Hostname: page-component-8448b6f56d-tj2md Total loading time: 0 Render date: 2024-04-23T17:37:42.082Z Has data issue: false hasContentIssue false

The pathogenesis of bornaviral diseases in mammals

Published online by Cambridge University Press:  23 May 2016

Ian Tizard*
Affiliation:
Department of Veterinary Pathobiology and the Schubot Exotic Bird Health Center, Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, College Station, Texas 77843, USA
Judith Ball
Affiliation:
Department of Veterinary Pathobiology and the Schubot Exotic Bird Health Center, Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, College Station, Texas 77843, USA
George Stoica
Affiliation:
Department of Veterinary Pathobiology and the Schubot Exotic Bird Health Center, Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, College Station, Texas 77843, USA
Susan Payne
Affiliation:
Department of Veterinary Pathobiology and the Schubot Exotic Bird Health Center, Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, College Station, Texas 77843, USA
*
*Corresponding author. E-mail: itizard@cvm.tamu.edu
Rights & Permissions [Opens in a new window]

Abstract

Natural bornavirus infections and their resulting diseases are largely restricted to horses and sheep in Central Europe. The disease also occurs naturally in cats, and can be induced experimentally in laboratory rodents and numerous other mammals. Borna disease virus-1 (BoDV-1), the cause of most cases of mammalian Borna disease, is a negative-stranded RNA virus that replicates within the nucleus of target cells. It causes severe, often lethal, encephalitis in susceptible species. Recent events, especially the discovery of numerous new species of bornaviruses in birds and a report of an acute, lethal bornaviral encephalitis in humans, apparently acquired from squirrels, have revived interest in this remarkable family of viruses. The clinical manifestations of the bornaviral diseases are highly variable. Thus, in addition to acute lethal encephalitis, they can cause persistent neurologic disease associated with diverse behavioral changes. They also cause a severe retinitis resulting in blindness. In this review, we discuss both the pathological lesions observed in mammalian bornaviral disease and the complex pathogenesis of the neurologic disease. Thus infected neurons may be destroyed by T-cell-mediated cytotoxicity. They may die as a result of excessive inflammatory cytokine release from microglia. They may also die as a result of a ‘glutaminergic storm’ due to a failure of infected astrocytes to regulate brain glutamate levels.

Type
Review Article
Copyright
Copyright © Cambridge University Press 2016 

Introduction

Bornaviruses have long been known to cause meningoencephalitis in horses and sheep in parts of Central Europe (Metzler et al., Reference Metzler, Frei and Danner1976; Durrwald and Ludwig, Reference Durrwald and Ludwig1997; Richt et al., Reference Richt, Grabner and Herzog2000). Subsequent studies have demonstrated that they induce a similar disease in experimentally infected laboratory rats and mice (Narayan et al., Reference Narayan, Herzog, Frese, Scheefers and Rott1983a, Reference Narayan, Herzog, Frese, Scheefers and Rottb; Kao et al., Reference Kao, Ludwig and Gosztonyi1984). The course of the rodent disease is however dependent upon age, such that adult rats develop an acute encephalitis while newborns develop multiple neurodevelopmental disorders. Other bornavirus-infected rodents such as gerbils fail to show these age-related differences. Additionally, a bornavirus from variegated squirrels (Variegated squirrel bornavirus, VSBV-1) can cause an acute lethal encephalitis in humans (Hoffmann et al., Reference Hoffmann, Tappe, Hoper, Herden, Boldt, Mawrin, Niederstrasser, Muller, Jenckel, Van Der Grinten, Lutter, Abendroth, Teifke, Cadar, Schmidt-Chanasit, Ulrich and Beer2015). More significantly, it was believed for many years that bornaviruses contributed to human mental illness (Bode and Ludwig, Reference Bode and Ludwig2003). This concept has been largely discredited. In contrast, avian bornaviral infection of parrots and waterbirds results in proventricular dilatation disease as well as encephalitis (Honkavuori et al., Reference Honkavuori, Shivaprasad, Williams, Quan, Hornig, Street, Palacios, Hutchison, Franca, Egholm, Briese and Lipkin2008; Kistler et al., Reference Kistler, Gancz, Clubb, Skewes-Cox, Fischer, Sorber, Chiu, Lublin, Mechani, Farnoushi, Greninger, Wen, Karlene, Ganem and Derisi2008; Rubbenstroth et al., Reference Rubbenstroth, Schmidt, Rinder, Legler, Corman and Staeheli2014). An uncharacterized bornavirus also appears to cause a naturally occurring acute paralytic syndrome in ostriches (Malkinson et al., Reference Malkinson, Weisman, Ashash, Bode and Ludwig1993; Ashash et al., Reference Ashash, Malkinson, Meir, Perl and Weisman1996). The goal of this review is to provide an overview of the pathogenesis of bornavirus-mediated neurologic disease in mammals in light of these recent findings.

Bornaviruses

Bornaviruses (order Mononegavirales family Bornaviridae,) are enveloped non-segmented, single stranded, negative sense RNA viruses whose prototype member is Borna disease virus 1 (BoDV-1) (Lipkin et al., Reference Lipkin, Briese and Hornig2011). BoDV-1 primarily infects mammals but has been detected in birds (Malkinson et al., Reference Malkinson, Weisman, Ashash, Bode and Ludwig1993; Berg et al., Reference Berg, Johansson, Montell and Berg2001). Bornaviruses have a unique genome organization among the Mononegavirales. The 8.9 kb genome contains six open reading frames that encode six viral proteins, N, P, M, G and L plus a small X protein that overlaps the P reading frame. To maximize the use of the genome, BoDV-1 employs the RNA splicing machinery for gene expression (Cubitt et al., Reference Cubitt, Oldstone, Valcarcel and Carlos de la Torre1994; Schneider et al., Reference Schneider, Schneemann and Lipkin1994). Three transcription start sites and four termination sites have been identified as well as the use of splicing to generate additional mRNAs (Schneemann et al., Reference Schneemann, Schneider, Kim and Lipkin1994; Ludwig, Reference Ludwig2008). N, P and L proteins together with the viral RNA form a ribonucleoprotein (RNP) complex (Fig. 1). Once within the cytoplasm, the RNP complex translocates into the nucleus (Jamali et al., Reference Jamali, Jamali, Mehrbod and Mofrad2011; Honda and Tomonaga, Reference Honda and Tomonaga2013). Viral replication and transcription occur within the nucleus. Only a small number of infectious particles are released from bornavirus-infected cells (Gonzalez-Dunia et al., Reference Gonzalez-Dunia, Cubitt and de la Torre1998; Tomonaga et al., Reference Tomonaga, Kobayashi and Ikuta2002).

Fig. 1. The intranuclear replication of bornaviruses. The viral ribonucleoprotein (RNP) complex is imported into the nucleus. It binds to the nuclear chromatin and generates more of these RNP complexes. The virus rarely forms complete virions and as a result, it probably spreads between cells in the form of the RNP.

For many years, BoDV-1 was the only known member of the Bornaviridae. It shows little genetic variation with 4.1% diversity in nucleotides and 1.5–3% diversity in the amino acid sequences of its N- and P-proteins (Formella et al., Reference Formella, Jehle, Sauder, Staeheli and Schwemmle2000; Lipkin et al., Reference Lipkin, Briese and Hornig2011). The finding of multiple bornavirus species in parrots in 2008 changed that situation. Thus the known Bornaviridae have expanded from a single conserved mammalian virus to a highly diverse family of at least six viral species (Kuhn et al., Reference Kuhn, Dürrwald, Bào, Briese, Carbone, Clawson, Derisi, Garten, Jahrling, Kolodziejek, Rubbenstroth, Schwemmle, Stenglein, Tomonaga, Weissenböck and Nowotny2015). Mammalian 1 bornavirus encompasses classical Borna disease virus- BoDV-1 and BoDV-2. Recently, VSBV-1 has been identified and may be a new species. Parrot 1 bornavirus encompasses psittacine bornaviruses 1, 2, 3, 4 and 7; Parrot 2 bornavirus, encompasses psittacine bornavirus 5; Passeriform 1 bornavirus (canary bornaviruses); Passeriform 2 bornavirus (estrilidid finch bornavirus); waterbird 1 bornavirus (aquatic bird bornaviruses -1 and -2) have also been described. At least two bornaviruses have also been identified in snakes (Elapid 1 bornavirus and an unclassified virus) and multiple endogenous bornaviral sequences have been identified in mammals including people (Horie et al., Reference Horie, Honda, Suzuki, Kobayashi, Daito, Oshida, Ikuta, Jern, Gojobori, Coffin and Tomonaga2010, Reference Horie, Kobayashi, Suzuki and Tomonaga2013).

Borna disease in mammals

Natural Borna disease affects horses and sheep in central Europe. It occasionally affects other domestic mammals such as donkeys, goats and cattle. It has been recorded in rabbits, some zoo animals and dogs (Staeheli et al., Reference Staeheli, Sauder, Hausmann, Ehrensperger and Schwemmle2000). Cases of ‘staggering disease’ in cats have also been attributed to infection by BoDV-1 (Wensman et al., Reference Wensman, Jaderlund, Holst and Berg2014). It is believed that these naturally occurring cases result from infection acquired from the urine of shrews.

The bicolored white-toothed shrew (Crocidura leucodon) is a reservoir host species for BoDV-1 within endemic regions of central Europe and develops an asymptomatic infection (Sprankel et al., Reference Sprankel, Richarz, Ludwig and Rott1978; Puorger et al., Reference Puorger, Hilbe, Muller, Kolodziejek, Nowotny, Zlinszky and Ehrensperger2010; Nobach et al., Reference Nobach, Bourg, Herzog, Lange-Herbst, Encarnacao, Eickmann and Herden2015). Virus can be detected, not only in the brain and other nervous tissues but also in hepatocytes, Leydig cells in the testes and epithelial cells of the respiratory and urinary tracts. Shrews express large amounts of virus in their oral epithelial cells, as well as skin keratinocytes (Durrwald et al., Reference Durrwald, Kolodziejek, Weissenbock and Nowotny2014). Infectious virus and viral RNA can be demonstrated in saliva, urine, skin, tears and feces (Nobach et al., Reference Nobach, Bourg, Herzog, Lange-Herbst, Encarnacao, Eickmann and Herden2015). There is also evidence for a wild reservoir of BoDV-1 in bank voles (Myodes glareolus) in northern Europe (Kinnunen et al., Reference Kinnunen, Billich, Ek-Kommonen, Henttonen, Kallio, Niemimaa, Palva, Staeheli, Vaheri and Vapalahti2007, Reference Kinnunen, Palva, Vaheri and Vapalahti2013).

Horses

Naturally occurring Borna disease has been recognized in horses in eastern Germany since the 18th Century. However, it gained notoriety (and its name) when in 1894–1896 it immobilized a regiment of cavalry based in the town of Borna in Saxony (Richt et al., Reference Richt, Grabner and Herzog2000). Equine BoDV-1 infection occurs naturally only in Germany, Switzerland, Lichtenstein and Austria where on average about 12% of horses are seropositive.

The typical disease course in horses is characterized by an acute encephalitis that develops following an incubation period of 4 weeks to 3 months (Richt et al., Reference Richt, Pfeuffer, Christ, Frese, Bechter and Herzog1997). Non-specific clinical findings such as depression (apathy, somnolence, stupor), fever and anorexia precede ataxia. Horses spread their legs or cross them, and support themselves by pressing their head against the manger or wall. Repetitive behaviors including circling, vacuous slow motion chewing and severe tooth grinding may occur. Eventually, the horses become paretic, develop neurogenic torticollis, or blindness, coma and death. Lethality exceeds 80% (Ludwig and Bode, Reference Ludwig and Bode2000; Lipkin et al., Reference Lipkin, Briese and Hornig2011). Death usually occurs 1–4 weeks after the onset of clinical signs. A chronic recurring form of the disease may develop in up to 10% of cases and thus some horses may become persistently infected carriers. Serologic surveys indicate that infection without obvious clinical disease is common (Richt et al., Reference Richt, Grabner and Herzog2000).

On necropsy, horses show no gross lesions. Histopathology shows a non-purulent meningoencephalomyelitis with randomly scattered inflammatory foci in the hippocampus and along the central axis including the mesencephalon and the hypothalamus (Fig. 2). These foci consist of extensive lymphocytic perivascular infiltrates involving primarily the gray matter (Rott and Becht, Reference Rott and Becht1995; Bilzer et al., Reference Bilzer, Grabner and Stitz1996). Most of the inflammatory cells in these foci are CD3+ T cells (Caplazi and Ehrensperger, Reference Caplazi and Ehrensperger1998). Of these, CD4+ cells outnumber CD8+ T cells. Natural killer (NK) cells are likely present in the infiltrates as well (Hatalski et al., Reference Hatalski, Hickey and Lipkin1998). Macrophages are fewer and B cells/plasma cells fewer still. Polymorphonuclear leukocytes are rarely present. The highest viral titers occur in the hippocampus and the piriform cortex reflecting viral invasion from the olfactory bulbs. The lowest viral titers are in the cerebellum (Gosztonyi, Reference Gosztonyi2008). The virus is primarily located within neuronal nuclei (Richt et al., Reference Richt, Grabner and Herzog2000). Less consistently, smaller amounts of viral antigen may be detected in the neuronal perikaryon, dendrites and axons (Bilzer et al., Reference Bilzer, Grabner and Stitz1996). There is usually a correlation between viral titer and the severity of the brain lesions and the inflammatory exudate is associated with the presence of viral antigen, but the inflammatory cells never appear to contain viral antigens (Gosztonyi and Ludwig, Reference Gosztonyi and Ludwig1984). In chronic infections, viral antigen is demonstrable in astrocytes and the meningeal surface is infiltrated with mononuclear cells (Richt et al., Reference Richt, Grabner and Herzog2000). Inflammation may spread from the gray matter into the adjacent white matter and involve nerve roots and the spinal ganglia (Johnson, Reference Johnson1980).

Fig. 2. The major sites within the equine brain that BoDV-1 appears to favor. Infected neurons are also scattered diffusely throughout the cerebrum. The arrows indicate what is believed to be the ‘natural’ route of viral invasion, originating in the olfactory bulb.

Sheep

Natural Borna disease in sheep resembles that in horses although large numbers of sheep may be affected within a flock unlike horses where the disease usually affects few animals (Richt et al., Reference Richt, Pfeuffer, Christ, Frese, Bechter and Herzog1997). Clinical signs vary from minor behavioral changes to severe encephalomyelitis reflecting the intensity of the inflammatory response in the brain (Vahlenkamp et al., Reference Vahlenkamp, Konrath, Weber and Muller2002). A short period of depression precedes overt disease including somnolence, ataxia and multiple deficits (Metzler et al., Reference Metzler, Frei and Danner1976; Waelchli et al., Reference Waelchli, Ehrensperger, Metzler and Winder1985). As in horses, the majority of BoDV-1 infections in sheep remain asymptomatic. The location of the virus within the brain is identical to that in horses. Lethality in clinically affected animals is at least 50%. Some sheep recover completely while others survive but fail to thrive (Metzler et al., Reference Metzler, Minder, Wegmann and Zindel1979).

Cats

BoDV-1 causes staggering disease in cats (Lundgren et al., Reference Lundgren, Zimmermann, Bode, Czech, Gosztonyi, Lindberg and Ludwig1995b; Wensman et al., Reference Wensman, Jaderlund, Holst and Berg2014; Lutz et al., Reference Lutz, Addie, Boucraut-Baralon, Egberink, Frymus, Gruffydd-Jones, Hartmann, Horzinek, Hosie, Lloret, Marsilio, Pennisi, Radford, Thiry, Truyen and Mostl2015). This begins with fever, apathy and a reduced appetite. Cats eventually develop ataxia, gait disturbances, blindness, lower back pain, behavioral changes, loss of postural reactions and hind-leg paralysis (Wensman et al., Reference Wensman, Jaderlund, Holst and Berg2014). Some may be unable to retract their claws (Lutz et al., Reference Lutz, Addie, Boucraut-Baralon, Egberink, Frymus, Gruffydd-Jones, Hartmann, Horzinek, Hosie, Lloret, Marsilio, Pennisi, Radford, Thiry, Truyen and Mostl2015). A similar disease can be induced by experimental challenge with BoDV-1 (Lundgren et al., Reference Lundgren, Johannisson, Zimmermann, Bode, Rozell, Muluneh, Lindberg and Ludwig1997). Some cats recover. There are anecdotal reports of cats surviving the acute infection and later developing extreme obesity (Wensman et al., Reference Wensman, Jaderlund, Holst and Berg2014). The lesions in the cat brain are similar to those observed in horses although plasma cells may be more prominent (Lundgren et al., Reference Lundgren, Johannisson, Zimmermann, Bode, Rozell, Muluneh, Lindberg and Ludwig1997). While it is possible that cats become infected by eating small infected mammals or birds (Berg et al., Reference Berg, Johansson, Montell and Berg2001) it is just as likely that it results from exposure to the urine of infected shrews (see below). It has been suggested that as in horses and sheep, the virus probably enters the cat through the olfactory epithelium and the oropharyngeal mucosa and then gains access to the brain by intra-axonal spread (Wensman et al., Reference Wensman, Jaderlund, Holst and Berg2014).

BoDV-1 triggers an intense T cell reaction in the cat brain. Lundgren et al. observed elevated T cell numbers in the blood and brains of experimentally challenged cats (Lundgren et al., Reference Lundgren, Lindberg, Ludwig and Gosztonyi1995a, Reference Lundgren, Johannisson, Zimmermann, Bode, Rozell, Muluneh, Lindberg and Ludwig1997). Berg et al. used flow cytometry to demonstrate that the CD8+ T cells in the brain of infected cats belong to a non-major histocompatibility complex (MHC) restricted population and suggested that they could be important in viral clearance from neurons (Berg et al., Reference Berg, Johannisson, Johansson, Hein, Berg and Dorries1999). Some cats may develop disease in the absence of gross encephalitis, a disease process similar to that seen in neonatal rats (See below). Infected cats express high levels of interferon (IFN)-γ in their brains and this may promote NK and T cell cytotoxicity and neuronal destruction (Wensman et al., Reference Wensman, Ilback, Hjertstrom, Blomstrom, Gustavsson, Jaderlund, Strom-Holst, Belak, Berg and Berg2011, Reference Wensman, Jaderlund, Holst and Berg2014). However BoDV-1 can survive in the presence of IFN-γ (Wensman et al., Reference Wensman, Jaderlund, Holst and Berg2014). CD8+ cells stimulated by BoDV-1 are found in blood, spleen and brain. Cats may also develop inflammatory changes in their intra-abdominal ganglia and the adrenal medulla (Wensman et al., Reference Wensman, Jaderlund, Gustavsson, Hansson-Hamlin, Karlstam, Lilliehook, Ostrom, Belak, Berg and Holst2012).

Humans

At least one bornaviral species can infect humans. In 2011–2013, three breeders of variegated squirrels (Sciurus variegatoides) in Germany developed a progressive meningoencephalitis and died within 2–4 months (Hoffmann et al., Reference Hoffmann, Tappe, Hoper, Herden, Boldt, Mawrin, Niederstrasser, Muller, Jenckel, Van Der Grinten, Lutter, Abendroth, Teifke, Cadar, Schmidt-Chanasit, Ulrich and Beer2015). Their clinical disease progressed from a fever, to progressive psychomotor slowing, confusion, ocular paresis, coma and death. On autopsy, their brains showed edema, gliosis, lymphocyte infiltration with perivascular cuffing, and necrosis. Subsequently a bornavirus was isolated from their brains as well as from the brains of one of their squirrels. Sequencing of this virus genome revealed that it was a previously unidentified mammalian bornavirus (VSBV-1). The distribution of virus in the brains of these patients and in the squirrel resembled that seen in equine Borna disease. There was a high viral RNA load in an oropharyngeal swab from the squirrel suggesting that squirrels may act as carriers and that the virus may have been accidentally transmitted by squirrel bites (Hoffmann et al., Reference Hoffmann, Tappe, Hoper, Herden, Boldt, Mawrin, Niederstrasser, Muller, Jenckel, Van Der Grinten, Lutter, Abendroth, Teifke, Cadar, Schmidt-Chanasit, Ulrich and Beer2015). One of these patients had high levels of autoantibodies to the Yo autoantigen, an antigen located within Purkinje cells. However other authors have considered the presence of these autoantibodies in other viral encephalitides to be a clinically irrelevant epiphenomenon (Jarius and Wildemann, Reference Jarius and Wildemann2015).

In 1985, it was suggested that Borna disease virus could cause mental illness in humans (Amsterdam et al., Reference Amsterdam, Winokur, Dyson, Herzog, Gonzalez, Rott and Koprowski1985). Thus Rott et al. using an indirect immunofluorescent focus assay, detected antibodies to BoDV-1 in 16 out of 979 psychiatric patients but none were found in 200 normal volunteers (Rott et al., Reference Rott, Herzog, Fleischer, Winokur, Amsterdam, Dyson and Koprowski1985). Fu et al. also reported that patients with mental illness had antibody titers against BoDV-1 (Fu et al., Reference Fu, Amsterdam, Kao, Shankar, Koprowski and Dietzschold1993). These, and subsequent reports stimulated extensive investigations into possible links between bornaviral infection and mental health. It was suggested that BoDV-1 was associated with human neuropsychiatric diseases including bipolar disorder, chronic fatigue syndrome, schizophrenia and unipolar depression. However, isolation of BoDV-1 from humans is rare, and the serologic results obtained are possibly mis- or over-interpreted (Lipkin et al., Reference Lipkin, Briese and Hornig2011). Positive reverse-transcriptase polymerase chain reaction results may have been a result of inadvertent laboratory contamination, because the human-derived sequences showed marked similarity to animal-derived laboratory strains (Durrwald et al., Reference Durrwald, Kolodziejek, Herzog and Nowotny2007). It is now generally accepted that bornaviruses are not a significant cause of human neuropsychiatric disease (Lipkin et al., Reference Lipkin, Briese and Hornig2011; Hornig et al., Reference Hornig, Briese, Licinio, Khabbaz, Altshuler, Potkin, Schwemmle, Siemetzki, Mintz, Honkavuori, Kraemer, Egan, Whybrow, Bunney and Lipkin2012).

Rodents

Experimental BoDV-1 infection of laboratory rats and mice has revealed many key features of bornaviral pathogenesis. One of the most significant features of these infections is the difference between the nature of the disease in adult and newborn rats.

Adult rats

Experimental infection of immunocompetent adult rats with BoDV-1 results in the development of an encephalitis similar to that observed in horses and sheep. The lesions that develop depend on the age and immune status of animals as well as their genetic background, the route of inoculation and on the passage number of the virus (Wu et al., Reference Wu, Schulz, Lin, Saar, Patone, Fischer, Hubner, Heimrich and Schwemmle2013). Many infected adult rats die within 1–4 months as a result of the encephalitis but 50–80% may survive and develop behavioral abnormalities or an obesity syndrome (Hirano et al., Reference Hirano, Kao and Ludwig1983; Narayan et al., Reference Narayan, Herzog, Frese, Scheefers and Rott1983a).

BoDV-1-inoculated adult rats develop illness after an incubation period of 17–90 days, the time required for the virus to spread in dendritic-axonal processes from the inoculation site to the hippocampus (Carbone et al., Reference Carbone, Duchala, Griffin, Kincaid and Narayan1987). Intranasal inoculation of virus results in spread to the olfactory bulb by 4–6 days, and to the rest of the brain in 20 days. Inoculation into the footpads results in intraaxonal spread towards the brain. The virus then migrates from the dorsal root ganglia adjacent to the lumbar spinal cord, to the gracilis nucleus in the medulla, the pyramidal cells in the hippocampus and eventually, results in clinical disease. This progression takes 50–60 days (Carbone et al., Reference Carbone, Duchala, Griffin, Kincaid and Narayan1987). Intravenous inoculation of rat foot veins fails to cause infection suggesting that the virus does not cross the vascular endothelium (Carbone et al., Reference Carbone, Rubin, Nishino and Pletnikov2001). Sectioning of the foot nerve within 1 day of footpad inoculation also prevents viral migration to the brain but not if the nerve is cut later. Once introduced into the rat central nervous system, BoDV-1 persists in the brain and spinal cord (Herzog et al., Reference Herzog, Kompter, Frese and Rott1984). In immunocompetent adult rats neither infectious virus nor viral antigens can be detected in lung, spleen, kidney, muscle, peritoneal macrophages or blood leukocytes (Stitz et al., Reference Stitz, Bilzer and Planz2002).

Infected adult Lewis rats show increased alertness at 20 days post-infection. Eventually this develops into frenzied behavior (exaggerated motor responses to minor stimuli), aggression and ataxia (Narayan et al., Reference Narayan, Herzog, Frese, Scheefers and Rott1983a). The onset of these behaviors coincides with the development of encephalitis and retinitis that reaches maximum intensity 30–40 days after infection. The cessation of this active phase coincides with a decline in inflammation and the onset of blindness and results in a change to a passive behavior with apathy, somnolence and depression. The diminution of inflammation may be associated with the development of static hydrocephalus. By 200 days, there is much virus but minimal inflammation in the brain (Narayan et al., Reference Narayan, Herzog, Frese, Scheefers and Rott1983b).

Neonatal rats

In neonatal rats, experimental infection with BoDV-1 results in transient mild inflammation, and there are no immediate clinical signs of disease. However the virus does cause glial activation that eventually leads to significant changes in brain development, behavioral abnormalities and a life-long persistent infection (Hornig et al., Reference Hornig, Weissenbock, Horscroft and Lipkin1999). This infection is initiated at a time when the rat brain is continuing to develop and its neuronal connections are being tuned and adapted to environmental influences (Gonzalez-Dunia et al., Reference Gonzalez-Dunia, Volmer, Mayer and Schwemmle2005). Herzog et al. compared BoDV-1 distribution following challenge in adult and neonatal rats. In adults, the virus was restricted to the central nervous system, but in neonates it was also found in the heart, adrenal, stomach and intestine but not blood (Herzog et al., Reference Herzog, Kompter, Frese and Rott1984).

This persistent infection of newborn rats results in selective injury to those areas of the brain undergoing significant post-natal development (de la Torre, Reference de la Torre2002). Thus, there is neuronal loss in the cortex, hippocampus and cerebellum (Fig. 3). The lesions in the hippocampus are concentrated in the granule cells of the dentate gyrus. This neuronal degeneration results from progressive granule cell apoptosis (Hornig et al., Reference Hornig, Weissenbock, Horscroft and Lipkin1999). Neonatal neuronal plasticity is accompanied by formation of new synapses and an increase in dendritic arborization (Engert and Bonhoeffer, Reference Engert and Bonhoeffer1999). Synaptic density is altered in BoDV-1-infected neonatal rats and in BoDV-1-P transgenic mice (Gonzalez-Dunia et al., Reference Gonzalez-Dunia, Watanabe, Syan, Mallory, Masliah and de la Torre2000; Kamitani et al., Reference Kamitani, Ono, Yoshino, Kobayashi, Taharaguchi, Lee, Yamashita, Kobayashi, Okamoto, Taniyama, Tomonaga and Ikuta2003)

Fig. 3. A schematic diagram showing the multiple alterations in neurogenesis induced by BoDV-1 in neonatal mice.

Eisenman et al. showed reduced cerebellar size but normal lamellar organization in BoDV-1-infected neonatal rats (Eisenman et al., Reference Eisenman, Brothers, Tran, Kean, Dickson, Dietzschold and Hooper1999). Gaps eventually develop in the Purkinje cell layer of the cerebellum and it has been estimated that up to 75% of Purkinje cells may be lost in these animals (Bautista et al., Reference Bautista, Rubin, Moran, Schwartz and Carbone1995; Eisenman et al., Reference Eisenman, Brothers, Tran, Kean, Dickson, Dietzschold and Hooper1999).

Persistently infected neonatal rats also develop an astrocytosis (Ovanesov et al., Reference Ovanesov, Ayhan, Wolbert, Moldovan, Sauder and Pletnikov2008a). These astrocytes control homeostasis around synapses and have a key role in removing excess neurotransmitters such as glutamate (Coulter and Eid, Reference Coulter and Eid2012). Bornavirus-infected astrocytes have a reduced ability to take up glutamate (de la Torre, Reference de la Torre2002) (see below).

Persistently infected neonatal rats have impaired cognitive functions, and deficiencies in fear conditioning (Carbone et al., Reference Carbone, Rubin, Nishino and Pletnikov2001; Pletnikov et al., Reference Pletnikov, Rubin, Vogel, Moran and Carbone2002). Despite their cerebellar abnormalities, they do not show ataxia, but they do exhibit novelty-induced hyperactivity, chronic anxiety and have abnormal sleep-wake cycles, as well as decreased play behavior (Hornig et al., Reference Hornig, Solbrig, Horscroft, Weissenbock and Lipkin2001; Pletnikov et al., Reference Pletnikov, Rubin, Vogel, Moran and Carbone2002). These neonatal rat behaviors resembles some human neuropsychiatric disorders especially autism spectrum disorder (Lancaster et al., Reference Lancaster, Dietz, Moran and Pletnikov2007).

Mice

The results of experimental BoDV-1 infection in adult mice are strain specific (Rubin et al., Reference Rubin, Waltrip, Bautista and Carbone1993). It may result in non-symptomatic infection (Hallensleben et al., Reference Hallensleben, Schwemmle, Hausmann, Stitz, Volk, Pagenstecher and Staeheli1998) or fatal encephalitis depending on the strain of mice used (Hausmann et al., Reference Hausmann, Hallensleben, de la Torre, Pagenstecher, Zimmermann, Pircher and Staeheli1999). For example, 13% of infected C57BL/6 mice show mild transient symptoms. In contrast, 80% of infected MRL mice develop severe disease although the amounts of virus in their brains are comparable (Rubin et al., Reference Rubin, Waltrip, Bautista and Carbone1993). Intracerebral inoculation of BoDV-1 into β2-Microglobulin-deficient (CD8-) newborn mice of both strains does not result in disease (Hallensleben et al., Reference Hallensleben, Schwemmle, Hausmann, Stitz, Volk, Pagenstecher and Staeheli1998).

Ackermann et al. generated a mouse-adapted BoDV-1 that expressed green fluorescent protein, and then infected mice intracerebrally. By 28 days after challenge, labeled astrocytes were detected in the lower hippocampus (the subiculum). Eventually infected neurons were found throughout the hippocampus. The virus was found in Purkinje cells and in the inner granule layer of the cerebellum by day 65. The virus was also expressed in cerebral neurons and from day 65 onward was found in the neurons of the spinal cord. By day 120 the virus was detectable in the sciatic nerve (Ackermann et al., Reference Ackermann, Guelzow, Staeheli, Schneider and Heimrich2010). BoDV-1 infection of neonatal mice causes neurologic disease 4–6 weeks after intracerebral infection. The animals show abnormal hind leg positioning and eventual paraparesis (Narayan et al., Reference Narayan, Herzog, Frese, Scheefers and Rott1983a).

Gerbils

Experimental BoDV-1 infection of neonatal gerbils (Meriones unguiculatus), unlike neonatal rats, resulted in their death within 30 days. Virus was readily detected in their brains associated with the development of acute inflammatory lesions (Nakamura et al., Reference Nakamura, Nakaya, Hagiwara, Momiyama, Kagawa, Taniyama, Ishihara, Sata, Kurata and Ikuta1999). Despite severe symptoms and high levels of virus, there was no apparent neuronal loss (Watanabe et al., Reference Watanabe, Lee, Kamitani, Kobayashi, Taniyama, Tomonaga and Ikuta2001). In gerbils that had not sickened, the virus was detected in the cerebral cortex and the hippocampus. As disease progressed, viral expression increased in the lower brain stem and cerebellum, especially within Purkinje cells (Watanabe et al., Reference Watanabe, Lee, Kamitani, Kobayashi, Taniyama, Tomonaga and Ikuta2001). However while infected newborn gerbils developed fatal neurologic disease those infected 14 days after birth survived (Lee et al., Reference Lee, Watanabe, Kamitani, Baba, Yamashita, Kobayashi, Tomonaga and Ikuta2003). Very low levels of virus were detected in their brains. Additionally, neonatal gerbils treated with cyclosporine A were not protected against fatal disease. The cyclosporine did however prevent brain inflammation and significantly reduced brain cytokines (except interleukin (IL)-1β) (Watanabe et al., Reference Watanabe, Lee, Yamashita, Kamitani, Kobayashi, Tomonaga and Ikuta2003).

Other species

BoDV-1 has been used to induce experimental infections in many other mammals. The clinical disease and outcome vary between species. Rabbits, for example, develop a fatal paralytic disease similar to that seen in horses (Richt and Rott, Reference Richt and Rott2001); tree shrews (Tupaia glis) develop aberrant neurologic behaviors (Richt et al., Reference Richt, Vandewoude, Zink, Clements, Herzog, Stitz, Rott and Narayan1992); and rhesus monkeys develop severe paralytic disease with retinopathy (Stitz et al., Reference Stitz, Krey and Ludwig1981; Richt et al., Reference Richt, Vandewoude, Zink, Clements, Herzog, Stitz, Rott and Narayan1992). BoDV-1 has been detected in two dogs with neurologic symptoms (Weissenbock et al., Reference Weissenbock, Nowotny, Caplazi, Kolodziejek and Ehrensperger1998; Okamoto et al., Reference Okamoto, Kagawa, Kamitani, Hagiwara, Kirisawa, Iwai, Ikuta and Taniyama2002). Cattle can also develop Borna disease but it is uncommon and sporadic (Bode et al., Reference Bode, Durrwald and Ludwig1994; Caplazi et al., Reference Caplazi, Waldvogel, Stitz, Braun and Ehrensperger1994). Bornaviral encephalitis has also been recorded in captive alpacas and wild deer (Jacobsen et al., Reference Jacobsen, Algermissen, Schaudien, Venner, Herzog, Wentz, Hewicker-Trautwein, Baumgartner and Herden2010).

The pathogeneses of Borna diseases

When viruses invade the brain a consistent defensive response is mounted. Thus virus-infected cells are detected by resident microglia and recruited macrophages. These responding cells become activated and release a mixture of antiviral cytokines and chemokines (Russo and McGavern, Reference Russo and Mcgavern2015). These cytokines in turn, attract T cells, NK cells and other mononuclear cells to the site of invasion. Antigen presentation by the antigen-processing cells, release of cytokines and other mediators from microglia and astrocytes stimulates a type 1 antiviral T cell response (Hatalski et al., Reference Hatalski, Hickey and Lipkin1998). For many, but not all viruses, this T cell response is sufficient to eliminate the virus. This is not the case in Borna diseases.

Encephalitis

In BoDV-1 infected rats, the most severely affected brain regions include the olfactory bulb, the dentate gyrus, the caudate nucleus and the hippocampus as well as adjacent structures such as the mesencephalon, central gray matter, substantia nigra and hypothalamus (Tomonaga et al., Reference Tomonaga, Kobayashi and Ikuta2002). BoDV-1 thus exhibits a preferential tropism for the rodent limbic system (de la Torre, Reference de la Torre2002). Unfortunately, the viral cellular receptor(s) have yet to be identified, so the biochemical basis of this tropism is unknown.

BoDV-1 enters cells by receptor-mediated endocytosis (Gonzalez-Dunia et al., Reference Gonzalez-Dunia, Cubitt and de la Torre1998). The primary route of natural invasion in most mammals is most likely through the nasal epithelium (Morales et al., Reference Morales, Herzog, Kompter, Frese and Rott1988; Sauder and Staeheli, Reference Sauder and Staeheli2003). The olfactory bulbs of experimentally infected horses show inflammation and edema early in disease (Solbrig and Koob, Reference Solbrig and Koob2003). The virus replicates in the neurons at the initial entry site and then migrates intra-axonally towards the brain (Carbone et al., Reference Carbone, Duchala, Griffin, Kincaid and Narayan1987; Salinas et al., Reference Salinas, Schiavo and Kremer2010). The viral surface glycoprotein is required for cell to cell transfer (Bajramovic et al., Reference Bajramovic, Munter, Syan, Nehrbass, Brahic and Gonzalez-Dunia2003; Lennartz et al., Reference Lennartz, Bayer, Czerwonka, Lu, Kehr, Hirz, Steinmetzer, Garten and Herden2016). The viral RNP complex spreads by axonal and polysynaptic neuronal transmission. Once within the axon, intracellular microtubules transport the viral RNP (Clemente et al., Reference Clemente, Sisman, Aza-Blanc and de la Torre2010). The protein dynein forms motor complexes with BoDV-1 RNP and transport it from the nerve terminal to the cell body (Gosztonyi et al., Reference Gosztonyi, Dietzschold, Kao, Rupprecht, Ludwig and Koprowski1993; Clemente et al., Reference Clemente, Sisman, Aza-Blanc and de la Torre2010). Subsequently, RNP spreads to other cells such as astrocytes, oligodendroglia, ependymal cells and possibly Schwann cells. Eventually viral RNA can be detected in all peripheral nerves (Enbergs et al., Reference Enbergs, Vahlenkamp, Kipar and Muller2001). As inflammation develops, perivascular cuffs form. These cuffs contain CD4+ and CD8+ T cells and macrophages (Hatalski et al., Reference Hatalski, Hickey and Lipkin1998).

BoDV-1 is not cytotoxic in cultured cells (Stitz et al., Reference Stitz, Bilzer and Planz2002; Matsumoto et al., Reference Matsumoto, Hayashi, Omori, Honda, Daito, Horie, Ikuta, Fujino, Nakamura, Schneider, Chase, Yoshimori, Schwemmle and Tomonaga2012). The virus is strongly cell associated and produces very few infectious virions per cell even though large quantities of RNP are present (de la Torre, Reference de la Torre2002; Tomonaga et al., Reference Tomonaga, Kobayashi and Ikuta2002). Virions are not detected in infected brain either, suggesting that virus spreads as an RNP complex (Zimmermann et al., Reference Zimmermann, Breter, Rudolph and Ludwig1994). Transgenic mice expressing BoDV P-protein in their astrocytes show major behavioral abnormalities such as aggressiveness, hyperexcitability and special reference memory deficit resembling those seen in bornavirus-infected mice (Honda et al., Reference Honda, Fujino, Okuzaki, Ohtaki, Matsumoto, Horie, Daito, Itoh and Tomonaga2011). These behaviors are associated with alterations in the expression of genes associated with the transforming growth factor (TGF)-β pathway (Kamitani et al., Reference Kamitani, Shoya, Kobayashi, Watanabe, Lee, Zhang, Tomonaga and Ikuta2001; Nishino et al., Reference Nishino, Murakami and Funaba2015).

Wu et al. infected hippocampal slice cultures of several rat strains with BoDV-1 (Wu et al., Reference Wu, Schulz, Lin, Saar, Patone, Fischer, Hubner, Heimrich and Schwemmle2013). Cultures from some strains such as Lewis (LEW) showed disrupted architecture while others, such as Sprague Dawley (SD) did not. The efficacy of viral replication was however identical in cultures from different rat strains. These strain differences also occur in vivo. Media harvested from uninfected LEW or SD cultures could prevent BoDV-1-induced damage in LEW cultures. Infection with BoDV-1 reduced the availability of this inhibitory factor in LEW but not SD cultures. Genetic analysis indicated that a bornaviral resistance locus is present on rat chr6q16 and a susceptibility locus on chr3q21–23 (Wu et al., Reference Wu, Schulz, Lin, Saar, Patone, Fischer, Hubner, Heimrich and Schwemmle2013).

Retinitis

BoDV-1 spreads to the retina from the brain along the optic nerve. Krey et al. blocked the optic nerves of rabbits by xenon coagulation and then infected them intracerebrally with BoDV-1 (Krey et al., Reference Krey, Ludwig and Rott1979). Retinopathy did not develop, and viral antigen could not be detected in animals with blocked nerves. Blindness is regularly observed in equine Borna disease as a result of retinal degeneration with lymphoplasmacytic infiltration (Bilzer et al., Reference Bilzer, Grabner and Stitz1996). Lymphocytic infiltrates may also be observed within the optic nerve. There is a great diversity in its severity, and not all horses have detectable bornavirus in their retinas (Dietzel et al., Reference Dietzel, Kuhrt, Stahl, Kacza, Seeger, Weber, Uhlig, Reichenbach, Grosche and Pannicke2007).

Muller cells are retinal glial cells (Kacza et al., Reference Kacza, Vahlenkamp, Enbergs, Richt, Germer, Kuhrt, Reichenbach, Muller, Herden, Stahl and Seeger2000, Reference Kacza, Mohr, Pannicke, Kuhrt, Dietzel, Fluss, Richt, Vahlenkamp, Stahl, Reichenbach and Seeger2001). The neuron:Muller cell ratio in the retina is significantly reduced in diseased horses as compared with controls. This appears to be due to a concomitant loss of neurons and an increase in glia. The neuronal degeneration begins in the outer retinal layer where the photoreceptors are located but all retinal layers show reduced thickness (Kacza et al., Reference Kacza, Vahlenkamp, Enbergs, Richt, Germer, Kuhrt, Reichenbach, Muller, Herden, Stahl and Seeger2000).

Neurons are also lost in the retinas of BoDV-1 infected rats and rabbits (Narayan et al., Reference Narayan, Herzog, Frese, Scheefers and Rott1983b). Four weeks after intracerebral inoculation of rats there is a significant thinning of the retina due to a loss of photoreceptor segments and ganglion cells. At the same time, there is a great increase in the number of glial cells in the ganglion cell and inner plexiform layers (Kacza et al., Reference Kacza, Vahlenkamp, Enbergs, Richt, Germer, Kuhrt, Reichenbach, Muller, Herden, Stahl and Seeger2000). Microglia and macrophages are involved in the neuronophagocytosis that accompanies this neurodegeneration (Kacza et al., Reference Kacza, Vahlenkamp, Enbergs, Richt, Germer, Kuhrt, Reichenbach, Muller, Herden, Stahl and Seeger2000). Muller cells show moderate changes (Iandiev et al., Reference Iandiev, Biedermann, Bringmann, Reichel, Reichenbach and Pannicke2006). Kacza et al. infected Lewis rats with BoDV-1 intracerebrally (Kacza et al., Reference Kacza, Mohr, Pannicke, Kuhrt, Dietzel, Fluss, Richt, Vahlenkamp, Stahl, Reichenbach and Seeger2001). Within months their retinal thickness had declined to a third of that in control animals. Photoreceptor segments were completely destroyed and the number of neurons reduced. There were many active microglia and macrophages undertaking neuronophagocytosis. Muller cells showed signs of gliosis, alterations in glutamate synthetase, altered K+ currents and thickened stem processes (Kacza et al., Reference Kacza, Mohr, Pannicke, Kuhrt, Dietzel, Fluss, Richt, Vahlenkamp, Stahl, Reichenbach and Seeger2001).

Narayan et al. attributed bornaviral retinitis to a transient attack by cytotoxic T cells (Narayan et al., Reference Narayan, Herzog, Frese, Scheefers and Rott1983b). For example, Krey et al. treated BoDV-1 infected rabbits with immunosuppressive drugs that delayed the onset of retinitis (Krey et al., Reference Krey, Ludwig and Gierend1981). The treatment reduced the confluency of the retinal lesions and some treated animals either lacked eye lesions or showed nonprogression. Subsequently, Stahl et al. showed that the retinal T cell infiltration consisted of αβTCR+, CD4+, CD8+ cells (Stahl et al., Reference Stahl, Mohr, Kacza, Reimers, Pannicke, Sauder, Reichenbach and Seeger2003). B cells were rarely found by Stahl et al. but Hatalski et al. found them to be plentiful (Hatalski et al., Reference Hatalski, Hickey and Lipkin1998). This may reflect the use of different reagents. Cytokine transcripts in affected retinas included raised IL-1β, IL-6, IFNγ and tumor necrosis factor (TNF)-α, as well as CXCL10, chemokine ligand (CCL)2 and CCL4. By day 36 the levels of these transcripts had returned to normal. These cytokines were probably derived from the infiltrating T cells and NK cells (Sauder and de la Torre, Reference Sauder and de la Torre1999). Chronic bornaviral infections may result from a switch in the brain-infiltrating T cells from generating a Th1 response to a Th2 response (Hatalski et al., Reference Hatalski, Hickey and Lipkin1998).

Alterations in neurogenesis

In the developing rodent brain, neurogenesis occurs predominantly in the subventricular zone of the lateral ventricles and the subgranular zone of the dentate gyrus in the hippocampus (Katsumoto et al., Reference Katsumoto, Lu, Miranda and Ransohoff2014). It is unclear how neuronal loss occurs in persistently infected neonatal rats but it is probably due to induced apoptosis (Hornig et al., Reference Hornig, Weissenbock, Horscroft and Lipkin1999; Zocher et al., Reference Zocher, Czub, Schulte-Monting, de la Torre and Sauder2000; Ovanesov et al., Reference Ovanesov, Moldovan, Smith, Vogel and Pletnikov2008b). Damage to synaptic structures precedes neuronal loss in persistently infected rats and this may impair trafficking of growth factors (Volmer et al., Reference Volmer, Prat, Le Masson, Garenne and Gonzalez-Dunia2007). Bornaviral-induced disturbances in the neurotrophin system also contribute to this neurodegeneration. Thus Zocher et al. found reduced levels of neurotrophin 3, brain-derived neurotropic factor (BDNF) and nerve growth factor in the hippocampus 14-days post-infection in newborn rats and in the cerebellum by 21 days post-infection (Zocher et al., Reference Zocher, Czub, Schulte-Monting, de la Torre and Sauder2000) (Fig. 3). They also detected reduced levels of neurotrophin receptors in both the hippocampus and cerebellum. Neonatal persistently-infected rats fail to gain weight but lack of nourishment does not account for the brain lesions or the lack of weight gain (Dietz and Pletnikov, Reference Dietz and Pletnikov2003). There are increased concentrations of serotonin in the hippocampus of bornavirus- infected neonates (Dietz and Pletnikov, Reference Dietz and Pletnikov2003). Hans et al. examined the effect of bornaviral infection on the response of hippocampal neurons to neurotrophin BDNF. Persistent infection blocked BDNF-induced extracellular signal-regulated kinases-1/2 (ERK) phosphorylation even although the expression of the BDNF receptor was normal (Hans et al., Reference Hans, Bajramovic, Syan, Perret, Dunia, Brahic and Gonzalez-Dunia2004). As a result, BDNF-induced expression of synaptic vesicle proteins is blocked, potentially causing defective synaptic organization. Persistently-infected newborn rats show a progressive decline in the expression of synaptic markers (growth-associated protein 43, a presynaptic membrane phosphoprotein and synaptophysin, a calcium-binding protein found in presynaptic vesicles) followed by a loss of up to 30% of cortical neurons (Gonzalez-Dunia et al., Reference Gonzalez-Dunia, Watanabe, Syan, Mallory, Masliah and de la Torre2000). This decline in synaptic density and neuronal plasticity occurs primarily within the cerebrum and the hippocampus.

Obesity

Some adult rats that survive acute BoDV-1 disease begin to overeat and as a result, become obese ((Narayan et al., Reference Narayan, Herzog, Frese, Scheefers and Rott1983a; Wensman et al., Reference Wensman, Jaderlund, Holst and Berg2014). Obesity also develops in some persistently infected neonatal rats (Lyons et al., Reference Lyons, Nagashima and Zabriskie2002). This uncontrolled appetite may result from damage to hunger control centers in the brain (Nagashima et al., Reference Nagashima, Zabriskie and Lyons1992; Gosztonyi and Ludwig, Reference Gosztonyi and Ludwig1995). The neonatal rats develop inflammation in their pituitary stalk (Gosztonyi and Ludwig, Reference Gosztonyi and Ludwig1995). As the obesity syndrome develops, the number of virus-infected cells grows and leads to progressive involution of the hypothalamus as well as vacuolar degeneration of neurons in the hypothalamic paraventricular nucleus. Herden et al. compared the brain lesions of a pathogenic BoDV-1 strain with that of an obesity-inducing strain (Herden et al., Reference Herden, Herzog, Richt, Nesseler, Christ, Failing and Frese2000). The obesity-inducing-strain lesions were restricted to the septum, hippocampus, ventromedial hypothalamus and amygdala. Herden et al. also examined the levels of neuropeptides in BoDV-1-infected brains and found that expression of melanocyte-stimulating hormone (MSH) was reduced in infected animals (Herden et al., Reference Herden, Herzog, Richt, Nesseler, Christ, Failing and Frese2000; Herden et al., Reference Herden, Schluesener and Richt2005). α-MSH reduces appetite so its deficiency may have an opposite effect and bornaviral obesity could therefore be due to lesions in the melanocortin feeding center within the hypothalamus.

Pathogenic processes

Three major pathogenic processes collectively cause the neuronal damage associated with bornaviral encephalitis (Fig. 4). Initially, the virus replicates almost exclusively in neurons (Gosztonyi and Ludwig, Reference Gosztonyi and Ludwig1995). This triggers immune-mediated attack by virus specific CD8+ T cells directed against viral antigens such as bornaviral N-protein or autoimmune attack against neoantigens on the neuronal surface (Amor et al., Reference Amor, Peferoen, Vogel, Breur, Van Der Valk, Baker, Van and ORT2014). In the later stages of infection, the virus infects microglia and astrocytes (Carbone et al., Reference Carbone, Moench and Lipkin1991; Gosztonyi and Ludwig, Reference Gosztonyi and Ludwig1995). These plus the results of neuronal destruction leads to the prolonged activation of microglia and a consequent increase in tissue cytokines and reactive nitrogen species (Gonzalez et al., Reference Gonzalez, Elgueta, Montoya and Pacheco2014). Thirdly, astrocyte dysfunction due to viral invasion disrupts glutamate regulation. Excess glutamate generated as a result kills neurons through a process called excitotoxicity.

Fig. 4. The three major mechanisms of neuronal destruction mediated by BoDV-1. These are, T-cell-mediated cytotoxicity, microglial activation resulting in cytokine-mediated neurotoxicity, and loss of astrocyte function resulting in glutamate accumulation and excitotoxic destructions of neurons.

Immune-mediated T cell attack

The primary mechanism of neuronal damage in bornaviral encephalitis is T cell-mediated neuronal cytotoxicity. These T cells are activated by viral infection and as a result, mount a type 1 response characterized by the production of both IFN-γ and TNF-α (Baruch and Schwartz, Reference Baruch and Schwartz2013). The neurotransmitters glutamine and acetylcholine also favor T cell activation (Pacheco et al., Reference Pacheco, Riquelme and Kalergis2010; Pikor et al., Reference Pikor, Astarita, Summers-Deluca, Galicia, Qu, Ward, Armstrong, Dominguez, Malhotra, Heiden, Kay, Castanov, Touil, Boon, O'Connor, Bar-Or, Prat, Ramaglia, Ludwin, Turley and Gommerman2015) as do activated Th17 cells (Nouri et al., Reference Nouri, Bredberg, Westrom and Lavasani2014).

In bornaviral lesions, the perivascular cellular infiltrates predominantly consist of CD4+ T cells while CD8+ cells predominate within the brain parenchyma. The development of neuronal lesions is specifically associated with invasion by CD8+ cells (Sobbe et al., Reference Sobbe, Bilzer, Gommel, Noske, Planz and Stitz1997). The importance of these T cell infiltrates can be demonstrated by adoptive transfer of lymphocytes from infected to uninfected rats. For example, Narayan et al. could transmit the disease using spleen cells from 4-week-old rats transferred into cyclosporine-treated recipients (Narayan et al., Reference Narayan, Herzog, Frese, Scheefers and Rott1983a). Rott et al. were able to establish a virus-specific, CD4+ T cell line that induced typical bornaviral lesions when administered to recipient rats of the corresponding MHC class II haplotype (Rott et al., Reference Rott, Herzog, Richt and Stitz1988). Sobbe et al. induced typical bornaviral brain lesions in rats by adoptive transfer of CD8+ brain T cells (Sobbe et al., Reference Sobbe, Bilzer, Gommel, Noske, Planz and Stitz1997). Additionally, lymphocytes obtained from rats early in bornaviral infection could transfer the disease but not late in infection implying that the cytotoxic cells were generated early in the disease process (Narayan et al., Reference Narayan, Herzog, Frese, Scheefers and Rott1983a). Planz et al. identified a peptide, ASYAQMTTY, from BoDV-1 nucleoprotein that was recognized by CD8+ T cells in association with the rat MHC class I molecule, RT1.A, and, as a result, triggered T cell cytotoxicity (Planz et al., Reference Planz, Dumrese, Hulpusch, Schirle, Stevanovic and Stitz2001). Transgenic mice expressing the nucleoprotein are resistant to disease, presumably because they are immunologically tolerant to it (Schwemmle et al., Reference Schwemmle, Salvatore, Shi, Richt, Lee and Lipkin1998). Lymphocytes from the brains of rats with acute bornaviral disease show MHC class I restricted cytotoxic T cell activity. MHC class I expression occurs on astrocytes and on some neurons in BoDV-1-infected brains. MHC class I production by neurons is normally minimal but can be induced (Planz et al., Reference Planz, Bilzer, Sobbe and Stitz1993).

Support for the essential role of T cells is also provided by immunosuppressive studies. For example, Rott et al. demonstrated that T cell elimination prevented inflammation and the development of clinical disease. A single intraperitoneal injection of cyclophosphamide given to adult rats prevented the development of bornaviral encephalitis and clinical disease. Stitz et al. were able to prevent the development of bornaviral disease by administering cyclosporine prior to infection (Stitz et al., Reference Stitz, Soeder, Deschl, Frese and Rott1989). Stitz et al. also treated bornavirus-infected adult rats with monoclonal antibodies against N-protein-specific CD4+ and CD8+ T cells (Stitz et al., Reference Stitz, Sobbe and Bilzer1992). Both types of antibody suppressed the encephalitis but anti-CD8 was more effective than anti-CD4. These monoclonal antibodies worked best if given before or shortly after infection and they did not prevent encephalitis or disease when given more than 4 days after infection. BoDV-1 infection will not result in disease in athymic nude rats (Herzog et al., Reference Herzog, Wonigeit, Frese, Hedrich and Rott1985). Thus the lesions of bornaviral encephalitis in rats are mediated primarily by N-protein-specific CD8+ T cells.

In persistently infected neonatal rats, unlike adult rats, T cells may be protective. Thus persistent BoDV-1 infection could be prevented by prior administration of a virus-specific CD4+ T cell line. Recipient rats developed a transient, mild encephalitis that lasted for only a few days. This T cell line had no cytotoxic properties but virus clearance was accompanied by the appearance of CD8+ cytotoxic T cells in the recipients (Noske et al., Reference Noske, Bilzer, Planz and Stitz1998). It is interesting to note however, that in adult rats infected with BoDV-1, the virus is found exclusively in the brain. In cyclosporine-treated rats the virus can be detected in peripheral nerve fibers and adjacent tissues such as lung, liver and spleen (Stitz et al., Reference Stitz, Schilken and Frese1991). This suggests that T cells are responsible for containing BoDV-1 within the central nervous system (CNS). In subsequent studies, Stitz et al. examined the role of antibodies in restricting BoDV-1 to the CNS. Immune serum transfer into cyclosporine-treated or newborn rats resulted in restriction of the virus to the CNS (Stitz et al., Reference Stitz, Noske, Planz, Furrer, Lipkin and Bilzer1998).

Hatalski et al. followed the evolution of the immune response in the brain of BoDV-1-infected Lewis rats (Hatalski et al., Reference Hatalski, Hickey and Lipkin1998). Thus at the peak of the acute infection, the perivascular infiltrates contained both CD4+ and CD8+ T cells as well as a significant population of NK cells. The NK cells could be identified in the brain lesions 3.5 weeks post-infection before the onset of clinical disease. As the disease progressed and became chronic, the numbers of all cell types dropped and the authors suggested that this reflected a switch from a type 1 to a type 2 immune response. This suggestion was supported by a rise in serum IgE levels. Hatalski et al. also measured cytokine mRNA expression in infected brains. They showed a significant increase in the levels of the proinflammatory cytokines, IL-1α, IL-2, IL-6, TNF-α and IFN-γ that peaked at 5 weeks post-infection when acute inflammation was maximal. On the other hand, the anti-inflammatory cytokine, IL-4 increased to reach maximal levels at the end of the study at 15 weeks. The regulatory cytokine, TGF-β in contrast, peaked at 5 weeks and then stabilized (Hatalski et al., Reference Hatalski, Hickey and Lipkin1998). It is possible that the decline in inflammation seen in long-term bornaviral infections may also be due to the development of neuroprotective Treg cells (Walsh et al., Reference Walsh, Watson and Kipnis2014). Nishino et al. investigated this upregulation of TGF-β in BoDV-1-infected rats. They found that signal receptors for TGF-β1 were also upregulated as were inhibitin/activin βC, two components of the TGF-β pathway (Nishino et al., Reference Nishino, Ooishi, Kurokawa, Fujino, Murakami, Madarame, Hashimoto, Sugiyama and Funaba2009). This may reflect the ongoing immunosuppressive effects of bornaviral P-protein (Nishino et al., Reference Nishino, Murakami and Funaba2015).

Microglia and the role of chronic inflammation

The second important pathogenic process in bornaviral disease results from prolonged activation of microglia (Weissenbock et al., Reference Weissenbock, Hornig, Hickey and Lipkin2000; Ovanesov et al., Reference Ovanesov, Sauder, Rubin, Richt, Nath, Carbone and Pletnikov2006, Reference Ovanesov, Vogel, Moran and Pletnikov2007, Reference Ovanesov, Ayhan, Wolbert, Moldovan, Sauder and Pletnikov2008a, Reference Ovanesov, Moldovan, Smith, Vogel and Pletnikovb). In the healthy brain, microglia remodel neuronal synapses and secrete neurotrophic proteins that help maintain effective neuronal network functions (Zocher et al., Reference Zocher, Czub, Schulte-Monting, de la Torre and Sauder2000). When activated however, microglia flood the brain with the inflammatory cytokines, TNF-α, IFN-β and IL-10 as well as induced nitric oxide synthase (iNOS), and reactive oxygen and nitrogen species (ROS and RNS) (Gonzalez et al., Reference Gonzalez, Elgueta, Montoya and Pacheco2014). These molecules induce neuronal dysfunction and death (Heneka et al., Reference Heneka, Kummer and Latz2014; Papageorgiou et al., Reference Papageorgiou, Lewen, Galow, Cesetti, Scheffel, Regen, Hanisch and Kann2015). Zheng et al. have demonstrated that the severity of the neurologic signs and of the corresponding encephalitis correlated well with iNOS and cNOS mRNA expression in rat bornaviral disease and that the distribution of iNOS-positive cells in the basolateral cortex and the hippocampus correlated with sites of BoDV-1 infected cells (Zheng et al., Reference Zheng, Schafer, Weihe, Sheng, Corisdeo, Fu, Koprowski and Dietzschold1993). This cannot however be the whole explanation for bornaviral pathogenicity since Hausmann et al. demonstrated that BoDV-1 caused neurologic disease in mice that lacked IFN-γ, Fas, iNOS or the chemokine receptor, CXCR3 (Hausmann et al., Reference Hausmann, Sauder, Wasmer, Lu and Staeheli2004).

Microglia are activated by BoDV-1 (Plata-Salaman et al., Reference Plata-Salaman, Ilyin, Gayle, Romanovitch and Carbone1999; Ovanesov et al., Reference Ovanesov, Sauder, Rubin, Richt, Nath, Carbone and Pletnikov2006) (Fig. 5). In BoDV-1-infected adult rats, these activated microglia can be detected in the dentate gyrus at 10 days post-infection but detectable loss of granule cells is not seen until 30 days. Thus the virus activates the microglia long before neuronal loss and it is therefore unlikely that the activated microglia alone trigger neuronal loss and dysfunction (Ovanesov et al., Reference Ovanesov, Moldovan, Smith, Vogel and Pletnikov2008b). Nevertheless, increased levels of brain IL-6, TNF-α and iNOS mRNAs do correlate with the severity of the inflammatory lesions in infected brains (de la Torre, Reference de la Torre2002).

Fig. 5. The role of microglial activation in the pathogenesis of bornaviral encephalitis. M1 activation of the microglia results in the flooding of the brain with multiple cytokines as well as potent oxidants resulting in neuronal death. Should the microglial phenotype change to M2, the resulting cytokines will reduce inflammation and cell destruction and promote repair although destroyed neurons unlikely to be replaced.

Classically activated microglia (M1 cells) have two alternative fates. They may differentiate into regulatory cells (M2 cells) and so reduce inflammation and promote tissue repair or alternatively, they can undergo uncontrolled activation and trigger chronic inflammation resulting in the production of neurotoxic factors and progressive neural loss (Gonzalez et al., Reference Gonzalez, Elgueta, Montoya and Pacheco2014). M2 activation of microglia enhances their release of neurotrophic factors, proteases, IL-4, TGFβ and arginase 1 and stimulates their phagocytic activity. M2 activation thus reduces inflammation and promotes tissue repair. The anti-inflammatory cytokines, IL-4 and TGFβ are produced during the later stages of bornaviral encephalitis (Hatalski et al., Reference Hatalski, Hickey and Lipkin1998). While they are mainly derived from T cells, they may also be produced by microglia (Heneka et al., Reference Heneka, Kummer and Latz2014).

Morimoto et al. used dexamethasone to inhibit bornavirus-induced inflammation in rats. They suggested that bornaviral disease involved an early inflammatory reaction mediated by resident microglia leading to sensitization (antigen processing and presentation), and an influx of primed T cells. Restimulation of these infiltrating T cells led to the local production of a cytokine mixture amplifying the reaction. Subsequently, recruitment and activation of microglia continued this process. Morimoto et al. suggested that the initial expression of pro-inflammatory cytokines was directly mediated by microglial BoDV-1 (Morimoto et al., Reference Morimoto, Hooper, Bornhorst, Corisdeo, Bette, Fu, Schafer, Koprowski, Weihe and Dietzschold1996).

Astrocytes and glutamate toxicity

The third major mechanism of bornaviral-induced neuronal destruction results from astrocyte-mediated disturbances in glutamate levels.

Astrocytes are the major microglial cell population in the brain (Rossi and Volterra, Reference Rossi and Volterra2009). Under normal conditions, they supply glucose to neurons and regulate the composition of extracellular fluid. Importantly, they remove excess potassium ions and neurotransmitters, especially glutamate (Coulter and Eid, Reference Coulter and Eid2012). Astrocytes take up extracellular glutamate, transform this into glutamine using glutamine synthetase and subsequently shuttle the glutamine back to neurons using transporters. Between 80 and 90% of extracellular glutamate uptake in the brain is through these astrocytic glutamine transporters (Vesce et al., Reference Vesce, Rossi, Brambilla and Volterra2007; Coulter and Eid, Reference Coulter and Eid2012) (Fig. 6). Once within neurons glutamine is reconverted to glutamate. This glutamate may act as a neurotransmitter directly or it is decarboxylated to form γ-amino butyric acid (GABA), which also acts as a neurotransmitter.

Fig. 6. The role of glutamate in BoDV-1 encephalitis. The normal glutamate cycle requires that astrocytes remove excess glutamate from the extracellular fluid, convert it to glutamine and return it to the neurons. If astrocytes are damaged glutamate accumulates and kills neurons. GABAergic neurons release GABA that inhibits glutamate excitotoxicity and microglial activation. If these neurons are also damaged by BoDV-1 then additional neuronal loss will be expected.

Excitotoxicity

Excitatory synaptic transmission in the brain is mainly mediated by glutamate (Bondy and Purdy, Reference Bondy and Purdy1977; Choi, Reference Choi1988). If the extracellular concentration of glutamate is excessive, neurons will be damaged. This process is called excitotoxicity and the excessive production of glutamate within the brain is called a glutaminergic storm. Neurons die as a result of excessive influx of calcium through glutamate receptor channels leading to mitochondrial damage and the production of ROS and RNS (Gudino-Cabrera et al., Reference Gudino-Cabrera, Urena-Guerrero, Rivera-Cervantes, Feria-Velasco and Beas-Zarate2014). Excitotoxicity has been associated with neurodegeneration (Jacobs et al., Reference Jacobs, Aden, Mathisen, Khuong, Gaarder, Loberg, Lomo, Maehlen and Paulsen2006). Excessive glutamate may also impair the blood-brain barrier (Gudino-Cabrera et al., Reference Gudino-Cabrera, Urena-Guerrero, Rivera-Cervantes, Feria-Velasco and Beas-Zarate2014).

In the healthy brain, homeostasis is maintained by the removal of excess glutamate by astrocytes. If these astrocytes are lost and replaced by microglia, the glutamine-glutamate cycle is down-regulated (Schwartz et al., Reference Schwartz, Shaked, Fisher, Mizrahi and Schori2003). TNF-α upregulates the production of glutamate in activated microglia and at the same time, inhibits glutamate uptake by astrocytes (Takeuchi et al., Reference Takeuchi, Jin, Wang, Zhang, Kawanokuchi, Kuno, Sonobe, Mizuno and Suzumura2006).

Billaud et al. showed that BoDV-1 inhibits glutamate uptake by feline primary cortical astrocytes (Billaud et al., Reference Billaud, Ly, Phillips and de la Torre2000; de la Torre, Reference de la Torre2002). This failure to remove glutamate could result in neuronal excitotoxicity. This is supported by the studies of Ovanesov et al. who measured extracellular glutamate in the striatum of Fischer 344 rats (Ovanesov et al., Reference Ovanesov, Vogel, Moran and Pletnikov2007). BoDV-1 infection increased the extracellular levels of glutamate. This elevated extracellular glutamate was associated with reduced neuron numbers and volume in the striatum.

Bergmann glial cells are astrocytes that surround Purkinje cells where they sequester neuronally-produced glutamate. Purkinje cells disappear from the cerebellum of BoDV-1-infected neonatal mice although they themselves may not be infected. The Bergmann glia are however infected and it is possible that damage to these cells could result in secondary loss of Purkinje cells (Bordey and Sontheimer, Reference Bordey and Sontheimer2003).

On the other hand, Richter et al. showed that two glutamate receptor antagonists failed to prevent bornaviral-induced neuronal loss in IFNγ-deficient mice. While there was a trend towards protection it was not statistically significant. Richter et al. also demonstrated that neither the frequency of virus-infected cells, nor the composition of the T cell infiltrate was altered by these antagonists and they concluded that glutamate excitotoxicity was an unlikely cause of neuronal damage (Richter et al., Reference Richter, Hausmann and Staeheli2009). They did not however exclude toxic effects mediated through the third glutamate receptor – kainite. The kainite receptor (KA-1) has been suggested to be a BoDV-1 receptor or target (Gosztonyi, Reference Gosztonyi2008).

Zhang et al. analyzed metabolites in bornavirus-infected areas of the equine hippocampus (Zhang et al., Reference Zhang, Lei, Liu, Wang, Liu, Li, Zheng, Zhang, Chen and Xie2014). They found that infected tissues had lower levels of D-myo-inositol-1-phosphate, glutamate, phosphoethanolamine, heptadecanoic acid and linoleic acid but higher levels of ammonia. These results also confirm that there are disturbances in glutamate metabolism within these tissues.

Effects of glutamate on microglia

Glutamate release by astrocytes is influenced by inflammatory mediators such as TNF-α and prostaglandins. Thus inflammation may disrupt astrocyte-neuronal interactions (Ovanesov et al., Reference Ovanesov, Ayhan, Wolbert, Moldovan, Sauder and Pletnikov2008a; Rossi and Volterra, Reference Rossi and Volterra2009). Activation of microglia by BoDV-1 in vitro requires the presence of astrocytes (Ovanesov et al., Reference Ovanesov, Ayhan, Wolbert, Moldovan, Sauder and Pletnikov2008a). Activated microglia also release large amounts of glutamate in response to TNF-α by upregulating glutaminase (Takeuchi et al., Reference Takeuchi, Jin, Wang, Zhang, Kawanokuchi, Kuno, Sonobe, Mizuno and Suzumura2006).

Effects of glutamate on T cell function

T cells have receptors for glutamate and glutamate enhances T cell adhesion, chemotactic migration and proliferation (Schwartz et al., Reference Schwartz, Shaked, Fisher, Mizrahi and Schori2003; Levite, Reference Levite2008). Glutamate also protects them against antigen-induced apoptosis (Ganor and Levite, Reference Ganor and Levite2014). Thus glutamate alters surface receptor expression, and may enhance T cell cytotoxicity. It is possible therefore that elevated glutamate in sites of bornaviral invasion might attract T cells and promote T-cell-mediated brain damage. It may be relevant to note that many neurotransmitters other than glutamate also activate T cells (Levite, Reference Levite2008). For example, resting T cells can also be activated by dopamine, serotonin and some neuropeptides

The role of gamma-aminobutyric acid

Gamma-aminobutyric acid (GABA) is an inhibitory neurotransmitter synthesized from glutamate using L- glutamic acid decarboxylase. GABA reduces neuronal excitability by acting at inhibitory synapses. GABA also acts as a growth factor for the developing brain where it stimulates neuronal branching (Chen and Kriegstein, Reference Chen and Kriegstein2015). Microglia possess GABA receptors and the GABAergic system can regulate microgial activities. These microglia in turn may regulate GABAergic transmission by neighboring neurons. T cells and dendritic cells contain glutamic acid decarboxylase and express GABA receptors. GABA also suppresses both proinflammatory cytokine production and immune cell proliferation. Thus an increase in GABA may suppress T cell function and inflammation (Peng et al., Reference Peng, Yan, Zhu, Wang, Yan, Lu, Li, Hu, Wei, Mu, Chen, Feng, Gong, Wu, Zhang, Zhang, Zhu and Wu2008).

Bornaviral P-protein binds directly to GABA-receptor-associated protein (Peng et al., Reference Peng, Yan, Zhu, Wang, Yan, Lu, Li, Hu, Wei, Mu, Chen, Feng, Gong, Wu, Zhang, Zhang, Zhu and Wu2008). It is then transported to the cell nuclei where it disrupts the trafficking of GABA-receptors (Scordel et al., Reference Scordel, Huttin, Cochet-Bernoin, Szelechowski, Poulet, Richardson, Benchoua, Gonzalez-Dunia, Eloit and Coulpier2015). The P-protein also reduces neurogenesis, and specifically targets GABAergic neurogenesis. This results from inhibition of the production of pro-neuronal factors such as ApoE, Noggin, TH and Scg10/Strathmin2. BoDV-1 can disrupt the GABA-glutamate cycle reducing its inhibitory effect and thus enhancing excitotoxicity and T cell activity (Fig. 6). Neonatal rats persistently infected with BoDV-1 undergo a selective loss of GABAergic cortical neurons (Bautista et al., Reference Bautista, Rubin, Moran, Schwartz and Carbone1995; Eisenman et al., Reference Eisenman, Brothers, Tran, Kean, Dickson, Dietzschold and Hooper1999; Gonzalez-Dunia et al., Reference Gonzalez-Dunia, Watanabe, Syan, Mallory, Masliah and de la Torre2000; Pletnikov et al., Reference Pletnikov, Rubin, Vogel, Moran and Carbone2002). It is perhaps no coincidence that aberrations in GABAergic neurogenesis have been associated with human neuropsychiatric disorders (Scordel et al., Reference Scordel, Huttin, Cochet-Bernoin, Szelechowski, Poulet, Richardson, Benchoua, Gonzalez-Dunia, Eloit and Coulpier2015).

Other effects on neurotransmission

Bornaviral encephalitis in Lewis rats is associated with a decline in the cholinergic activity of the brain (Gies et al., Reference Gies, Bilzer, Stitz and Staiger1998, Reference Gies, Gorcs, Mulder, Planz, Stitz, Bilzer, Luiten and Harkany2001). This decline appears to be due to a loss of choline acetyltransferase activity in the cerebral cortex and hippocampus and a reduction in acetylcholinesterase in these regions. These declines parallel the loss of neurons in these areas. The decline in choline acetyltransferase occurs in the pre-encephalitic stage of bornaviral infection prior to T cell infiltration into the brains (Gies et al., Reference Gies, Bilzer, Stitz and Staiger1998).

BoDV-1 also affects the dopamine system. Neonatal rats experimentally infected with BoDV-1 develop a hyperactive movement disorder (Solbrig et al., Reference Solbrig, Koob, Joyce and Lipkin1996). Since locomotor activity is regulated through the dopamine system, Solbrig et al. examined the dopamine receptors in these rats (Solbrig et al., Reference Solbrig, Koob, Joyce and Lipkin1996). They found that there was reduced binding to several dopamine receptors in the nucleus accumbens. The nucleus accumbens is the site where the limbic system and motor information interact. It plays a role in motivational, appetite and locomotor behaviors. Bornaviral-induced alterations have also been recorded in other neurotransmitters such as cholecystokinin and somatostatin (de la Torre, Reference de la Torre2002).

Conclusions

The diversity of clinical manifestations of BoDV-1 infections reflect its complex pathogenicity. Even the development of acute encephalitis reflects the results of multiple disturbances in many neuronal and immune pathways. There appear to be three primary mechanisms involved in this pathology, namely T cell cytotoxicity; microglial release of inflammatory cytokines and ROS; and astrocyte-induced disturbances in glutaminergic signaling. It is unlikely however that these are the only important pathways involved in the disease process. Thus it is recognized that bornaviruses disrupt the NF-kB pathway (Makino et al., Reference Makino, Fujino, Parrish, Honda and Tomonaga2015), the RIG-1/MAVS pathway (Reuter et al., Reference Reuter, Ackermann, Kothlow, Rinder, Kaspers and Staeheli2010) as well as HMGB-1 signaling (Kamitani et al., Reference Kamitani, Shoya, Kobayashi, Watanabe, Lee, Zhang, Tomonaga and Ikuta2001) and can cause epigenetic changes (Liu et al., Reference Liu, Liu, Bode, Liu, Zhang, Wang, Li, Lei, Peng, Cheng and Xie2015). These effects collectively permit the virus to survive innate immune attack and contribute to the disease process. Even in the absence of acute encephalitis BoDV-1 has a profound effect on neurodevelopmental pathways in neonatal rats. This may be of relevance to neurodevelopmental disease in humans.

During the 1990s, interest in BoDV-1 peaked as a result of assertions that it was an unusually common infection in humans with neuropsychiatric disorders. This claim has been refuted. Nevertheless, given the diversity of molecular and epigenetic changes mediated by bornaviruses in mammalian brains, it is conceivable that they can induce alterations in brain function well short of the lethal encephalitis observed in squirrel breeders. It is perhaps time to reopen the investigation into possible links between bornaviral infection and mental illness.

References

Ackermann, A, Guelzow, T, Staeheli, P, Schneider, U and Heimrich, B (2010). Visualizing viral dissemination in the mouse nervous system, using a green fluorescent protein-expressing Borna disease virus vector. Journal of Virology 84: 54385442.Google Scholar
Amor, S, Peferoen, LA, Vogel, DY, Breur, M, Van Der Valk, P, Baker, D and Van, NO ORT, JM (2014). Inflammation in neurodegenerative diseases–an update. Immunology 142: 151166.Google Scholar
Amsterdam, JD, Winokur, A, Dyson, W, Herzog, S, Gonzalez, F, Rott, R and Koprowski, H (1985). Borna disease virus. A possible etiologic factor in human affective disorders? Archives of General Psychiatry 42: 10931096.Google Scholar
Ashash, E, Malkinson, M, Meir, R, Perl, S and Weisman, Y (1996). Causes of losses including a Borna disease paralytic syndrome affecting young ostriches of one breeding organization over a five-year period (1989–1993). Avian Diseases 40: 240245.Google Scholar
Bajramovic, JJ, Munter, S, Syan, S, Nehrbass, U, Brahic, M and Gonzalez-Dunia, D (2003). Borna disease virus glycoprotein is required for viral dissemination in neurons. Journal of Virology 77: 1222212231.Google Scholar
Baruch, K and Schwartz, M (2013). CNS-specific T cells shape brain function via the choroid plexus. “Brain, Behavior, and Immunity” 34: 1116.Google Scholar
Bautista, JR, Rubin, SA, Moran, TH, Schwartz, GJ and Carbone, KM (1995). Developmental injury to the cerebellum following perinatal Borna disease virus infection. Brain Research Developmental Brain Research 90: 4553.Google Scholar
Berg, AL, Johannisson, A, Johansson, M, Hein, A, Berg, M and Dorries, R (1999). Peripheral and intracerebral T cell immune response in cats naturally infected with Borna disease virus. Veterinary Immunology and Immunopathology 68: 241253.Google Scholar
Berg, M, Johansson, M, Montell, H and Berg, AL (2001). Wild birds as a possible natural reservoir of Borna disease virus. Epidemiology and Infection 127: 173178.Google Scholar
Billaud, JN, Ly, C, Phillips, TR and de la Torre, JC (2000). Borna disease virus persistence causes inhibition of glutamate uptake by feline primary cortical astrocytes. Journal of Virology 74: 1043810446.Google Scholar
Bilzer, T, Grabner, A and Stitz, L (1996). Immunopathology of Borna disease in the horse: clinical, virological and neuropathologic findings. Tierarztl Prax 24: 567576.Google ScholarPubMed
Bode, L and Ludwig, H (2003). Borna disease virus infection, a human mental-health risk. Clinical Microbiology Reviews 16: 534545.Google Scholar
Bode, L, Durrwald, R and Ludwig, H (1994). Borna virus infections in cattle associated with fatal neurological disease. Veterinary Record 135: 283284.Google Scholar
Bondy, SC and Purdy, JL (1977). Putative neurotransmitters of the avian visual pathway. Brain Research 119: 417426.CrossRefGoogle ScholarPubMed
Bordey, A and Sontheimer, H (2003). Modulation of glutamatergic transmission by bergmann glial cells in rat cerebellum in situ. Journal of Neurophysiology 89: 979988.CrossRefGoogle ScholarPubMed
Caplazi, P and Ehrensperger, F (1998). Spontaneous Borna disease in sheep and horses: immunophenotyping of inflammatory cells and detection of MHC-I and MHC-II antigen expression in Borna encephalitis lesions. Veterinary Immunology and Immunopathology 61: 203220.Google Scholar
Caplazi, P, Waldvogel, A, Stitz, L, Braun, U and Ehrensperger, F (1994). Borna disease in naturally infected cattle. Journal of Comparative Pathology 111: 6572.CrossRefGoogle ScholarPubMed
Carbone, KM, Duchala, CS, Griffin, JW, Kincaid, AL and Narayan, O (1987). Pathogenesis of Borna disease in rats: evidence that intra-axonal spread is the major route for virus dissemination and the determinant for disease incubation. Journal of Virology 61: 34313440.Google Scholar
Carbone, KM, Moench, TR and Lipkin, WI (1991). Borna disease virus replicates in astrocytes, Schwann cells and ependymal cells in persistently infected rats: location of viral genomic and messenger RNAs by in situ hybridization. Journal of Neuropathology and Experimental Neurology 50: 205214.Google Scholar
Carbone, KM, Rubin, SA, Nishino, Y and Pletnikov, MV (2001). Borna disease: virus-induced neurobehavioral disease pathogenesis. Current Opinion in Microbiology 4: 467475.CrossRefGoogle ScholarPubMed
Chen, J and Kriegstein, AR (2015). A GABAergic projection from the zona incerta to cortex promotes cortical neuron development. Science 350: 554558.Google Scholar
Choi, DW (1988). Glutamate neurotoxicity and diseases of the nervous system. Neuron 1: 623634.CrossRefGoogle ScholarPubMed
Clemente, R, Sisman, E, Aza-Blanc, P and de la Torre, JC (2010). Identification of host factors involved in borna disease virus cell entry through a small interfering RNA functional genetic screen. Journal of Virology 84: 35623575.CrossRefGoogle ScholarPubMed
Coulter, DA and Eid, T (2012). Astrocytic regulation of glutamate homeostasis in epilepsy. Glia 60: 12151226.CrossRefGoogle ScholarPubMed
Cubitt, B, Oldstone, C, Valcarcel, J and Carlos de la Torre, J (1994). RNA splicing contributes to the generation of mature mRNAs of Borna disease virus, a non-segmented negative strand RNA virus. Virus Research 34: 6979.Google Scholar
de la Torre, JC (2002). Bornavirus and the brain. Journal of Infectious Diseases 186 (suppl. 2): S241S247.Google Scholar
Dietz, DM and Pletnikov, MV (2003). Postnatal weight gain inhibition does not account for neurobehavioral consequences of neonatal Borna disease virus infection. Physiology & Behavior 80: 359366.Google Scholar
Dietzel, J, Kuhrt, H, Stahl, T, Kacza, J, Seeger, J, Weber, M, Uhlig, A, Reichenbach, A, Grosche, A and Pannicke, T (2007). Morphometric analysis of the retina from horses infected with the Borna disease virus. Veterinary Pathology 44: 5763.Google Scholar
Durrwald, R and Ludwig, H (1997). Borna disease virus (BDV), a (zoonotic?) worldwide pathogen. A review of the history of the disease and the virus infection with comprehensive bibliography. Zentralbl Veterinarmed B 44: 147184.Google Scholar
Durrwald, R, Kolodziejek, J, Herzog, S and Nowotny, N (2007). Meta-analysis of putative human bornavirus sequences fails to provide evidence implicating Borna disease virus in mental illness. Reviews in Medical Virology 17: 181203.Google Scholar
Durrwald, R, Kolodziejek, J, Weissenbock, H and Nowotny, N (2014). The bicolored white-toothed shrew Crocidura leucodon (HERMANN 1780) is an indigenous host of mammalian Borna disease virus. PLoS ONE 9: e93659.CrossRefGoogle ScholarPubMed
Eisenman, LM, Brothers, R, Tran, MH, Kean, RB, Dickson, GM, Dietzschold, B and Hooper, DC (1999). Neonatal Borna disease virus infection in the rat causes a loss of Purkinje cells in the cerebellum. Journal of Neurovirology 5: 181189.CrossRefGoogle ScholarPubMed
Enbergs, HK, Vahlenkamp, TW, Kipar, A and Muller, H (2001). Experimental infection of mice with Borna disease virus (BDV): replication and distribution of the virus after intracerebral infection. Journal of Neurovirology 7: 272277.Google ScholarPubMed
Engert, F and Bonhoeffer, T (1999). Dendritic spine changes associated with hippocampal long-term synaptic plasticity. Nature 399: 6670.Google Scholar
Formella, S, Jehle, C, Sauder, C, Staeheli, P and Schwemmle, M (2000). Sequence variability of Borna disease virus: resistance to superinfection may contribute to high genome stability in persistently infected cells. Journal of Virology 74: 78787883.Google Scholar
Fu, ZF, Amsterdam, JD, Kao, M, Shankar, V, Koprowski, H and Dietzschold, B (1993). Detection of Borna disease virus-reactive antibodies from patients with affective disorders by western immunoblot technique. Journal of Affective Disorders 27: 6168.CrossRefGoogle ScholarPubMed
Ganor, Y and Levite, M (2014). The neurotransmitter glutamate and human T cells: glutamate receptors and glutamate-induced direct and potent effects on normal human T cells, cancerous human leukemia and lymphoma T cells, and autoimmune human T cells. Journal of Neural Transmission 121: 9831006.Google Scholar
Gies, U, Bilzer, T, Stitz, L and Staiger, JF (1998). Disturbance of the cortical cholinergic innervation in Borna disease prior to encephalitis. Brain Pathology 8: 3948.Google Scholar
Gies, U, Gorcs, TJ, Mulder, J, Planz, O, Stitz, L, Bilzer, T, Luiten, PG and Harkany, T (2001). Cortical cholinergic decline parallels the progression of Borna virus encephalitis. Neuroreport 12: 37673772.CrossRefGoogle ScholarPubMed
Gonzalez, H, Elgueta, D, Montoya, A and Pacheco, R (2014). Neuroimmune regulation of microglial activity involved in neuroinflammation and neurodegenerative diseases. Journal of Neuroimmunology 274: 113.CrossRefGoogle ScholarPubMed
Gonzalez-Dunia, D, Cubitt, B and de la Torre, JC (1998). Mechanism of Borna disease virus entry into cells. Journal of Virology 72: 783788.CrossRefGoogle ScholarPubMed
Gonzalez-Dunia, D, Watanabe, M, Syan, S, Mallory, M, Masliah, E and de la Torre, JC (2000). Synaptic pathology in Borna disease virus persistent infection. Journal of Virology 74: 34413448.CrossRefGoogle ScholarPubMed
Gonzalez-Dunia, D, Volmer, R, Mayer, D and Schwemmle, M (2005). Borna disease virus interference with neuronal plasticity. Virus Research 111: 224234.Google Scholar
Gosztonyi, G (2008). Natural and experimental Borna disease virus infections--neuropathology and pathogenetic considerations. APMIS. Supplementum 124: 5357.CrossRefGoogle Scholar
Gosztonyi, G and Ludwig, H (1984). Borna disease of horses. An immunohistological and virological study of naturally infected animals. Acta Neuropathologica 64: 213221.Google Scholar
Gosztonyi, G and Ludwig, H (1995). Borna disease–neuropathology and pathogenesis. Current Topics in Microbiology and Immunology 190: 3973.Google Scholar
Gosztonyi, G, Dietzschold, B, Kao, M, Rupprecht, CE, Ludwig, H and Koprowski, H (1993). Rabies and borna disease. A comparative pathogenetic study of two neurovirulent agents. Laboratory Investigation 68: 285295.Google ScholarPubMed
Gudino-Cabrera, G, Urena-Guerrero, ME, Rivera-Cervantes, MC, Feria-Velasco, AI and Beas-Zarate, C (2014). Excitotoxicity triggered by neonatal monosodium glutamate treatment and blood-brain barrier function. Archives of Medical Research 45: 653659.Google Scholar
Hallensleben, W, Schwemmle, M, Hausmann, J, Stitz, L, Volk, B, Pagenstecher, A and Staeheli, P (1998). Borna disease virus-induced neurological disorder in mice: infection of neonates results in immunopathology. Journal of Virology 72: 43794386.Google Scholar
Hans, A, Bajramovic, JJ, Syan, S, Perret, E, Dunia, I, Brahic, M and Gonzalez-Dunia, D (2004). Persistent, noncytolytic infection of neurons by Borna disease virus interferes with ERK 1/2 signaling and abrogates BDNF-induced synaptogenesis. Faseb Journal 18: 863865.Google Scholar
Hatalski, CG, Hickey, WF and Lipkin, WI (1998). Evolution of the immune response in the central nervous system following infection with Borna disease virus. Journal of Neuroimmunology 90: 137142.Google Scholar
Hausmann, J, Hallensleben, W, de la Torre, JC, Pagenstecher, A, Zimmermann, C, Pircher, H and Staeheli, P (1999). T cell ignorance in mice to Borna disease virus can be overcome by peripheral expression of the viral nucleoprotein. Proceedings of the National Academy of Sciences of the United States of America 96: 97699774.Google Scholar
Hausmann, J, Sauder, C, Wasmer, M, Lu, B and Staeheli, P (2004). Neurological disorder after Borna disease virus infection in the absence of either interferon-gamma, Fas, inducible NO synthase, or chemokine receptor CXCR3. Viral Immunology 17: 7985.Google Scholar
Heneka, MT, Kummer, MP and Latz, E (2014). Innate immune activation in neurodegenerative disease. Nature Reviews. Immunology 14: 463477.Google Scholar
Herden, C, Herzog, S, Richt, JA, Nesseler, A, Christ, M, Failing, K and Frese, K (2000). Distribution of Borna disease virus in the brain of rats infected with an obesity-inducing virus strain. Brain Pathology 10: 3948.Google Scholar
Herden, C, Schluesener, HJ and Richt, JA (2005). Expression of allograft inflammatory factor-1 and haeme oxygenase-1 in brains of rats infected with the neurotropic Borna disease virus. Neuropathology and Applied Neurobiology 31: 512521.CrossRefGoogle ScholarPubMed
Herzog, S, Kompter, C, Frese, K and Rott, R (1984). Replication of Borna disease virus in rats: age-dependent differences in tissue distribution. Medical Microbiology and Immunology 173: 171177.Google Scholar
Herzog, S, Wonigeit, K, Frese, K, Hedrich, HJ and Rott, R (1985). Effect of Borna disease virus infection on athymic rats. Journal of General Virology 66 (Pt 3): 503508.Google Scholar
Hirano, N, Kao, M and Ludwig, H (1983). Persistent, tolerant or subacute infection in Borna disease virus-infected rats. Journal of General Virology 64 (Pt 7): 15211530.Google Scholar
Hoffmann, B, Tappe, D, Hoper, D, Herden, C, Boldt, A, Mawrin, C, Niederstrasser, O, Muller, T, Jenckel, M, Van Der Grinten, E, Lutter, C, Abendroth, B, Teifke, JP, Cadar, D, Schmidt-Chanasit, J, Ulrich, RG and Beer, M (2015). A variegated squirrel bornavirus associated with fatal human encephalitis. New England Journal of Medicine 373: 154162.Google Scholar
Honda, T and Tomonaga, K (2013). Nucleocytoplasmic shuttling of viral proteins in borna disease virus infection. Viruses 5: 19781990.Google Scholar
Honda, T, Fujino, K, Okuzaki, D, Ohtaki, N, Matsumoto, Y, Horie, M, Daito, T, Itoh, M and Tomonaga, K (2011). Upregulation of insulin-like growth factor binding protein 3 in astrocytes of transgenic mice that express Borna disease virus phosphoprotein. Journal of Virology 85: 45674571.Google Scholar
Honkavuori, KS, Shivaprasad, HL, Williams, BL, Quan, PL, Hornig, M, Street, C, Palacios, G, Hutchison, SK, Franca, M, Egholm, M, Briese, T and Lipkin, WI (2008). Novel borna virus in psittacine birds with proventricular dilatation disease. Emerging Infectious Diseases 14: 18831886.Google Scholar
Horie, M, Honda, T, Suzuki, Y, Kobayashi, Y, Daito, T, Oshida, T, Ikuta, K, Jern, P, Gojobori, T, Coffin, JM and Tomonaga, K (2010). Endogenous non-retroviral RNA virus elements in mammalian genomes. Nature 463: 8487.Google Scholar
Horie, M, Kobayashi, Y, Suzuki, Y and Tomonaga, K (2013). Comprehensive analysis of endogenous bornavirus-like elements in eukaryote genomes. “Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences” 368: 20120499.Google Scholar
Hornig, M, Weissenbock, H, Horscroft, N and Lipkin, WI (1999). An infection-based model of neurodevelopmental damage. Proceedings of the National Academy of Sciences of the United States of America 96: 1210212107.Google Scholar
Hornig, M, Solbrig, M, Horscroft, N, Weissenbock, H and Lipkin, WI (2001). Borna disease virus infection of adult and neonatal rats: models for neuropsychiatric disease. Current Topics in Microbiology and Immunology 253: 157177.Google Scholar
Hornig, M, Briese, T, Licinio, J, Khabbaz, RF, Altshuler, LL, Potkin, SG, Schwemmle, M, Siemetzki, U, Mintz, J, Honkavuori, K, Kraemer, HC, Egan, MF, Whybrow, PC, Bunney, WE and Lipkin, WI (2012). Absence of evidence for bornavirus infection in schizophrenia, bipolar disorder and major depressive disorder. Molecular Psychiatry 17: 486493.Google Scholar
Iandiev, I, Biedermann, B, Bringmann, A, Reichel, MB, Reichenbach, A and Pannicke, T (2006). Atypical gliosis in Muller cells of the slowly degenerating rds mutant mouse retina. Experimental Eye Research 82: 449457.Google Scholar
Jacobs, CM, Aden, P, Mathisen, GH, Khuong, E, Gaarder, M, Loberg, EM, Lomo, J, Maehlen, J and Paulsen, RE (2006). Chicken cerebellar granule neurons rapidly develop excitotoxicity in culture. Journal of Neuroscience Methods 156: 129135.Google Scholar
Jacobsen, B, Algermissen, D, Schaudien, D, Venner, M, Herzog, S, Wentz, E, Hewicker-Trautwein, M, Baumgartner, W and Herden, C (2010). Borna disease in an adult alpaca stallion (Lama pacos). Journal of Comparative Pathology 143: 203208.Google Scholar
Jamali, T, Jamali, Y, Mehrbod, M and Mofrad, MR (2011). Nuclear pore complex: biochemistry and biophysics of nucleocytoplasmic transport in health and disease. International Review of Cell and Molecular Biology 287: 233286.Google Scholar
Jarius, S and Wildemann, B (2015). ‘Medusa head ataxia’: the expanding spectrum of Purkinje cell antibodies in autoimmune cerebellar ataxia. Part 3: Anti-Yo/CDR2, anti-Nb/AP3B2, PCA-2, anti-Tr/DNER, other antibodies, diagnostic pitfalls, summary and outlook. Journal of Neuroinflammation 12: 168.Google Scholar
Johnson, RT (1980). Selective vulnerability of neural cells to viral infections. Brain 103: 447472.Google Scholar
Kacza, J, Vahlenkamp, TW, Enbergs, H, Richt, JA, Germer, A, Kuhrt, H, Reichenbach, A, Muller, H, Herden, C, Stahl, T and Seeger, J (2000). Neuron-glia interactions in the rat retina infected by Borna disease virus. Archives of Virology 145: 127147.Google Scholar
Kacza, J, Mohr, C, Pannicke, T, Kuhrt, H, Dietzel, J, Fluss, M, Richt, JA, Vahlenkamp, TW, Stahl, T, Reichenbach, A and Seeger, J (2001). Changes of the organotypic retinal organization in Borna virus-infected Lewis rats. Journal of Neurocytology 30: 801820.Google Scholar
Kamitani, W, Shoya, Y, Kobayashi, T, Watanabe, M, Lee, BJ, Zhang, G, Tomonaga, K and Ikuta, K (2001). Borna disease virus phosphoprotein binds a neurite outgrowth factor, amphoterin/HMG-1. Journal of Virology 75: 87428751.Google Scholar
Kamitani, W, Ono, E, Yoshino, S, Kobayashi, T, Taharaguchi, S, Lee, BJ, Yamashita, M, Kobayashi, T, Okamoto, M, Taniyama, H, Tomonaga, K and Ikuta, K (2003). Glial expression of Borna disease virus phosphoprotein induces behavioral and neurological abnormalities in transgenic mice. Proceedings of the National Academy of Sciences of the United States of America 100: 89698974.Google Scholar
Kao, M, Ludwig, H and Gosztonyi, G (1984). Adaptation of Borna disease virus to the mouse. Journal of General Virology 65 (Pt 10): 18451849.Google Scholar
Katsumoto, A, Lu, H, Miranda, AS and Ransohoff, RM (2014). Ontogeny and functions of central nervous system macrophages. Journal of Immunology 193: 26152621.Google Scholar
Kinnunen, PM, Billich, C, Ek-Kommonen, C, Henttonen, H, Kallio, RK, Niemimaa, J, Palva, A, Staeheli, P, Vaheri, A and Vapalahti, O (2007). Serological evidence for Borna disease virus infection in humans, wild rodents and other vertebrates in Finland. Journal of Clinical Virology 38: 6469.Google Scholar
Kinnunen, PM, Palva, A, Vaheri, A and Vapalahti, O (2013). Epidemiology and host spectrum of Borna disease virus infections. Journal of General Virology 94: 247262.Google Scholar
Kistler, AL, Gancz, A, Clubb, S, Skewes-Cox, P, Fischer, K, Sorber, K, Chiu, CY, Lublin, A, Mechani, S, Farnoushi, Y, Greninger, A, Wen, CC, Karlene, SB, Ganem, D and Derisi, JL (2008). Recovery of divergent avian bornaviruses from cases of proventricular dilatation disease: identification of a candidate etiologic agent. Virology Journal 5: 88.Google Scholar
Krey, H, Ludwig, H and Rott, R (1979). Spread of infectious virus along the optic nerve into the retina in Borna disease virus-infected rabbits. Archives of Virology 61: 283288.Google Scholar
Krey, H, Ludwig, H and Gierend, M (1981). Borna disease virus-induced retinouveitis treated with immunosuppressive drugs. Albrecht Von Graefes Arch Klin Exp Ophthalmol 216: 111119.Google Scholar
Kuhn, J, Dürrwald, R, Bào, Y, Briese, T, Carbone, K, Clawson, A, Derisi, J, Garten, W, Jahrling, P, Kolodziejek, J, Rubbenstroth, D, Schwemmle, M, Stenglein, M, Tomonaga, K, Weissenböck, H and Nowotny, N (2015). Taxonomic reorganization of the family Bornaviridae. Archives of Virology 160: 621632.Google Scholar
Lancaster, K, Dietz, DM, Moran, TH and Pletnikov, MV (2007). Abnormal social behaviors in young and adult rats neonatally infected with Borna disease virus. Behavioural Brain Research 176: 141148.Google Scholar
Lee, BJ, Watanabe, M, Kamitani, W, Baba, S, Yamashita, M, Kobayashi, T, Tomonaga, K and Ikuta, K (2003). Age- and host-dependent control of Borna disease virus spread in the developing brains of gerbils and rats. Microbes and Infection 5: 11951204.CrossRefGoogle ScholarPubMed
Lennartz, F, Bayer, K, Czerwonka, N, Lu, Y, Kehr, K, Hirz, M, Steinmetzer, T, Garten, W and Herden, C (2016). Surface glycoprotein of Borna disease virus mediates virus spread from cell to cell. Cellular Microbiology 18: 340354.Google Scholar
Levite, M (2008). Neurotransmitters activate T-cells and elicit crucial functions via neurotransmitter receptors. Current Opinion in Pharmacology 8: 460471.Google Scholar
Lipkin, WI, Briese, T and Hornig, M (2011). Borna disease virus - fact and fantasy. Virus Research 162: 162172.Google Scholar
Liu, X, Liu, S, Bode, L, Liu, C, Zhang, L, Wang, X, Li, D, Lei, Y, Peng, X, Cheng, Z and Xie, P (2015). Persistent human Borna disease virus infection modifies the acetylome of human oligodendroglia cells towards higher energy and transporter levels. Virology 485: 5878.Google Scholar
Ludwig, H (2008). The biology of bornavirus. APMIS. Supplementum 124: 1420.Google Scholar
Ludwig, H and Bode, L (2000). Borna disease virus: new aspects on infection, disease, diagnosis and epidemiology. Revue Scientifique et Technique 19: 259288.Google Scholar
Lundgren, AL, Lindberg, R, Ludwig, H and Gosztonyi, G (1995a). Immunoreactivity of the central nervous system in cats with a Borna disease-like meningoencephalomyelitis (staggering disease). Acta Neuropathologica 90: 184193.Google Scholar
Lundgren, AL, Zimmermann, W, Bode, L, Czech, G, Gosztonyi, G, Lindberg, R and Ludwig, H (1995b). Staggering disease in cats: isolation and characterization of the feline Borna disease virus. Journal of General Virology 76 (Pt 9): 22152222.Google Scholar
Lundgren, AL, Johannisson, A, Zimmermann, W, Bode, L, Rozell, B, Muluneh, A, Lindberg, R and Ludwig, H (1997). Neurological disease and encephalitis in cats experimentally infected with Borna disease virus. Acta Neuropathologica 93: 391401.Google Scholar
Lutz, H, Addie, DD, Boucraut-Baralon, C, Egberink, H, Frymus, T, Gruffydd-Jones, T, Hartmann, K, Horzinek, MC, Hosie, MJ, Lloret, A, Marsilio, F, Pennisi, MG, Radford, AD, Thiry, E, Truyen, U and Mostl, K (2015). Borna disease virus infection in cats: ABCD guidelines on prevention and management. Journal of Feline Medicine and Surgery 17: 614616.Google Scholar
Lyons, MJ, Nagashima, K and Zabriskie, JB (2002). Animal models of postinfectious obesity: hypothesis and review. Journal of Neurovirology 8: 15.Google Scholar
Makino, A, Fujino, K, Parrish, NF, Honda, T and Tomonaga, K (2015). Borna disease virus possesses an NF-kB inhibitory sequence in the nucleoprotein gene. Scientific Reports 5: 8696.Google Scholar
Malkinson, M, Weisman, Y, Ashash, E, Bode, L and Ludwig, H (1993). Borna disease in ostriches. Veterinary Record 133: 304.Google Scholar
Matsumoto, Y, Hayashi, Y, Omori, H, Honda, T, Daito, T, Horie, M, Ikuta, K, Fujino, K, Nakamura, S, Schneider, U, Chase, G, Yoshimori, T, Schwemmle, M and Tomonaga, K (2012). Bornavirus closely associates and segregates with host chromosomes to ensure persistent intranuclear infection. Cell Host & Microbe 11: 492503.Google Scholar
Metzler, A, Frei, U and Danner, K (1976). Virologically confirmed outbreak of Borna's disease in a Swiss herd of sheep. Schweizer Archiv fur Tierheilkunde 118: 483492.Google Scholar
Metzler, A, Minder, HP, Wegmann, C and Zindel, W (1979). Borna disease, a veterinary problem of regional significance. Schweizer Archiv fur Tierheilkunde 121: 207213.Google Scholar
Morales, JA, Herzog, S, Kompter, C, Frese, K and Rott, R (1988). Axonal transport of Borna disease virus along olfactory pathways in spontaneously and experimentally infected rats. Medical Microbiology and Immunology 177: 5168.Google Scholar
Morimoto, K, Hooper, DC, Bornhorst, A, Corisdeo, S, Bette, M, Fu, ZF, Schafer, MK, Koprowski, H, Weihe, E and Dietzschold, B (1996). Intrinsic responses to Borna disease virus infection of the central nervous system. Proceedings of the National Academy of Sciences of the United States of America 93: 1334513350.Google Scholar
Nagashima, K, Zabriskie, JB and Lyons, MJ (1992). Virus-induced obesity in mice: association with a hypothalamic lesion. Journal of Neuropathology and Experimental Neurology 51: 101109.Google Scholar
Nakamura, Y, Nakaya, T, Hagiwara, K, Momiyama, N, Kagawa, Y, Taniyama, H, Ishihara, C, Sata, T, Kurata, T and Ikuta, K (1999). High susceptibility of Mongolian gerbil (Meriones unguiculatus) to Borna disease virus. Vaccine 17: 480489.Google Scholar
Narayan, O, Herzog, S, Frese, K, Scheefers, H and Rott, R (1983a). Behavioral disease in rats caused by immunopathological responses to persistent borna virus in the brain. Science 220: 14011403.Google Scholar
Narayan, O, Herzog, S, Frese, K, Scheefers, H and Rott, R (1983b). Pathogenesis of Borna disease in rats: immune-mediated viral ophthalmoencephalopathy causing blindness and behavioral abnormalities. Journal of Infectious Diseases 148: 305315.Google Scholar
Nishino, Y, Ooishi, R, Kurokawa, S, Fujino, K, Murakami, M, Madarame, H, Hashimoto, O, Sugiyama, K and Funaba, M (2009). Gene expression of the TGF-beta family in rat brain infected with Borna disease virus. Microbes and Infection 11: 737743.Google Scholar
Nishino, Y, Murakami, M and Funaba, M (2015). Expression and role of the TGF-beta family in glial cells infected with Borna disease virus. Microbes and Infection 18: 128136.Google Scholar
Nobach, D, Bourg, M, Herzog, S, Lange-Herbst, H, Encarnacao, JA, Eickmann, M and Herden, C (2015). Shedding of Infectious Borna Disease Virus-1 in Living Bicolored White-Toothed Shrews. PLoS ONE 10: e0137018.Google Scholar
Noske, K, Bilzer, T, Planz, O and Stitz, L (1998). Virus-specific CD4+ T cells eliminate Borna disease virus from the brain via induction of cytotoxic CD8+ T cells. Journal of Virology 72: 43874395.Google Scholar
Nouri, M, Bredberg, A, Westrom, B and Lavasani, S (2014). Intestinal barrier dysfunction develops at the onset of experimental autoimmune encephalomyelitis, and can be induced by adoptive transfer of auto-reactive T cells. PLoS ONE 9: e106335.Google Scholar
Okamoto, M, Kagawa, Y, Kamitani, W, Hagiwara, K, Kirisawa, R, Iwai, H, Ikuta, K and Taniyama, H (2002). Borna disease in a dog in Japan. Journal of Comparative Pathology 126: 312317.Google Scholar
Ovanesov, MV, Sauder, C, Rubin, SA, Richt, J, Nath, A, Carbone, KM and Pletnikov, MV (2006). Activation of microglia by borna disease virus infection: in vitro study. Journal of Virology 80: 1214112148.Google Scholar
Ovanesov, MV, Vogel, MW, Moran, TH and Pletnikov, MV (2007). Neonatal Borna disease virus infection in rats is associated with increased extracellular levels of glutamate and neurodegeneration in the striatum. Journal of Neurovirology 13: 185194.Google Scholar
Ovanesov, MV, Ayhan, Y, Wolbert, C, Moldovan, K, Sauder, C and Pletnikov, MV (2008a). Astrocytes play a key role in activation of microglia by persistent Borna disease virus infection. Journal of Neuroinflammation 5: 50.Google Scholar
Ovanesov, MV, Moldovan, K, Smith, K, Vogel, MW and Pletnikov, MV (2008b). Persistent Borna Disease Virus (BDV) infection activates microglia prior to a detectable loss of granule cells in the hippocampus. Journal of Neuroinflammation 5: 16.Google Scholar
Pacheco, R, Riquelme, E and Kalergis, AM (2010). Emerging evidence for the role of neurotransmitters in the modulation of T cell responses to cognate ligands. Central Nervous System Agents in Medicinal Chemistry 10: 6583.Google Scholar
Papageorgiou, IE, Lewen, A, Galow, LV, Cesetti, T, Scheffel, J, Regen, T, Hanisch, UK and Kann, O (2015). TLR4-activated microglia require IFN-gamma to induce severe neuronal dysfunction and death in situ. Proceedings of the National Academy of Sciences of the United States of America 113: 212217.CrossRefGoogle ScholarPubMed
Peng, G, Yan, Y, Zhu, C, Wang, S, Yan, X, Lu, L, Li, W, Hu, J, Wei, W, Mu, Y, Chen, Y, Feng, Y, Gong, R, Wu, K, Zhang, F, Zhang, X, Zhu, Y and Wu, J (2008). Borna disease virus P protein affects neural transmission through interactions with gamma-aminobutyric acid receptor-associated protein. Journal of Virology 82: 1248712497.Google Scholar
Pikor, NB, Astarita, JL, Summers-Deluca, L, Galicia, G, Qu, J, Ward, LA, Armstrong, S, Dominguez, CX, Malhotra, D, Heiden, B, Kay, R, Castanov, V, Touil, H, Boon, L, O'Connor, P, Bar-Or, A, Prat, A, Ramaglia, V, Ludwin, S, Turley, SJ and Gommerman, JL (2015). Integration of Th17- and Lymphotoxin-derived signals initiates meningeal-resident stromal cell remodeling to propagate Neuroinflammation. Immunity 43: 11601173.Google Scholar
Planz, O, Bilzer, T, Sobbe, M and Stitz, L (1993). Lysis of major histocompatibility complex class I-bearing cells in Borna disease virus-induced degenerative encephalopathy. Journal of Experimental Medicine 178: 163174.Google Scholar
Planz, O, Dumrese, T, Hulpusch, S, Schirle, M, Stevanovic, S and Stitz, L (2001). A naturally processed rat major histocompatibility complex class I-associated viral peptide as target structure of borna disease virus-specific CD8+ T cells. Journal of Biological Chemistry 276: 1368913694.Google Scholar
Plata-Salaman, CR, Ilyin, SE, Gayle, D, Romanovitch, A and Carbone, KM (1999). Persistent Borna disease virus infection of neonatal rats causes brain regional changes of mRNAs for cytokines, cytokine receptor components and neuropeptides. Brain Research Bulletin 49: 441451.Google Scholar
Pletnikov, MV, Rubin, SA, Vogel, MW, Moran, TH and Carbone, KM (2002). Effects of genetic background on neonatal Borna disease virus infection-induced neurodevelopmental damage. II. Neurochemical alterations and responses to pharmacological treatments. Brain Research 944: 108123.Google Scholar
Puorger, ME, Hilbe, M, Muller, JP, Kolodziejek, J, Nowotny, N, Zlinszky, K and Ehrensperger, F (2010). Distribution of Borna disease virus antigen and RNA in tissues of naturally infected bicolored white-toothed shrews, Crocidura leucodon, supporting their role as reservoir host species. Veterinary Pathology 47: 236244.Google Scholar
Reuter, A, Ackermann, A, Kothlow, S, Rinder, M, Kaspers, B and Staeheli, P (2010). Avian bornaviruses escape recognition by the innate immune system. Viruses 2: 927938.Google Scholar
Richt, JA and Rott, R (2001). Borna disease virus: a mystery as an emerging zoonotic pathogen. Veterinary Journal 161: 2440.Google Scholar
Richt, JA, Vandewoude, S, Zink, MC, Clements, JE, Herzog, S, Stitz, L, Rott, R and Narayan, O (1992). Infection with Borna disease virus: molecular and immunobiological characterization of the agent. Clinical Infectious Diseases 14: 12401250.Google Scholar
Richt, JA, Pfeuffer, I, Christ, M, Frese, K, Bechter, K and Herzog, S (1997). Borna disease virus infection in animals and humans. Emerging Infectious Diseases 3: 343352.Google Scholar
Richt, JA, Grabner, A and Herzog, S (2000). Borna disease in horses. The Veterinary Clinics of North America. Equine Practice 16: 579595, xi.Google Scholar
Richter, K, Hausmann, J and Staeheli, P (2009). Interferon-gamma prevents death of bystander neurons during CD8 T cell responses in the brain. American Journal of Pathology 174: 17991807.Google Scholar
Rossi, D and Volterra, A (2009). Astrocytic dysfunction: insights on the role in neurodegeneration. Brain Research Bulletin 80: 224232.Google Scholar
Rott, R and Becht, H (1995). Natural and experimental Borna disease in animals. Current Topics in Microbiology and Immunology 190: 1730.Google Scholar
Rott, R, Herzog, S, Fleischer, B, Winokur, A, Amsterdam, J, Dyson, W and Koprowski, H (1985). Detection of serum antibodies to Borna disease virus in patients with psychiatric disorders. Science 228: 755756.Google Scholar
Rott, R, Herzog, S, Richt, J and Stitz, L (1988). Immune-mediated pathogenesis of Borna disease. Zentralbl Bakteriol Mikrobiol Hyg A 270: 295301.Google Scholar
Rubbenstroth, D, Schmidt, V, Rinder, M, Legler, M, Corman, VM and Staeheli, P (2014). Discovery of a new avian bornavirus genotype in estrildid finches (Estrildidae) in Germany. Veterinary Microbiology 168: 318323.Google Scholar
Rubin, SA, Waltrip, RW II, Bautista, JR and Carbone, KM (1993). Borna disease virus in mice: host-specific differences in disease expression. Journal of Virology 67: 548552.Google Scholar
Russo, MV and Mcgavern, DB (2015). Immune surveillance of the CNS following infection and injury. Trends in Immunology 36: 637650.Google Scholar
Salinas, S, Schiavo, G and Kremer, EJ (2010). A hitchhiker's guide to the nervous system: the complex journey of viruses and toxins. Nature Reviews. Microbiology 8: 645655.Google Scholar
Sauder, C and de la Torre, JC (1999). Cytokine expression in the rat central nervous system following perinatal Borna disease virus infection. Journal of Neuroimmunology 96: 2945.Google Scholar
Sauder, C and Staeheli, P (2003). Rat model of borna disease virus transmission: epidemiological implications. Journal of Virology 77: 1288612890.Google Scholar
Schneemann, A, Schneider, PA, Kim, S and Lipkin, WI (1994). Identification of signal sequences that control transcription of borna disease virus, a nonsegmented, negative-strand RNA virus. Journal of Virology 68: 65146522.Google Scholar
Schneider, PA, Schneemann, A and Lipkin, WI (1994). RNA splicing in Borna disease virus, a nonsegmented, negative-strand RNA virus. Journal of Virology 68: 50075012.Google Scholar
Schwartz, M, Shaked, I, Fisher, J, Mizrahi, T and Schori, H (2003). Protective autoimmunity against the enemy within: fighting glutamate toxicity. Trends in Neurosciences 26: 297302.Google Scholar
Schwemmle, M, Salvatore, M, Shi, L, Richt, J, Lee, CH and Lipkin, WI (1998). Interactions of the borna disease virus P, N, and X proteins and their functional implications. Journal of Biological Chemistry 273: 90079012.Google Scholar
Scordel, C, Huttin, A, Cochet-Bernoin, M, Szelechowski, M, Poulet, A, Richardson, J, Benchoua, A, Gonzalez-Dunia, D, Eloit, M and Coulpier, M (2015). Borna disease virus phosphoprotein impairs the developmental program controlling neurogenesis and reduces human GABAergic neurogenesis. PLoS Pathogens 11: e1004859.CrossRefGoogle ScholarPubMed
Sobbe, M, Bilzer, T, Gommel, S, Noske, K, Planz, O and Stitz, L (1997). Induction of degenerative brain lesions after adoptive transfer of brain lymphocytes from Borna disease virus-infected rats: presence of CD8+ T cells and perforin mRNA. Journal of Virology 71: 24002407.Google Scholar
Solbrig, MV and Koob, GF (2003). Neuropharmacological sequelae of persistent CNS viral infections: lessons from Borna disease virus. “Pharmacology, Biochemistry, and Behavior” 74: 777787.Google Scholar
Solbrig, MV, Koob, GF, Joyce, JN and Lipkin, WI (1996). A neural substrate of hyperactivity in borna disease: changes in brain dopamine receptors. Virology 222: 332338.Google Scholar
Sprankel, H, Richarz, K, Ludwig, H and Rott, R (1978). Behavior alterations in tree shrews (Tupaia glis, Diard 1820) induced by Borna disease virus. Medical Microbiology and Immunology 165: 118.Google Scholar
Staeheli, P, Sauder, C, Hausmann, J, Ehrensperger, F and Schwemmle, M (2000). Epidemiology of Borna disease virus. Journal of General Virology 81: 21232135.Google Scholar
Stahl, T, Mohr, C, Kacza, J, Reimers, C, Pannicke, T, Sauder, C, Reichenbach, A and Seeger, J (2003). Characterization of the acute immune response in the retina of Borna disease virus infected Lewis rats. Journal of Neuroimmunology 137: 6778.Google Scholar
Stitz, L, Krey, H and Ludwig, H (1981). Borna disease in rhesus monkeys as a models for uveo-cerebral symptoms. Journal of Medical Virology 6: 333340.Google Scholar
Stitz, L, Soeder, D, Deschl, U, Frese, K and Rott, R (1989). Inhibition of immune-mediated meningoencephalitis in persistently Borna disease virus-infected rats by cyclosporine A. Journal of Immunology 143: 42504256.Google Scholar
Stitz, L, Schilken, D and Frese, K (1991). Atypical dissemination of the highly neurotropic Borna disease virus during persistent infection in cyclosporine A-treated, immunosuppressed rats. Journal of Virology 65: 457460.Google Scholar
Stitz, L, Sobbe, M and Bilzer, T (1992). Preventive effects of early anti-CD4 or anti-CD8 treatment on Borna disease in rats. Journal of Virology 66: 33163323.Google Scholar
Stitz, L, Noske, K, Planz, O, Furrer, E, Lipkin, WI and Bilzer, T (1998). A functional role for neutralizing antibodies in Borna disease: influence on virus tropism outside the central nervous system. Journal of Virology 72: 88848892.Google Scholar
Stitz, L, Bilzer, T and Planz, O (2002). The immunopathogenesis of Borna disease virus infection. Frontiers in Bioscience 7: d541d555.Google Scholar
Takeuchi, H, Jin, S, Wang, J, Zhang, G, Kawanokuchi, J, Kuno, R, Sonobe, Y, Mizuno, T and Suzumura, A (2006). Tumor necrosis factor-alpha induces neurotoxicity via glutamate release from hemichannels of activated microglia in an autocrine manner. Journal of Biological Chemistry 281: 2136221368.Google Scholar
Tomonaga, K, Kobayashi, T and Ikuta, K (2002). Molecular and cellular biology of Borna disease virus infection. Microbes and Infection 4: 491500.Google Scholar
Vahlenkamp, TW, Konrath, A, Weber, M and Muller, H (2002). Persistence of Borna disease virus in naturally infected sheep. Journal of Virology 76: 97359743.Google Scholar
Vesce, S, Rossi, D, Brambilla, L and Volterra, A (2007). Glutamate release from astrocytes in physiological conditions and in neurodegenerative disorders characterized by neuroinflammation. International Review of Neurobiology 82: 5771.Google Scholar
Volmer, R, Prat, CM, Le Masson, G, Garenne, A and Gonzalez-Dunia, D (2007). Borna disease virus infection impairs synaptic plasticity. Journal of Virology 81: 88338837.Google Scholar
Waelchli, RO, Ehrensperger, F, Metzler, A and Winder, C (1985). Borna disease in a sheep. Veterinary Record 117: 499500.Google Scholar
Walsh, JT, Watson, N and Kipnis, J (2014). T cells in the central nervous system: messengers of destruction or purveyors of protection? Immunology 141: 340344.Google Scholar
Watanabe, M, Lee, BJ, Kamitani, W, Kobayashi, T, Taniyama, H, Tomonaga, K and Ikuta, K (2001). Neurological diseases and viral dynamics in the brains of neonatally borna disease virus-infected gerbils. Virology 282: 6576.Google Scholar
Watanabe, M, Lee, BJ, Yamashita, M, Kamitani, W, Kobayashi, T, Tomonaga, K and Ikuta, K (2003). Borna disease virus induces acute fatal neurological disorders in neonatal gerbils without virus- and immune-mediated cell destructions. Virology 310: 245253.Google Scholar
Weissenbock, H, Nowotny, N, Caplazi, P, Kolodziejek, J and Ehrensperger, F (1998). Borna disease in a dog with lethal meningoencephalitis. Journal of Clinical Microbiology 36: 21272130.Google Scholar
Weissenbock, H, Hornig, M, Hickey, WF and Lipkin, WI (2000). Microglial activation and neuronal apoptosis in Bornavirus infected neonatal Lewis rats. Brain Pathology 10: 260272.Google Scholar
Wensman, JJ, Ilback, C, Hjertstrom, E, Blomstrom, AL, Gustavsson, MH, Jaderlund, KH, Strom-Holst, B, Belak, S, Berg, AL and Berg, M (2011). Expression of interferon gamma in the brain of cats with natural Borna disease virus infection. Veterinary Immunology and Immunopathology 141: 162167.Google Scholar
Wensman, JJ, Jaderlund, KH, Gustavsson, MH, Hansson-Hamlin, H, Karlstam, E, Lilliehook, I, Ostrom, IL, Belak, S, Berg, M and Holst, BS (2012). Markers of Borna disease virus infection in cats with staggering disease. Journal of Feline Medicine and Surgery 14: 573582.Google Scholar
Wensman, JJ, Jaderlund, KH, Holst, BS and Berg, M (2014). Borna disease virus infection in cats. Veterinary Journal 201: 142149.Google Scholar
Wu, YJ, Schulz, H, Lin, CC, Saar, K, Patone, G, Fischer, H, Hubner, N, Heimrich, B and Schwemmle, M (2013). Borna disease virus-induced neuronal degeneration dependent on host genetic background and prevented by soluble factors. Proceedings of the National Academy of Sciences of the United States of America 110: 18991904.Google Scholar
Zhang, L, Lei, Y, Liu, X, Wang, X, Liu, Z, Li, D, Zheng, P, Zhang, L, Chen, S and Xie, P (2014). Glutamate and lipid metabolic perturbation in the hippocampi of asymptomatic borna disease virus-infected horses. PLoS ONE 9: e99752.Google Scholar
Zheng, YM, Schafer, MK, Weihe, E, Sheng, H, Corisdeo, S, Fu, ZF, Koprowski, H and Dietzschold, B (1993). Severity of neurological signs and degree of inflammatory lesions in the brains of rats with Borna disease correlate with the induction of nitric oxide synthase. Journal of Virology 67: 57865791.Google Scholar
Zimmermann, W, Breter, H, Rudolph, M and Ludwig, H (1994). Borna disease virus: immunoelectron microscopic characterization of cell-free virus and further information about the genome. Journal of Virology 68: 67556758.Google Scholar
Zocher, M, Czub, S, Schulte-Monting, J, de la Torre, JC and Sauder, C (2000). Alterations in neurotrophin and neurotrophin receptor gene expression patterns in the rat central nervous system following perinatal Borna disease virus infection. Journal of Neurovirology 6: 462477.Google Scholar
Figure 0

Fig. 1. The intranuclear replication of bornaviruses. The viral ribonucleoprotein (RNP) complex is imported into the nucleus. It binds to the nuclear chromatin and generates more of these RNP complexes. The virus rarely forms complete virions and as a result, it probably spreads between cells in the form of the RNP.

Figure 1

Fig. 2. The major sites within the equine brain that BoDV-1 appears to favor. Infected neurons are also scattered diffusely throughout the cerebrum. The arrows indicate what is believed to be the ‘natural’ route of viral invasion, originating in the olfactory bulb.

Figure 2

Fig. 3. A schematic diagram showing the multiple alterations in neurogenesis induced by BoDV-1 in neonatal mice.

Figure 3

Fig. 4. The three major mechanisms of neuronal destruction mediated by BoDV-1. These are, T-cell-mediated cytotoxicity, microglial activation resulting in cytokine-mediated neurotoxicity, and loss of astrocyte function resulting in glutamate accumulation and excitotoxic destructions of neurons.

Figure 4

Fig. 5. The role of microglial activation in the pathogenesis of bornaviral encephalitis. M1 activation of the microglia results in the flooding of the brain with multiple cytokines as well as potent oxidants resulting in neuronal death. Should the microglial phenotype change to M2, the resulting cytokines will reduce inflammation and cell destruction and promote repair although destroyed neurons unlikely to be replaced.

Figure 5

Fig. 6. The role of glutamate in BoDV-1 encephalitis. The normal glutamate cycle requires that astrocytes remove excess glutamate from the extracellular fluid, convert it to glutamine and return it to the neurons. If astrocytes are damaged glutamate accumulates and kills neurons. GABAergic neurons release GABA that inhibits glutamate excitotoxicity and microglial activation. If these neurons are also damaged by BoDV-1 then additional neuronal loss will be expected.