD8⫹ T cells in
immune response to coccidial infection. Much of this has come protective immunity to E. tenella infections is also suggested by
from work on model infections with E. vermiformis in geneti- the observation of a sharp transitory increase in the number of
60 ALLEN AND FETTERER CLIN. MICROBIOL. REV.

these cells in the peripheral blood (19) during primary infec- generates superoxide (O2䡠), and in the levels of NO2⫺ ⫹
tion. NO3⫺, stable metabolites of NO䡠, occur in a qualitatively dif-
In fact, CD8⫹ T cells seem to play dual role in the immune ferent time pattern in chickens infected with E. maxima (days
response to coccidial infections in chickens, as determined by 1, 3, and 6 postinfection [p.i.] for NADPH oxidase, day 6 PI for
two-color immunofluorescence staining of infected gut from NO2⫺ ⫹ NO3⫺) (2). The differences in the time courses of
chicken strains differing genetically in susceptibility to coccid- change in these parameters during primary infection suggest
iosis (59, 60). First, during primary infections with E. acervulina the participation of separate cell types in their production.
and E. tenella, the number of T cells coexpressing TCR␥␦ and Plasma NO2⫺ ⫹ NO3⫺ levels are significantly elevated at
CD8 markers increases in the epithelium. There they appear to about 6 days PI during primary infections with E. acervulina
be invaded by sporozoites. It is thought that these are the cells and E. tenella, as well as E. maxima (3, 9), but not during
that transport sporozoites (1, 49, 95, 97) through the lamina homologous challenge infections of well-immunized chickens.
propria to the crypt epithelium, where they exit and invade the This observation suggests that NO䡠 production may be associ-
crypt cells. Populations of CD8⫹ cells also increase in the ated more closely with immune responses associated with in-
mucosa following challenge infection, where they frequently nate resistance than with acquired immunity to coccidiosis.
can be seen in close contact with infected cells. It is theorized There appears to be a genetic link between NO䡠 production
that they are functioning as cytotoxic cells, helping to eliminate during primary infection and resistance to E. tenella (9). A

Downloaded from cmr.asm.org at MAHIDOL UNIV FAC OF MED on June 25, 2007
the parasite challenge by killing the infected cells (59). recent report (36) of in vitro studies suggests that macrophages
Coccidia invade other leukocyte types in the gut mucosa. For stimulated by IFN-␥ produce NO, which inhibits the replica-
example, sporozoites of E. tenella have been found in het- tion of E. tenella within these cells.
erophils (75) and merozoites of E. tenella have been found in
goblet cells and mast cells (34). The relevance of these obser- CONTROL OF COCCIDIOSIS
vations to immune control of avian coccidiosis is not yet clear.
Populations of cells bearing markers (low-level CD8⫹ and Role of Poultry House Management
asialo-GM1) for natural killer (NK) cells increase early during
Because coccidial oocysts are ubiquitous and easily dissem-
primary infections (28, 52). These cells exhibit NK acitivity in
inated in the poultry house environment and have such a large
vitro (23) and thus may be involved in immune surveillance.
reproduction potential, it is very difficult to keep chickens
Mucosal macrophages phagocytize sporozoites during pri-
coccidia free, especially under current intensive rearing condi-
mary and challenge infections, but this activity does not seem
tions. Oocysts sporulate readily in poultry house litter. How-
enhanced in the immune chicken (97). Macrophages may also
ever, they can be damaged by bacteria, other organisms, and
function in sporozoite transport (99). Activated macrophages
ammonia that are also present, and their viability can begin to
are the source of various inflammatory cytokines that can mod-
diminish after 3 weeks (106). Many producers in the United
ulate cellular immune responses.
States basically remove caked litter, let the houses air out for
Increases in the numbers of mucosal mast cells have been
2 to 3 weeks, and top dress with fresh litter before placing a
noted early and late following primary and challenge infections
new flock (H. D. Danforth, private communication). On the
with E. acervulina (76), but the contribution of these cells to
other hand, it is common practice in most European countries
effective immunity and resistance is not known.
and Canada to do a thorough cleanout between flocks. This
Cytokines synthesized and secreted by leukocytes play im-
practice may become more widespread in the United States as
portant regulatory roles during the immune response to infec-
the effectiveness of anticoccidials continues to decrease and
tion. In avian coccidiosis, it is clear that IFN-␥ is produced by
the use of live vaccines increases. Biocontrol measures such as
the host at sites of infection (78, 112), and IFN-␥ release has
requiring caretakers to change clothes between houses can
been used as a measure of the T-cell response to coccidial
minimize the spread of infective oocysts. The practice of strict
antigens (68, 73). Treatment of coccidia-infected chickens with
biosecurity is essential in the care of broiler breeders.
recombinant chicken IFN-␥ (89) improved weight gains with
respect to uninfected controls (55, 64). Tumor necrosis factor
Prophylactic Control with Anticoccidials
(TNF), an inflammatory cytokine, is secreted by activated mac-
rophages. TNF-␣-like activity can be detected in stimulated The effective use of anticoccidial feed additives over the past
macrophages from infected chickens (22, 114) and in sera from 50 years has played a major role in the growth of the poultry
infected chickens (113). Treatment of infected chickens with industry and has allowed the increased availability of high-
TNF-␣ exacerbated the suppression of weight gain due to quality, affordable poultry products to the consumer. These
infection, whereas treatment with polyclonal antibodies to anticoccidials can be classified as (i) chemicals which have
TNF-␣ partially reversed the weight gain depression (114). specific modes of action against parasite metabolism, such as
These results suggest that TNF-␣ may play an important role amprolium, clopidol decoquinate, halofuginone, or (ii) poly-
in the pathophysiology of coccidiosis infections in chickens. ether ionophores such as monensin lasalocid, salinomycin,
Free radicals, including reactive oxygen species and nitric narasin, and maduramycin, which act through general mecha-
oxide (NO䡠), are products of activated macrophages and other nisms of altering ion transport and disrupting osmotoic bal-
phagocytic leukocytes and are known to be toxic to bacteria ance. These latter compounds are now the mainstay of coccid-
and some parasites (77). They are thus implicated as playing iosis control (41). A compendium of the most frequently used
significant roles in resistance and immunity to infectious dis- anticoccidials is available through Janssen Pharmaceutica (40).
eases (65, 71). It is quite clear, however, (25, 26, 79), that some degree of
Peaks in the mucosal activity of NADPH oxidase, which resistance to all anticoccidial drugs, including ionophores, has
VOL. 15, 2002 BIOLOGY AND IMMUNOBIOLOGY OF EIMERIA 61

developed. To minimize the effects of resistance, poultry pro- the highest T-cell proliferation and macrophage-activating ac-
ducers rotate the use of various anticoccidials with successive tivity, when administered as a vaccine, also yielded chickens
flocks, combine chemical and ionophore treatments, or employ with the lowest cecal lesion scores after challenge (20).
shuttle programs during a flock growout. Application of these Live vaccines. Live vaccines for coccidiosis control have
treatment programs depends on seasonal conditions and prev- been used to a limited degree by the poultry industry for about
alence of various species of coccidia (107). In recent years, 50 years. Their effectiveness hinges on the recycling of initially
pharmaceutical companies have not brought new anticoccidials very low doses of oocyst, and the gradual buildup of solid
to market. However, two potential drug targets, enzymes of the immunity. They have been used primarily to protect breeder
sporozoite mannitol cycle (12, 80) and trophozoite histone and layer flocks (85). However, their use, particularly in broiler
deacetylase, have been recently identified (80). flocks, is increasing.
Live vaccines contain either attenuated or virulent coccidial
Vaccination strains. Paracox (consisting of precocious strains of all seven
species of chicken coccidia) and Livacox (consisting of preco-
Avian coccidia are highly immunogenic, and primary infec- cious and egg-passaged lines) are attenuated vaccines. All coc-
tions can stimulate solid immunity to homologous challenges. cidial strains in these vaccines are drug sensitive. Coccivac and
Therefore, it would seem obvious that vaccines could offer Immucox are each composed of several virulent species. All

Downloaded from cmr.asm.org at MAHIDOL UNIV FAC OF MED on June 25, 2007
excellent alternatives to drugs as a means of controlling coc- four vaccines are commercially available. An in-depth compar-
cidiosis. Efforts to develop various types of vaccines (57, 58, 61) ison of these vaccines is provided by Bedrnik et al. (17). All of
have been continuous over the past several decades, but these vaccines are administered during the first week after
progress has been slow. Highlights of research on vaccine de- hatch and will produce solid immunity when they are used
velopment are included here. carefully under good rearing conditions (30). Because live vac-
Recombinant vaccines. Over the past 10 years, much re- cines are multivalent (containing more than one species), tests
search effort has been spent on the development of recombi- of their efficacies require a different approach from tests of the
nant vaccines. Although none are in commercial use, this re- efficacies of anticoccidial compounds. A standard protocol that
search has served to highlight the complex nature of the avian is based on growth rates after challenge and that avoids lesion
host-coccidia interaction. A major hurdle to overcome in the scoring and oocyst output enumeration has been published
development of a recombinant vaccine is the lack of cross- recently (109).
species immune protection. Other factors impeding the devel- An advantage of attenuated vaccines is that they have low
opment of a successful vaccine have been recently reviewed reproductive potentials, thus avoiding crowding in the specific
(42, 100), the most important of which is the identification of mucosal areas of infection and resulting in the development of
protective antigens. Many potential coccidial antigens have optimal immunity with minimal tissue damage (105). It is be-
been characterized and cloned. A total of 29 DNA sequences lieved that the drug-sensitive, attenuated strains and wild, na-
encoding immunity-stimulating Eimeria proteins from various tive strains interbreed, reducing both virulence and drug re-
species and developmental stages have been listed in a recent sistance in local populations. Thus, the useful period of
review (42). Many of these antigens are surface proteins or anticoccidial drugs could be extended by rotating their appli-
internal antigens associated with organelles such as mi- cations with live vaccines (107).
cronemes (92, 93), rhoptries (91), and refractile bodies (101, Coccivac and Immucox have also been used to treat broiler
102). Recently (90) a low-molecular-weight immunogenic an- breeders, particularly in the United States and Canada. There
tigen with a single immunodominant epitope was reported to has been a general reluctance to try them in broilers and heavy
be present in all endogenous stages of E. tenella. Metabolic roaster birds, because of reduced weight gain and feed con-
antigens from developing sporozoites (44, 45, 46), merozoite version compared to those in prophylactically medicated birds
antigens (18, 100), and gamete antigens (103, 104) all elicit (30). Additionally, there is a risk of introducing unwanted
various degrees of protective immunity. Eimeria species into the environment. However, recent battery,
A delivery mechanism for coccidial vaccines that produces floor pen, and broiler house trials (30) have shown that this
optimum resistance to challenge infection has yet to be deter- type of vaccine (e.g., Immucox) can provide equal or superior
mined. Immunogenic Eimeria antigens have been administered performance, compared with prophylactic medication, when
as isolated proteins with adjuvants (18, 100), as recombinant given in gel form at 1 day of age, because this method ensures
antigens in live vectors such as nonpathogenic strains of Esch- the synchronous exposure of all birds to small uniform num-
erichia coli, Salmonella enterica serovar Typhimurium, poxvi- bers of oocysts. (32, 33).
ruses, fowlpox virus, and turkey herpesvirus (93), and by direct Antigenicity of coccidial strains can vary geographically (39,
plasmid DNA injection (43, 48, 54), with various degrees of 66). Therefore, it is important to screen live vaccines against
success. More research is needed to determine how to target local populations before administering them to large flocks.
vaccines to the cellular immune effector systems that operate Immunovariant strains can be isolated and substituted to pro-
during natural primary and challenge infections and to deter- vide region-specific, autogenous vaccines (30). Live vaccines
mine the unique host immune responses to each parasite. can also be modified by incorporating drug-resistant strains. In
Because T-cell-stimulatory antigens appear to be the most floor pen trials, anticoccidially medicated broiler birds that had
relevant to protective immunity, T-cell proliferation assays (us- been immunized at 1 day of age with a drugresistant strain of
ing T cells from recovering chickens) and IFN-␥ production coccidia were protected almost completely against homologous
have been used to screen for protective antigens against E. challenge (30). When this vaccination regimen was applied to
tenella infections (20). The fraction of antigens that produced large flocks in two sets of paired broiler houses, vaccinated-
62 ALLEN AND FETTERER CLIN. MICROBIOL. REV.

medicated birds had consistently lower (more efficient) feed of oxidative stress (due to the high concentration of easily
conversions but slightly more variable final weights than did oxidized double bonds) that is detrimental to parasite devel-
the medicated controls (30). Thus, this type of vaccine treat- opment. Further support for the oxidative-stress hypothesis
ment has the potential to extend the life of anticoccidials include the observations that (i) n-3 FA diets effective in con-
cleared for use in geographical areas where drug resistance and trolling E. tenella reduce plasma carotenoid levels even in
lack of coccidial control is a problem (30). uninfected chickens (8), (ii) the effect on E. tenella appears
Use of cytokines as adjuvants. As mentioned above, cyto- related to the concentrations of double bonds in diets supple-
kines are major modulators of immune responses to infection, mented with n-3 FA ethyl esters (4), (iii) antioxidant-stabilized
and therefore they represent natural sources of immunostimu- diets supplemented with up to 10% flaxseed do not protect
lation that could be used as adjuvants in vaccines. IFN-␥ has against E. tenella (11) and (iv) diets supplemented with 15 and
received the most attention as a possible adjuvant for an an- 20% chia seed (another seed with high linolenic acid content)
ticoccidial vaccine. Chicken IFN-␥ has been cloned (35, 62, and containing no additional antioxidants reduce E. tenella
89). Treatment with recombinant IFN-␥ alone has been shown lesion scores (P. C. Allen and J. DeGroot, unpublished obser-
to moderate reductions in weight gain (55, 64) during E. acer- vations). Additionally, Michalski and Prowse (69) have shown
vulina and E. tenella infections and to moderate oocyst output sporulated oocysts and sporozoites of E. tenella to be deficient
(55) during E. acervulina infection. One problem with the in in superoxide dismutase, an enzyme that would protect them

Downloaded from cmr.asm.org at MAHIDOL UNIV FAC OF MED on June 25, 2007
vivo use of cytokines is their rapid degradation and clearance from damage by reactive oxygen species.
(63). Administering them in DNA form or in a viral or bacte- Artemisinin is a Chinese herb isolated from Artemisia an-
rial vector could overcome this problem and provide a more nua; it is a naturally occurring endoperoxide with antimalarial
practical method for treating large flocks. For example, a re- properties. It has been found effective in reducing oocyst out-
combinant protein (3-1E) from E. acervulina merozoites, put from both E. acervulina and E. tenella infections when fed
which is also found in E. tenella sporozoites and merozoites, at levels of 8.5 and 17 ppm in starter diets (10). The mode of
stimulated IFN-␥ production in chicken spleen lymphocytes. action is also thought to involve oxidative stress.
Direct injection of 3-1E cDNA induced protective immunity Most recently, extracts from 15 Asian herbs were tested for
against E. acervulina challenge. Simultaneous injection of
anticoccidial activity against E. tenella. Of the species tested,
cDNA encoding chicken IFN-␥ further enhanced immunity
extracts from Sophora flavescens Aiton was the most effective in
(56). Chickens treated with a fowl adenovurus chicken IFN-␥
reducing lesion scores, maintaining body weight gain, and re-
construct showed increased weight gains compared to un-
ducing oocyst production. (111)
treated controls and exhibited smaller weight gain reductions
Feed supplementation with antioxidants such as ␥-tocoph-
on challenge with E. acervulina (47).
erol (8 ppm), found plentifully in seed oils such as wheat, corn,
and soybean, and the spice tumeric (1%), as well as its main
Alternative Controls Including Natural-Product medicinal component, curcumin (0.05%), appear effective in
Feed Additives reducing upper- and mid-small-intestinal infections caused by
E. acervulina and E. maxima (5). They are not beneficial for E.
Many different types of substances have been investigated in
the search for alternative controls of coccidiosis. Peptidyl tenella infections, however. The osmoprotectant betaine (from
membrane-interactive molecules are known to have antibacte- sugar beets) has long been known to have beneficial effects on
rial activities. Three of seven tested produced ultrastructural livestock growth and performance. When fed at 0.15% along
damage to sporozoite pellicles after a 5- to 10-min exposure to with 66 ppm salinomycin in floor pen studies, betaine had
5 ␮M concentrations in vitro (67). Oral doses (10, 50, or 75 beneficial effects on weight gains and feed conversion and
␮M) of two of them, which were methylated to prevent pro- resulted in reduced mortality. In tissue culture, betaine and
teolysis, reduced lesion scores from challenge infections with salinomycin significantly reduced cell invasion by E. acervulina.
E. acervulina, E. tenella, and E. maxima, suggesting that they It is concluded that this combination may improve perfor-
have potential for coccidiosis control. mance in coccidially infected chickens directly by its action on
A number of natural products or feedstuffs have been tested E. acervulina development and indirectly by its action as an
as anticoccidial dietary additives. Some of this work has been osmoprotectant, supporting intestinal structure and function.
recently reviewed (5). Sources of fats containing high concen- (13, 15, 16).
trations of n-3 fatty acids (n-3 FA) (docosahexaenoic acid, These studies emphasize the unique biology of the individual
eicosapentaenoic acid, and linolenic acid), such as fish oils, Eimeria species that parasitize chickens, and a pattern of ef-
flaxseed oil, and whole flaxseed, when added to starter rations fectiveness of various dietary treatments seems apparent.
and fed to chicks from 1 day of age, effectively reduced lesions Products that can generate a state of oxidative stress, such as
resulting from challenge infections with E. tenella (6, 7) but not n-3 FA and artemisinin, are particularly effective against the
E. maxima (7). The fish oil and flaxseed oil diets significantly cecal parasite E. tenella. Products that have antioxidant prop-
reduced the degree of parasitization by and development of E. erties, such as ␥-tocopherol and curcumin, seem to be more
tenella (6) and caused ultrastructural degradation of both asex- effective against the mid- and upper-small intestinal species E.
ual and sexual stages, characterized by cytoplasmic vacuoliza- maxima and E. acervulina. The osmoprotectant betaine ap-
tion, chromatin condensation within the nucleus, and lack of pears to be most active against E. acervulina. Practical appli-
parasitophorous vacuole delineation (31). These results are cations of these findings, such as the use of the products in
consistent with reports of the effects of high n-3 FA diets on starter rations or combinations of them with current anticoc-
other parasites (5) and suggest that these diets induce a state cidials or vaccines, appear possible and need to be investigated.
VOL. 15, 2002 BIOLOGY AND IMMUNOBIOLOGY OF EIMERIA 63

DIRECTION OF FUTURE RESEARCH balanced and stabilized flaxmeal-based diets on Eimeria tenella infections in
chickens. Poult. Sci. 79:489–492.
12. Allocco, J. J., H. Profous-Juchelka, R. W. Myers, B. Nare, and D. M.
Over the past several decades, knowledge about the genetic Schmatz. 1999. Biosynthesis and catabolism of mannitol is developmentally
makeup of avian Eimeria spp. and the immunobiological inter- regulated in the protozoan parasite Eimeria tenella. J. Parasitol. 85:167–173.
13. Augustine, P. C. 1997. Effect of betaine on invasion and development of
action of the avian host with the coccidian parasites has in- avian coccidia and growth performance in coccidia-infected chickens. Aust.
creased dramatically. The impetus for this expansion has come Poult. Sci. Symp. 9:39–45.
from efforts to find new, effective ways to control coccidiosis in 14. Augustine, P. C., and H. D. Danforth. 1986. A study in the dynamics of the
invasion of immunized birds by Eimeria sporozoites. Avian Dis. 30:347–351.
the face of increased resistance of the parasites to traditional 15. Augustine, P. C., and H. D. Danforth. 1999. Influence of betaine and
anticoccidal pharmaceuticals. Much of the work on both the salinomycin on intestinal absorption of methionine and glucose and on the
parasites and the host has been aided by the development of ultrastructure of intestinal cells and parasite development stages in chicks
infected with Eimeria acervulina. Avian Dis. 43:89–97.
techniques in molecular biology and driven by the search for 16. Augustine, P. C., J. L. McNaughton, E. Virtanen, and L. Rosi. 1997. Effect
effective vaccines. of betaine on the growth performance of chicks inoculated with mixed
cultures of avian Eimeria species and on invasion and development of
The ultimate key to coccidiosis control may well come in Eimeria tenella and Eimeria acervulina in vivo and in vitro. Poult. Sci.
part from research directed toward a better understanding of 76:802–809.
parasite biology and biochemistry. Some of the questions that 17. Bedrnik, P., T. Hiepe, D. Mielke, and U. Drossigk. 1995. Antigens and
immunization procedures in the development of vaccines against poultry
need to be answered include the following. Why are avian coccidiosis, p. 176–189, In J. Eckert, R. Braun, M. W. Shirley, and F.

Downloaded from cmr.asm.org at MAHIDOL UNIV FAC OF MED on June 25, 2007
coccidia host and site specific? What are the important criteria Coudert (ed.), COST 89/820. Biotechnology: guidelines on techniques in
that allow parasites to invade a host cell? What are the bio- coccidiosis research. European Commission, Luxembourg; Luxembourg.
18. Brake, D. A., G. Strang, and J. E. Lineberger. 1997. Immunogenic charac-
chemical defense mechanisms that promote survival within the terization of a tissue culture-derived vaccine that affords partial protection
host? What genes control the development of coccidia within against avian coccidiosis. Poult. Sci. 76:974–983.
19. Breed, D. G., J. Dorrestein, and A. N. Vermeulen. 1996. Immunity to
the host? New developments or improvements in in vitro cul- Eimeria tenella in chickens: phenotypical and functional changes in periph-
tivation methods, the current worldwide push to unravel the eral blood T-cell subsets. Avian Dis. 40:37–48.
parasite genome, and the availability of genetic sequence in- 20. Breed, D. G., T. M. P. Schetters, N. A. P. Verhoeven, and A. N. Vermeulen.
1997. Characterization of phenotype related responsiveness of peripheral
formation in publicly accessible databases, along with new blood lymphocytes from Eimeria tenella infected chickens. Parasite Immu-
techniques in proteomic research, should help to answer these nol. 19:563–569.
questions. 21. Bucy, R. P., C. L. Chen, J. Cihak, U. Lasch, and M. D. Cooper. 1988. Avian
T cells expressing receptors localize in the splenic sinusoids and the intes-
Further insight into the control of coccidiosis will also come tinal epithelium. J. Immunol. 141:2200–2205.
from research on the avian host that is directed to the following 22. Byrnes, S., R. Eaton, and M. Kogut. 1993. In vitro interleukin-1 and tumor
necrosis factor alpha production by macrophages from chickens infected
questions. What are the host genetic components associated with either Eimeria maxima or Eimeria tenella. Int. J. Parasitol. 23:639–645.
with innate and acquired resistance to coccidial infections (i.e., 23. Chai, J. Y., and H. S. Lillehoj. 1988. Isolation and functional characteriza-
identification of qualitative trait loci) (115)? What are the tion of chicken intestinal intraepithelial lymphocytes showing natural killer
cell activity against tumor target cells. Immunology 63:111–117.
immunological restrictions that limit cross-protection among 24. Chan, M. M., C. L. Chen, L. L. Ager, and M. D. Cooper. 1988. Identification
species? What are the differences in host cellular responses of the avian homologues of mammalian CD4 and CD8 antigens. J. Immu-
between live infection and vaccination? Current and future nol. 140:2133–2138.
25. Chapman, H. D. 1994. Sensitivity of field isolates of Eimeria to monensin
research using microarray and proteomic technologies will be- following the use of a coccidiosis vaccine in broiler chickens. Poult. Sci.
gin to answer some of these questions. It is anticipated that 73:476–478.
large advances will be made in the understanding and control 26. Chapman, H. D. 1998. Evaluation of the efficacy of anticoccidial drugs
against Eimeria species in the fowl. Int. J. Parasitol. 28:1141–1144.
of avian coccidiosis within the next decade. 27. Chen, C. H., J. Cihak, U. Losch, and M. D. Cooper. 1988. Differential
expression of two T cell receptors, TCR1 and TCR2 on chicken lympho-
cytes. Eur. J. Immunol. 18:539–543.
REFERENCES
28. Cheung, K., and H. S. Lillehoj. 1991. Characterization of monoclonal an-
1. Al-Attar, M. A., and M. A Fernando. 1987. Transport of Eimeria necatrix tibodies detecting avian macrophages and NK cells. Vet. Immunol. Immu-
sporozoites in the chicken: effects of irritants injected intraperitonally. J. nopath. 28:351–363.
Parasitol. 73:494–502. 29. Conway, D. P., and M. E. McKenzie. 1991. Poultry coccidiosis diagnosis and
2. Allen, P. C. 1996. Production of free radical species during Eimeria maxima testing procedures, 2nd ed. Pfizer, Inc., New York, N.Y.
infections in chickens. Poult. Sci. 76:814–821. 30. Danforth, H. D. 1998. Use of live oocyst vaccines in the control of avian
3. Allen, P. C. 1997. Nitric oxide production during Eimeria tenella infections coccidiosis: experimental studies and field trials. Int. J. Parasitol. 28:1099–
in chickens. Poult. Sci. 76:810–813. 1109.
4. Allen, P. C., and H. D. Danforth. 1998. Effects of dietary supplementation 31. Danforth, H. D., P. C. Allen, and O. A. Levander. 1997. The effect of high
with n-3 fatty acid ethyl esters on coccidiosis in chickens. Poult. Sci. 77: n-3 fatty acid diets on the ultrastructural development of E. tenella. Para-
1631–1635. sitol. Res. 83:440–444.
5. Allen, P. C., H. D. Danforth, and P. C. Augustine. 1998. Dietary modulation 32. Danforth, H. D., E.-H. Lee, A. Martin, and M. Dekich. 1997. Evaluation of
of avian coccidiosis. Int. J. Parasitol. 28:1131–1140. a gel-immunization technique used with two different Immucox vaccine
6. Allen, P. C., H. D. Danforth, and O. A. Levander. 1996. Diets high in n-3 formulations in battery and floor-pen trials with broiler chickens. Parasitol.
fatty acids reduce cecal lesion scores in broiler chickens infected with Res. 83:445–451.
Eimeria tenella. Poult. Sci. 75:179–185. 33. Danforth, H. D., K. Watkins, A. Martin, and M. Dekich. 1997. Evaluation
7. Allen, P. C., H. D. Danforth, and O. A. Levander. 1997. Interaction of of the efficacy of Eimeria maxima oocysts immunization with different
dietary flaxseed with coccidia infections in chickens. Poult. Sci. 76:822–827. strains of day-old broiler and roaster chickens. Avian Dis. 41:792–801.
8. Allen, P. C., H. D. Danforth, V. L. Morris, and O. A. Levander. 1996. 34. Daszak, P., S. J. Ball, R. M. Pittilo, and C. C. Norton. 1993. Ultrastructural
Association of lowered plasma carotenoids with protection against cecal observations on cecal epithelial cells invaded by first generation merozoites
coccidiosis by diets high in n-3 fatty acids. Poult. Sci. 75:966–973. of Eimeria tenella in vivo. Ann. Trop. Med. Parasitol. 87:259–364.
9. Allen, P. C., and H. S. Lillehoj. 1998. Genetic influence of nitric oxide 35. Digby, M. R., and J. W. Lowenthal. 1995. Cloning and expression of the
production during Eimeria tenella infections in chickens. Avian Dis. 42:397– chicken interferon-gamma gene. J. Interferon Cytokine Res. 15:939–945.
403. 36. Dimier-Poisson, I. H., Z. Soundous, M. Naciri, D. T. Bout, and P. Quere.
10. Allen, P. C., J. Lydon, and H. D. Danforth. 1997. Effects of components of 1999. Mechanisms of the Eimeria tenella growth inhibitory activity induced
Artemisia annua on coccidia infections in chickens. Poult. Sci. 76:1156– by concanavalin A and reticuloendotheliosis virus supernatants with inter-
1163. feron gamma activity in chicken macrophages and fibroblasts. Avian Dis.
11. Allen, P. C., H. D. Danforth, and P. A. Stitt. 2000. Effects of nutritionally 43:65–74.
64 ALLEN AND FETTERER CLIN. MICROBIOL. REV.

37. Ellis, J., and J. Bumstead. 1990. Eimeria species: studies using rRNA and 64. Lowenthal, J. W., J. J. York, T. E. O’Neil, S. Rhodes, S. J. Prowse, D. G.
rDNA probes. Parasitology 101:1–6. Strom, and M. R. Digby. 1997. In vivo effects of chicken interferon-gamma
38. Fernando, A. M. 1990. Eimeria: Infections of the intestine, p. 63–75. in P. L. during infection with Eimeria. J. Interferon Cytokine Res. 17:551–558.
Long (ed.), Coccidiosis of man and domestic animals. CRC Press, Inc., 65. MacMicking, J., Q.-W. Xie, and K. Nathan. 1997. Nitric oxide and macro-
Boca Raton, Fla. phage function. Annu. Rev. Immunol. 15:323–350.
39. Fitz-Coy, S. H. 1992. Antigenic variation among strains of Eimeria maxima 66. Martin, A. G., H. D. Danforth, J. R. Barta, and M. A. Fernando. 1997.
and E. tenella of the chicken. Avian Dis. 36:40–43. Analysis of immunological cross protection and sensitivities to anticoccidial
40. Fowler, N. G. 1995. Anticoccidial compendium. Janssen Pharmaceutica, drugs among five geographical and temporal strains of Eimeria maxima. Int.
Animal Health, Beerse, Belgium. J. Parasitol. 5:527–533.
41. Jeffers, T. 1997. Tyzzer to tomorrow: control of avian coccidiosis into the 67. Martin, A. G., H. D. Danforth, J. M. Jaynes, and J. E. Thornton. 1999.
next millenium, p 16. In M. W. Shirley, F. M. Tomley, and B. M. Freeman Evaluation of the effect of peptidyl membrane-interactive molecules on
(ed.), Control of coccidiosis into the next millennium. Proc. VII Int. Coc- avian coccidia. Parasitol. Res. 85:331–336.
cidiosis Conf. 68. Martin, A. G., H. S. Lillehoj, B. Kaspers, and L. Bacon. 1994. Mitogen-
42. Jenkins, M. C. 1998. Progress on developing a recombinant coccidiosis induced lymphocyte proliferation and interferon production following coc-
vaccine. Int J. Parasitol. 28:1111–1119 cidia infection. Avian Dis. 38:262–268.
43. Jenkins, M. C., P. Allen, H. Danforth, and P. Augustine. 1998. Immuniza- 69. Michalski, W. P., and S. J. Prowse. 1991. Superoxide dismutases in Eimeria
tion of chickens with plasmid DNA encoding recombinant Eimeria acervu- tenella. Mol. Biochem. Parasitol. 47:189–195.
lina antigen via jet-gun injection elicits immunity against coccidiosis, p. 47. 70. Ouarzane, M., M. Labbe, and P. Pery. 1998. Eimeria tenella: cloning and
Proc. 43rd Annu. Meet. AAVP. characterization of cDNA encoding a s3a ribosomal protein. Gene 28:125–
44. Jenkins, M. C., P. C. Augustine, J. R. Barta, and M. D. Castle. 1991. 130.
Development of resistance to coccidiosis in the absence of merogonic 71. Ovington, K. S., and N. C. Smith. 1992. Cytokines, Free radicals and
development using X-irradiated Eimeria acervulina oocysts. Exp. Parasitol. resistance to Eimeria. Parasitol. Today 8:422–426.

Downloaded from cmr.asm.org at MAHIDOL UNIV FAC OF MED on June 25, 2007
72:285–293. 72. Procunier, J. D., M. A. Fernando, and J. R. Barta. 1993. Species and strain
45. Jenkins, M. C., P. C. Augustine, H. D. Danforth, and J. R. Barta. 1991. differentiation of Eimeria spp. of the domestic fowl using DNA polymor-
X-irradiation of Eimeria tenella oocysts provides direct evidence that sporo- phisms amplified by arbitrary primers. Parasitol. Res. 79:98–102.
zoite invasion and early schizont development induce protective immune 73. Prowse, S. J., and J. Pallister. 1989. Interferon release as a measure of the
response(s). Infect. Immun. 59:4042–4048. T-cell response to coccidial antigens in chickens. Avian Pathol. 18:619–630.
46. Jenkins, M. C., P. G. Sefarian, P. C. Augustine, and H. D. Danforth. 1993. 74. Rose, M. E. 1987. Eimeria, Isospora, and Cryptosporidium, p. 275–312. In
Protective immunity against coccidiosis elicited by radiation-attenuated Ei- E. J. L. Soulsby (ed.), Immune responses in parasitic infections: immunol-
meria maxima sporozoites that are incapable of asexual development. Avian ogy, immunopathology and immunoprophylaxis. CRC Press, Inc., Boca
Dis. 37:74–82. Raton, Fla.
47. Johnson, M. A., C. Pooley, and J. W. Lowenthal. 2000. Delivery of avian 75. Rose, M. E., and D. L. Lee. 1977. Interactions in vitro between sporozoites
cytokines by adenovirus vectors. Dev. Comp. Immunol. 24:343–354. of Eimeria tenella and host peritoneal exudate cells: electron microscopal
48. Kopko, S. H., D. S. Martin, and J. R. Barta. 2000. Responses of chickens to observations. Z. Parasitenkd. 54:1–7.
a recombinant refractile body antigen of Eimeria tenella administered using 76. Rose, M. E., B. M. Ogilvie, and J. W. Bradley. 1980. Intestinal mast cell
various immunizing strategies. Poult. Sci. 79:336–342. response in rats and chickens to coccidiosis, with some properties of
49. Lawn, A. M., and M. E. Rose. 1982. Mucosal transport of Eimeria tenella in chicken mast cells. Int. Arch. Allergy Appl. Immunol. 63:21–29.
the cecum of the chicken. J. Parasitol. 68:1117–1123. 77. Rosen, G. M., S. Pou, C. L. Ramos, M. Cohen, and B. E. Britigan. 1995.
50. Lillehoj, H. S. 1986. Immune response during coccidiosis in SC and FP Free radicals and phagocytic cells. FASEB J. 9:200–209.
chickens. I. In vitro assessment of T cell proliferation response to stage 78. Rothwell, L., W. Muir, and P. Kaiser. 2000. Interferon-gamma is expressed
specific parasite antigens. Vet. Immunol. Immunopathol. 13:321–330. in both gut and spleen during Eimeria tenella infection. Avian Pathol.
51. Lillehoj, H. S. 1987. Effects of immunosuppression of avian coccidiosis: 29:333–342.
cyclosporin A but not hormonal bursectomy abrogates host protective im- 79. Ruff, M. D., and H. D. Danforth. 1966. Resistance of coccidia to medica-
munity. Infect. Immun. 55:1616–1621. tions, p. 427–430. In Proc. XX World’s Poult. Congr., vol. II.
52. Lillehoj, H. S. 1989. Intestinal intraepithelial and splenic natural killer cell 80. Schmatz, D. M. 1997. Anticoccidial drug discovery and design, p. 20–21, In
responses to Eimeria infections in inbred chickens. Infect. Immun. 57:1879– M. W. Shirley, F. M. Tomley, and B. M. Freeman (ed.), Control of coccid-
1884. iosis into the next millennium. Proc. VII Int. Coccidiosis Conf.
53. Lillehoj, H. S. 1994. Analysis of Eimeria acervulina induced changes in 81. Schnitzler, B. E., P. Thebo, F. M. Tomley, M. W. Shirley, and A. Uggla.
intestinal T lymphocyte subpopulations in two inbred chickens showing 1997. Identification of Eimeria sp in poultry by in vitro amplification of the
different levels of disease susceptibility to coccidia. Res. Vet. Sci. 56:1–7. internal transcribed spacer 1, p. 64. In M. W. Shirley, F. M. Tomley, and
54. Lillehoj, H. S., K. D. Chai., D. Heller, M. C. Jenkins, and V. N. Vakharia. B. M. Freeman (ed.), Control of coccidiosis into the next millennium. Proc.
1997. Naked DNA immunization with a plasmid encoding an Eimeria VII Int. Coccidiosis Conf.
surface sporozoite protein and oral feeding of baculovirus-expressed Eime- 82. Shirley, M. W. 1975. Enzyme variation in Eimeria species of the chicken.
ria protein elicit serum antibodies and reduced oocyst output, p. 105. Proc. Parasitology 71:369– 376.
VII Int. Coccidiosis Conf. 83. Shirley, M. W. 1986. New methods for the identification of species and
55. Lillehoj, H. S., and K. D. Choi. 1998. Recombinant chicken interferon- strains of Eimeria, p. 13–35. In L. R. McDougald, P. L. Long, and L. P.
gamma inhibits in vitro Eimeria tenella development, and reduces in vivo Joyner (ed.), Research in avian coccidiosis. University of Georgia, Athens.
oocyst production and body weight loss following challenge infection with 84. Shirley, M. W. 1994. Coccidial parasites from the chicken: discrimination of
Eimeria acervulina. Avian Dis. 42:307–314. different populations of Eimeria tenella by DNA hybridisation. Res. Vet.
56. Lillehoj, H. S., K. D. Choi, M. C. Jenkins, V. N. Vakharia, K. D. Song, J. Y. Sci. 57:10–14.
Han, and E. P. Lillehoj. 2000. A recombinant Eimeria protein inducing 85. Shirley, M. W., A. C. Bushell, J. E. Bushell, V. McDonald, and B. Roberts.
interferon-gamma production: comparison of different gene expression sys- 1995. A live attenuated vaccine for control of avian coccidiosis: trials in
tems and immunization strategies for vaccination against coccidiosis. Avian broiler breeders and replacement layer flocks in the United Kingdom. Vet.
Dis. 44:379–389. Rec. 137:453–457.
57. Lillehoj, H. S., and E. P. Lillehoj. 2000. Avian coccidiosis. A review of 86. Shirley, M. W., and D. A. Harvey. 1996. Eimeria tenella: genetic recombi-
acquired intestinal immunity and vaccination strategies. Avian Dis. 44:408– nation of markers for precoccious development and arprinocid resistance.
425. Appl. Parasitol. 37:293–299.
58. Lillehoj, H. S., and J. M. Trout. 1993. Coccidia: a review of recent advances 87. Shirley, M. W., and D. A. Harvey. 2000. A genetic linkage map of the
on immunity and vaccine development. Avian Pathol. 22:3–21. apicomplexan protozoan parasite Eimeria tenella. Genome Res. 10:1587–
59. Lillehoj, H. S., and J. M. Trout. 1994. CD8⫹ T cell-coccidia interactions. 1593.
Parasitol. Today 10:10–14. 88. Smith, A. L., and A. C. Hayday. 1998. genetic analysis of the essential
60. Lillehoj, H. S., and J. M. Trout. 1996. Avian gut-associated lymphoid tissues components of the immunoprotective response to infection with Eimeria
and intestinal immune responses to Eimeria parasites. Clin. Microbiol. Rev. vermiformis. Int. J. Parasitol. 28:1060– 1069.
9:349–360. 89. Song, K. D., H. S. Lillehoj, K. D. Choi, D. Zarlenga, and J. Y. Han. 1997.
61. Lillehoj, E. P., C. H. Yun, and H. S. Lillehoj. 2000. Vaccines against the Expression and functional characterization of recombinant chicken inter-
avian enteropathogens Eimeria, Cryptosporidium and Salmonella. Anim. feron-gamma. Vet. Immunol. Immunopathol. 58:321–333.
Health Res. Rev. 1:47–65. 90. Tennyson, S. A., and J. R. Barta. 2000. Localization and immunogenicity of
62. Lowenthal, J. W., M. R. Digby, and J. J. York. 1995. Production of inter- a low molecular weight antigen of Eimeria tenella. Parasitol. Res. 86:453–
feron-gamma by chicken T-cells. J. Interferon Cytokine Res. 15:933–938. 460.
63. Lowenthal, J. W., T. E. O’Neil, A. David, G. Strom, and M. E. Andrew. 1999. 91. Tomley, F. M. 1994. Characterization of rhoptry proteins of Eimeria tenella
Cytokine therapy: a natural alternative for disease control. Vet Immunol. sporozoites: antigenic diversity of rhoptry epitopes within species of the
Immunopathol. 72:183–188. genus Eimeria and among three asexual generations of a single species, E.
VOL. 15, 2002 BIOLOGY AND IMMUNOBIOLOGY OF EIMERIA 65

tenella. Infect. Immun. 62:4656–4658. 104. Wallach, M., N. C. Smith, M. Petracca, C. M. D. Miller, J. Eckert, and R.
92. Tomley, F. M., J. M. Bumstead, K. J. Billington, and P. P. J. Dunn. 1996. Braun. 1995. Eimeria maxima gametocyte antigens: potential use as a sub-
Molecular cloning and characterization of a novel acidic microneme protein unit maternal vaccine against coccidiosis in chickens. Vaccine 13:347–354.
(Etmic-2) from the apicomplexan protozoan parasite, E. tenella. Mol. Bio- 105. Williams, R. B. 1994. Safety of the attenuated anticoccidial vaccine “Para-
chem. Parasitol. 79:195–206. cox” in broiler chickens isolated from extraneous coccidial infection. Vet.
93. Tomley, F. M., L. E. Clark, U. Kawazoe, R. Dijkema, and J. J. Kok. 1991. Res. Commun. 18:189–198.
Sequence of gene encoding an immunodominant microneme protein of E. 106. Williams, R. B. 1995. Epidemiological studies of coccidiosis in the domestic
tenella. Mol. Biochem. Parasitol. 49:277–288. fowl (Gallus gallus). II. Physical condition and survival of Eimeria acervulina
94. Tomley, F., A. N. Vermeulen, and P. van den Boogart. 1993. The use of live oocysts in poultry house liter. Appl. Parasitol. 36:90–96.
vectors for presentation of recombinant antigens to chickens, p. 124–126. In 107. Williams, R. B. 1998. Epidemiological aspects of the use of live anticoc-
Proc. VI Int. Coccidiosis Conf. cidial vaccines for chickens. Int. J. Parasitol. 28:1089–1098
95. Trout, J. M., and H. S. Lillehoj. 1993. Transport of Eimeria acervulina 108. Williams, R. B. 1999. Three enzymes newly identified from the genus
sporozoites, evidence of a role for intestinal CD8⫹ lymphocytes and mac- Eimeria and two more newly identified from E. maxima, leading to the
rophages. J. Parasitol. 79:790–792. discovery of some aliphatic acids with activity against coccidia of the do-
96. Trout, J. M., and H. S. Lillehoj. 1996. T lymphocyte roles during Eimeria
mestic fowl. Vet. Res. Commun. 23:151–163.
acervulina and Eimeria acervulina infections. Veterinary immnuology and
109. Williams, R. B., and J. Catchpole. 2000. A new protocol for a challenge test
immunopathology 53:163–172.
to assess the efficacy of live anticoccidial vaccines for chickens. Vaccine
97. Trout, J. M., and H. S. Lillehoj. 1995. Eimeria acervulina infection: evidence
18:1178–1185.
for the involvement of CD8⫹ T lymphocytes in sporozoite transport and
110. Woods, W. G., K. G. Whithear, D. G. Richards, G. R. Anderson, W. K.
host protection. Poult. Sci. 74:1117–1125
Jorgensen, and R. B. Gasser. 2000. Single-strand restriction fragment
98. Tsuji, N., S. Kawazu, M. Ohta, T. Kamio, T. Isobe, K. Shimura, and K.
length polymorphism analysis of the second internal transcribed spacer
Fujisaki. 1997. Discrimination of eight chicken Eimeria species using the
(ribosomal DNA) for six species of Eimeria from chickens in Australia. Int.

Downloaded from cmr.asm.org at MAHIDOL UNIV FAC OF MED on June 25, 2007
two-step polymerase chain reaction. J. Parasitol. 83:966–970.
99. Van Doornick, W. M., and E. R. Becker. 1957. Transport of Eimeria necatrix J. Parasitol. 30:1019–1023.
in macrophages. J. Parasitol. 43:40–44. 111. Youn, H. J., and J. W. Noh. 2001. Screening of the anticoccidial effects of
100. Vermeulen, A. N. 1998. Progress in recombinant vaccine development herb extracts against Eimeria tenella. Vet. Parasitol. 96:257–263.
against coccidiosis. A review and prospects into the next millenium. Int J. 112. Yun, C. H., H. S. Lillehoj, J. Zhu, and W. Min. 2000. Kinetic differences in
Parasitol. 28:1121–1130. intestinal and systemic interferon-gamma and antigen-specific antibodies in
101. Vermeulen, A., R. Dijkema, and J. J. Kok. Mar. 1994. Coccidiosis vaccine. chickens experimentally infected with Eimeria maxima. Avian Dis. 44:305–
European patent 0349071B1. 312.
102. Vermeulen, A. N., J. J. Kok, P. van den Boogart, R. Dijkema, and J. A. J. 113. Zhang, S., H. S. Lillehoj, and M. D. Ruff. 1995. Chicken tumor necrosis-like
Claessens. 1993. Eimeria refractile body proteins contain 2 potentially func- factor. I. In vitro production by macrophages stimulated with Eimeria
tional characteristice-transhydrogenase and carbohydrate transport. FEMS tenella or bacterial lipopolysaccharide. Poult. Sci. 74:1304–1310.
Microbiol. Lett. 1993:223–230. 114. Zhang, S., H. S. Lillehoj, and M. D. Ruff. 1995. In vivo role of tumor
103. Wallach, M., A. Halabi, G. Pillemer, O. Sar Shalom, D. Mencher, M. Gilad, necrosis-like factor in Eimeria tenella infection. Avian Dis. 39:859–866.
U. Bendheim, H. D. Danforth, and P. C. Augustine. 1992. Maternal immu- 115. Zhu, J., H. S. Lillehoj, P. Allen, C. H. Yun, D. Pollock, and E. Emara. 2000.
nization with gametocyte antigens as a means of providing immunity against Analysis of disease resistance associated parameters in broiler chickens
Eimeria maxima in chickens. Infect. Immun. 60:2036– 2039. challenged with Eimeria maxima. Poult. Sci. 79:619–625.

You might also like