Veterinary Immunology and Immunopathology: A. Beineke, C. Puff, F. Seehusen, W. Baumga Rtner

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

Veterinary Immunology and Immunopathology 127 (2009) 1–18

Contents lists available at ScienceDirect

Veterinary Immunology and Immunopathology


journal homepage: www.elsevier.com/locate/vetimm

Review paper

Pathogenesis and immunopathology of systemic and nervous canine


distemper
A. Beineke, C. Puff, F. Seehusen, W. Baumgärtner *
Department of Pathology, University of Veterinary Medicine, Bünteweg 17, 30559 Hannover, Germany

A R T I C L E I N F O A B S T R A C T

Article history: Canine distemper is a worldwide occurring infectious disease of dogs, caused by a
Received 9 May 2008
morbillivirus, closely related to measles and rinderpest virus. The natural host range
Received in revised form 17 September 2008
comprises predominantly carnivores. Canine distemper virus (CDV), an enveloped,
Accepted 18 September 2008
negative-sense RNA virus, infects different cell types, including epithelial, mesenchymal,
neuroendocrine and hematopoietic cells of various organs and tissues. CDV infection of
Keywords:
Canine distemper virus dogs is characterized by a systemic and/or nervous clinical course and viral persistence in
Cell tropism selected organs including the central nervous system (CNS) and lymphoid tissue. Main
Lymphoid tissues manifestations include respiratory and gastrointestinal signs, immunosuppression and
Central nervous system demyelinating leukoencephalomyelitis (DL). Impaired immune function, associated with
Immunopathology depletion of lymphoid organs, consists of a viremia-associated loss of lymphocytes,
Neuroimmunology especially of CD4+ T cells, due to lymphoid cell apoptosis in the early phase. After clearance
of the virus from the peripheral blood an assumed diminished antigen presentation and
altered lymphocyte maturation cause an ongoing immunosuppression despite repopula-
tion of lymphoid organs. The early phase of DL is a sequel of a direct virus-mediated
damage and infiltrating CD8+ cytotoxic T cells associated with an up-regulation of pro-
inflammatory cytokines such as interleukin (IL)-6, IL-8, tumor necrosis factor (TNF)-a and
IL-12 and a lacking response of immunomodulatory cytokines such as IL-10 and
transforming growth factor (TGF)-b. A CD4+-mediated delayed type hypersensitivity and
cytotoxic CD8+ T cells contribute to myelin loss in the chronic phase. Additionally, up-
regulation of interferon-g and IL-1 may occur in advanced lesions. Moreover, an altered
balance between matrix metalloproteinases and their inhibitors seems to play a pivotal
role for the pathogenesis of DL. Summarized, DL represents a biphasic disease process
consisting of an initial direct virus-mediated process and immune-mediated plaque
progression. Immunosuppression is due to early virus-mediated lymphocytolysis
followed by still poorly understood mechanisms affecting antigen presentation and
lymphocyte maturation.
ß 2008 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Viral properties . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
3. Natural host range and different strains of CDV . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2

* Corresponding author. Tel.: +49 511 953 8620; fax: +49 511 953 8675.
E-mail address: wolfgang.baumgaertner@tiho-hannover.de (W. Baumgärtner).

0165-2427/$ – see front matter ß 2008 Elsevier B.V. All rights reserved.
doi:10.1016/j.vetimm.2008.09.023
2 A. Beineke et al. / Veterinary Immunology and Immunopathology 127 (2009) 1–18

4. Protective immunity in CDV infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3


5. Pathogenesis of distemper. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
5.1. Route of infection and virus spread. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
5.2. Clinical manifestations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
6. Pathology of non-nervous tissues . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
7. Immunosuppression in canine distemper. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
7.1. Lymphotropism of CDV . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
7.2. Morphological findings in lymphoid organs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
7.3. Phenotypical changes in lymphoid organs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
7.4. Mechanisms of immunosuppression in canine distemper . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
8. Pathogenesis of nervous distemper. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
8.1. Virus entry, spread and expression within the CNS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
8.2. Neurotropism of CDV . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
8.2.1. Oligodendroglial infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
8.2.2. Astrocytic infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
8.2.3. Neuronal infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
8.3. Pathology of nervous distemper . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
8.4. Immunopathology of demyelinating leukoencephalomyelitis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
8.5. Role of cytokines in the pathogenesis of demyelinating leukoencephalitis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
8.6. Role of extracellular matrix in the pathogenesis of demyelinating leukoencephalitis . . . . . . . . . . . . . . . . . . . . . . . . . 12
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14

1. Introduction manifestation forms of distemper, the pathogenesis of


demyelination in the central nervous system (CNS) and
Canine distemper is a worldwide occurring infectious mechanisms of immunosuppression in CDV infected dogs.
disease, caused by a morbillivirus of the family Paramyx- In addition, the perpetuating role of the misdirected
oviridae. The natural host spectrum of canine distemper immune response especially for the development of
virus (CDV) comprises all families of the order Carnivora nervous white matter lesions is detailed.
(Deem et al., 2000). Clinical and morbid anatomical signs of
canine distemper were already described in detail at the 2. Viral properties
beginning of the 19th century by Jenner (Lauder et al.,
1954). He also commented on the high incidence of CDV belongs to the family Paramyxoviridae (Lamb and
nervous complications in affected dogs. Kolakofsky, 2001). Beside CDV, the genus morbillivirus
Recent canine distemper epidemics have been observed comprises measles virus, dolphin morbillivirus, peste-des-
in France, Germany, USA, Japan and Finland demonstrating petits-ruminants virus, rinderpest virus, porpoise morbil-
the importance of regular vaccination as a highly efficient livirus, and phocine distemper virus (Pringle, 1999; Lamb
and protective tool (Adelus-Neveu et al., 1991; Mori et al., and Kolakofsky, 2001).
1994; Johnson et al., 1995; Gemma et al., 1996; Ek- CDV is an enveloped, negative-sense, single-stranded
Kommonen et al., 1997; Moritz et al., 2000). Furthermore, RNA virus (Lamb and Kolakofsky, 2001), which contains six
occasional disease outbreaks can be observed in vacci- structural proteins, the nucleocapsid (N), the phospho- (P),
nated dog cohorts, possible due to the introduction of the large (L), the matrix- (M), the hemagglutinin (H), and
genetically different CDV strains (Mori et al., 1994; the fusion (F) protein (Hall et al., 1980; Örvell, 1980; Diallo,
Mochizuki et al., 1999; Simon-Martı́nez et al., 2007). 1990; Lamb and Kolakofsky, 2001). Additional accessory
Though infection of dogs may result in a variety of genes, the C and V protein, are found mostly as extra
clinical forms affecting the respiratory and gastrointestinal transcriptional units within the P gene (Lamb and
tract, skin and other organs and tissues, immunosuppres- Kolakofsky, 2001). The lipid envelope, surrounding the
sion and demyelinating leukoencephalitis (DL) represent virion, contains the two surface glycoproteins F and H,
the main sequel in this species (Krakowka et al., 1985; which mediate viral entry and exit from the host cell.
Appel, 1987). Due to the morphological similarities of Furthermore, the helical nucleocapsid core, containing the
neuropathological changes between DL and human N, P and L protein, initiates intracellular replication and is
demyelinating diseases, such as multiple sclerosis (MS), located within the envelope (Lamb and Kolakofsky, 2001).
the canine disease represents one of the few spontaneously The viral M protein connects the surface glycoproteins and
occurring animal models to study the pathogenesis of nucleocapsid during viral maturation.
myelin loss associated with immune-mediated mechan-
isms (Baumgärtner and Alldinger, 2005). Furthermore, 3. Natural host range and different strains of CDV
lymphoid depletion and long-lasting impaired immune
functions in distemper are comparable with those The host spectrum of CDV, which traditionally included
described for measles virus (MV) infection, a closely numerous families within the order of Carnivora like
related morbillivirus in humans. It is the aim of the present Canidae (dogs, dingos, foxes), Procyonidae (raccoons,
communication to provide an overview about the different kinkajous, lesser panda), Mustelidae (ferret, mink, badger),
A. Beineke et al. / Veterinary Immunology and Immunopathology 127 (2009) 1–18 3

Mephitidae (skunk), Hyaenidae, Ailuridae, Viverridae and recognize viral envelope proteins (Krakowka et al.,
Felidae, has changed in the last years and distemper-like 1975b). At this, especially anti-viral H protein specific
diseases have been observed in large felids in the antibodies prevent the development of CNS lesions in
Tanzanian Serengeti National Park in 1994 and in North infected dogs (Rima et al., 1991). However, a temporary
American zoos in 1991 und 1992 (Montali et al., 1987; lack of antibodies against the viral M protein as well as a
Appel et al., 1994; Roelke-Parker et al., 1996), collared delayed or diminished complement-fixing antibody
peccaries (Tayassu tajacu) in Arizona (Appel et al., 1991) response to viral envelope proteins results in persistent
and in a non-human primate (Macaca fuscata) in Japan neurological disease (Krakowka et al., 1975b; Miele and
(Gorham, 1966; Budd, 1981; Yoshikawa et al., 1989; Krakowka, 1983). Additionally, neutralizing antibodies and
Machida et al., 1992, 1993; van Moll et al., 1995; complement-mediated humoral cytotoxicity represent
Baumgärtner et al., 2003). Furthermore, besides infection critical factors for the elimination of free viral particles
with the closely related phocine distemper virus, seals can and for the prediction of the clinical outcome in CDV-
become infected by CDV (Mahy et al., 1988; Kennedy et al., infected animals (Ho and Babiuk, 1980; Appel et al., 1982,
1989; Likhoshway et al., 1989; Bostock et al., 1990; Curran 1984). Neutralizing antibodies prevent intracellular and
et al., 1990; McCullough et al., 1991; Pringle, 1999; Müller extracellular viral spread; however, prolonged antibody
et al., 2000, 2002; Seibel et al., 2007; Wohlsein et al., 2007). exposure results in an internalization of viral surface
In some CDV outbreaks including the mass mortalities antigens from the cell membrane and subsequently in their
among Baikal and Caspian seals and large felids in the disappearance from the membrane of infected cells. Thus,
Serengeti Park, terrestrial carnivores including dogs and despite a reduction of viral spread, antibody-mediated
wolves have been suspected as vectors for the infectious modulation of viral antigen expression might represent a
agent (Kennedy et al., 2000; Kreutzer et al., 2008). contributing factor for persistent infection in dogs,
So far, only one serotype of CDV is recognized; however, possibly due to a diminished antigen recognition and
several co-circulating CDV genotypes of different virulence resultant inadequate complement-mediated humoral
and cell tropism have been found (McCullough et al., cytotoxicity (Ho and Babiuk, 1979b, 1980; Alldinger
1974a,b,c; Summers et al., 1984; Shapshak et al., 1987; et al., 1993b). Following CDV infection, a virus-specific
Mori et al., 1994; Frisk et al., 1999a; Haas et al., 1999). humoral immune response can be detected throughout
Some strains are associated with a polioencephalitis (e.g. life, while a cellular immune response can only be detected
Snyder Hill-CDV strain) while others induce a demyelinat- for a short time period in convalescent dogs (Appel et al.,
ing leukoencephalomyelitis (DL) (e.g. R252- and A75-17- 1982). However, the importance of the cellular immune
CDV strain; Krakowka and Koestner, 1977; Summers and response in canine distemper is highlighted by the
Appel, 1994; Orlando et al., 2008). Though sequence data demonstration of a protective cellular immunity in the
analysis of field isolates of CDV revealed several clusters of absence of a detectable antibody-mediated immune
distemper strains, overall a considerable genetic stability response (Gerber and Marron, 1976). Additionally, vacci-
of CDV even in recent outbreaks was observed. Still, nated and infected dogs exhibit a T cell-mediated CDV-
cocirculation of more than one genotype of CDV must be specific immune response, independently of the antibody
considered (Haas et al., 1997; Frisk et al., 1999a). Similarly, titer (Krakowka et al., 1978). Cytotoxic T cells (Krakowka
different genotypes of CDV have been identified in recent and Wallace, 1979; Shek et al., 1980; Appel et al., 1982),
isolates from dogs in disease outbreaks in Japan (Mori Killer cells (Ho and Babiuk, 1979a), Natural Killer (NK) cells
et al., 1994; Mochizuki et al., 1999). (Ringler and Krakowka, 1985) and macrophage function
(Appel et al., 1984) have also been investigated in distemper
4. Protective immunity in CDV infection and revealed that a sustained antiviral lymphocyte-
mediated cytotoxicity determines the clinical outcome in
An intact humoral and cell-mediated immune response CDV-infected dogs. Thereby, a strong cellular immunity
is crucial for the elimination of CDV. The humoral immune causes viral elimination, while a lacking or a delayed
response in canine distemper has been investigated in lymphocyte-mediated cytotoxicity correlates with viral
naturally and experimentally infected animals (Krakowka persistence in the CNS (Appel et al., 1982). However, NK
et al., 1980; Krakowka, 1982; Rima et al., 1991). Anti-CDV cell function seems to play a minor role in the host immune
IgM antibodies occur within the first 2 weeks of infection defense against CDV (Ringler and Krakowka, 1985).
(Vandevelde and Zurbriggen, 2005). Here, the magnitude
of the humoral immune response correlates with the 5. Pathogenesis of distemper
outcome of the disease. In general, protective humoral
immunity in canine distemper is due to the production of Great variations in duration, severity and clinical
anti-viral nucleoprotein antibodies, followed by the presentation of distemper have been found in experimen-
appearance of a virus envelope protein-specific humoral tally infected dogs as well as in animals suffering from this
immune response (Miele and Krakowka, 1983; Rima et al., world-wide spontaneously occurring disease. The incuba-
1991). The lack of an effective humoral immune response tion period may vary from 1 to 4 weeks and depends on
leads to an acute, often fatal clinical course, while a viral strain, age of the animal at the time of infection and
detectable virus-specific antibody titer at 10–14 days post- immune status of the host. Disease manifestation ranges
infection (pi) promotes viral elimination or persistence, from virtually no clinical signs to severe disease with
respectively. Referring to this, viral clearance significantly approximately 50% mortality (Appel, 1970, 1987; Kra-
depends upon the specificity of immunoglobulins to kowka et al., 1980; Moritz et al., 2000; Schwab et al., 2007).
4 A. Beineke et al. / Veterinary Immunology and Immunopathology 127 (2009) 1–18

5.1. Route of infection and virus spread 2002; Vandevelde and Zurbriggen, 2005; Amude et al.,
2007).
The virus is shed primarily by oro-nasal secretion, In some cases, an improved immune response espe-
however, any discharge and secretion can carry the virus. cially an increased production of virus-specific neutraliz-
CDV infects susceptible dogs primarily by inhalation of ing antibodies can promote the recovery of the animal.
airborne viruses or via infective aerosol droplets (Kra- However, despite elimination of the virus from several
kowka et al., 1980) followed by virus replication in organs and the peripheral blood, CDV can persist in certain
lymphoid tissues of the respiratory tract. Tissue macro- tissues including uvea, CNS, lymphoid organs and footpads
phages and monocytes located in or along the respiratory (Appel, 1970, 1987; Zurbriggen et al., 1995a,b; Greene and
epithelium and tonsils represent the first cell type to pick Appel, 1998; Gröne et al., 2003a; Schobesberger et al.,
up and propagate the virus (Appel, 1970). Following this 2005). Moreover, some infected animals display a delayed
local burst of virus replication the pathogen is then progression of the disease and a moderate immune
disseminated by lymphatics and blood to distant hema- response with subtle early clinical signs. Later, as a
topoietic tissues during the first viremic phase. consequence of viral persistence in the CNS, overt CNS
First clinical signs are characterized by lethargy, disturbances can be observed resulting in the nervous form
dehydration, anorexia, and weight loss followed by a more of canine distemper. Dogs with nervous signs usually die,
pronounced clinical manifestation depending on the pre- but some recover, and may display lifelong residual signs
dominantly affected organ. Development of a biphasic fever such as a persistent myoclonus.
represents another characteristic clinical finding (Wright
et al., 1974). A transient fever and the onset of lymphopenia 6. Pathology of non-nervous tissues
can be observed 3–6 days pi (Krakowka et al., 1980) and
coincide with the first viremia that results in a generalized Following pathogen spread, virus antigen and asso-
infection of all lymphoid tissues including spleen, thymus, ciated lesions including cytoplasmic and intranuclear
lymph nodes, bone marrow, mucosa-associated lymphatic inclusion bodies can be found in various organs and
tissues (MALT) and macrophages in the lamina propria of the tissues. Respiratory manifestation results in serous to
gastrointestinal tract (Appel, 1970; Wright et al., 1974), as mucopurulent rhinitis, interstitial pneumonia and necro-
well as hepatic Kupffer cells (Appel, 1987). Viremia occurs tizing bronchiolitis, which is often complicated by a
by spread of cell free virus as well as leukocyte and suppurative bronchopneumonia due to secondary bacter-
thrombocyte associated infectious pathogens. The second ial infection (Caswell and Williams, 2007). Enteral infec-
viremia follows several days later, frequently associated tion leads to catarrhal enteritis with depletion of Peyer’s
with high fever, and results in infection of parenchymal and patches (Krakowka et al., 1985; Greene and Appel, 1998;
tissue cells throughout the body (Appel, 1969; Appel and Decaro et al., 2004). In naturally infected dogs, a pustular
Gillespie, 1972; Blixenkrone-Møller, 1989; Blixenkrone- dermatitis, also termed distemper exanthema, of thighs,
Møller et al., 1989; Okita et al., 1997). Thus, CDV can be ventral abdomen and the inner surface of ear pinnae can be
found in cells of the respiratory, gastrointestinal and urinary found. Histologically, hyper- and parakeratosis with
tract, endocrine system, lymphoid tissues, central nervous vesicle and pustule formation as well as multinucleated
system and vasculature including keratinocytes, fibroblasts, syncytial giant cells are present (Baumgärtner, 1993;
thrombocytes and different lymphoid cell subsets, as well as Maeda et al., 1994). Hard pad disease represents an
bronchial, endothelial, epithelial and neuroectodermal cells uncommon cutaneous manifestation of distemper and is
(Baumgärtner et al., 1989; Gröne et al., 2004a,b; Koutinas characterized by hyperkeratosis of the footpads and nasal
et al., 2004). planum (MacIntyre et al., 1948; Moritz et al., 2000).
Though the pathogenesis of this unusual manifestation
5.2. Clinical manifestations remains undetermined, it seems that CDV causes a
disturbance of keratinocyte differentiation and it has been
According to clinical features a catarrhal and nervous speculated whether epidermal infection is also a conse-
form or a combination of both, also termed acute systemic quence of a restricted viral infection (Gröne et al., 2003b,
form, and a chronic nervous manifestation can be distin- 2004a). During permanent tooth development CDV also
guished. In addition, various unusual manifestations, infects tooth buds and ameloblasts causing enamel
including old dog encephalitis and hard pad disease are hypoplasia (Dubielzig et al., 1981; Bittegeko et al., 1995).
recognized (Krakowka et al., 1985; Baumgärtner, 1993; A persistence of the primary spongiosa in the meta-
Moritz et al., 1998, 2000, 2003). At the acute stage, virus is physes of long bones also termed metaphyseal osteo-
found in every secretion and excretion of the body. This sclerosis or growth retardation lattice has been described
phase is accompanied by various dramatic clinical signs in young dogs suffering from systemic distemper (Baum-
including onset of a cutaneous rash, serous nasal and ocular gärtner et al., 1995a,b, 1996). Pathogenetically, osteoclast
discharge, conjunctivitis and anorexia, followed by gastro- necrosis due to direct virus infection has been assumed to
intestinal and respiratory signs, which are often compli- represent the primary defect resulting in loss of bone
cated by secondary bacterial infections and neurological modeling and subsequent persistence of the primary
disturbances (Krakowka et al., 1985). Nervous signs are spongiosa. Morphological similarities between this CDV-
diverse and progressive (Parker, 1978; Greene and Appel, associated lesion and the human Paget’s disease of bone
1998) and include myoclonus, nystagmus, ataxia, postural have been pointed out and a common etiology has been
reaction deficits and tetraparesis or plegia (Koutinas et al., assumed (Gordon et al., 1991, 1992; Mee, 1999). However,
A. Beineke et al. / Veterinary Immunology and Immunopathology 127 (2009) 1–18 5

these findings could not be substantiated by others humans, SLAM is permanently expressed on immature
(Ralston et al., 1991; Helfrich et al., 2000; Ooi et al., 2000). thymocytes, memory T cells and a portion of B cells.
Cytoplasmic and intranuclear inclusion bodies are Furthermore, its surface expression is rapidly induced on
found frequently in many organs in distemper. peripheral T and B lymphocytes, monocytes and mature
Additionally, a generalized depletion of lymphoid dendritic cells upon activation. Similar to other morbilli-
organs and an associated immunosuppression represents viruses, the selective destruction or impairment of infected
an important and common manifestation of canine SLAM-expressing cells might be an important mechanism
distemper and will be discussed in the following chapter of immunosuppression induced by CDV. Since SLAM is
(Krakowka et al., 1975a; Krakowka, 1982). expressed on mature dendritic cells and activated mono-
cytes, antigen presentation might also be affected (Tatsuo
7. Immunosuppression in canine distemper et al., 2001; Tatsuo and Yanagi, 2002; Lan et al., 2005).
Referring to this, SLAM recognition is essential for
Similar to other morbilliviruses, such as measles and lymphocyte infection, viral dissemination and induction
rinderpest virus, CDV is a lymphotrophic and highly of immunosuppression in CDV-infected ferrets (von
immunosuppressive infectious agent. However, exacerba- Messling et al., 2006). In vitro experiments have demon-
tion of disease depends upon the virus strain as well as the strated an infection and subsequent induction of apoptosis
age and immune status of the animal at the time of of SLAM-expressing canine lymphoid cell lines. In this
infection. As described above, adequate antiviral immunity context, infection of canine neoplastic B and T cells by CDV
leads to an abortive clinical course or recovery, respec- might represent a potential novel treatment of canine
tively. However, once manifested, infection causes a long lymphoma by attenuated CDV strains (Suter et al., 2005;
lasting and profound inhibition and impairment of cellular Yamaguchi et al., 2005; Puff et al., 2008).
and humoral immune functions characterized by immu- The role of the CD46 surface antigen in CDV infection,
nosuppression, lymphocyte loss, and leukopenia, render- an alternative viral receptor in SLAM-negative canine cells,
ing animals highly susceptible to opportunistic infections as described for measles, remains to be determined. So far,
(Krakowka et al., 1975a; Cerruti-Sola et al., 1983). CD46 has only been detected in neoplastic lymphoid cells
Experimental infection of gnotobiotic dogs has substan- of dogs (Nussbaum et al., 1995; Suter et al., 2005).
tially increased our understanding of the CDV-induced CD9, a tetraspan transmembrane protein is associated
immune response and immunosuppression (Krakowka with CDV-induced cell–cell fusion and syncytial cell
et al., 1980). Additionally, natural and experimental CDV formation but not virus–cell fusion (Schmid et al., 2000).
infection causes an acute systemic, often fatal and severely However, since no direct binding of this transmembrane
immunosuppressive disease in ferrets. Leukopenia and an protein by the virus has been demonstrated, CD9 is
inhibited cellular immune response, such as suppressed regarded as a cofactor for viral infection, possibly as a part
delayed type hypersensitivity and a decreased lymphocyte of the receptor complex for CDV or due to intracellular
proliferation activity as well as a reduced humoral immune signaling, enhancing the expression or activity of a
response have been reported in experimentally CDV- receptor molecule (Löffler et al., 1997; Schmid et al.,
infected ferrets. Thus, CDV infection of ferrets represents a 2000; Singethan et al., 2006).
reliable animal model for the investigation of morbilli-
virus-induced immunosuppression and the development 7.2. Morphological findings in lymphoid organs
of new vaccine strategies for human measles and canine
distemper, respectively (Kauffman et al., 1982; Stephensen Systemic CDV infection causes gross changes in multi-
et al., 1997; Welter et al., 2000; von Messling et al., 2003). ple lymphoid tissues of affected dogs, such as lymph node
swelling, depletion of MALT and reduced thymus size. In
7.1. Lymphotropism of CDV the acute phase, lesions are microscopically characterized
by a generalized depletion of T and B cell compartments in
Several studies have focused on the tropism of CDV for spleen, lymph nodes, MALT and tonsils as well as
lymphocytes (Appel et al., 1982; Cerruti-Sola et al., 1983). hyperplasia of reticular cells in the medullary region of
Experimental infection of ferrets with recombinant CDV lymph nodes (Fig. 1). Thymic atrophy is associated with a
expressing green fluorescent protein and dogs with decreased corticomedullary ratio, indistinct demarcation
virulent CDV strains, demonstrated an initial infection of between cortex and medulla as well as a reduction of
lymphoid cells in spleen, lymph nodes, MALT and thymus Hassall’s corpuscles (Krakowka et al., 1980). Furthermore,
as well as circulating T and B lymphocytes preceding syncytia formation and cell death of immune cells is
epithelial cell invasion (Appel, 1969, 1970; von Messling predominantly observed in lymphoid follicles (Iwatsuki
et al., 2004). Current in vitro investigations demonstrated et al., 1995), leading to a complete loss of secondary
that lymphotropism of CDV is presumably based on the follicles in dogs suffering from acute distemper infection
binding of CD150 (signaling lymphocyte activation molecule, (Fig. 1; Jacoby and Griesemer, 1970; Krakowka and
SLAM) by the viral H protein, followed by cell entry of the Koestner, 1977; Iwatsuki et al., 1995). Characteristic
agent. SLAM is constitutively expressed in a variety of eosinophilic cytoplasmic viral inclusion bodies are present
organs in healthy dogs. However, upon CDV infection, in reticular cells and lymphocytes of affected dogs.
SLAM is markedly up-regulated in lymphoid cells of Immunosuppression during the acute phase of distemper
affected dogs, indicating a possible strategy to increase is further accompanied by a lack of recirculating T
virus amplification in the host (Wenzlow et al., 2007). In lymphocytes in the marginal sinus of lymph nodes
6 A. Beineke et al. / Veterinary Immunology and Immunopathology 127 (2009) 1–18

Fig. 1. Histological and phenotypical characteristics found in the spleen of control animals (I, a–c) and dogs with acute (II, d–f) and chronic distemper (III, g–
i). (I) Normal lymphocyte distribution and compartment formation of B (follicle) and T cell (PALS) areas (a, HE). B lymphocytes (IgG) are the predominant
cell type in the follicle and marginal zone (b). Nearly all lymphocytes in the PALS and single cells in the marginal zone and follicle represent T lymphocytes
(c, CD3). (II) Severe splenic depletion consisting of a severe loss of most cell types (d, HE) including B lymphocytes (e, IgG) in lymphoid follicles and T cell (f,
CD3) in the PALS. (III) Repopulation of the spleen (g, HE) is associated with increasing numbers of B lymphocytes (h, IgG) and T cells (i, CD3) in the PALS.
F = lymphoid follicle; G = germinal center; M = marginal zone, P = PALS; C = corona; Z = central artery; HE = hematoxylin-eosin stain (a, d, g, HE) and
immunohistochemistry, ABC method (IgG, CD3, b, c, e, f, h, i; B and T cell specific antibodies). Bars = 100 mm.

(Krakowka et al., 1980). However, morphological altera- infection, regeneration of lymphoid organs in the chronic
tions of the immune system during the acute disease phase disease phase leads to the observed CD4- and CD8-
are partially reversed by repopulation and germinal center dominated inflammatory response in the CNS (McCullough
formation of lymphoid tissues in persistently infected and et al., 1974a,c; Krakowka et al., 1980; Krakowka and
convalescent dogs, respectively (Fig. 1; McCullough et al., Koestner, 1977; Wünschmann et al., 1999, 2000). Never-
1974b; Krakowka et al., 1980; Friedlander et al., 1985). theless, morphologically intact appearing compartments
Overall, the degree of the lymphoid depletion is positively after lymphoid repopulation and virus clearance from
correlated to the amount of CDV antigen in the affected lymphoid tissues will not result in complete functional
organ (Wünschmann et al., 1999; Kumagai et al., 2004). regeneration of the immune response (Krakowka et al.,
Histological examination of the bone marrow of CDV 1975a, 1980; Cerruti-Sola et al., 1983). Lymphoid repo-
infected dogs revealed no or only non-specific changes, pulation in dogs with chronic distemper is characterized
such as hypercellularity and granulocytic hyperplasia by a complete reconstitution of CD3-, CD4- and CD8-
(Breuer et al., 1998) or necrosis of hematopoietic cells expressing lymphocyte subsets in T cell areas (Fig. 1).
(Baumgärtner et al., 1995a,b). However, phenotypically, the numbers of CD5- and IgG-
expressing cells are still reduced and viral antigen can be
7.3. Phenotypical changes in lymphoid organs found in dendritic cells and rarely in lymphocytes
(Wünschmann et al., 2000). In the chronic stage, dendritic
The diminished immune function in the early phase of cells seem to serve as the primary host cells for the virus
the disease is associated with viremia and partially a (Fig. 2). This change of cell tropism is suspected to be a
consequence of lysis of lymphocytes and macrophages. consequence of the immune response and might represent a
Viral antigen is located in T-cell-dependent areas (Fig. 2) mechanism of viral persistence as described for neurons and
and in the follicles during the acute phase (Iwatsuki et al., oligodendrocytes (Wünschmann et al., 2000). Furthermore,
1995). Although different lymphocyte subsets, such as T terminal differentiation of dendritic cells into mature
and B cells as well as macrophages become infected, CD4+ effector cells might be inhibited by the virus, possibly
lymphocytes are most frequently affected by CDV during leading to a persistent immune depressive effect by a
the acute disease phase (Iwatsuki et al., 1995). Accordingly, delayed repopulation of peripheral lymphoid tissues with
the loss of CD4+ cells in lymphoid organs prevails the functional dendritic cells. In addition, CDV infection of
depletion of CD8+ cells (Wünschmann et al., 2000). After thymic epithelial dendritic cells may result in a compro-
initial lymphoid depletion in the incipient stage of CDV mised maturation and selection of T cells, promoting the
A. Beineke et al. / Veterinary Immunology and Immunopathology 127 (2009) 1–18 7

Fig. 2. Distribution of canine distemper virus (CDV) in the spleen of dogs with acute (a) and chronic distemper (b). During the acute disease phase, viral
antigen can be detected in many lymphoid cells (a), while CDV infection seems to be restricted to dendritic cells in follicles in chronic distemper (b). CDV-N-
specific monoclonal antibody, ABC-method. Bars = 50 mm.

release of immature CD5-negative T cells, including function by inhibition of interleukin (IL)-1, while prosta-
potentially autoreactive cells (Wünschmann et al., 2000). glandin E2 release is increased in this cell population of
viremic dogs (Krakowka et al., 1987b). Therefore, besides
7.4. Mechanisms of immunosuppression in canine distemper viral infection of B cells, an impaired antigen presentation by
monocytes might contribute to a diminished B cell
During the acute phase of distemper, lymphopenia is differentiation, plasma cell formation and immunoglobulin
characterized by a transient depletion of CD4+ T helper, production in CDV infected dogs (Fig. 3; Krakowka et al.,
CD8+ cytotoxic T and CD21+ B cells from the peripheral 1987b). Additionally, immunomodulatory properties of the
blood. Reduced numbers of circulating immune cells in CDV CDV N protein by engagement of the Fcg receptor (CD32)
might be a sequel of an impaired cellular output from expressed on B cells also leads to a suppression of uninfected
primary and secondary lymphoid organs as well as cells (Fig. 3; Kerdiles et al., 2006). Furthermore, diminished T
apoptosis of peripheral blood leukocytes. As demonstrated helper cell function in persistently infected dogs might
in lymphoid tissues, programmed cell death can be detected contribute to disturbed germinal center formation and a
in a considerable number of uninfected lymphocytes, reduced class switch from IgM to IgG (Winters et al., 1983).
indicating the existence of additional virus-independent Possibly, CDV N protein acts indirectly on T cell function by
mechanisms of apoptosis (Moro et al., 2003a; Kumagai et al., modulating antigen presentation of dendritic cells via
2004; Schobesberger et al., 2005). Therefore, mechanisms interaction with CD32 and subsequent diminished IL-12
other than directly virus-induced apoptosis, such as an over- production, as described for measles virus (Schneider-
activation of the innate immune system or Fas-mediated Schaulies and Dittmer, 2006). Using recombinant CDV in
activation-induced apoptosis of lymphoid cells must be ferrets, it has been demonstrated, that the viral V protein
considered as possible mechanisms of lymphocyte cell sustains viral replication in lymphocytes and T cell
death in canine distemper (Moro et al., 2003b; Schobes- depletion. Furthermore, the viral V protein is an essential
berger et al., 2005; Kajita et al., 2006). In addition, remaining interferon antagonist and cytokine response inhibitor in
peripheral blood lymphocytes of naive dogs suffering from CDV-infected ferrets (von Messling et al., 2006). Further-
CDV infection showed an extensively suppressed lympho- more, a lack of detectable cytokine expression in peripheral
proliferative response to mitogens (Krakowka et al., 1975a; blood leukocytes is associated with a high viral load and
Krakowka and Wallace, 1979). The mechanisms of CDV- viremia, indicating that an overwhelming virus infection
induced immunosuppression are not completely under- may suppress cytokine production in lymphoid cells (Gröne
stood and are most likely caused by various factors. Firstly, et al., 1998). Similarly, severe immunosuppression and the
only a proportion of cells are infected and, secondly, long lethal outcome in CDV-infected ferrets are correlated with a
lasting immunologic abnormalities are still present after lack of cytokine expression in peripheral blood leukocytes.
viral clearance from peripheral blood leukocytes in con- In comparison, surviving ferrets exhibited a robust and
valescent dogs. Accordingly, mechanisms of immune cell sustained cytokine production with an initial Th1- and later
impairment that do not involve direct viral infection of Th2-biased immune response (Svitek and von Messling,
lymphoid cells must be present (Krakowka et al., 1975a, 2007).
1980; Krakowka and Koestner, 1977). Consequently, Further studies have to focus on the importance of
Krakowka (1982) discussed the existence of a mononuclear virus-dependent and -independent mechanisms leading to
suppressor cell population leading to the observed long a reduced humoral and cellular immune response. This
lasting immunosuppression in canine distemper. However, includes questions upon mechanisms of viral persistence
the phenotypical classification of this cell type and in lymphoid organs with special emphasis upon the role of
mechanisms leading to immune cell inhibition remain to dendritic cells as a potential mediator for the long lasting
be determined. Furthermore, CDV modulates monocyte immunosuppression.
8 A. Beineke et al. / Veterinary Immunology and Immunopathology 127 (2009) 1–18

Fig. 3. Mechanisms of immunosuppression in CDV-infected dogs. Viral infection and viral N protein/CD32 engagement leads to diminished antigen
presentation as well as disturbed dendritic cell and B cell maturation within germinal centers. Subsequently, plasma cell formation and immunoglobulin
production is significantly reduced.

8. Pathogenesis of nervous distemper associated virus spread have been described (Axthelm and
Krakowka, 1987; Summers and Appel, 1987; Higgins et al.,
Occurrence of CNS lesions depend on virus strain, age 1982a,b). However, free infectious plasma virus cannot be
and immune status of the affected animal and can be observed after the appearance of virus-neutralizing anti-
classified in different subtypes (Campbell, 1957; Krakowka bodies (Rockborn, 1958). The leukocyte-associated viremia
and Koestner, 1976; Summers et al., 1984; Pearce-Kelling is believed to represent the major source of hematogenous
et al., 1990; Raw et al., 1992). Generally, a polio- and a infectivity (Rockborn, 1958; Summers et al., 1979). Viral
leukoencephalitis, characterized by different distribution antigen is first detected within CNS capillaries and venular
patterns of the lesions and pathogenesis, can be distin- endothelia at 5 and 6 days pi and/or in perivascular
guished. lymphocytes, astrocytic foot processes and pericytes at 8
days pi (Axthelm and Krakowka, 1987; Summers and
8.1. Virus entry, spread and expression within the CNS Appel, 1987). Furthermore, a productive CDV infection of
the choroid plexus epithelium with release of progeny
Neuroinvasion of CDV occurs predominantly via the virus into the cerebrospinal fluid followed by ependymal
hematogenous route (Fig. 4; Krakowka et al., 1987a; infection and spread of the virus to the subependymal
Krakowka, 1989). Studies tracking the route of CDV within white matter can be observed at 10 days pi (Appel and
the brain showed ependymal and subependymal white Gillespie, 1972; Higgins et al., 1982a; Vandevelde et al.,
matter infection indicating CNS spread along the cere- 1985b).
brospinal fluid (CSF) pathway (Vandevelde et al., 1985b). In Detailed studies on virus spread within the CNS indicate
addition, virus spread via the olfactory nerve has been that a rare and brief phase of gray matter disease prior to
described in experimentally infected ferrets. Whether this the development of DL can be noticed (Vandevelde and
applies to natural infection of dogs needs to be confirmed Kristensen, 1977; Summers et al., 1984). Based on this
(Rudd et al., 2006). In addition, a direct spread from observation, it has been proposed that any CDV infection
meningeal cells of the pia mater has been assumed (Fig. 4; may pass through a sequence of CNS entry causing gray
Baumgärtner et al., 1989). Both a cell-free viremia during matter and later white matter disease. The endpoint is
the first days following experimental infection and a cell- mainly determined by the virus strain (Summers et al.,
A. Beineke et al. / Veterinary Immunology and Immunopathology 127 (2009) 1–18 9

Fig. 4. Canine distemper virus (CDV) spread within the CNS. Cross-section of the cerebellum: blue arrows: viral spread via infected meningeal cells; black
circles: viral spread via infected leukocytes and endothelial cells; yellow arrows: viral spread via infected choroid plexus epithelial cells; red arrows: viral
spread via infected ependymal cells. Inserts display detection of CDV antigen. CDV-N-specific monoclonal antibody, ABC-method. (For interpretation of the
references to color in this figure legend, the reader is referred to the web version of the article.)

1984). This is supported by findings of subpial lesions in as indicated by most recent findings, aldynoglia can get
the cerebellum and the presence of CDV antigen positive infected (Orlando et al., 2008). The term aldynolgia has
cells within the pia mater and subjacent gray matter in the been used to define a family of growth-promoting glia and
early phase of infection (Fig. 4). includes several specialized macroglia with Schwann cell-
The presence of viral core proteins in cell processes like properties. Recent in vitro studies indicated a
suggests a possible spread of the virus along this pathway preferential infection of these cells by various CDV strains
in order to escape immune surveillance (Alldinger et al., (Orlando et al., 2008).
1993a,b) as has been shown in cell culture studies
(Zurbriggen et al., 1995a,b). Furthermore, antigenic 8.2.1. Oligodendroglial infection
modulation due to viral protein specific antibodies Studies focusing on the role of CDV infection and
may contribute to restricted expression of viral oligodendrocytes revealed inconsistent and controversial
proteins and virus persistence in DL (Alldinger et al., results. Using light and transmission electron microscopy,
1993a,b). In the gray matter, however, more CDV-RNA a direct oligodendrocyte infection, albeit only few cells
than protein is found, suggestive of impaired viral harbored the virus, was found (McCullough et al., 1974a;
translation and a possible mode of viral persistence Raine, 1976; Blakemore et al., 1989). In contrast to these
(Müller et al., 1995). findings, various in vitro and in vivo investigations failed to
Viral persistence seems to be an important factor for the demonstrate oligodendrocyte infection (Vandevelde et al.,
induction of immune-mediated mechanisms in the chronic 1983, 1985a; Zurbriggen and Vandevelde, 1983; Zurbrig-
phase of DL (Gaedke et al., 1995, 1997, 1999). In this gen et al., 1986, 1987a,b). However, Zurbriggen et al.
regard, factors favoring persistence include non-cytolytic (1993) demonstrated a restricted infection of oligoden-
infection and little shedding of progeny virus, hereby drocytes, characterized by the presence of CDV nucleic acid
limiting the exposure of virus antigen to local immune sequences and lack of viral antigen. Recently, it has been
cells (Zurbriggen et al., 1995a,b). shown, that a significant amount of oligodendrocytes is
still present in chronic lesions, and thus demyelination
8.2. Neurotropism of CDV seems to precede oligodendrocyte loss in DL (Schobes-
berger et al., 1999, 2002). However, in contrast to
Within the CNS, astrocytes, microglia, oligodendro- multipolar mature oligodendrocytes that are rarely
cytes, neurons, ependymal cells, choroid plexus cells and, infected a subset of bipolar oligodendrocyte precursor
10 A. Beineke et al. / Veterinary Immunology and Immunopathology 127 (2009) 1–18

cells expressing galactocerebroside exhibited a high restricted gray matter infection as a mechanism of viral
susceptibility for some CDV strains at least in vitro persistence (Nesseler et al., 1999).
(Pearce-Kelling et al., 1990, 1991). Similarly, another In contrast to polioencephalitis, DL represents the most
bipolar cell population obtained from adult canine brains, common CNS manifestation of distemper. Due to simila-
expressing the neurotrophin receptor p75NTR revealed an rities distemper represents a useful model to study the
increased susceptibility for CDV in in vitro studies (Orlando pathogenesis of human demyelinating immune mediated
et al., 2008). A phenotypically similar cell type can be diseases including MS (Koestner, 1975; Cook et al., 1978;
detected within CNS lesions of DL (W. Baumgärtner, Dal Canto and Rabinowitz, 1982; Hodge and Wolfson,
personal observation). It seems CDV infection of oligoden- 1997; Baumgärtner and Alldinger, 2005).
drocytes represents a still poorly understood process that DL lesions are found in the cerebellar and, less
at least in part, depends upon the stage of cellular frequently, in the cerebral white matter and in the spinal
differentiation at the time of infection. cord (Baumgärtner et al., 1989; Bathen-Nöthen et al.,
2008). Changes are present most frequently in fiber tracts
8.2.2. Astrocytic infection in close proximity to the ventricles, within the cerebellar
Astrocytes are the main cell population infected by CDV velum, cerebellar peduncles and optic tracts (Summers
in early plaques. In non-inflammatory lesions 64% of and Appel, 1994). Changes in DL can be categorized as
astrocytes within a lesion are infected and this cell acute, subacute non-inflammatory, subacute inflamma-
population represents 95% of all infected cells (Mutinelli tory, chronic and sclerotic lesions (McCullough et al.,
et al., 1989). A recent study revealed the importance of 1974a,b; Raine, 1976; Summers et al., 1979; Higgins et al.,
vimentin-positive cells during the spatio-temporal devel- 1982b; Krakowka et al., 1985; Summers and Appel, 1987;
opment of distemper lesions. Vimentin-positive astrocyte- Gaedke et al., 1999). Several plaque types may occur
like cells are present in the center and towards the simultaneously in the brain of affected dogs (Baumgärtner
periphery of advanced lesions, whereas reactive astro- et al., 1989; Alldinger et al., 1993a,b; Gaedke et al., 1999;
gliosis of GFAP-positive cells was only present at the Wünschmann et al., 1999).
periphery of the lesion. Vimentin-positive astrocyte-like Plaque genesis has been intensely studied in naturally
cells represent the main cell type harboring the pathogen and experimentally CDV-infected dogs (McCullough et al.,
in advanced lesions, indicating a change in cell tropism 1974a,b; Raine, 1976; Summers et al., 1979; Higgins et al.,
and/or susceptibility of glial cells during lesion progression 1982b; Summers and Appel, 1987). Acute lesions consist of
in DL. In addition, in vitro studies showed a higher focal vacuolization of the white matter, mild gliosis with few
vulnerability and reduced susceptibility of mature, activated astrocytes and macrophages/microglia. In experi-
GFAP-positive astrocytes for CDV compared to immature mentally infected dogs, primary changes such as vacuoliza-
astrocytes. Alternatively, these in vivo and in vitro findings tion due to an intramyelin edema of the subependymal and
could indicate a reversion of a mature to an immature subpial white matter are observed as early as 24 days. These
phenotype (Seehusen et al., 2007). vacuoles grow in number and size to form a demyelinated
plaque with micro- and astrogliosis and formation of
8.2.3. Neuronal infection multinucleated giant cells. However, mononuclear infil-
CDV antigen and RNA expression in neurons, which trates are rare in the initial phase. Remyelination of denuded
seems to occur in any type of nervous distemper during the axons is a rare event in older demyelinating lesions
early phase and most prominent in distemper polioence- (Wisniewski et al., 1972; McCullough et al., 1974c; Raine,
phalitis is associated with a disproportional low amount of 1976; Higgins et al., 1982b; Summers and Appel, 1987).
viral protein compared to mRNA (Nesseler et al., 1999). A Subsequent inflammatory changes are characterized by
reduced amount of detectable viral M protein indicated a progressive perivascular mononuclear infiltrations. Sclero-
restricted CDV infection similar to findings in oligoden- tic changes, so-called ‘‘burned out’’ plaques consist of
drocytes in DL and subacute sclerosing panencephalitis, a astrocytic scars characterized by isomorphic astrogliosis
rare complication of human measles virus infection (Vandevelde et al., 1981, 1982). In spite of a strong virus
(Nesseler et al., 1997). specific intrathecal immune response early lesions without
overt inflammation can be observed in the vicinity of
8.3. Pathology of nervous distemper chronic changes with prominent inflammation and reduced
or lacking virus expression (Alldinger et al., 1993b).
Polioencephalitis including old dog encephalitis, inclu- However, recent immunohistochemical studies revealed
sion body encephalitis and postvaccinal encephalitis is a the presence of immune cells already in acute and subacute
rare manifestation of CDV infection and is predominantly non-inflammatory lesions, indicating that this subdivision
located in the cortical areas and nuclei of the brain stem. of DL lesions represents a rather arbitrary approach to
Neurons and protoplasmic astrocytes represent the pre- classify distemper plaques (Tipold et al., 1999; Wünsch-
dominantly affected cell populations (Nesseler et al., mann et al., 1999).
1999). Histologically, lesions consist of neuronal necrosis
and neuronophagia characterized by infiltrating microglia/ 8.4. Immunopathology of demyelinating
macrophages and T lymphocytes, perivascular cuffs as well leukoencephalomyelitis
as intranuclear inclusion bodies in neurons and astrocytes
(Nesseler et al., 1997). Studies upon the pathogenesis of DL represents be a biphasic event (Baumgärtner and
distemper polioencephalitis support the hypothesis of a Alldinger, 2005). The initiation of this process is ascribed to
A. Beineke et al. / Veterinary Immunology and Immunopathology 127 (2009) 1–18 11

a direct virus-mediated action, whereas plaque progres- response and autoantibodies in the pathogenesis of CNS
sion seems to be an immunopathological process (Baum- demyelination remains undetermined. In other words,
gärtner et al., 1989; Summers and Appel, 1994; Alldinger compared to autoimmune demyelinating diseases includ-
et al., 1996). Studies about the cellular immune response ing experimental allergic encephalitis (EAE), the anti-
revealed only a minimal but still substantial influx of myelin response might represent an epiphenomenon
inflammatory cells in the CNS during the early phase of rather than a contributing factor for myelin loss in DL
infection (acute and subacute, non-inflammatory lesions), (Krakowka et al., 1973; Cerruti-Sola et al., 1983).
and a dramatic increase of blood-borne inflammatory cells, Coinciding with the recovery of the immune system at
simultaneously with the recovery and repopulation of the approximately 7 weeks pi, perivascular mononuclear
peripheral lymphoid tissues during the chronic phase cellular infiltrates are found within brain lesions. During
(McCullough et al., 1974a,b; Vandevelde et al., 1981; the recovery of the immune system, infiltration of CD4+
Summers and Appel, 1994; Wünschmann et al., 1999). In cells is followed by plasma cells and a strong intrathecal
early lesions, infiltrating lymphocytes displayed predomi- antibody synthesis (Vandevelde and Zurbriggen, 2005).
nantly the CD8+ phenotype indicating that antibody- Here, B cells producing predominately IgG antibodies are
independent cytotoxicity is maybe involved in viral mainly found in perivascular infiltrates and scattered
clearance and possibly early lesion development (Wünsch- throughout the lesions (Vandevelde et al., 1981, 1982).
mann et al., 1999). Initial T cell migration is suspected to be Furthermore, a prominent anti-CDV-specific humoral
mediated by microglia-derived chemokines, such as IL-8 immune response can be found in the serum and
(Tipold et al., 1999). An accumulation of immune cells in cerebrospinal fluid. The decline of the intrathecal antiviral
the CNS during the early stages of the disease may facilitate antibody production and the simultaneously occurring
the later development of an intrathecal immune response clinical improvement suggests a deleterious role of the
and associated immunopathological complications (Tipold humoral immune response on the disease process.
et al., 1999). An up-regulation of major histocompatibility Increased intrathecal antibody production due to intracer-
complex class II (MHC class II) molecules during the ebral B cells is suspected to accelerate myelin destruction
progression of DL lesions, and a simultaneous reduction of in chronic DL (Vandevelde et al., 1981, 1982, 1986;
virus protein expression within the CNS, indicates an Alldinger et al., 1996). Hence, a complement-dependent
involvement of non-viral antigens as a trigger of immune- antibody mediated humoral cytotoxicity is discussed as a
mediated processes in the chronic phase of demyelination possible mechanism for demyelination (Vandevelde et al.,
(Alldinger et al., 1996). MHC class II is mainly expressed on 1982, 1986). Furthermore, the simultaneous occurrence of
microglia, endothelial, meningeal, choroid plexus epithe- a high number of intralesional and perivascular CD8+
lial, ependymal and intravascular cells as well as most lymphocytes and perivascular B and CD4+ cells is
perivascular blood-borne cells. Activated macrophages/ suggestive of an antibody-dependent T-cell mediated
microglial cells exhibit beneficial and detrimental effects cytotoxicity (Vandevelde et al., 1982; Alldinger et al.,
during lesion development. Though myelin loss in early 1996; Wünschmann et al., 1999). Furthermore, immuno-
lesions has been ascribed to directly or indirectly virus- phenotypical changes are indicative of a delayed-type
mediated processes, demyelination in chronic lesions may hypersensitivity reaction in advanced lesions (Wünsch-
occur as collateral damage and could develop due to mann et al., 1999).
overactive macrophages/microglia. Chronic inflammatory In addition, anti-myelin specific antibodies have been
demyelination may be due to so-called bystander mechan- detected in the CSF (Krakowka et al., 1973; Vandevelde
isms resulting from interactions between macrophages/ et al., 1986; Rima et al., 1991). However, the significance of
microglia and antiviral antibodies (Vandevelde and anti-myelin antibodies remained uncertain as high titers of
Zurbriggen, 1995). The term ‘‘bystander demyelination’’ these antibodies are also detected in dogs with resolving
is used to describe the myelin damaging effect of lesions (Vandevelde et al., 1986).
proteolytic enzymes released by stimulated macro-
phages/microglia in the absence of obvious oligodendro- 8.5. Role of cytokines in the pathogenesis of demyelinating
cyte infection (Cammer et al., 1978). Activation of these leukoencephalitis
cells characterized by increased MHC class II and adhesion
molecule expression, results in release of toxic factors, Cytokines were investigated to clarify cell-to-cell
increased phagocytic activity and oxygen radical produc- communication and the interaction between immune
tion after induction of the respiratory burst (Bürge et al., cells and resident brain cells in DL. It seems that cytokine
1989; Griot et al., 1989a,b, 1990). Therefore, CDV-induced expression can be directly induced by the virus or due to
phagocyte activation could contribute to oligodendrocyte autocrine and paracrine regulatory loops during CDV
and/or myelin damage (Vandevelde and Zurbriggen, 2005; infection (Bencsik et al., 1996; Gröne et al., 2002; Markus
Stein et al., 2006, 2007). Additionally, a humoral antiviral et al., 2002). Tsai et al. (1982) found interferon (IFN) in the
immune response could lead to the destruction of CSF of dogs with CNS CDV infection, indicating IFN as a
oligodendrocytes as ‘‘innocent bystanders’’. Also auto- valid marker to determine CDV persistence in the CNS.
reactive T cells might be responsible for the observed However, the type of IFN playing such an essential role
induction of myelin-specific cellular immunity via epitope during DL remains to be determined. Referring to this, IFN-
spreading secondary to myelin damage in the CNS g is not expressed in peripheral blood leukocytes of dogs
(Krakowka et al., 1973; Wünschmann et al., 2000). with canine distemper, whereas IL-8 can be found in blood
However, the precise role of this self-reactive immune samples of CDV-infected animals with and without
12 A. Beineke et al. / Veterinary Immunology and Immunopathology 127 (2009) 1–18

viremia as well as in non-infected dogs (Gröne et al., 1998). inflammatory cytokines in the presence of an intense up-
Though there is no obvious correlation between cytokine regulation of pro-inflammatory cytokines might contri-
mRNA transcripts and respiratory or gastrointestinal tract bute to initiation and progression of lesions, possibly due
manifestation, CNS demyelination is frequently associated to a derailment of the immune response in the early phase
with increased expression of IL-1, IL-12, tumor necrosis of DL (Markus et al., 2002).
factor (TNF)-a and transforming growth factor (TGF)-b in In addition, increased IL-1 and IL-6 expression can be
the blood of dogs with natural CDV infection (Gröne et al., found in all types of distemper lesions and is predomi-
1998). Furthermore, and indicative of disease initiation, IL- nately detected in CD3+ T lymphocytes and macrophages/
6 transcripts can be detected in blood samples of animals microglia in early plaques as well as in cells comprising
with early CNS lesions, while peripheral TGF-b expression perivascular cuffs in chronic lesions (Fig. 6). TNF-a is
seems to play a role in chronic demyelination (Gröne et al., predominantly expressed by astrocytes and seems to play
1998). The simultaneous occurrence of pro- and anti- a crucial role in the pathogenesis of early demyelination, as
inflammatory cytokines in the blood of dogs with demonstrated for MS (Gröne et al., 2000). Furthermore,
distemper indicates a complex, most likely disease TNF-a as well as IL-1 and IL-6 represent crucial factors for
stage-dependent orchestrated or dysregulated cytokine endothelial cell adhesion molecules induction and sub-
expression (Fig. 5; Gröne et al., 1998). Within the CNS, the sequent leukocyte extravasation. Therefore, initial produc-
chemokine IL-8 might be crucial for early T cell attraction tion of TNF-a by astrocytes may drive a vicious circle of
and antiviral cytotoxicity during acute distemper (Tipold attracting inflammatory cells to CNS lesions, leading to the
et al., 1999). In the CSF of naturally infected dogs, TNF-a synthesis of more cytokines and development of the
and IL-6 mRNA, associated with disease exacerbation, and chronic stage of DL. IL-12 production within demyelinating
IL-10 and TGF-b mRNA, indicative of remission, are often lesions in CDV-infected brains is supposed to promote a
observed simultaneously and cannot be assigned to a Th1-biased immune responses (Gröne et al., 2000;
specific disease stage. However, detection of IL-10 mRNA Wünschmann et al., 2000).
in the CSF might represent a stage of inactivity in dogs with
DL (Frisk et al., 1999b). Moreover, IFN-g can be found in 8.6. Role of extracellular matrix in the pathogenesis of
association with chronic lesions. A semiquantitative RT- demyelinating leukoencephalitis
PCR study displayed a prominent up-regulation of the pro-
inflammatory cytokines IL-6, IL-8, IL-12 and TNF-a in early In addition to cytokines, disruption of the blood–brain-
distemper CNS lesions. In contrast, immunomodulatory barrier by proteolytic enzymes is regarded as a pivotal
cytokines such as IL-10 and TGF-b were not significantly factor for the influx of inflammatory cells and lesion
up-regulated following CDV infection. In addition, IL-1, IL- progression in demyelinating diseases. Referring to this,
2 and IFN-g were not detected (Markus et al., 2002). astrocytes represent the main target cells of CDV (Mutinelli
Therefore, the lack of or an inappropriate response of anti- et al., 1989), form the blood–brain-barrier and are a major

Fig. 5. Schematic display of the proposed pathogenesis of early lesions in demyelinating distemper leukoencephalitis. Following virus infection of astrocytes
and macrophages/microglia an increased influx of CD8+ lymphocytes associated with increased expression of IL-6, IL-8, IL-12 and TNF-a is observed. In
contrast, immunomodulatory cytokines including IL-10 and TGF-b remain unchanged.
A. Beineke et al. / Veterinary Immunology and Immunopathology 127 (2009) 1–18 13

Fig. 6. Schematic display of the proposed pathogenesis of chronic lesions in demyelinating distemper leukoencephalitis. Following virus infection, reduced
or lacking virus expression and an increased influx of CD8+, CD4+ and B cells can be noticed. CD4+ and B cells are found predominantly in the perivascular
space. In addition, infiltrating peripheral or resident brain cells exhibit an increased MHC class II expression and a moderate to severe up-regulation of IL-6,
IL-8-, IL-12 and TNF-a. The role of IL-1 and IFN-g remains to be determined.

source of extracellular matrix (ECM) proteins and cell astrocytes and macrophages/microglia as demonstrated by
mediators. Furthermore, astrocytes play an essential role immunohistochemistry (Miao et al., 2003). It seems that
in the maintenance of structural integrity of the CNS they play a crucial role in lesion initiation. In advanced
(Montgomery, 1994). The importance of the ECM is plaques, the expression of MMPs and TIMPs declined apart
stressed by the observation, that a plaque-associated from MMP-11, MMP-12 and MMP-13 and invading
CD44 up-regulation, paralled by astrocyte activation is perivascular mononuclear cells express also MMPs and
found in acute and subacute demyelinating distemper TIMPs in addition to astrocytes and macrophages/micro-
lesions. CD44 plays a dual role for the progression of the glia (Miao et al., 2002, 2003), indicating a disease phase-
demyelination process. Ligation of this cell surface dependent expression of MMPs and TIMPs in DL. Similarly,
receptor for hyaluronate might induce chemokine and in situ hybridization studies revealed a MMP-9, MMP-14
cytokine activation and hence initiates and perpetuates the and TIMP-1 expression predominantly in astrocytes and
inflammatory process. In chronic lesions, a reduction of activated macrophages/microglial cells in early lesions,
CD44 associated with a loss of astrocytes is noticed. whereas activated macrophages/microglia and infiltrating
However, CD44 can still be found on the cell membrane of lymphocytes are the main source of these enzymes and
perivascular mononuclear cells. Therefore, CD44 might proteins in advanced DL (Gröters et al., 2005). Overall the
also represent a marker for the activation of immune cells number of cells expressing MMPs seems to exceed the
in the late phase of the infection (Alldinger et al., 2000, amount of cells with TIMP expression. These findings favor
2006). the hypothesis of an altered MMP–TIMP balance as an
Matrix metalloproteinases (MMPs) are zinc- and additional key factor for lesion initiation and progression
calcium-dependent endopeptidases that are capable to (Miao et al., 2003; Gröters et al., 2005; Alldinger et al.,
cleave the extracellular matrix, and thus are able to open 2006).
the blood–brain-barrier and affect myelin (Lo et al., 2002;
Ulrich et al., 2006; Puff et al., 2008). Furthermore, certain
MMPs are crucial for the activation of cytokine and Acknowledgments
chemokine functions, as demonstrated for MS (Chandler
et al., 1997; van Lint and Libert, 2007). Their inhibitors, The authors wish to thank Annette Artelt, Petra Grünig,
TIMPs (tissue inhibitors of matrix metalloproteinases), are Danuta Waschke and Julia Schirrmeier for excellent
most prominently up-regulated in acute and subacute technical support. Dr. Frauke Seehusen received a scholar-
non-inflammatory lesions and are expressed mostly by ship from Bayer AG, Animal Health Care, Leverkusen,
14 A. Beineke et al. / Veterinary Immunology and Immunopathology 127 (2009) 1–18

Germany. Dr. Christina Puff received an award from the Baumgärtner, W., Alldinger, S., Beineke, A., Gröters, S., Herden, C., Kaim, U.,
Müller, G., Seeliger, F., van Moll, P., Wohlsein, P., 2003. Das staupe-
Frauenförderplan of the University of Veterinary Medicine, virus–ein erreger auf der suche nach neuen Wirten. Dtsch. Tierärztl.
Hannover. Studies were supported by the Deutsche Wochenschr. 110, 137–142.
Forschungsgemeinschaft and the Hertie-Foundation. Baumgärtner, W., Alldinger, S., 2005. The pathogenesis of canine dis-
temper virus induced demyelination—a biphasic process. In: Lavi, E.,
Constantinescu, C.S. (Eds.), Experimental Models of Multiple Sclero-
References sis. Springer, New York, pp. 871–887.
Bencsik, A., Malcus, C., Akaoka, H., Giraudon, P., Belin, M.F., Bernard, A.,
Adelus-Neveu, F., Saint-Gérand, A.L., Fayet, G., Wiedemann, C., 1991. 1996. Selective induction of cytokines in mouse-brain infected with
Hundestaupe: lehren aus einer epizootie in frankreich. Prakt. Tierarzt. canine distemper virus—structural, cellular and temporal expression.
10, 866–871. J. Neuroimmunol. 65, 1–9.
Alldinger, S., Baumgärtner, W., van Moll, P., Örvell, C., 1993a. In vivo and in Bittegeko, S.B., Arnbjerg, J., Nkya, R., Tevik, A., 1995. Multiple dental
vitro expression of canine distemper viral proteins in dogs and non- developmental abnormalities following canine distemper infection.
domestic carnivores. Arch. Virol. 132, 421–428. J. Am. Anim. Hosp. Assoc. 31, 42–45.
Alldinger, S., Baumgärtner, W., Örvell, C., 1993b. Restricted expression of Blakemore, W.F., Summers, B.A., Appel, M.G., 1989. Evidence of oligoden-
viral surface proteins in canine distemper encephalitis. Acta Neuro- drocyte infection and degeneration in canine distemper encephalitis.
pathol. 85, 635–645. Acta Neuropathol. 77, 550–553.
Alldinger, S., Wünschmann, A., Baumgärtner, W., Voss, C., Kremmer, E., Blixenkrone-Møller, M., 1989. Detection of intracellular canine distemper
1996. Up-regulation of major histocompatibility complex class II virus antigen in mink inoculated with an attenuated or a virulent
antigen expression in the central nervous system of dogs with spon- strain of canine distemper virus. Am. J. Vet. Res. 50, 1616–1620.
taneous canine distemper virus encephalitis. Acta Neuropathol. 92, Blixenkrone-Møller, M., Svansson, V., Have, P., Botner, A., Nielsen, J., 1989.
273–280. Infection studies in mink with seal-derived morbillivirus. Arch. Virol.
Alldinger, S., Fonfara, S., Kremmer, E., Baumgärtner, W., 2000. Up-regula- 106, 165–170.
tion of hyaluronate receptor CD44 in canine distemper demyelinated Bostock, C.J., Barrett, T., Crowther, J.R., 1990. Characterisation of the
plaques. Acta Neuropathol. 99, 138–146. European seal morbillivirus. Vet. Microbiol. 23, 351–360.
Alldinger, S., Gröters, S., Miao, Q., Fonfara, S., Kremer, E., Baumgärtner, W., Breuer, W., Stahr, K., Majzoub, M., Hermanns, W., 1998. Bone-marrow
2006. Roles of an extracellular matrix (ECM) receptor and ECM changes in infectious diseases and lymphohaemopoietic neoplasias in
processing enzymes in demyelinating canine distemper encephalitis. dogs and cats—a retrospective study. J. Comp. Pathol. 119, 57–66.
Dtsch. Tierärztl. Wochenschr. 113, 154–156. Budd, J., 1981. Distemper. In: Davis, J.W., Karstad, L.H., Trainer, D.O.
Amude, A.M., Alfieri, A.A., Alfieri, A.F., 2007. Clinicopathological findings (Eds.), Infectious Diseases of Wild Animals. 2nd edition. The Iowa
in dogs with distemper encephalomyelitis presented without char- State University Press, Iowa, pp. 33–44.
acteristic signs of the disease. Res. Vet. Sci. 82, 416–422. Bürge, T., Griot, C., Vandevelde, M., Peterhans, E., 1989. Antiviral anti-
Appel, M.J., 1969. Pathogenesis of canine distemper. Am. J. Vet. Res. 30, bodies stimulate production of reactive oxygen species in cultured
1167–1182. canine brain cells infected with canine distemper virus. J. Virol. 63,
Appel, M.J., 1970. Distemper pathogenesis in dogs. J. Am. Vet. Med. Assoc. 2790–2797.
156, 1681–1684. Cammer, W., Bloom, B.R., Norten, W.T., Gorden, S., 1978. Degradation of
Appel, M.J.G., Gillespie, J.H., 1972. Canine distemper virus. In: Gard, S., basic protein in myelin by neutral proteases secreted by stimulated
Hallauer, C., Meyer, K.F. (Eds.), Virology Monographs, vol. 11. macrophages: a possible mechanism of inflammatory demyelination.
Springer, New York, pp. 1–96. Proc. Natl. Acad. Sci. U.S.A. 75, 1554–1558.
Appel, M.J., Shek, W.R., Summers, B.A., 1982. Lymphocyte-mediated Campbell, R.S.F., 1957. Encephalitis in canine distemper. Brit. Vet. J. 113,
immune cytotoxicity in dogs infected with virulent canine distemper 143–162.
virus. Infect. Immun. 37, 592–600. Caswell, J.L., Williams, K.J., 2007. Respiratory system. In: Grant Maxie, M.
Appel, M.J., Mendelson, S.G., Hall, W.W., 1984. Macrophage Fc receptors (Ed.), Jubb, Kennedy and Palmer’s Pathology of Domestic Animals. 5th
control infectivity and neutralization of canine distemper virus-anti- edition. Elsevier Saunders, Edinburgh, London, New York, Oxford,
body complexes. J. Virol. 51, 643–649. Philadelphia, St Louis, Sydney, Toronto, pp. 635–636.
Appel, M.J., 1987. Canine distemper virus. In: Horzinek, M.C.M. (Ed.), Cerruti-Sola, S., Kristensen, F., Vandevelde, M., Bichsel, P., Kihm, U., 1983.
Virus Infections of Vertebrates, vol. 1. Elsevier Science Publishers Lymphocyte responsiveness to lectin and myelin antigens in canine
B.V., Amsterdam, Oxford, New York, Tokyo, pp. 133–159. distemper infection in relation to the development of demyelinating
Appel, M.J., Reggiardo, C., Summers, B.A., Pearce-Kelling, S., Maré, C.J., lesions. J. Neuroimmunol. 4, 77–90.
Noon, T.H., Reed, R.E., Shively, J.N., Örvell, C., 1991. Canine distemper Chandler, S., Miller, K.M., Clements, J.M., Lury, J., Corkill, D., Anthony, D.C.,
virus infection and encephalitis in javelinas (collared peccaries). Arch. Adams, S.E., Gearing, A.J., 1997. Matrix metalloproteinases, tumor
Virol. 119, 147–152. necrosis factor and multiple sclerosis: an overview. J. Neuroimmunol.
Appel, M.J., Yates, R.A., Foley, G.L., Bernstein, J.J., Santinelli, S., Spelman, 72, 155–161.
L.H., Miller, L.D., Arp, L.H., Anderson, M., Barr, M., 1994. Canine Cook, S.D., Dowling, P.C., Russell, W.C., 1978. Multiple sclerosis and canine
distemper epizootic in lions, tigers, and leopards in North America. distemper. Lancet 1, 605–606.
J. Vet. Diagn. Invest. 6, 277–288. Curran, M.D., ÓLoan, D., Rima, B.K., Kennedy, S., 1990. Nucleotide
Axthelm, M., Krakowka, S., 1987. Canine distemper virus: the early blood- sequenze analysis of phocine distemper virus reveals its distinctness
brain barrier lesion. Acta Neuropathol. 75, 27–33. from canine distemper virus. Vet. Rec. 127, 430–431.
Bathen-Nöthen, A., Stein, V.M., Puff, C., Baumgärtner, W., Tipold, A., 2008. Dal Canto, M.C., Rabinowitz, S.G., 1982. Experimental models of virus-
Magnetic resonance imaging findings in acute canine distemper virus induced demyelination of the central nervous system. Ann. Neurol.
infection. J. Small Anim. Pract. 49, 460–467. 11, 109–127.
Baumgärtner, W., Örvell, C., Reinacher, M., 1989. Naturally occurring Decaro, N., Camero, M., Greco, G., Zizzo, N., Tinelli, A., Campolo, M.,
canine distemper virus encephalitis: distribution and expression of Pratelli, A., Buonavoglia, C., 2004. Canine distemper and related
viral polypeptides in nervous tissues. Acta Neuropathol. 78, 504–512. diseases: report of a severe outbreak in a kennel. New Microbiol.
Baumgärtner, W., 1993. Virale Infektionskrankheiten bei Welpen und 27, 177–181.
Junghunden unter besonderer Berücksichtigung der Staupevirusin- Deem, S.L., Spelman, L.H., Yates, R.A., Montali, R.J., 2000. Canine distemper
fektion. Prakt. Tierarzt. 74, 26–32. in terrestrial carnivores: a review. J. Zoo Wildl. Med. 31, 441–451.
Baumgärtner, W., Boyce, R.W., Weisbrode, S.E., Alldinger, S., Axthelm, Diallo, A., 1990. Morbillivirus group: genome organisation and proteins.
M.K., Krakowka, S., 1995a. Histologic and immunocytochemical char- Vet. Microbiol. 23, 155–163.
acterization of canine distemper-associated metaphyseal bone Dubielzig, R.R., Higgins, R.J., Krakowka, S., 1981. Lesions of the enamel
lesions in young dogs following experimental infection. Vet. Pathol. organ of developing dog teeth following experimental inoculation of
32, 702–709. gnotobiotic puppies with canine distemper virus. Vet. Pathol. 18,
Baumgärtner, W., Boyce, R.W., Alldinger, S., Axthelm, M.K., Weisbrode, 684–689.
S.E., Krakowka, S., Gaedke, K., 1995b. Metaphyseal bone lesions in Ek-Kommonen, C., Sihvonen, L., Pekkanen, K., Rikula, U., Nuotio, L., 1997.
young dogs with systemic canine distemper virus infection. Vet. Outbreak of canine distemper in vaccinated dogs in Finland. Vet. Rec.
Microbiol. 44, 201–209. 141, 380–383.
Baumgärtner, W., Alldinger, S., Gaedke, K., Moritz, A., 1996. Metaphyseal Friedlander, J.M., Summers, B.A., Appel, M.J., 1985. Persistence of virulent
osteosclerosis in young dogs with naturally occurring distemper. Eur. canine distemper virus in lymphoblastoid cell lines. Arch. Virol. 86,
J. Vet. Pathol. 2, 23–32. 47–62.
A. Beineke et al. / Veterinary Immunology and Immunopathology 127 (2009) 1–18 15

Frisk, A.L., König, M., Moritz, A., Baumgärtner, W., 1999a. Detection of gene of wild-type and vaccine canine distemper viruses. Vet. Micro-
canine distemper virus nucleoprotein RNA by reverse transcription- biol. 69, 15–18.
PCR using serum, whole blood, and cerebrospinal fluid from dogs with Hall, W.W., Lamb, R.A., Choppin, P.W., 1980. Polypeptides of canine
distemper. J. Clin. Microbiol. 37, 3634–3643. distemper virus: synthesis in infected cells and relatedness to the
Frisk, A.L., Baumgärtner, W., Gröne, A., 1999b. Dominating interleukin-10 polypeptides of other morbilliviruses. Virology 100, 433–449.
mRNA expression induction in cerebrospinal fluid cells of dogs with Helfrich, M.H., Hobson, R.P., Grabowski, P.S., Zurbriggen, A., Cosby, S.L.,
natural canine distemper virus induced demyelinating and non- Dickson, G.R., Fraser, W.D., Ooi, C.G., Selby, P.L., Crisp, A.J., Wallace,
demyelinating CNS lesions. J. Neuroimmunol. 97, 102–109. R.G., Kahn, S., Ralston, S.H., 2000. A negative search for a paramyx-
Gaedke, K., Teifke, J., Hardt, M., Alldinger, S., Baumgärtner, W., 1995. oviral etiology of Paget’s disease of bone: molecular, immunological,
Nachweis von Staupevirus-N-Protein-RNS im Gehirn von Hunden mit and ultrastructural studies in UK patients. J. Bone Miner. Res. 15,
spontaner staupeenzephalitis mittels einer digoxigenin-markierten, 2315–2329.
doppelsträngigen DNS-Sonde in der in situ hybridisierung. Berl. Higgins, R.J., Krakowka, S.G., Metzler, A.E., Koestner, A., 1982a. Experi-
Muench. Tierarztl. Wochenschr. 108, 51–54. mental canine distemper encephalomyelitis in neonatal gnotobiotic
Gaedke, K., Zurbriggen, A., Baumgärtner, W., 1997. In vivo and in vitro dogs. A sequential ultrastructural study. Acta Neuropathol. 57, 287–
detection of canine distemper virus nucleoprotein gene with digox- 295.
igenin-labelled RNA, double-stranded DNA probes and oligonucleo- Higgins, R.J., Krakowka, S.G., Metzler, A.E., Koestner, A., 1982b. Primary
tides by in situ hybridization. Zentralbl. Veterinarmed. B 44, 329–340. demyelination in experimental canine distemper virus induced ence-
Gaedke, K., Zurbriggen, A., Baumgärtner, W., 1999. Lack of correlation phalomyelitis in gnotobiotic dogs. Sequential immunologic and mor-
between virus nucleoprotein and mRNA expression and the inflam- phologic findings. Acta Neuropathol. 58, 1–8.
matory response in demyelinating distemper encephalitis indicates a Ho, C.K., Babiuk, L.A., 1979a. Immune mechanisms against canine dis-
biphasic disease progress. Eur. Vet. Pathol. 5, 9–20. temper. I. Identification of K cell against canine distemper virus
Gemma, T., Iwatsuki, K., Shin, Y.S., Yoshida, E., Kai, C., Mikami, T., 1996. infected target cells in vitro. Immunology 37, 231–239.
Serological analysis of canine distemper virus using an immunocap- Ho, C.K., Babiuk, L.A., 1979b. Immune mechanisms against canine dis-
ture ELISA. J. Vet. Med. Sci. 58, 791–794. temper. II. Role of antibody in antigen modulation and prevention of
Gerber, J.D., Marron, A.E., 1976. Cell-mediated immunity and age at intercellular and extracellular spread of canine distemper virus.
vaccination associated with measles inoculation and protection of Immunology 38, 765–772.
dogs against canine distemper. Am. J. Vet. Res. 37, 133–138. Ho, C.K., Babiuk, L.A., 1980. Immune mechanisms against canine distem-
Gordon, M.T., Anderson, D.C., Sharpe, P.T., 1991. Canine distemper virus per. III. Role of complement lysis in the immunity and persistent
localised in bone cells of patients with Paget’s disease. Bone 12, 195– infection of canine distemper virus. Immunology 39, 231–237.
201. Hodge, M.J., Wolfson, C., 1997. Canine distemper virus and multiple
Gordon, M.T., Mee, A.P., Anderson, D.C., Sharpe, P.T., 1992. Canine dis- sclerosis. Neurology 49, 62–69.
temper virus transcripts sequenced from pagetic bone. Bone Miner. Iwatsuki, K., Okita, M., Ochikubo, F., Gemma, T., Shin, Y.S., Miyashita, N.,
19, 159–174. Mikami, T., Kai, C., 1995. Immunohistochemical analysis of the lym-
Gorham, J.R., 1966. The epizootiology of distemper. J. Am. Vet. Med. Assoc. phoid organs of dogs naturally infected with canine distemper virus. J.
149, 610–622. Comp. Pathol. 113, 185–190.
Greene, C.E., Appel, M.J., 1998. Canine distemper. In: Greene, C.E. (Ed.), Jacoby, R.O., Griesemer, R.A., 1970. Effect of adrenalectomy on the lym-
Infectious Diseases of the Dog and Cat. WB Saunders Company, phoid lesions in dogs with experimentally induced canine distemper.
Philadelphia, London, Toronto, Montreal, Sydney, Tokyo, pp. 9–22. Am. J. Vet. Res. 31, 1825–1833.
Griot, C., Burge, T., Brigger, S., Richard, A., Peterhans, E., Vandevelde, M., Johnson, R., Glickman, L.T., Emerick, T.J., Patronek, G.J., 1995. Canine
1989a. Macrophages in central nervous canine distemper: friends or distemper infection in pet dogs. I. Surveillance in Indiana during a
foes? Schweiz. Arch. Tierheilkd. 131, 351–359. suspected outbreak. J. Am. Anim. Hosp. Assoc. 31, 223–229.
Griot, C., Burge, T., Vandevelde, M., Peterhans, E., 1989b. Antibody- Kajita, M., Katayama, H., Murata, T., Kai, C., Hori, M., Ozaki, H., 2006.
induced generation of reactive oxygen radicals by brain macrophages Canine distemper virus induces apoptosis through caspase-3 and -8
in canine distemper encephalitis: a mechanism for bystander demye- activation in Vero cells. J. Vet. Med. B 53, 273–277.
lination. Acta Neuropathol. 78, 396–403. Kauffman, C.A., Bergman, A.G., O’Connor, R.P., 1982. Distemper virus
Griot, C., Vandevelde, M., Peterhans, E., Stocker, R., 1990. Selective degen- infection in ferrets: an animal model of measles-induced immuno-
eration of oligodendrocytes mediated by reactive oxygen species. suppression. Clin. Exp. Immunol. 47, 617–625.
Free Rad. Res. Commun. 14, 181–193. Kennedy, S., Smyth, J.A., Cush, P.F., Duignan, P., Platten, M., McCullough,
Gröne, A., Frisk, A.L., Baumgärtner, W., 1998. Cytokine mRNA expression S.J., Allan, G.M., 1989. Histopathologic and immunocytochemical
in whole blood samples from dogs with natural canine distemper studies of distemper in seals. Vet. Pathol. 26, 97–103.
virus infection. Vet. Immunol. Immunopathol. 65, 11–27. Kennedy, S., Kuiken, T., Jepson, P.D., Deaville, R., Forsyth, M., Barrett, T.,
Gröne, A., Alldinger, S., Baumgärtner, W., 2000. Interleukin-1b, -6-12 and van de Bildt, M.W.G., Osterhaus, A.D.M.E., Eybatov, T., Duck, C.,
tumor necrosis factor-a expression in brains of dogs with canine Kydyrmanov, A., Mitrofanov, I., Wilson, S., 2000. Mass die-off of
distemper virus infection. J. Neuroimmunol. 110, 20–30. Caspian seals caused by canine distemper virus. Emerg. Infect. Dis.
Gröne, A., Fonfara, S., Baumgärtner, W., 2002. Cell type-dependent cyto- 6, 637–639.
kine expression after canine distemper virus infection. Viral Immu- Kerdiles, Y.M., Cherif, B., Marie, J.C., Tremillon, N., Blanquier, B., Libeau, G.,
nol. 15, 493–505. Diallo, A., Wild, T.F., Villiers, M.B., Horvat, B., 2006. Immunomodula-
Gröne, A., Gröters, S., Koutinas, A., Saridomichelakis, M., Baumgärtner, W., tory properties of morbillivirus nucleoproteins. Viral Immunol. 324–
2003a. Non-cytocidal infection of keratinocytes by canine distemper 334.
virus in the so-called hard pad disease of canine distemper. Vet. Koestner, A., 1975. Animal model of human disease: subacute sclerosing
Microbiol. 96, 157–163. panencephalitis, multiple sclerosis; animal model: distemper-
Gröne, A., Engelhardt, P., Zurbriggen, A., 2003b. Canine distemper virus associated demyelinating encephalomyelitis. Am. J. Pathol. 78,
infection: proliferation of canine footpad keratinocytes. Vet. Pathol. 361–364.
40, 574–578. Koutinas, A.F., Polizopoulou, Z.S., Baumgärtner, W., Lekkas, S., Kontos, V.,
Gröne, A., Doherr, M.G., Zurbriggen, A., 2004a. Canine distemper virus 2002. Relation of clinical signs to pathological changes in 19 cases of
infection of canine footpad epidermis. Vet. Dermatol. 15, 159–167. canine distemper encephalomyelitis. J. Comp. Pathol. 126, 47–56.
Gröne, A., Doherr, M.G., Zurbriggen, A., 2004b. Up-regulation of cytoker- Koutinas, A.F., Baumgärtner, W., Tontis, D., Polizopoulou, Z., Saridomi-
atin expression in canine distemper virus-infected canine footpad chelakis, M.N., Lekkas, S., 2004. Histopathology and immunohisto-
epidermis. Vet. Dermatol. 15, 168–174. chemistry of canine distemper virus-induced footpad hyperkeratosis
Gröters, S., Alldinger, S., Baumgärtner, W., 2005. Up-regulation of mRNA (hard pad disease) in dogs with natural canine distemper. Vet. Pathol.
for matrix metalloproteinases-9 and -14 in advanced lesions of 41, 2–9.
demyelinating canine distemper leukoencephalitis. Acta Neuro- Krakowka, S., McCullough, B., Koester, A., Olsen, R., 1973. Myelin-specific
pathol. 110, 369–382. autoantibodies associated with central nervous system demyelina-
Haas, L., Martens, W., Greiser-Wilke, I., Mamaev, L., Butina, T., Maack, D., tion in canine distemper virus infection. Infect. Immun. 8, 819–827.
Barrett, T., 1997. Analysis of the haemagglutinin gene of current wild- Krakowka, S., Cockerell, G., Koestner, A., 1975a. Effects of canine dis-
type canine distemper virus isolates from Germany. Virus Res. 48, temper virus infection on lymphoid functions in vitro and in vivo.
165–171. Infect. Immun. 11, 1069–1078.
Haas, L., Liermann, H., Harder, T.C., Barrett, T., Löchelt, M., von Messling, Krakowka, S., Olsen, R., Confer, A., Koestner, A., McCullough, B., 1975b.
V., Baumgärtner, W., Greiser-Wilke, I., 1999. Analysis of the H gene, Serologic response to canine distemper viral antigens in gnotobiotic
the central untranslated region and the proximal coding part of the F dogs infected with canine distemper virus. J. Infect. Dis. 132, 384–392.
16 A. Beineke et al. / Veterinary Immunology and Immunopathology 127 (2009) 1–18

Krakowka, S., Koestner, A., 1976. Age-related susceptibility to infection McCullough, B., Krakowka, S., Koestner, A., Shadduck, J., 1974c. Demye-
with canine distemper virus in gnotobiotic dogs. J. Infect. Dis. 134, linating activity of canine distemper virus isolates in gnotobiotic
629–632. dogs. J. Infect. Dis. 130, 343–350.
Krakowka, S., Koestner, A., 1977. Comparison of canine distemper virus McCullough, S.J., McNeilly, F., Allan, G.M., Kennedy, S., Smyth, J.A., Cosby,
strains in gnotobiotic dogs: effects on lymphoid tissues. Am. J. Vet. S.L., McQuaid, S., Rima, B.K., 1991. Isolation and characterisation of a
Res. 38, 1919–1922. porpoise morbillivirus. Arch. Virol. 118, 247–252.
Krakowka, S., Wallace, A.L., Koestner, A., 1978. Syncytia inhibition by Mee, A.P., 1999. Paramyxoviruses and Paget’s disease: the affirmative
immune lymphocytes: in vitro test for immunity to canine distemper. view. Bone 24, 19–21.
J. Clin. Microbiol. 7, 292–297. Miao, Q., Baumgärtner, W., Failing, K., Alldinger, S., 2002. Up-regulation of
Krakowka, S., Wallace, A.L., 1979. Lymphocyte-associated immune matrix-metalloproteinases and their inhibitors in demyelinating
responses to canine distemper and measles viruses in distemper- canine distemper encephalitis. Acta Neuropathol. 104, 560.
infected gnotobiotic dogs. Am. J. Vet. Res. 40, 669–672. Miao, Q., Baumgärtner, W., Failing, K., Alldinger, S., 2003. Phase-depen-
Krakowka, S., Higgins, R.J., Koestner, A., 1980. Canine distemper virus: dent expression of matrix metalloproteinases and their inhibitors in
review of structural and functional modulations in lymphoid tissues. demyelinating canine distemper encephalitis. Acta Neuropathol. 106,
Am. J. Vet. Res. 41, 284–292. 486–494.
Krakowka, S., 1982. Mechanisms of in vitro immunosuppression in canine Miele, J.A., Krakowka, S., 1983. Antibody responses to virion polypeptides
distemper virus infection. J. Clin. Lab. Immunol. 8, 187–196. in gnotobiotic dogs infected with canine distemper virus. Infect.
Krakowka, S., Axthelm, M.K., Johnson, G.C., 1985. Canine distemper virus. Immun. 41, 869–871.
In: Olsen, R.G., Krakowka, S., Blakeslee, J.R. (Eds.), Comparative Mochizuki, M., Hashimoto, M., Hagiwara, S., Yoshida, Y., Ishiguro, S., 1999.
Pathobiology of Viral Diseases, vol. 2. CRC Press, Boca Raton, pp. Genotypes of canine distemper virus determined by analysis of the
137–164. hemagglutinin genes of recent isolates from dogs in Japan. J. Clin.
Krakowka, S., Cork, L.C., Winkelstein, J.A., Axthelm, M.K., 1987a. Establish- Microbiol. 37, 2936–2942.
ment of central nervous system infection by canine distemper virus: Montali, R.J., Bartz, C.R., Bush, M., 1987. Canine distemper virus. In: Appel,
breach of the blood-brain barrier and facilitation by antiviral anti- M. (Ed.), Virus Infections of Carnivores. Elsevier Science Publishers
body. Vet. Immunol. Immunopathol. 17, 471–482. BV, Amsterdam, pp. 437–443.
Krakowka, S., Ringler, S.S., Lewis, M., Olsen, R.G., Axthelm, M.K., 1987b. Montgomery, D.L., 1994. Astrocytes: form, functions, and roles in disease.
Immunosuppression by canine distemper virus: modulation Vet. Pathol. 31, 145–167.
of in vitro immunoglobulin synthesis, interleukin release and pros- Mori, T., Shin, Y.S., Okita, M., Hirayama, N., Miyashita, N., Gemma, T., Kai,
taglandin E2 production. Vet. Immunol. Immunopathol. 15, 181– C., Mikami, T., 1994. The biological characterization of field
201. isolates of canine distemper virus from Japan. J. Gen. Virol. 75,
Krakowka, S., 1989. Canine distemper virus infectivity of various blood 2403–2408.
fractions for central nervous system vasculature. J. Neuroimmunol. Moritz, A., Frisk, A.L., Baumgärtner, W., 1998. Beurteilung diagnostischer
21, 75–80. Möglichkeiten bei der Staupevirusinfektion des Hundes. Kleintier-
Kreutzer, M., Kreutzer, R., Siebert, U., Müller, G., Reijnders, P., Brasseur, S., praxis 43, 153–172.
Härkönen, T., Dietz, R., Sonne, C., Born, E.W., Baumgärtner, W., 2008. Moritz, A., Frisk, A., Baumgärtner, W., 2000. The evaluation of diagnostic
In search for virus carriers of the 1988 and 2002 phocine distemper procedures for the detection of canine distemper virus infection. Eur.
virus outbreaks in European harbour seals. Arch. Virol. 153, 187– J. Comp. Anim. Pract. 10, 37–47.
192. Moritz, A., Baumgärtner, W., Frisk, A.L., König, M., 2003. Sensitivität und
Kumagai, K., Yamaguchi, R., Uchida, K., Tateyama, S., 2004. Lymphoid spezifität der CDV-PR-PCR zur diagnose der kaninen staupe. Tierärz-
apoptosis in acute canine distemper. J. Vet. Med. Sci. 66, 175–181. tliche Praxis 31(K) 60–66.
Lamb, R.A., Kolakofsky, D., 2001. Paramyxoviridae: the viruses and their Moro, L., de Sousa Martins, A., de Moraes Alves, C., de Araujo Santos, F.G.,
replication. In: Knipe, D.M., Howley, P.M. (Eds.), Fields of Virology, dos Santos Nunes, J.E., Carneiro, R.A., Carvalho, R., Vasconcelos, A.C.,
4th ed., vol. 1. Lippincott Williams&Wilkins, Philadelphia, pp. 1305– 2003a. Apoptosis in canine distemper. Arch. Virol. 148, 153–164.
1443. Moro, L., Martins, A.S., Alves, C.M., Santos, F.G., Del Puerto, H.L., Vascon-
Lan, N.T., Yamaguchi, R., Uchida, K., Sugano, S., Tateyama, S., 2005. Growth celos, A.C., 2003b. Apoptosis in the cerebellum of dogs with distem-
profiles of recent canine distemper isolates on Vero cells expressing per. J. Vet. Med. B 50, 221–225.
canine signalling lymphocyte activation molecule (SLAM). J. Comp. Müller, C.F., Fatzer, R.S., Beck, K., Vandevelde, M., Zurbriggen, A., 1995.
Pathol. 133, 77–81. Studies on canine distemper virus persistence in the central nervous
Lauder, I.M., Martin, W.B., Gordon, E.B., Lawson, D.D., Campbell, R.S.F., system. Acta Neuropathol. 89, 438–445.
Watrach, A.M., 1954. Vet. Rec. 66, 607–611. Müller, G., Siebert, U., Wünschmann, A., Artelt, A., Baumgärtner, W., 2000.
Likhoshway, Y.E.V., Grachev, M.A., Kumarev, V.P., Solodun, Y.U.V., Gold- Immunohistological and serological investigation of morbillivirus
berg, O.A., Belykh, O.I., Nagieva, F.G., Nikulina, V.G., Kolesnik, B.S., infection in harbour porpoises (Phocoena phocoena) from the German
1989. Baikal seal virus. Nature 339, 266. Baltic and North Sea. Vet. Microbiol. 75, 17–25.
Löffler, S., Lottspeich, F., Lanza, F., Azorsa, D.O., ter Meulen, V., Schneider- Müller, G., Wünschmann, A., Baumgärtner, W., Birkun, A., Komakhidze, A.,
Schaulies, J., 1997. CD9, a tetraspan transmembrane protein, renders Stanev, T., Joiris, C.R., 2002. Immunohistochemical and serological
cells susceptible to canine distemper virus. J. Virol. 71, 42–49. investigations of morbillivirus infection in Black Sea harbour por-
Lo, E.H., Wang, X., Cuzner, M.L., 2002. Extracellular proteolysis in brain poises (Phocoena phocoena). Vet. Microbiol. 2332, 183–190.
injury and inflammation: role for plasminogen activators and matrix Mutinelli, F., Vandevelde, M., Griot, C., Richard, A., 1989. Astrocytic
metalloproteinases. J. Neurosci. Res. 69, 1–9. infection in canine distemper virus-induced demyelination. Acta
Machida, N., Izumisawa, N., Nakamura, T., Kiryu, K., 1992. Canine dis- Neuropathol. 77, 333–335.
temper virus infection in a masked palm civet (Paguma larvata). J. Nesseler, A., Baumgärtner, W., Gaedke, K., Zurbriggen, A., 1997. Abundant
Comp. Pathol. 107, 439–443. expression of viral nucleoprotein mRNA and restricted translation of
Machida, N., Kiryu, K., Oh-ishi, K., Kanada, E., Izumisawa, N., Nakamura, T., the corresponding viral protein in inclusion body polioencephalitis of
1993. Pathology and epidemiology of canine distemper in raccoon canine distemper. J. Comp. Pathol. 116, 291–301.
dogs (Nyctereutes procyonoides). J. Comp. Pathol. 108, 383–392. Nesseler, A., Baumgärtner, W., Zurbriggen, A., Örvell, C., 1999. Restricted
MacIntyre, A.B., Trevan, D.J., Montgomerie, R.F., 1948. Observations on virus protein translation in canine distemper virus inclusion body
canine encephalitis. Vet. Rec. 41, 635–644. polioencephalitis. Vet. Microbiol. 69, 23–28.
Maeda, H., Ozaki, K., Takagi, Y., Sawashima, K., Narama, I., 1994. Dis- Nussbaum, O., Broder, C.C., Moss, B., Stern, L.B., Rozenblatt, S., Berger, E.A.,
temper skin lesions in a dog. Zentralbl. Veterinärmed. A 41, 247–250. 1995. Functional and structural interactions between measles virus
Mahy, B.W.J., Barrett, T., Evans, S., Anderson, E.C., Bostock, C.J., 1988. hemagglutinin and CD46. J. Virol. 69, 3341–3349.
Characterization of a seal morbillivirus. Nature 336, 115. Örvell, C., 1980. Structural polypeptides of canine distemper virus. Arch.
Markus, S., Failing, K., Baumgärtner, W., 2002. Increased expression of Virol. 66, 193–206.
pro-inflammatory cytokines and lack of up-regulation of anti-inflam- Okita, M., Yanai, T., Ochikubo, F., Gemma, T., Mori, T., Maseki, T., Yama-
matory cytokines in early distemper CNS lesions. J. Neuroimmunol. nouchi, K., Mikami, T., Kai, C., 1997. Histopathological features of
125, 30–41. canine distemper recently observed in Japan. J. Comp. Pathol. 116,
McCullough, B., Krakowka, S., Koestner, A., 1974a. Experimental canine 403–408.
distemper virus-induced demyelination. Lab. Invest. 31, 216–222. Ooi, C.G., Walsh, C.A., Gallagher, J.A., Fraser, W.D., 2000. Absence of
McCullough, B., Krakowka, S., Koestner, A., 1974b. Experimental canine measles virus and canine distemper virus transcripts in long-term
distemper virus-induced lymphoid depletion. Am. J. Pathol. 74, 155– bone marrow cultures from patients with Paget’s disease of bone.
170. Bone 27, 417–421.
A. Beineke et al. / Veterinary Immunology and Immunopathology 127 (2009) 1–18 17

Orlando, E., Imbschweiler, I., Gerhauser, I., Baumgärtner, W., Wewetzer, Simon-Martı́nez, J., Ulloa-Arvizu, R., Soriano, V.E., Fajardo, R., 2007. Iden-
K., 2008. In vitro characterisation and preferential infection by canine tification of a genetic variant of canine distemper virus from clinical
distemper virus of glial precursors with Schwann cell characteristics cases in two vaccinated dogs in Mexico. Vet. J. 175, 423–426.
from adult canine brains. Neuropathol. Appl. Neurobiol., in press. Singethan, K., Topfstedt, E., Schubert, S., Duprex, W.P., Rima, B.K., Schnei-
Parker, A.J., 1978. The many faces of canine distemper. Canine Pract. 14, der-Schaulies, J., 2006. CD9-dependent regulation of Canine distem-
25–28. per virus-induced cell–cell fusion segregates with the extracellular
Pearce-Kelling, S., Mitchell, W.J., Summers, B.A., Appel, M.J., 1990. Growth domain of the haemagglutinin. J. Gen. Virol. 87, 1635–1642.
of canine distemper virus in cultured astrocytes: relationship to in Stein, V., Baumgärtner, W., Kreienbrock, L., Zurbriggen, A., Vandervelde,
vivo persistence and disease. Microb. Pathog. 8, 71–82. M., Tipold, A., 2006. Canine microglial cells: stereotype in immuno-
Pearce-Kelling, S., Mitchell, W.J., Summers, B.A., Appel, M.J., 1991. Viru- phenotype and specficity in function? Vet. Immunol. Immunopathol.
lent and attenuated canine distemper virus infects multiple dog brain 113, 277–287.
cell types in vitro. Glia 4, 408–416. Stein, V., Baumgärtner, W., Tipold, A., 2007. Differential expression of
Pringle, C.R., 1999. Virus taxonomy at the XI International Congress of CD45 on canine microglial cells. J. Vet. Med. A 54, 314–320.
Virology, Sydney, Australia. Arch. Virol. 144, 2065–2070. Stephensen, C.B., Welter, J., Thaker, S.R., Taylor, J., Tartaglia, J., Paoletti, E.,
Puff, C., Krudewig, C., Imbschweiler, I., Baumgärtner, W., Alldinger, S., 1997. Canine distemper virus (CDV) infection of ferrets as a model for
2008. Influence of persistent canine distemper virus infection on testing Morbillivirus vaccine strategies: NYVAC- and ALVAC-based
expression of RECK, matrix-metalloproteinases and their inhibitors CDV recombinants protect against symptomatic infection. J. Virol. 71,
in a canine macrophage/monocytic tumour cell line (DH82). Vet. J., in 1506–1513.
press. Summers, B.A., Greisen, H.A., Appel, M.J., 1979. Early events in canine
Raine, C.S., 1976. On the development of CNS lesions in natural canine distemper demyelinating encephalomyelitis. Acta Neuropathol. 46,
distemper encephalomyelitis. J. Neurol. Sci. 30, 13–28. 1–10.
Ralston, S.H., Digiovine, F.S., Gallacher, S.J., Boyle, I.T., Duff, G.W., 1991. Summers, B.A., Greisen, H.A., Appel, M.J., 1984. Canine distemper ence-
Failure to detect paramyxovirus sequences in Paget’s disease of bone phalomyelitis: variation with strain. J. Comp. Pathol. 94, 65–75.
using the polymerase chain reaction. J. Bone Miner. Res. 11, 1243– Summers, B.A., Appel, M.J., 1987. Demyelination in canine distemper
1248. encephalomyelitis: an ultrastructural analysis. J. Neurocytol. 16,
Raw, M.E., Pearson, G.R., Brown, P.J., Baumgärtner, W., 1992. Canine 871–881.
distemper infection associated with acute nervous signs in dogs. Summers, B.A., Appel, M.J., 1994. Aspects of canine distemper virus and
Vet. Rec. 130, 291–293. measles encephalomyelitis. Neuropathol. Appl. Neurobiol. 20, 525–
Rima, B.K., Duffy, N., Mitchell, W.J., Summers, B.A., Appel, M.J., 1991. 534.
Correlation between humoral immune responses and presence of Suter, S.E., Chein, M.B., von Messling, V., Yip, B., Cattaneo, R., Vernau, W.,
virus in the CNS in dogs experimentally infected with canine dis- Madewell, B.R., London, C.A., 2005. In vitro canine distemper virus
temper virus. Arch. Virol. 121, 1–8. infection of canine lymphoid cells: a prelude to oncolytic therapy for
Ringler, S.S., Krakowka, S., 1985. Effects of canine distemper virus on lymphoma. Clin. Cancer Res. 11, 1579–1587.
natural killer cell activity in dogs. Am. J. Vet. Res. 46, 1781–1786. Svitek, N., von Messling, V., 2007. Early cytokine mRNA expression
Rockborn, G., 1958. Further studies on viremia and neutralizing antibo- profiles predict Morbillivirus disease outcome in ferrets. Virology
dies in naturally acquired distemper in dogs. Arch. Gesamte Virus- 362, 404–410.
forsch. 8, 500–510. Tatsuo, H., Ono, N., Yanagi, Y., 2001. Morbilliviruses use signaling lym-
Roelke-Parker, M.E., Munson, L., Packer, C., Kock, R., Cleaveland, S., Car- phocyte activation molecules (CD150) as cellular receptors. J. Virol.
penter, M., O‘Brien, S.J., Pospischil, A., Hofmann-Lehmann, R., Lutz, H., 75, 5842–5850.
1996. A canine distemper virus epidemic in Serengeti lions (Panthera Tatsuo, H., Yanagi, Y., 2002. The morbillivirus receptor SLAM (CD150).
leo). Nature 379, 441–445. Microbiol. Immunol. 46, 135–142.
Rudd, P.A., Cattaneo, R., von Messling, V., 2006. Canine distemper virus Tipold, A., Moore, P., Zurbriggen, A., Burgener, I., Barben, G., Vandevelde,
uses both the anterograde and the hematogenous pathway for neu- M., 1999. Early T cell response in the central nervous system in canine
roinvasion. J. Virol. 80, 9361–9370. distemper virus infection. Acta Neuropathol. 97, 45–56.
Schmid, E., Zurbriggen, A., Gassen, U., Rima, B., ter Meulen, V., Schneider- Tsai, S.C., Summers, B.A., Appel, M.J., 1982. Interferon in cerebrospinal
Schaulies, J., 2000. Antibodies to CD9, a tetraspan transmembrane fluid. A marker for viral persistence of canine distemper encephalo-
protein, inhibits canine distemper virus-induced cell-cell fusion but myelitis. Arch. Virol. 72, 257–265.
not virus-cell fusion. J. Virol. 74, 7554–7561. Ulrich, R., Baumgärtner, W., Gerhauser, I., Seeliger, F., Haist, V., Deschl, U.,
Schneider-Schaulies, S., Dittmer, U., 2006. Silencing T cells or T-cell Alldinger, S., 2006. MMP-12, MMP-3, and TIMP-1 are markedly
silencing: concepts in virus-induced immunosuppression. J. Gen. upregulated in chronic demyelinating Theiler murine encephalomye-
Virol. 87, 1423–1438. litis. J. Neuropathol. Exp. Neurol. 65, 783–793.
Schobesberger, M., Zurbriggen, A., Summerfield, A., Vandevelde, M., Griot, van Lint, P., Libert, C., 2007. Chemokine and cytokine processing by matrix
C., 1999. Oligodendroglial degeneration in distemper: apoptosis or metalloproteinases and its effect on leukocyte migration and inflam-
necrosis? Acta Neuropathol. 97, 279–287. mation. J. Leukoc. Biol. 82, 1375–1381.
Schobesberger, M., Zurbriggen, A., Doherr, M.G., Weissenböck, H., Van- van Moll, P., Alldinger, S., Baumgärtner, W., Adami, M., 1995. Distemper in
develde, M., Lassmann, H., Griot, C., 2002. Demyelination precedes wild carnivores: an epidemiological, histological and immunocyto-
oligodendrocyte loss in canine distemper virus-induced encephalitis. chemical study. Vet. Microbiol. 44, 193–199.
Acta Neuropathol. 103, 11–19. Vandevelde, M., Kristensen, B., 1977. Observations on the distribution of
Schobesberger, M., Summerfield, A., Doherr, M.G., Zurbriggen, A., Griot, C., canine distemper virus in the central nervous system of dogs with
2005. Canine distemper virus-induced depletion of uninfected lym- demyelinating encephalitis. Acta Neuropathol. 40, 233–236.
phocytes is associated with apoptosis. Vet. Immunol. Immunopathol. Vandevelde, M., Fankhauser, R., Kristensen, F., Kristensen, B., 1981.
104, 33–44. Immunoglobulins in demyelinating lesions in canine distemper ence-
Schwab, S., Herden, C., Seeliger, F., Papaioannou, N., Psalla, D., Poliozopou, phalitis. An immunohistological study. Acta Neuropathol. 54, 31–41.
Z., Baumgärtner, W., 2007. Non-suppurative meningoencephalitis of Vandevelde, M., Kristensen, F., Kristensen, B., Steck, A.J., Kihm, U., 1982.
unknown origin in cats and dogs: an immunohistochemical study. J. Immunological and pathological findings in demyelinating encepha-
Comp. Pathol. 136, 96–110. litis associated with canine distemper virus infection. Acta Neuro-
Seehusen, F., Orlando, E., Wewetzer, K., Baumgärtner, W., 2007. pathol. 56, 1–8.
Vimentin-positive astrocytes in canine distemper—a target for canine Vandevelde, M., Bichsel, P., Cerruti-Sola, S., Steck, A., Kristensen, F.,
distemper virus especially in chronic lesions? Acta Neuropathol. 114, Higgins, R.J., 1983. Glial proteins in canine distemper virus-induced
597–608. demyelination. A sequential immunocytochemical study. Acta Neu-
Seibel, H., Baumgärtner, W., Müller, G., Wohlsein, P., Siebert, U., 2007. ropathol. 59, 269–276.
Retrospektive analyse der zwei seehundstaupe-epidemien in der Vandevelde, M., Zurbriggen, A., Dumas, M., Palmer, D., 1985a. Canine
nord- und ostesee 1988 und 2002. Dtsch. Tierärztl. Wochenschr. distemper virus does not infect oligodendrocytes in vitro. J. Neurol.
114, 284–293. Sci. 69, 133–137.
Shapshak, P., Graves, M.C., Imagawa, D.T., 1987. Autologous and allo- Vandevelde, M., Zurbriggen, A., Higgins, R.J., Palmer, D., 1985b. Spread and
geneic antibody responses to canine distemper virus isolates from distribution of viral antigen in nervous canine distemper. Acta Neu-
dogs with chronic neurological diseases. Viral Immunol. 1, 45–54. ropathol. 67, 211–218.
Shek, W.R., Schultz, R.D., Appel, M.J., 1980. Natural and immune cytolysis Vandevelde, M., Zurbriggen, A., Steck, A., Bichsel, P., 1986. Studies on the
of canine distemper virus-infected target cells. Infect. Immun. 28, intrathecal humoral immune response in canine distemper encepha-
724–734. litis. J. Neuroimmunol. 11, 41–51.
18 A. Beineke et al. / Veterinary Immunology and Immunopathology 127 (2009) 1–18

Vandevelde, M., Zurbriggen, A., 1995. The neurobiology of canine dis- Wünschmann, A., Alldinger, S., Kremmer, E., Baumgärtner, W., 1999.
temper virus infection. Vet. Microbiol. 44, 271–280. Identification of CD4+ and CD8+ cell subsets and B cells in the brain
Vandevelde, M., Zurbriggen, A., 2005. Demyelination in canine distemper of dogs with spontaneous acute, subacute-, and chronic-demyelinat-
virus infection: a review. Acta Neuropathol. 109, 56–68. ing distemper encephalitis. Vet. Immunol. Immunopathol. 67, 101–
von Messling, V., Springfeld, C., Devaux, P., Cattaneo, R., 2003. A ferret 116.
model of canine distemper virus virulence and immunosuppression. J. Wünschmann, A., Kremmer, E., Baumgärtner, W., 2000. Phenotypical
Virol. 77, 12579–12591. characterization of T and B cell areas in lymphoid tissues of dogs with
von Messling, V., Milosevic, D., Cattaneo, R., 2004. Tropism illuminated: spontaneous distemper. Vet. Immunol. Immunopathol. 73, 83–98.
lymphocyte-based pathways blazed by lethal morbillivirus through Yamaguchi, R., Kojimoto, A., Sakai, H., Uchida, K., Sugano, S., Tateyama, S.,
the host immune system. Proc. Natl. Acad. Sci. U.S.A. 101, 14216– 2005. Growth characteristics of canine distemper virus in a new cell
14221. line CCT cells originated from canine malignant histiocytosis. J. Vet.
von Messling, V., Svitek, N., Cattaneo, R., 2006. Receptor (SLAM [CD150]) Med. Sci. 67, 203–206.
recognition and the V protein sustain swift lymphocyte-based inva- Yoshikawa, Y., Ochikubo, F., Matsubara, Y., Tsuruoka, H., Ishii, M., Shirota,
sion of mucosal tissue and lymphatic organs by a morbillivirus. J. K., Nomura, Y., Sugiyama, M., Yamanouchi, K., 1989. Natural infection
Virol. 80, 6084–6092. with canine distemper virus in a Japanese monkey (Macaca fuscata).
Welter, J., Taylor, J., Tartaglia, J., Paoletti, E., Stephensen, C.B., 2000. Vet. Microbiol. 20, 193–205.
Vaccination against canine distemper virus infection in infant ferrets Zurbriggen, A., Vandevelde, M., 1983. Canine distemper virus-induced
with and without maternal antibody protection, using recombinant glial cell changes in vitro. Acta Neuropathol. 62, 51–58.
attenuated poxvirus vaccines. J. Virol. 74, 6358–6367. Zurbriggen, A., Vandevelde, M., Dumas, M., 1986. Secondary degeneration
Wenzlow, N., Plattet, P., Wittek, R., Zurbriggen, A., Gröne, A., 2007. of oligodendrocytes in canine distemper virus infection in vitro. Lab.
Immunohistochemical demonstration of the putative canine distem- Invest. 54, 424–431.
per virus receptor CD150 in dogs with and without distemper. Vet. Zurbriggen, A., Vandevelde, M., Bollo, E., 1987a. Demyelinating, non-
Pathol. 44, 943–948. demyelinating and attenuated canine distemper virus strains induce
Winters, K.A., Mathes, L.E., Krakowka, S., Olsen, R.G., 1983. Immunoglo- oligodendroglial cytolysis in vitro. J. Neurol. Sci. 79, 33–41.
bulin class response to canine distemper virus in gnotobiotic dogs. Zurbriggen, A., Vandevelde, M., Dumas, M., Griot, C., Bollo, E., 1987b.
Vet. Immunol. Immunopathol. 5, 209–215. Oligodendroglial pathology in canine distemper virus infection in
Wisniewski, H., Raine, C.S., Kay, W.J., 1972. Observations on viral demye- vitro. Acta Neuropathol. 74, 366–373.
linating encephalomyelitis, Canine distemper. Lab. Invest. 26, 589– Zurbriggen, A., Yamawaki, M., Vandevelde, M., 1993. Restricted canine
599. distemper virus infection of oligodendrocytes. Lab. Invest. 68, 277–
Wohlsein, P., Müller, G., Haas, L., Siebert, U., Harder, T.C., Baumgärtner, W., 284.
2007. Antigenic characterization of phocine distemper virus causing Zurbriggen, A., Graber, H.U., Vandevelde, M., 1995a. Selective spread and
mass mortality in 2002 and its relationship to other morbilliviruses. reduced virus release leads to canine distemper virus persistence in
Arch. Virol. 152, 1559–1564. the nervous system. Vet. Microbiol. 44, 281–288.
Wright, N.G., Cornwell, A.J., Thompson, H., Lauder, I.M., 1974. Canine Zurbriggen, A., Graber, H.U., Wagner, A., Vandevelde, M., 1995b. Canine
distemper: current concepts in laboratory and clinical diagnosis. Vet. distemper virus persistence in the nervous system is associated with
Rec. 94, 86–92. noncytolytic selective virus spread. J. Virol. 69, 1678–1686.

You might also like