<<

Journal of NeuroVirology (1999) 5, 604 ± 612 ã 1999 Journal of NeuroVirology, Inc. http://www.jneurovirology.com

Bornavirus immunopathogenesis in : models for human neurological diseases

Thomas Briese1, Mady Hornig1 and W Ian Lipkin*,1

1Laboratory for the Study of Emerging Diseases, Department of Neurology, 3101 Gillespie Neuroscience Research Facility, University of California, Irvine, California, CA 92697-4292, USA

Although the question of human BDV infection remains to be resolved, burgeoning interest in this unique has provided tools for exploring the pharmacology and neurochemistry of neuropsychiatric disorders poten- tially linked to BDV infection. Two animal models have been established based on BDV infection of adult or neonatal Lewis rats. Analyis of these models is already yielding insights into mechanisms by which neurotropic agents and/or immune factors may impact developing or mature CNS circuitry to effect complex disturbances in movement and behavior.

Keywords: ; neurotropism; humoral and cellular immune

response; Th1 ±Th2 shift; apoptosis; dopamine; cytokines

Introduction

Borna disease virus (BDV), the prototype of a new disorders and schizophrenia (Amsterdam et al, family, , within the nonsegmented 1985; Bode et al, 1988, 1992, 1993; Fu et al, 1993; negative-strand RNA , infects the central Kishi et al, 1995; Waltrip II et al, 1995), others have nervous system (CNS) of warmblooded animals to not succeeded in replicating these ®ndings (Iwata et cause behavioral disturbances reminiscent of au- al, 1998; Kubo et al, 1997; Lieb et al, 1997; Richt et tism, schizophrenia, and mood disorders (Lipkin et al, 1997). Here we review two models of al, 1995). BDV is not lytic in vitro or in vivo, BDV infection that provide insight into mechanisms replicates at lower levels than most known viruses by which neurotropic viruses may impact CNS and is dissimilar in nucleic acid and development and function to effect complex dis- sequence to other infectious agents (de la Torre, turbances in behavior. 1994; Schneemann et al, 1995). The molecular biology of BDV is unusual in many respects including a nuclear localization for replication and Neurotropism and persistence transcription, overlap of open reading frames and transcription units, posttranscriptional modi®cation Neurotropism of BDV is likely to be multifactorial. of subgenomic , and marked conservation of The integrity of the humoral immune response is coding sequence across a wide variety of animal critical to restriction of virus to neural compart- species and tissue culture systems. Natural infec- ments (Stitz et al, 1998); however, replication is still tion has been con®rmed in horses, sheep, cattle, higher in limbic structures in animals with dogs, birds and cats. can be infected compromised humoral immunity; thus, additional experimentally (Stitz et al, 1980). This wide host factors must contribute to neurotropism. One means range suggests that humans are likely to be by which preferential replication of BDV in limbic susceptible to BDV infection; however, there is no structures might occur is through restricted dis- consensus concerning the role of BDV in human tribution of the enzymatic machinery required for disease. Although some investigators report an its lifecycle. The phosphoprotein of BDV (P) is increased prevalence of BDV infection in mood predicted by analogy to phosphoproteins of other nonsegmented negative-strand RNA viruses to serve as a transcriptional activator (de la Torre, 1994; *Correspondence: WI Lipkin Received 3 March 1999; revised 21 May 1999; accepted 29 June Schneemann et al, 1995). It also contains potent 1999 nuclear localization signals (Shoya et al, 1998; Bornavirus rodent models of human CNS diseases TBrieseet al 605 Schwemmle et al, 1999) and interacts with two work by Stitz et al where passive transfer of other BDV , the nucleoprotein and X- neutralizing antibodies resulted in limitation of protein to affect their intracellular distributions viral replication within the CNS (Stitz et al, 1998). (Schwemmle et al, 1998). Thus, P may also have Similar results have been reported in other viral additional functions related to nucleocasid assem- systems; for example, passive transfer of virus- bly and/or protein traf®cking within the . BDV P speci®c antibodies limits viral replication in the is phosphorylated primarily by the epsilon isotype CNS following infection with murine hepatitis virus of protein kinase C (PKCe) (Schwemmle et al, 1997). type-4 (Buchmeier et al, 1984) or virus Interestingly, the regional distributions of PKCe (Liebert et al, 1990), and induces clearance of virus (Saito et al, 1993) and BDV in rat brain (Narayan following infection with (Dietzschold et al, et al, 1983a) are similar, suggesting the possibility 1992) or (Levine et al, 1991). that the localization of PKCe, through its phosphor- ylation effects, may in¯uence the tropism of BDV for limbic circuitry. Adult Lewis rat infection In many CNS viral infections, the presence of an intact immune response results in either viral Bornavirus neuropathogenesis has been studied clearance or host mortality. This is not the case in primarily in adult immunocompetent rodents and BDV, where infection may persist in the presence of ungulates where infection results in dramatic a transient but robust immune response. Persistence disturbances in behavior, limbic circuitry, and may be due to induction of Th1 tolerance. Whereas monoamine neurotransmitter systems. These mod- lymphocytes isolated from brains of acutely in- els are intriguing; however, they are associated with fected rats have potent cytolytic activity, lympho- marked CNS in¯ammation, loss of brain mass and cytes from brains of chronically infected rats do not gliosis and may be less relevant to neuropsychiatric lyse BDV-infected target cells (Sobbe et al, 1997). diseases than those in neonatally infected rats Induction of BDV-speci®c tolerance in chronic where BDV induces subtle disturbances of behavior infection may re¯ect the timecourse for presentation and dysgenesis of the cerebellum and hippocampus of viral antigens in the thymus (Rubin et al, 1995). without robust in¯ammatory cell in®ltration.

Alternatively, Th1 cells may become anergic or As in autism (Anderson, 1994; Ernst et al, 1997), undergo apoptosis due to presentation of BDV schizophrenia (Cooper et al, 1991), and mood antigens in brain without essential costimulatory disorders (Hamner and Diamond, 1996; Kelsoe et signals (Karpas et al, 1994; Khoury et al, 1995; al, 1996; Partonen, 1996), disorders of movement Schwartz, 1992). Support for the latter hypothesis is and behavior in adult BD rats are linked to distinct found in the observation that apoptosis of perivas- changes in CNS dopamine systems (Solbrig et al, cular in¯ammatory cells is most apparent at 5 ± 6 1994, 1995, 1996a,b, 1998) and may be further weeks post infection (p.i.), coincident with the onset linked to serotonin abnormalities (Solbrig et al, of decline in (Hatalski et al, 1998a). 1995). The immune-mediated disorder in adult In an effort to explore differences in host gene infected rats presents clinically as hyperactivity expression during persistent infection that might be and exaggerated startle responses 10 ± 14 days after linked to tolerance, subtractive cloning methods intracerebral infection (Narayan et al, 1983a). The were applied to analysis of RNA from brains of acute phase coincides with in®ltration of mono- acute and persistent adult infected rats. The cytes into the brain, particularly in areas of high persistently infected rats had a dramatic increase viral burden including the hippocampus, amygdala in mRNAs encoding immunoglobulin ± a ®nding and other limbic structures (Carbone et al, 1987). that suggested that Th1 tolerance might re¯ect a Two to three weeks later, rats show high grade

Th1 ±Th2 shift in the immune response. This stereotyped motor behaviors (the continuous repeti- hypothesis was con®rmed by RNase protection tion of behavioral elements such as snif®ng, assays that demonstrated a shift in the brain chewing, scratching, grooming, and self-biting), cytokine mRNA pro®le from Th1 ±Th2 cytokines dyskinesias, dystonias, and ¯exed seated postures and an isotype switch in peripheral blood from IgG (Solbrig et al, 1994), in parallel with the widespread to IgE (Hatalski et al, 1998a,b). Although the distribution of virus in limbic and prefrontal enhanced humoral immune response in chronic circuits. Five to ten per cent of animals become disease does not promote viral clearance, it may obese, achieving body weights up to 300% of play an important role in limiting viral gene normal (Ludwig et al, 1988). expression (Hatalski et al, 1998a). Complementary Central dopamine systems of adult-infected BD work by Hatalski et al (1998b) in the chronic phase animals are more sensitive to dopamine agonists of BD reported increases in intra-CNS production of and antagonists than normal rats. Infected animals IgG antibodies that parallel increases of antibodies have increased locomotor and stereotypic behavior with neutralizing activity against BDV in peripheral following administration of the mixed-acting dopa- blood. De®nitive evidence that humoral immunity mine agonist, dextroamphetamine (Solbrig et al, contributes to BDV tropism emerged from recent 1994). Similarly, enhanced locomotion and stereo- Bornavirus rodent models of human CNS diseases TBrieseet al 606 typies are seen in response to the dopaminergic tor abnormalities have been found. Levels of mRNA reuptake inhibitory effects of cocaine, indicating for somatostatin, cholecystokinin, and glutamic dose-dependent potentiation of dopamine neuro- acid decarboxylase are reduced during the acute transmission (Solbrig et al, 1998). The movement phase and recover toward normal in the chronic and behavior disorder is improved following phase of disease (Lipkin et al, 1988). The choliner- treatment with selective dopamine antagonists; gic system, a major participant in sensorimotor whereas D2-selective antagonists (e.g., raclopride) processing, learning, and memory, also appears to do not affect locomotor responses in BD rats, high be affected in adult infection. A decrease in the doses of selective D1 antagonists (e.g., SCH23390) number of choline acetyltransferase-positive ®bers and atypical dopamine blocking agents with mixed has been observed to begin as early as day 6 p.i. and D1 and D2 antagonist activity such as clozapine progress to nearly complete loss of cholinergic selectively reduce locomotor activity in BD rats but ®bers in hippocampus and neocortex by day 15 not in controls (Solbrig et al, 1994). p.i. (Gies et al, 1998). Preliminary work on HPLC analyses reveal abnormal neurochemistry dysregulation of serotonin and norepinephrine in adult-infected BD rats. Levels of dopamine and systems suggests metabolic hyperactivity of seroto- its major metabolite, dihydroxyphenylacetic acid nin (as evidenced by modest increase in the (DOPAC), are reduced in structures that receive metabolite 5-hydroxyindoleacetic acid [5HIAA]) in dopaminergic terminals including striatum, nu- striatum and of norepinephrine (as evidenced by a cleus accumbens, and olfactory tubercle (Solbrig small increase in 3-methoxy-4-hydroxyphenethyle- et al, 1994). Interestingly, although levels of neglycol [MHPG]) in prefrontal and anterior cingu- dopamine and DOPAC are also reduced in pre- late cortex regions (Solbrig et al, 1995). These frontal cortex, the ratio of DOPAC to dopamine is changes may re¯ect compensatory upregulation or increased suggesting enhanced turnover of dopa- heterotypic sprouting following partial loss of mine in this structure (Solbrig et al, 1996a). dopaminergic afferents to these brain regions. Tyrosine hydroxylase-immunoreactive cells are Selective effects of BDV on serotonin and norepi- depleted in substantia nigra and ventral tegmental nephrine pre- or postsynaptic receptors have not yet area (tyrosine hydroxylase is the rate limiting been investigated. Pharmacological and neurotrans- enzyme in dopamine synthesis and may be used mitter-speci®c molecular probes have also been to identify cells that produce dopamine) (Solbrig et used to characterize endogenous opioid systems in al, 1994). Taken together, these results suggest the adult rat model. Infected animals respond partial dopaminergic deafferentation with compen- abnormally to the opiate antagonist, naloxone, with satory metabolic hyperactivity in nigrostriatal and hyperkinesis and seizures, and also demonstrate mesolimbic dopamine systems. At the receptor increases in striatal preproenkephalin mRNA at 14 level, both pre- and postsynaptic sites of the and 21 days (Fu et al, 1993b), and 45 days after BDV dopamine transmitter system appear to be damaged infection (Solbrig and Lipkin, personal communica- in striatum (caudate-putamen and nucleus accum- tion). However, the mechanisms by which these bens). Dopamine uptake sites, as measured by changes in endogenous opioid systems occur are binding of mazindol, are reduced in nucleus unclear. The marked CNS in¯ammation in adult- accumbens (Solbrig et al, 1996b) and caudate- infected rats makes it dif®cult to determine whether putamen (Solbrig et al, 1998). D2 (but not D1) monoamine, cholinergic, and opiatergic dysfunc- receptor binding is markedly reduced in caudate- tion in BD results from direct effects of the virus, putamen; D2 and D3 receptor binding are reduced virus effects on resident cells of the CNS, or a in nucleus accumbens (Solbrig et al, 1994, 1996a,b). cellular immune response to viral gene products. In contrast, postsynaptic dopamine receptors (D1, It is dif®cult to establish a direct parallel D2, D3) remain intact in prefrontal cortex (Solbrig et between the disturbances observed in adult al, 1996a). The basis for this targeted pathogenesis infected rats and a single human CNS disorder remains unclear; however, within caudate-putamen because infected rats have an evolving syndrome. and nucleus accumbens the postsynaptic dopamine Early phases of disease are reminiscent of the lesions appear to be con®ned to receptors expressed hyperactivity observed in bipolar disorder or from spliced messages (D2, D3) rather than un- attention de®cit disorder. Such interpretations spliced messages (D1). Given that BDV requires the are interesting historically for their in¯uence on host cell splicing machinery for expression of its investigators pursuing connections between BDV genome it has been proposed that competition for and human diseases; however, the most compel- splicing machinery may contribute to selective ling aspects of the syndrome from the vantage pathology within dopamine circuits (Solbrig et al, point of establishing models for human disease 1994). instead focus on movement disorders. These Although the increased locomotor activity, simple and complex behaviors clinically and stereotypic behaviors and dyskinesias of the adult pharmacologically resemble tardive dyskinesia, a BD model are linked to distinct disturbances in common iatrogenic condition that follows use of dopaminergic pathways, additional neuromodula- psychotropic medications, the `on-off' phenomen- Bornavirus rodent models of human CNS diseases TBrieseet al 607 on found in late stages of Parkinsonism, and are Granite and Herdon, 1985). Con®rmation of subtle consistent with some descriptions of postence- abnormalities in motor coordination in neonatally- phalitic Parkinsonism (Solbrig et al, 1999). infected rats would provide a functional correlate to anatomic alterations in cerebellum. However, further studies are needed to evaluate the mechan- Neonatal Lewis rat infection isms by which early postnatal exposure to BDV induces functional damage in either cerebellar or Neonatal rat infection may provide an even more limbic circuitry. intriguing model for neuropsychiatric disorders Early reports indicated that there was no cellular than adult rat infection. Indeed, the cerebellar and in¯ammatory response following neonatal infection hippocampal dysgenesis that is observed in neona- (Carbone et al, 1991; Stitz et al, 1995; Gosztonyi and tally-infected animals (Carbone et al, 1991; Narayan Ludwig, 1995), a phenomenon ascribed to the et al, 1983a) is consistent with the more subtle immaturity of the rat immune system in the neurodevelopmental abnormalities reported by postnatal period. Humoral immune response to some investigators in autism (Kemper and Bauman, BDV in neonatally-infected animals has also been 1993), schizophrenia (Altshuler et al, 1987; Fish et reported to be restricted, with anti-BDV antibody al, 1992), and affective disorders (Soares and Mann, titers remaining below 1 : 10 through 133 days p.i. 1997). Neonatally infected animals display a wide (Carbone et al, 1991). However, marked astrocytosis range of physiologic and neurobehavioral distur- has been noted (Carbone et al, 1991; Gonzalez- bances. A study of behavioral and cognitive changes Dunia et al, 1996; Bautista et al, 1995) in dentate in neonatally infected Wistar rats found spatial and gyrus and cerebellum, suggesting alternate, nonin- aversive learning de®cits, increased motor activity, ¯ammatory pathways for glial activation. Higher and decreased anxiety responses (Dittrich et al, levels of message for tissue factor (TF) are found in 1989). Similar de®cits in spatial learning and infected hippocampus. TF is a member of the class memory were reported in neonatally infected Lewis II cytokine receptor family primarily produced by rats 23 ± 73 days p.i. (Carbone et al, 1996). More astrocytes that plays important roles in cellular recently, play behavior has been reported to be signal transduction, brain function, and neural abnormal in the neonatal model, with decreases in development through its effects on coagulation both initiation of nondominance-related play inter- protease cascades. Although this may be one actions and in response to initiation of play by mechanism by which BDV may alter CNS develop- noninfected, age-matched control animals or by ment (Gonzalez-Dunia et al, 1996), cerebellar infected littermates (Pletnikov et al, 1999). Neona- changes cannot be explained by this mechanism, tally infected animals are smaller than uninfected as TF upregulation is not observed in cerebellum littermates (Carbone et al, 1991; Bautista et al, despite prominent astrocytosis. Furthermore, BDV 1994). The basis for runting is unclear as levels of infection of astrocytes appears to be required for TF glucose, growth hormone, and insulin-like growth upregulation (Gonzalez-Dunia et al, 1996), and factor-1 (Bautista et al, 1994) are normal. The cerebellar astrocytes are reported to be spared from amount of food ingested (Bautista et al, 1995) is BDV infection, at least through 30 days following similar in uninfected and infected rats although the neonatal infection (Bautista et al, 1995). latter have a heightened taste preference for salt Persistent tolerant BDV infection of neonatal rats solutions (Bautista et al, 1994). is linked to hippocampal and cerebellar disorgani- CNS dysfunction in neonatally infected animals zation (Narayan et al, 1983b; Stitz et al, 1995); has been proposed to be linked to direct viral effects however, cytoarchitectonic anomalies in other on morphogenesis of the hippocampus and cere- limbic regions have not been extensively explored. bellum, two structures in rodents that continue to Dentate gyrus involution is evident along with the develop after birth. Carbone and colleagues found a appearance of reactive glial cells (Carbone et al, quantitative relationship of limbic pathology to 1991), suggesting more direct pathways of viral behavioral abnormalities in the neonatal infection cytopathic injury. Cerebellar size is reduced, and model; the extent of neuronal loss in dentate gyrus there is evidence of reactive astrocytosis as demon- appeared to be correlated with the severity of spatial strated by glial ®brillary acidic protein (GFAP) learning and memory de®ciencies in neonatally- reactivity as early as 3 days p.i., preceding the infected Lewis rats (Carbone et al, 1996). Although identi®cation of BDV proteins in the cerebellum. overt ambulatory or cerebellar dysfunction has not Furthermore, reactivity of cerebellar astrocytes and been reported (Carbone et al, 1991), we found loss of cerebellar granule cells occurs without signs impairments in balance and coordination during a of infection in those cell populations at all time- sensitive dowel-walking task (Hatalski, 1996). points through to 30 days p.i. Curiously, Purkinje Because the cerebellum undergoes substantial cells appeared to be the predominant cerebellar cell postnatal development in many mammals, it is population demonstrating BDV proteins, although particularly vulnerable to injury from perinatal these cells did not appear to be selectively lost virus infection (Monjan et al, 1971, 1973; Oster- through day 30 p.i. (Bautista et al, 1995). The Bornavirus rodent models of human CNS diseases TBrieseet al 608 mechanism by which astrocytes are activated in the elements; activation or suppression of second absence of infection, be it directly by BDV or messenger/intracellular signaling pathways; induc- indirectly through elaboration of soluble factors by tion of shifts in excitotoxic elements such as other cell types, is not known. Nonetheless, given quinolinic acid or acute phase proteins such as the role of astrocytes in guiding migration of granule neopterin or b-2-microglobulin; direct alterations of cells during cerebellar development, an assessment neuronal function (e.g., inhibition of long-term of the frequency of astrocyte reactivity in conjunc- potentiation in hippocampus); activation or sup- tion with studies of apoptosis in limbic structures pression of glial cells; or alteration of glial cell may elucidate the relative contributions of migra- proliferation or differentiation (including expres- tional failure and programmed cell death in sion of adhesion molecules such as the integrins) pathogenesis of neonatal infection. (Benveniste, 1997; Mehler et al, 1996). Given that Although previous work suggests subtle func- the postnatal expression of neuronotrophic cyto- tional disturbances of limbic circuitry based upon kine and cytokine receptor mRNAs in brain differs analysis of complex learning behaviors, memory for each cytokine (Benveniste, 1997), and that the capacities, and emotional responses, the evolution sensitivity of neuronal populations to the trophic or of such disturbances and the mechanisms by which apoptosis-inducing effects of cytokines changes BDV induces their underlying neuropathology during development, wide variation in the patterns without invoking in®ltrating in¯ammatory ele- of virus-induced, cytokine-related damage would ments remains to be determined. In an effort to be expected, depending on the relative maturity of more fully de®ne the nature and unfolding of the the evolving nervous system at the time of infection. neurologic syndrome in neonatally-infected Lewis In addition, cell loss induced by either BDV or rats, and to understand the mechanisms of neuro- developmentally-programmed changes may alter pathogenesis in the neonatal model, we established the capacity of resident CNS cells to both produce neonatal infection in Lewis rats and serially and respond to neuronotrophic cytokines. One assessed shifts in neuroanatomy, neurobehavior, means by which a virus might disrupt neural and regional gene expression (Hornig et al, 1999). function and development in the absence of Locomotor activity and stereotypes were assessed in¯ammation is through the induction of neurono- in neonatally infected animals 4, 6 and 12 weeks p.i. trophic cytokines. Neuronotrophic cytokines com- Results of locomotor activity analyses indicated an prise a burgeoning set of immunoregulatory overall signi®cant increase in neonatally infected molecules, including the hematolymphoietic fac- groups relative to controls at all timepoints tested. tors (e.g., interleukins, tumor necrosis factor family, Analysis of data extrapolated from previous studies ), the TGF-b superfamily factors (includ- of locomotor activity in adult infected animals ing TGF-b1, 2, 3; GDNF), and the classic neuro- (Solbrig et al, 1994, 1996b, 1998) revealed that the trophic factors (NGF, BDNF, NT3, NT4/5). A large degree of heightened exploratory locomotor activity subset of the neuronotrophic, hematolymphoietic found at baseline in neonatally infected animals at 6 cytokines may be categorized according to their

and 12 weeks postinfection is greater than in adult origin from one of two types of T-helper cells: Th1 infected animals. Locomotor activity across the (cell-mediated immunity and stimulation of anti-

90 min observation period differed for all neurally gen-presenting cells) or Th2 (humoral or B-cell infected groups relative to noninfected controls. At mediated immunity). 4 weeks following neonatal infection, animals had Brains of infected and noninfected animals were prolonged behavioral inhibition upon introduction removedat2,4,6,12,and24weeksp.i.and to the novel environment (®rst 30 min interval). dissected to collect hippocampus, amygdala, cere- These ®ndings are consistent with greater anxiety in bellum, prefrontal cortex, and nucleus accumbens. novel situations and suggests dysfunction of the RNA samples from individual brain regions were amygdala. At the 60 and 90 min intervals, infected subjected to RNase protection assay to quantitate animals had greater mean activity measures than level of transcripts encoding cytokines interleukin controls. Additionally, infected animals showed no (IL)-1a,IL-1b, IL-2, IL-3, IL-4, IL-5, IL-6, IL-10, TNF- attenuation in exploratory activity at 60 and 90 min, a,TNF-b,IFN-g, and TGF-b and housekeeping genes consistent with spatial memory de®cits and hippo- L32 and GAPDH. Infected rats had higher levels of campal dysfunction. Stereotypic behaviors were mRNAs for cytokine products of CNS macrophages/ also increased in neonatally infected groups relative microglia (IL-1a,IL-1b, IL-6, TNF-a) in hippocam- to noninfected controls. pus, amygdala, cerebellum, prefrontal cortex, and Serial analyses of differential gene expression of nucleus accumbens. Elevated levels of these proin- cytokines, neurotrophic factors, and apoptosis- ¯ammatory cytokines were ®rst apparent at 2 related proteins were pursued by RNase protection weeks, peaked at 4 weeks, then declined at 6 and assays to assess potential contribution of soluble 12 weeks. Alterations in other proin¯ammatory factors to neuropathogenesis. Possible mechanisms cytokines, including IL-2, IL-3, TNF-b,andIFN-g, of cytokine-mediated damage in the context of the were not observed. The fact that cell populations developing brain include: direct effects on neuronal other than macrophages or microglia ± T cells, B Bornavirus rodent models of human CNS diseases TBrieseet al 609 cells, mast cells, bone marrow stromal cells ± are the tions in brain of mRNAs encoding factors associated primary sources for the proin¯ammatory cytokines with apoptosis. Levels of mRNA for FAS and ICE that remained static following neonatal infection (caspase-1), two promoters of apoptosis, were suggests a selective effect of BDV on cells of increased. Levels of mRNA for bcl-x, a factor that microglial or macrophage lineage. inhibits apoptosis, were decreased. Maximal shifts Shifts were also observed in gene expression of were observed at 4 and 6 weeks p.i., closely neurotrophic factors; however, in contrast to the paralleling the increases in proin¯ammatory cyto- diffuse alterations found in gene expression of kines noted earlier. cytokine mRNAs, shifts in neurotrophic factor Anatomic studies were consistent with previous mRNAs were restricted to hippocampus. Decreased reports (Carbone et al, 1991; Narayan et al, 1983b) mRNAs for BDNF and NT3 were prominent in in demonstrating loss of dentate gyrus granule cells hippocampus by 4 weeks p.i., but were still evident and disorganization of cerebellar granule cell layer. by 12 weeks p.i. Although decreased NT3 mRNA However, new ®ndings included in¯ammatory may re¯ect loss of the granule cell population in cellular in®ltrates at week 4 and nearly complete dentate gyrus, the role of BDNF in maintaining loss of cerebellar Purkinje cells by week 6. viability of cells suggests that its downregulation Interestingly, the in¯ammatory in®ltrates were maybeamoreessentialstepinneonatalBDV restricted to the motor, parietal and temporal pathogenesis. cortex; only rare in¯ammatory cells were detected Apoptosis, or programmed cell death, is a in dentate gyrus or cerebellum, the regions where mechanism in which cells undergo chromosome architecture was most disturbed. Terminal deox- condensation, DNA degradation, and morphologic ynucleotidyl transferase dUTP-biotin nick end change in the nuclear membrane (Wyllie, 1995). labeling (TUNEL) was observed in cerebral cortex Apoptosis plays an important role in CNS develop- and dentate gyrus peaking at 4 weeks p.i. and in ment and response to neuronal injury (Bredesen, granule cell layer of cerebellum of neonatally 1995). It is conceivable that abnormal regulation of infected rats at weeks 4 and 6 p.i. Although apoptosis, either failure of normal apoptotic se- apoptosis is described in normally developing rat quences to proceed or excessive activity, may hippocampus as late as day 7 to 10 of postnatal life, contribute to abnormal CNS architecture in neona- it is not found at later timepoints (Toth et al, 1998). tal infections with BDV or other neurotropic The anatomic location of the apoptotic cells and the viruses. Furthermore, apoptosis of antigen-speci®c absence of in¯ammatory cells in hippocampus and lymphocytes might provide an explanation for the cerebellum suggests that at least some aspects of immunotolerant state following neonatal BDV neuropathology in neonatal infection re¯ect apop- infections. Anergy or apoptosis of T cells may result tosis rather than cell-mediated, speci®c immunity if their stimulation by antigen presenting cells to BDV. Efforts are underway to determine which resident to the CNS occurs in the absence of cells in hippocampus and cerebellum express costimulatory signals required for immune activa- soluble mediators that might promote apoptosis. tion, such as MHC Class II antigens (Munn et al, Microglial cells are candidates for this activity. 1996). Various stimuli, such as binding of TNF-a to Recent studies indicate that they are activated in its receptor, can trigger apoptosis; proteins such as hippocampus and cerebellum of neonatally in- the bcl-2 and bax proteins, NF-kappaB and ICE- fected rats (Hornig et al, 1999). related proteases have also been shown to play important roles in regulating apoptosis. TNF-a also stimulates apoptosis in a wide variety of cell types Summary (Benveniste, 1997). Furthermore, a host of excitants or neurotoxins including arachidonic acid, platelet- The issue of human BDV infection remains con- 7 activating factor, free radicals (NO, O2 ), glutamate, troversial. Nonetheless, recent focus on this intri- quinolinate, cysteine, cytokines (TNF-a,IL1-b,IL- guing agent has led to establishment of small 6), amines, and as yet unidenti®ed factors arising animal models for studies in BDV pathogenesis from stimulated macrophages and possibly reactive that may provide insights into human neuropsy- astrocytes may in¯uence apoptosis by excessive chiatric disorders. The two systems presented here, activation of N-methyl-D-aspartate (NMDA) recep- adult and neonatal Lewis rat infection, illustrate the tors (Lipton, 1996). Interestingly, Gosztonyi and complexities of analyzing neural:immune interac- Ludwig have proposed that the targeted pathology tions in a developmental context. Together, they of BDV for two hippocampal cell layers, stratum provide powerful tools for exploring the effects of oriens and stratum radiatum, may be due to their viral and immune factors on CNS development and rich concentration of glutamate and aspartate dynamic models for parsing the anatomy, pharma- receptors (Gosztonyi and Ludwig, 1995). Our RNase cology and neurochemistry of speci®c neuropsy- protection assay analyses revealed complex altera- chiatric syndromes. Bornavirus rodent models of human CNS diseases TBrieseet al 610 References Altshuler LL, Conrad A, Kovelman JA, Scheibel A Dietzschold B, Kao M, Zheng YM, Chen ZY, Maul G, Fu (1987). Hippocampal pyramidal cell orientation in ZF, Rupprecht CE, Koprowski H (1992). Delineation of schizophrenia: a controlled neurohistologic study of putative mechanisms involved in antibody-mediated the Yakovlev Collection. Arch Gen Psychiatry 44: clearance of rabies virus from the central nervous 1094 ± 1098. system. Proc Natl Acad Sci USA 89: 7252 ± 7256. Amsterdam J, Winokur A, Dyson W, Herzog S, Gonzalez Dittrich W, Bode L, Ludwig H, Kao M, Schneider K F, Rott R, Koprowski H (1985). : a (1989). Learning de®ciencies in Borna disease virus- possible etiologic factor in human affective disorders. infected but clinically healthy rats. Biol Psychiatry 26: Arch Gen Psychiatry 42: 1093 ± 1096. 818 ± 828. Anderson GM (1994). Studies on the neurochemistry of Ernst M, Zametkin AJ, Matochik JA, Pascualvaca D, autism. In: The Neurobiology of Autism. Bauman ML, Cohen RM (1997). Low medial prefrontal dopaminer- Kemper TL (eds). Johns Hopkins University Press: gic activity in autistic children. Lancet 350: 638. Baltimore, pp 227 ± 242. Fish B, Marcus J, Hans SL, Auerbach JG, Perdue S Bautista JR, Rubin SA, Moran TH, Schwartz GJ, Carbone (1992). Infants at risk for schizophrenia: sequelae of a KM (1995). Developmental injury to the cerebellum genetic neurointegrative defect: a review and replica- following perinatal Borna disease virus infection. Dev tion analysis of pandysmaturation in the Jerusalem Brain Res 90: 45 ± 53. Infant Development Study. Arch Gen Psychiatry 49: Bautista JR, Schwartz GJ, de la Torre JC, Moran TH, 221 ± 235. Carbone KM (1994). Early and persistent abnormal- Fu ZF, Amsterdam JD, Kao M, Shankar V, Koprowski H, ities in rats with neonatally acquired Borna disease Dietzschold B (1993). Detection of Borna disease virus infection. Brain Res Bull 34: 31 ± 40. virus-antibodies from patients with affective disorders Benveniste EN (1997). Cytokine expression in the by western immunoblot technique. J Affect Disord 27: nervous system. In: Immunology of the Nervous 61 ± 68. System. Keane RW, Hickey WF (eds). Oxford Uni- Fu ZF, Weihe E, Zheng YM, Schafer MKH, Sheng H, versity Press: New York, pp. 419 ± 459. Corisdeo S, Rauscher III FJ, Koprowski H, Dietzschold Bode L, Ferszt R, Czech G (1993). Borna disease virus B (1993b). Differential effects of rabies and Borna infection and affective disorders in man. Arch Virol 7: Disease viruses on immediate-early- and late-gene 159 ± 167. expression in brain tissues. J Virol 67: 6674 ± 6681. Bode L, Riegel S, Lange W, Ludwig H (1992). Human Gies U, Bilzer T, Stitz L, Staiger JF (1998). Disturbance of infections with Borna disease virus: seroprevalence in the cortical cholinergic innervation in Borna disease patients with chronic diseases and healthy indivi- prior to encephalitis. Brain Pathol 8: 39 ± 48. duals. J Med Virol 36: 309 ± 315. Gonzalez-Dunia D, Eddleston M, Mackman N, Carbone Bode L, Riegel S, Ludwig H, Amsterdam J, Lange W, KM, de la Torre JC (1996). Expression of tissue factor Koprowski H (1988). Borna disease virus-speci®c is increased in astrocytes within the central nervous antibodies in patients with HIV infection and with system during persistent infection with Borna disease mental disorders. Lancet ii: 689. virus. J Virol 70: 5812 ± 5820. Bredesen D (1995). Neural apoptosis. Ann Neurol 38: Gosztonyi G, Ludwig H (1995). Borna disease: neuro- 839 ± 851. pathology and pathogenesis. In: Current Topics in Buchmeier MJ, Lewicki HA, Talbot PJ, Knobler RL Microbiology & Immunology, Vol. 190: Borna Disease. (1984). Murine hepatitis virus-4 (strain JHM)-induced Koprowski H, Lipkin WI (eds). Springer-Verlag: neurologic disease is modulated in vivo by mono- Berlin, pp. 39 ± 73. clonal antibody. Virology 132: 261 ± 270. Hamner MB, Diamond BI (1996). Plasma dopamine and Carbone K, Park S, Rubin S, Waltrip R, Vogelsang G norepinephrine correlations with psychomotor retar- (1991). Borna disease: association with a maturation dation, anxiety, and depression in non-psychotic defect in the cellular immune response. J Virol 65: depressed patients: a pilot study. Psychiatry Res 64: 6154 ± 6164. 209 ± 211. Carbone KM, Duchala CS, Grif®n JW, Kincaid AL, Hatalski CG (1996). Alterations in the immune response Narayan O (1987). Pathogenesis of Borna disease in within the central nervous system of rats infected rats: evidence that intra-axonal spread is the major with Borna disease virus: potential mechanisms for route for virus dissemination and the determinant for viral persistence. Ph.D. Thesis, University of Califor- disease incubation. J Virol 61: 3431 ± 3440. nia Irvine. Carbone KM, Silvas PM, Rubin SA, Vogel M, Moran TH, Hatalski CG, Hickey WF, Lipkin WI (1998a). Evolution of Schwartz G (1996). Quantitative correlation of viral the immune response in the central nervous system induced damage to the hippocampus and spatial following infection with Borna disease virus. J learning and memory de®cits. J Neurovirol 2: 195. Neuroimmunol 90: 137 ± 142. Cooper JR, Bloom FE, Roth RH (1991). The Biochemical Hatalski CG, Hickey WF, Lipkin WI (1998b). Humoral Basis of Neuropharmacology. Oxford University Press: immunity in the central nervous system of Lewis rats New York, pp. 332 ± 334. infected with Borna disease virus. J Neuroimmunol de la Torre JC (1994). Molecular biology of Borna disease 90: 128 ± 136. virus: prototype of a new group of animal viruses. J Virol 68: 7669 ± 7675. Bornavirus rodent models of human CNS diseases TBrieseet al 611 Hornig M, Solbrig M, WeissenboÈck H, Lipkin WI (1999). Lipton SA (1996). Similarity of neuronal cell injury and Mechanisms of central nervous system dysfunction in death in AIDS dementia and focal cerebral ischemia: Bornavirus infected rats. Current Topics in Microbiol- potential treatment with NMDA open-channel block- ogy & Immunology, Mechanisms of Neuronal Damage ers and nitric oxide-related species. Brain Pathol 6: in Viral Infections of the Nervous System. Goztonyi G 507 ± 517. (ed). Springer-Verlag: Heidelberg, In Press. Ludwig H, Bode L, Gosztonyi G (1988). Borna disease: a Iwata Y, Takahashi K, Peng X, Fukuda K, Ohno K, persistent disease of the central nervous system. Prog Ogawa T, Gonda K, Mori N, Niwa S, Shigeta S (1998). Med Virol 35: 107 ± 151. Detection and sequence analysis of borna disease Mehler MF, Goldstein H, Kessler JA (1996). Effects of virus p24 RNA from peripheral blood mononuclear cytokines on CNS cells: neurons. In: Cytokines and cells of patients with mood disorders or schizophrenia the CNS. Ransohoff RM, Benveniste EN (eds). CRC and of blood donors. J Virol 72: 10044 ± 10049. Press: Boca Raton, pp 115 ± 150. Karpas WJ, Peterson JD, Miller SD (1994). Anergy in Monjan AA, Cole GA, Gilden DH, Nathanson N (1973). vivo: down regulation of antigen-speci®c CD4+ Th1 Pathogenesis of cerebellar hypoplasia produced by but not Th2 cytokine responses. Int Immunol 6: 721 ± lymphocytic choriomeningitis virus infection of neo- 730. natal rats: evolution of disease following infection at 4 Kelsoe JR, Sadovnick AD, Krisbjarnarson H, Bergesch P, days of age. J Neuropathol Exp Neurol 32: 110 ± 124. Mroczkowski-Parker Z, Drennan M, Rapaport MH, Monjan AA, Gilden DH, Cole GA, Nathanson N (1971). Flodman P, Spence MA, Remick RA (1996). Possible Cerebellar hypoplasia in neonatal rats caused by locus for bipolar disorder near the dopamine trans- lymphocytic choriomeningitis virus. Science 171: porter on chromosome 5. Am J Med Genet 67: 533 ± 194 ± 196. 540. Munn DH, Pressey J, Beall AC, Hudes R, Alderson MR Kemper TL, Bauman ML (1993). The contribution of (1996). Selective activation-induced apoptosis of neuropathologic studies to the understanding of peripheral T cells imposed by macrophages: a autism. Neurol Clin North Am 11: 175 ± 187. potential mechanism of antigen-speci®c peripheral Khoury SJ, Akalin E, Chandraker A, Turka LA, Linsley lymphocyte deletion. J Immunol 156: 523 ± 532. PS, Sayegh MH, Hancock WW (1995). CD28-B7 Narayan O, Herzog S, Frese K, Scheefers H, Rott R costimulatory blockade by CTLA4Ig prevents actively (1983a). Behavioral disease in rats caused by im- induced experimental autoimmune encephalomyelitis munopathological responses to persistent Borna virus and inhibits Th1 but spares Th2 cytokines in the in the brain. Science 220: 1401 ± 1403. central nervous system. J Immunol 155: 4521 ± 4524. Narayan O, Herzog S, Frese K, Scheefers H, Rott R Kishi M, Nakaya T, Nakamura Y, Zhong Q, Iketa K, (1983b). Pathogenesis of Borna disease in rats: Sengo M, Kakinuma M, Kato S, Ikuta K (1995). immune-mediated viral ophthalmoencephalopathy Demonstration of human Borna disease virus RNA causing blindness and behavioral abnormalities. J Inf in human peripheral blood mononuclear cells. FEBS Dis 148: 305 ± 315. Lett 364: 293 ± 297. Oster-Granite ML, Herdon RM (1985). The pathogenesis Kubo K, Fujiyoshi T, Yokoyama MM, Kamei K, Richt JA, of parvovirus-induced cerebellar hypoplasia in the Kitze B, Herzog S, Takigawa M, Sonoda S (1997). Syrian hamster, Mesociretus auratus: ¯uorescent anti- Lack of association of Borna disease virus and human body, foliation, cytoarchitectonic, golgi and electron T-cell leukemia virus type 1 infections with psychia- microscopic studies. J Comp Neurol 169: 481 ± 522. tric disorders among Japanese patients. Clin Diagn Partonen T (1996). Dopamine and circadian rhythms in Lab Immunol 4: 189 ± 194. seasonal affective disorder. Med Hyp 47: 191 ± 192. Levine B, Hardwick JM, Trapp BD, Crawford TO, Pletnikov MV, Rubin SA, Vasudevan K, Moran TH, Bollinger RC, Grif®n DE (1991). Antibody-mediated Carbone KM (1999). Developmental brain injury clearance of infection from neurons. associated with abnormal play behavior in neonatally Science 254: 856 ± 860. Borna disease virus (BDV)-infected rats: a model of Lieb K, Hallensleben W, Czygan M, Stitz L, Staeheli P autism. Behavioral Brain Res 100: 43 ± 50. (1997). No Borna disease virus-speci®c RNA detected Richt JA, Alexander RC, Herzog S, Hooper DC, Kean R, in blood from psychiatric patients in different regions Spitsin S, Bechter K, Schuttler R, Feldmann H, Heiske of Germany. The Bornavirus Study Group. Lancet 350: A, Fu ZF, Dietzschold B, Rott R, Koprowski H (1997). 1002. Failure to detect Borna disease virus infection in Liebert UG, Schneider-Schaulies S, Baczko K, ter Meulen peripheral blood leukocytes from humans with V (1990). Antibody-induced restriction of viral gene psychiatric disorders. J Neurovirol 3: 174 ± 178. expression in measles encephalitis in rats. J Virol 64: Rubin SA, Sierra-Honigmann AM, Lederman HM, 706 ± 713. Waltrip II RW, Eiden JJ, Carbone KM (1995). Lipkin WI, Carbone KM, Wilson MC, Duchala CS, Hematologic consequences of Borna disease virus Narayan O, Oldstone MBA (1988). Neurotransmitter infection of rat bone marrow and thymus stromal abnormalities in Borna disease. Brain Res 475: 366 ± cells. Blood 85: 2762 ± 2769. 370. Lipkin WI, Schneemann A, Solbrig MV (1995). Borna disease virus: implications for human neuropsychia- tric illness. Trends Microbiol 3: 64 ± 69. Bornavirus rodent models of human CNS diseases TBrieseet al 612 Saito N, Itouji A, Totani Y, Osawa I, Koide H, Fujisawa Solbrig MV, Koob GF, Fallon JH, Lipkin WI (1994). N, Ogita K, Tanaka C (1993). Cellular and intracellular Tardive dyskinetic syndrome in rats infected with localization of epsilon-subspecies of protein kinase C Borna disease virus. Neurobiol Dis 1: 111 ± 119. in the rat brain; presynaptic localization of the Solbrig MV, Koob GF, Fallon JH, Reid S, Lipkin WI epsilon-subspecies. Brain Res 607: 241 ± 248. (1996a). Prefrontal cortex dysfunction in Borna Schneemann A, Schneider PA, Lamb RA, Lipkin WI disease virus (BDV)-infected rats. Biol Psychiatry 40: (1995). The remarkable coding strategy of Borna 629 ± 636. disease virus: a new member of the nonsegmented Solbrig MV, Koob GF, Joyce JN, Lipkin WI (1996b). A negative strand RNA viruses. Virology 210: 1±8. neural substrate of hyperactivity in Borna disease: Schwartz RH (1992). Costimulation of T lymphocytes: changes in dopamine receptors. Virology 222: 332 ± the role of CD28, CTLA-4 and B7/BB1 in interleukin-2 338. production and immunotherapy. Cell 71: 1065 ± 1068. Solbrig MV, Koob GF, Lipkin WI (1998). Cocaine Schwemmle M, De B, Shi L, Banerjee A, Lipkin WI sensitivity in Borna disease virus infected rats. (1997). Borna Disease Virus P-protein is phosphory- Pharmacol Biochem Behav 59: 1047 ± 1052. lated by protein kinase Ce and casein kinase II. J Biol Solbrig MV, Koob GF, Lipkin WI (1999). Orofacial Chem 272: 21818 ± 21823. dyskinesia and dystonia in rats infected with Borna Schwemmle M, Salvatore M, Shi L, Richt J, Lee CH, disease virus: a model for tardive dyskinetic syn- Lipkin WI (1998). Interactions of the Borna disease dromes. Mol Psychiatry In Press. virus P, N, and X proteins and their functional Stitz L, Dietzschold B, Carbone KM (1995). Immuno- implications. J Biol Chem 273: 9007 ± 9012. pathogenesis of Borna disease. In: Current Topics in Schwemmle M, Jehle C, Shoemaker T, Lipkin WI (1999). Microbiology & Immunology, Vol. 190: Borna Disease. Characterization of the major nuclear localization Koprowski H, Lipkin WI (eds). Springer-Verlag: signal of the Borna disease virus phosphoprotein. J Heidelberg, pp 75 ± 92. Gen Virol 80: 97 ± 100. Stitz L, Krey H, Ludwig H (1980). Borna disease in Shoya Y, Kobayashi T, Koda T, Ikuta K, Kakinuma M, rhesus monkeys as a model for uveo-cerebral Kishi M (1998). Two proline-rich nuclear localization symptoms. J Med Virol 6: 333 ± 340. signals in the amino- and carboxyl-terminal regions of Stitz L, NoÈske K, Planz O, Furrer E, Lipkin WI, Bilzer T the Borna disease virus phosphoprotein. J Virol 72: (1998). A functional role for neutralizing antibodies in 9755 ± 9762. Borna disease: in¯uence on virus tropism outside the Soares JC, Mann JJ (1997). The anatomy of mood central nervous system. J Virol 72: 8884 ± 8892. disorders ± review of structural neuroimaging studies. Toth Z, Yan XX, Haftoglou S, Ribak CE, Baram TZ Biol Psychiatry 41: 86 ± 106. (1998). Seizure-induced neuronal injury: vulnerability Sobbe M, Bilzer T, Gommel S, Noske K, Planz O, Stitz L to febrile seizures in an immature rat model. J (1997). Induction of degenerative brain lesions after Neurosci 18: 4285 ± 4294. adoptive transfer of brain lymphocytes from Borna Waltrip II RW, Buchanan RW, Summerfelt A, Breier A, disease virus-infected rats: presence of CD8+ T cells Carpenter WT, Bryant NL, Rubin SA, Carbone K and perforin mRNA. J Virol 71: 2400 ± 2407. (1995). Borna disease virus and schizophrenia. Psy- Solbrig MV, Fallon JH, Lipkin WI (1995). Behavioral chiatry Res 56: 33 ± 44. disturbances and pharmacology of Borna disease Wyllie AH (1995). The genetic upregulation of apoptosis. virus. In: Current Topics in Microbiology & Immunol- Curr Opin Genet Dev 5: 97 ± 104. ogy, Vol. 190: Borna Disease. Koprowski H, Lipkin WI (eds). Springer-Verlag: Heidelberg, pp 93 ± 101.