Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
J Virol ; 91(14)2017 07 15.
Article in English | MEDLINE | ID: mdl-28446679

ABSTRACT

Central nervous system infection of neonatal and adult rats with Borna disease virus (BDV) results in neuronal destruction and behavioral abnormalities with differential immune-mediated involvement. Neuroactive metabolites generated from the kynurenine pathway of tryptophan degradation have been implicated in several human neurodegenerative disorders. Here, we report that brain expression of key enzymes in the kynurenine pathway are significantly, but differentially, altered in neonatal and adult rats with BDV infection. Gene expression analysis of rat brains following neonatal infection showed increased expression of kynurenine amino transferase II (KATII) and kynurenine-3-monooxygenase (KMO) enzymes. Additionally, indoleamine 2,3-dioxygenase (IDO) expression was only modestly increased in a brain region- and time-dependent manner in neonatally infected rats; however, its expression was highly increased in adult infected rats. The most dramatic impact on gene expression was seen for KMO, whose activity promotes the production of neurotoxic quinolinic acid. KMO expression was persistently elevated in brain regions of both newborn and adult BDV-infected rats, with increases reaching up to 86-fold. KMO protein levels were increased in neonatally infected rats and colocalized with neurons, the primary target cells of BDV infection. Furthermore, quinolinic acid was elevated in neonatally infected rat brains. We further demonstrate increased expression of KATII and KMO, but not IDO, in vitro in BDV-infected C6 astroglioma cells. Our results suggest that BDV directly impacts the kynurenine pathway, an effect that may be exacerbated by inflammatory responses in immunocompetent hosts. Thus, experimental models of BDV infection may provide new tools for discriminating virus-mediated from immune-mediated impacts on the kynurenine pathway and their relative contribution to neurodegeneration.IMPORTANCE BDV causes persistent, noncytopathic infection in vitro yet still elicits widespread neurodegeneration of infected neurons in both immunoincompetent and immunocompetent hosts. Here, we show that BDV infection induces expression of key enzymes of the kynurenine pathway in brains of newborn and adult infected rats and cultured astroglioma cells, shunting tryptophan degradation toward the production of neurotoxic quinolinic acid. Thus, our findings newly implicate this metabolic pathway in BDV-induced neurodegeneration. Given the importance of the kynurenine pathway in a wide range of human infections and neurodegenerative and neuropsychiatric disorders, animal models of BDV infection may serve as important tools for contrasting direct viral and indirect antiviral immune-mediated impacts on kynurenine pathway dysregulation and the ensuing neurodevelopmental and neuropathological consequences.


Subject(s)
Borna Disease/physiopathology , Borna disease virus/growth & development , Brain/pathology , Host-Pathogen Interactions , Kynurenine/metabolism , Metabolic Networks and Pathways , Quinolinic Acid/toxicity , Animals , Borna Disease/pathology , Cell Line , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Rats
2.
J Vet Med Sci ; 74(4): 419-21, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22123302

ABSTRACT

We investigated the influence of Borna disease virus (BDV) infection on the clinical state of dairy cows. Sera from 149 cows were examined using enzyme-linked immunosorbent assay and western blotting detect antibodies to the BDV-nucleoprotein antigen. Among 149 investigated cows, 25 (16.8%) showed a positive reaction to BDV antigen. No significant difference existed in milk production or medical history between seropositive and seronegative cows. Although the estrus cycle appeared normal even in the seropositive cows, the frequency of artificial insemination and calving-to-conception intervals significantly increased in seropositive cows. Therefore, fertilization failure was recognized in the BDV-antibody positive cows.


Subject(s)
Borna Disease/virology , Borna disease virus/isolation & purification , Cattle Diseases/physiopathology , Cattle Diseases/virology , Reproduction/physiology , Animals , Antibodies, Viral/blood , Borna Disease/epidemiology , Borna Disease/immunology , Borna Disease/physiopathology , Cattle , Cattle Diseases/epidemiology , Cattle Diseases/immunology , Chi-Square Distribution , Enzyme-Linked Immunosorbent Assay/veterinary , Female , Fertilization/physiology , Japan/epidemiology , Lactation , Male , Milk/metabolism , Pregnancy , Seroepidemiologic Studies
3.
J Virol ; 86(5): 2512-22, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22190725

ABSTRACT

Understanding the complex mechanisms by which infectious agents can disrupt behavior represents a major challenge. The Borna disease virus (BDV), a potential human pathogen, provides a unique model to study such mechanisms. Because BDV induces neurodegeneration in brain areas that are still undergoing maturation at the time of infection, we tested the hypothesis that BDV interferes with neurogenesis. We showed that human neural stem/progenitor cells are highly permissive to BDV, although infection does not alter their survival or undifferentiated phenotype. In contrast, upon the induction of differentiation, BDV is capable of severely impairing neurogenesis by interfering with the survival of newly generated neurons. Such impairment was specific to neurogenesis, since astrogliogenesis was unaltered. In conclusion, we demonstrate a new mechanism by which BDV might impair neural function and brain plasticity in infected individuals. These results may contribute to a better understanding of behavioral disorders associated with BDV infection.


Subject(s)
Borna Disease/physiopathology , Borna disease virus/physiology , Neurogenesis , Neurons/cytology , Stem Cells/cytology , Borna Disease/virology , Brain/cytology , Brain/virology , Cells, Cultured , Humans
4.
Cell Tissue Res ; 338(2): 179-90, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19806365

ABSTRACT

Granule cells are major targets of entorhinal afferents terminating in a laminar fashion in the outer molecular layer of the dentate gyrus. Since Borna disease virus (BDV) infection of newborn rats causes a progressive loss of granule cells in the dentate gyrus, entorhinal fibres become disjoined from their main targets. We have investigated the extent to which entorhinal axons react to this loss of granule cells. Unexpectedly, anterograde DiI tracing has shown a prominent layered termination of the entorhinal projection, despite an almost complete loss of granule cells at 9 weeks after infection. Combined light- and electron-microscopic analysis of dendrites at the outer molecular layer of the dentate gyrus at 6 and 9 weeks post-infection has revealed a transient increase in the synaptic density of calbindin-positive granule cells and parvalbuminergic neurons after 6 weeks. In contrast, synaptic density reaches values similar to those of uninfected controls 9 weeks post-infection. These findings indicate that, after BDV infection, synaptic reorganization processes occur at peripheral dendrites of the remaining granule cells and parvalbuminergic neurons, including the unexpected persistence of entorhinal axons in the absence of their main targets.


Subject(s)
Borna Disease/pathology , Borna disease virus , Entorhinal Cortex/pathology , Synapses/virology , Afferent Pathways , Animals , Axons/physiology , Axons/ultrastructure , Borna Disease/physiopathology , Calbindins , Dendrites/physiology , Dendrites/ultrastructure , Dentate Gyrus/pathology , Neurons/metabolism , Neurons/ultrastructure , Neurons/virology , Parvalbumins/metabolism , Rats , S100 Calcium Binding Protein G/metabolism , Synapses/physiology , Synapses/ultrastructure
5.
PLoS Pathog ; 5(5): e1000425, 2009 May.
Article in English | MEDLINE | ID: mdl-19424436

ABSTRACT

Understanding the pathogenesis of infection by neurotropic viruses represents a major challenge and may improve our knowledge of many human neurological diseases for which viruses are thought to play a role. Borna disease virus (BDV) represents an attractive model system to analyze the molecular mechanisms whereby a virus can persist in the central nervous system (CNS) and lead to altered brain function, in the absence of overt cytolysis or inflammation. Recently, we showed that BDV selectively impairs neuronal plasticity through interfering with protein kinase C (PKC)-dependent signaling in neurons. Here, we tested the hypothesis that BDV phosphoprotein (P) may serve as a PKC decoy substrate when expressed in neurons, resulting in an interference with PKC-dependent signaling and impaired neuronal activity. By using a recombinant BDV with mutated PKC phosphorylation site on P, we demonstrate the central role of this protein in BDV pathogenesis. We first showed that the kinetics of dissemination of this recombinant virus was strongly delayed, suggesting that phosphorylation of P by PKC is required for optimal viral spread in neurons. Moreover, neurons infected with this mutant virus exhibited a normal pattern of phosphorylation of the PKC endogenous substrates MARCKS and SNAP-25. Finally, activity-dependent modulation of synaptic activity was restored, as assessed by measuring calcium dynamics in response to depolarization and the electrical properties of neuronal networks grown on microelectrode arrays. Therefore, preventing P phosphorylation by PKC abolishes viral interference with neuronal activity in response to stimulation. Our findings illustrate a novel example of viral interference with a differentiated neuronal function, mainly through competition with the PKC signaling pathway. In addition, we provide the first evidence that a viral protein can specifically interfere with stimulus-induced synaptic plasticity in neurons.


Subject(s)
Borna disease virus/physiology , Neurons/virology , Protein Kinase C/genetics , Synapses/virology , Synaptic Transmission/physiology , Action Potentials , Animals , Blotting, Western , Borna Disease/physiopathology , Cells, Cultured , Fluorescent Antibody Technique , Mutation , Neuronal Plasticity/physiology , Patch-Clamp Techniques , Phosphoproteins , Phosphorylation , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Viral Proteins/genetics , Viral Proteins/metabolism
6.
J Chem Neuroanat ; 37(2): 118-27, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19038327

ABSTRACT

Neonatal Borna disease virus (BDV) infection of the Lewis rat leads to progressive degeneration of dentate gyrus granule cells, and cerebellar Purkinje neurons. Our aim here was to clarify whether BDV interfered with the formation of electrical synapses, and we, therefore, analysed expression of the neuronal gap junction protein connexin36 (Cx36) in the Lewis rat hippocampal formation, and cerebellar cortex, 4 and 8 weeks after neonatal infection. Semiquantitative RT-PCR, revealed a BDV-dependent decrease in Cx36 mRNA in the hippocampal formation 4 and 8 weeks post-infection (p.i.), and in the cerebellar cortex 8 weeks p.i. Correspondingly, immunofluorescent staining revealed reduced Cx36 immunoreactivity in both dentate gyrus, and ammons horn CA3 region, 4 and 8 weeks post-infection. In the cerebellar cortex, Cx36 immunoreactivity was detected only 8 weeks post-infection in the molecular layer, where it was down regulated by BDV. Our findings demonstrate, for the first time, distinct BDV-dependent reductions in Cx36 mRNA and protein in the rat hippocampal formation and cerebellar cortex, suggesting altered neuronal network properties to be an important feature of persistent viral brain infections.


Subject(s)
Borna Disease/metabolism , Cerebellar Cortex/metabolism , Connexins/genetics , Gap Junctions/metabolism , Hippocampus/metabolism , Nerve Degeneration/metabolism , Age Factors , Animals , Animals, Newborn , Borna Disease/pathology , Borna Disease/physiopathology , Cerebellar Cortex/physiopathology , Cerebellar Cortex/virology , Down-Regulation/physiology , Female , Fluorescent Antibody Technique , Gap Junctions/pathology , Gene Expression Regulation/physiology , Hippocampus/physiopathology , Hippocampus/virology , Nerve Degeneration/physiopathology , Nerve Degeneration/virology , Nerve Net/metabolism , Nerve Net/physiopathology , Nerve Net/virology , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Synaptic Transmission/physiology , Gap Junction delta-2 Protein
7.
J Neuroinflammation ; 5: 50, 2008 Nov 11.
Article in English | MEDLINE | ID: mdl-19014432

ABSTRACT

Neonatal Borna disease virus (BDV) infection of the rat brain is associated with microglial activation and damage to certain neuronal populations. Since persistent BDV infection of neurons is nonlytic in vitro, activated microglia have been suggested to be responsible for neuronal cell death in vivo. However, the mechanisms of activation of microglia in neonatally BDV-infected rat brains remain unclear. Our previous studies have shown that activation of microglia by BDV in culture requires the presence of astrocytes as neither the virus nor BDV-infected neurons alone activate microglia. Here, we evaluated the mechanisms whereby astrocytes can contribute to activation of microglia in neuron-glia-microglia mixed cultures. We found that persistent infection of neuronal cells leads to activation of uninfected astrocytes as measured by elevated expression of RANTES. Activation of astrocytes then produces activation of microglia as evidenced by increased formation of round-shaped, MHCI-, MHCII- and IL-6-positive microglia cells. Our analysis of possible molecular mechanisms of activation of astrocytes and/or microglia in culture indicates that the mediators of activation may be soluble heat-resistant, low molecular weight factors. The findings indicate that astrocytes may mediate activation of microglia by BDV-infected neurons. The data are consistent with the hypothesis that microglia activation in the absence of neuronal damage may represent initial steps in the gradual neurodegeneration observed in brains of neonatally BDV-infected rats.


Subject(s)
Astrocytes/immunology , Borna Disease/physiopathology , Brain/physiopathology , Encephalitis/physiopathology , Gliosis/physiopathology , Microglia/immunology , Animals , Astrocytes/virology , Biomarkers/analysis , Biomarkers/metabolism , Borna Disease/immunology , Borna Disease/pathology , Borna disease virus/physiology , Brain/immunology , Brain/virology , Cells, Cultured , Chemokine CCL5/immunology , Chemokine CCL5/metabolism , Chronic Disease , Coculture Techniques , Encephalitis/immunology , Encephalitis/virology , Gliosis/immunology , Gliosis/virology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Microglia/virology , Nerve Degeneration/immunology , Nerve Degeneration/physiopathology , Nerve Degeneration/virology , Neurons/immunology , Neurons/pathology , Neurons/virology , Rats , Rats, Inbred F344
9.
J Neurovirol ; 14(3): 252-60, 2008 May.
Article in English | MEDLINE | ID: mdl-18569459

ABSTRACT

A growing number of environmental and pharmacologic manipulations have been shown to influence adult neurogenesis. Borna disease virus (BDV) in rats causes cortical and subcortical infection with extrapyramidal motor symptoms, and hippocampal infection suppresses neurogenesis. Given the known effects of cannabinoids in promoting neural progenitor cell survival, the authors examined in vivo effects of chronic BDV infection in rats on BrdU-positive progenitor cells in striatum, together with neuroprotective actions of cannabinoids. Birth and survival of BrdU-positive progenitor cells in striatum of BDV-infected rats treated with a general cannabinoid agonist (WIN 55,212 1 mg/kg i.p. b.i.d. x 7 days) were examined, as well as anti-inflammatory, antiviral, and nutritional effects of cannabinoids. Cannabinoid treatment protected BrdU-positive progenitor cells in striatum that were susceptible to virus-induced injury (p < .01) through suppression of microglia activation (p < .001). As a consequence of their anti-inflammatory actions and support of neural progenitor cell survival, cannabinoids may be adjunctive treatment for encephalitides with microglial inflammation and neurodegeneration.


Subject(s)
Benzoxazines/administration & dosage , Benzoxazines/pharmacology , Borna Disease/drug therapy , Borna disease virus , Cannabinoids , Corpus Striatum/drug effects , Morpholines/administration & dosage , Morpholines/pharmacology , Naphthalenes/administration & dosage , Naphthalenes/pharmacology , Neurons/drug effects , Stem Cells/drug effects , Animals , Anti-Inflammatory Agents , Borna Disease/physiopathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Chronic Disease , Corpus Striatum/cytology , Drug Administration Schedule , Inflammation/drug therapy , Inflammation/pathology , Injections, Intraperitoneal , Male , Microglia/cytology , Neurons/physiology , Rats , Rats, Inbred Lew , Stem Cells/physiology
10.
Neurosci Lett ; 431(1): 81-5, 2008 Jan 24.
Article in English | MEDLINE | ID: mdl-18155836

ABSTRACT

Our previous studies have shown that the persistent expression of Borna disease virus phosphoprotein (BDV P) in mice leads to behavioral abnormalities resembling those in BDV-infected animals. In this study, we investigated whether the neurobehavioral abnormalities genetically induced by BDV P influence experimental prion disease. The effect of the phosphoprotein on prion diseases was evaluated based on the incubation time and survival curve, as well as the abnormal isoform of prion protein (PrP(Sc)) levels in brains of BDV P Tg mice treated with proteinase K (PK) treatment and subjected to western blotting. Increased expression of the BDV P transgene had no effect on the PrP(Sc) level, incubation time, or survival curve. The abnormalities induced by BDV P are different from those induced by prion diseases, indicating that the signaling cascades induced by the phosphoprotein differ from those induced by prion diseases.


Subject(s)
Borna Disease/genetics , Brain/metabolism , Brain/virology , PrPSc Proteins/metabolism , Prion Diseases/genetics , Viral Proteins/genetics , Animals , Behavior, Animal/physiology , Borna Disease/metabolism , Borna Disease/physiopathology , Borna disease virus/metabolism , Brain/physiopathology , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Cerebral Cortex/virology , Disease Models, Animal , Disease Progression , Endopeptidase K/metabolism , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphoproteins/genetics , Phosphoproteins/metabolism , Prion Diseases/metabolism , Prion Diseases/physiopathology , Signal Transduction/genetics , Survival Rate , Time Factors , Transgenes/genetics , Viral Proteins/metabolism
11.
J Virol ; 81(16): 8833-7, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17553893

ABSTRACT

The mechanisms whereby Borna disease virus (BDV) can impair neuronal function and lead to neurobehavioral disease are not well understood. To analyze the electrophysiological properties of neurons infected with BDV, we used cultures of neurons grown on multielectrode arrays, allowing a real-time monitoring of the electrical activity across the network shaped by synaptic transmission. Although infection did not affect spontaneous neuronal activity, it selectively blocked activity-dependent enhancement of neuronal network activity, one form of synaptic plasticity thought to be important for learning and memory. These findings highlight the original mechanism of the neuronal dysfunction caused by noncytolytic infection with BDV.


Subject(s)
Borna Disease/physiopathology , Borna disease virus , Cerebral Cortex/physiopathology , Neuronal Plasticity , Synapses/virology , Animals , Cerebral Cortex/virology , Electrophysiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Neurons/virology , Phosphoproteins/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Synaptic Transmission , Viral Structural Proteins/metabolism
12.
J Virol ; 81(11): 5940-8, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17376896

ABSTRACT

Borna disease virus (BDV) is a neurotropic virus that causes a persistent infection in the central nervous system (CNS) of many vertebrate species. Although a severe reactive gliosis is observed in experimentally BDV-infected rat brains, little is known about the glial reactions contributing to the viral persistence and immune modulation in the CNS. In this regard, we examined the expression of an astrocyte-derived factor, S100B, in the brains of Lewis rats persistently infected with BDV. S100B is a Ca(2+)-binding protein produced mainly by astrocytes. A prominent role of this protein appears to be the promotion of vascular inflammatory responses through interaction with the receptor for advanced glycation end products (RAGE). Here we show that the expression of S100B is significantly reduced in BDV-infected brains despite severe astrocytosis with increased glial fibrillary acidic protein immunoreactivity. Interestingly, no upregulation of the expression of S100B, or RAGE, was observed in the persistently infected brains even when incited with several inflammatory stimuli, including lipopolysaccharide. In addition, expression of the vascular cell adhesion molecule 1 (VCAM-1), as well as the infiltration of encephalitogenic T cells, was significantly reduced in persistently infected brains in which an experimental autoimmune encephalomyelitis was induced by immunization with myelin-basic protein. Furthermore, we demonstrated that the continuous activation of S100B in the brain may be necessary for the progression of vascular immune responses in neonatally infected rat brains. Our results suggested that BDV infection may impair astrocyte functions via a downregulation of S100B expression, leading to the maintenance of a persistent infection.


Subject(s)
Astrocytes/metabolism , Borna Disease/pathology , Borna disease virus/physiology , Brain/blood supply , Brain/pathology , Down-Regulation/physiology , Nerve Growth Factors/antagonists & inhibitors , S100 Proteins/antagonists & inhibitors , Vasculitis, Central Nervous System/pathology , Animals , Astrocytes/pathology , Borna Disease/metabolism , Borna Disease/physiopathology , Brain/virology , Chronic Disease , Nerve Growth Factors/biosynthesis , Rats , Rats, Inbred Lew , S100 Calcium Binding Protein beta Subunit , S100 Proteins/biosynthesis , Vasculitis, Central Nervous System/physiopathology , Vasculitis, Central Nervous System/virology
13.
J Virol ; 80(24): 12141-8, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17020949

ABSTRACT

Neonatal Borna disease virus (BDV) infection of the rat brain is associated with microglial activation and damage to the certain neuronal populations. Since persistent BDV infection of neurons in vitro is noncytolytic and noncytopathic, activated microglia have been suggested to be responsible for neuronal cell death in vivo. However, the mechanisms of activation of microglia in neonatally BDV-infected rat brain have not been investigated. To address these issues, activation of primary rat microglial cells was studied following exposure to purified BDV or to persistently BDV-infected primary cortical neurons or after BDV infection of primary mixed neuron-glial cultures. Neither purified virus nor BDV-infected neurons alone activated primary microglia as assessed by the changes in cell shape or production of the proinflammatory cytokines. In contrast, in the BDV-infected primary mixed cultures, we observed proliferation of microglia cells that acquired the round morphology and expressed major histocompatibility complex molecules of classes I and II. These manifestations of microglia activation were observed in the absence of direct BDV infection of microglia or overt neuronal toxicity. In addition, compared to uninfected mixed cultures, activation of microglia in BDV-infected mixed cultures was associated with a significantly greater lipopolysaccharide-induced release of tumor necrosis factor alpha, interleukin 1beta, and interleukin 10. Taken together, the present data are the first in vitro evidence that persistent BDV infection of neurons and astrocytes rather than direct exposure to the virus or dying neurons is critical for activating microglia.


Subject(s)
Borna Disease/physiopathology , Borna disease virus , Microglia/physiology , Microglia/virology , Animals , Cells, Cultured , Cytotoxicity Tests, Immunologic , Histocompatibility Antigens/metabolism , Immunohistochemistry , In Vitro Techniques , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Microglia/cytology , Microglia/metabolism , Rats , Tumor Necrosis Factor-alpha/metabolism
14.
Wei Sheng Wu Xue Bao ; 46(4): 676-9, 2006 Aug.
Article in Chinese | MEDLINE | ID: mdl-17037079

ABSTRACT

Borna disease virus (BDV) is highly neuronotropic. Recently, more and more investigations indicated that BDV infection was close related to human neuropsychic disorders. However, the mechanism underlying the disorders was unclear to date. Some investigators thought neuronal plasticity changes by BDV infection in the central nervous system may be it's cardinal basis. Many researchers have studied the molecular mechanisms, which might lead to disturbances in neuronal plasticity by BDV infection, through infection-based animal modes such as gerbils modes, rats modes, mice modes and transgenic mice modes. The results showed that neuronal plasticity was interfered by BDV infection through interference with trophic support from astrocytes to neurons, interference with amphoterin signaling and interference with neurotrophin signaling. The disturbances in neuronal plasticity would result in cerebral disorders and cause behavior alterations and psychic alterations of host. In the following years, the ability to manipulate the BDV genome may help uncover unrecognized aspects of the basic mechanisms operating in the regulation of neuronal plasticity of interference with neuronal physiology caused by BDV, give academic foundation for clinical prevention and therapy of BD.


Subject(s)
Borna Disease/pathology , Borna Disease/physiopathology , Borna disease virus/physiology , Neuronal Plasticity/physiology , Animals , Humans
15.
PLoS Pathog ; 2(3): e19, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16552443

ABSTRACT

Infection by Borna disease virus (BDV) enables the study of the molecular mechanisms whereby a virus can persist in the central nervous system and lead to altered brain function in the absence of overt cytolysis and inflammation. This neurotropic virus infects a wide variety of vertebrates and causes behavioral diseases. The basis of BDV-induced behavioral impairment remains largely unknown. Here, we investigated whether BDV infection of neurons affected synaptic activity, by studying the rate of synaptic vesicle (SV) recycling, a good indicator of synaptic activity. Vesicular cycling was visualized in cultured hippocampal neurons synapses, using an assay based on the uptake of an antibody directed against the luminal domain of synaptotagmin I. BDV infection did not affect elementary presynaptic functioning, such as spontaneous or depolarization-induced vesicular cycling. In contrast, infection of neurons with BDV specifically blocked the enhancement of SV recycling that is observed in response to stimuli-induced synaptic potentiation, suggesting defects in long-term potentiation. Studies of signaling pathways involved in synaptic potentiation revealed that this blockade was due to a reduction of the phosphorylation by protein kinase C (PKC) of proteins that regulate SV recycling, such as myristoylated alanine-rich C kinase substrate (MARCKS) and Munc18-1/nSec1. Moreover, BDV interference with PKC-dependent phosphorylation was identified downstream of PKC activation. We also provide evidence suggesting that the BDV phosphoprotein interferes with PKC-dependent phosphorylation. Altogether, our results reveal a new mechanism by which a virus can cause synaptic dysfunction and contribute to neurobehavioral disorders.


Subject(s)
Borna Disease/physiopathology , Borna Disease/virology , Borna disease virus/physiology , Presynaptic Terminals/physiology , Protein Kinase C/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Enzyme Activation , Molecular Sequence Data , Neurons/physiology , Rats , Synapses/physiology , Synaptic Vesicles/physiology
16.
Exp Neurol ; 194(2): 355-62, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16022863

ABSTRACT

Dyskinesias and seizures are both medically refractory disorders for which cannabinoid-based treatments have shown early promise as primary or adjunctive therapy. Using the Borna disease (BD) virus rat, an animal model of viral encephalopathy with spontaneous hyperkinetic movements and seizure susceptibility, we identified a key role for endocannabinoids in the maintenance of a balanced tone of activity in extrapyramidal and limbic circuits. BD rats showed significant elevations of the endocannabinoid anandamide in subthalamic nucleus, a relay nucleus compromised in hyperkinetic disorders. While direct and indirect cannabinoid agonists had limited motor effects in BD rats, abrupt reductions of endocannabinoid tone by the CB1 antagonist SR141716A (0.3 mg/kg, i.p.) caused seizures characterized by myoclonic jerks time-locked to periodic spike/sharp wave discharges on hippocampal electroencephalography. The general opiate antagonist naloxone (NLX) (1 mg/kg, s.c.), another pharmacologic treatment with potential efficacy in dyskinesias or L-DOPA motor complications, produced similar seizures. No changes in anandamide levels in hippocampus and amygdala were found in convulsing NLX-treated BD rats. In contrast, NLX significantly increased anandamide levels in the same areas of normal uninfected animals, possibly protecting against seizures. Pretreatment with the anandamide transport blocker AM404 (20 mg/kg, i.p.) prevented NLX-induced seizures. These findings are consistent with an anticonvulsant role for endocannabinoids, counteracting aberrant firing produced by convulsive agents, and with a functional or reciprocal relation between opioid and cannabinoid tone with respect to limbic convulsive phenomena.


Subject(s)
Borna Disease/drug therapy , Cannabinoid Receptor Modulators/pharmacology , Endocannabinoids , Movement Disorders/drug therapy , Seizures/drug therapy , Animals , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Arachidonic Acids/metabolism , Arachidonic Acids/pharmacology , Basal Ganglia/drug effects , Basal Ganglia/physiopathology , Basal Ganglia/virology , Borna Disease/physiopathology , Borna Disease/virology , Cannabinoid Receptor Modulators/therapeutic use , Convulsants/antagonists & inhibitors , Disease Models, Animal , Limbic System/drug effects , Limbic System/physiopathology , Limbic System/virology , Male , Movement Disorders/physiopathology , Movement Disorders/virology , Naloxone/antagonists & inhibitors , Narcotic Antagonists/pharmacology , Piperidines/antagonists & inhibitors , Polyunsaturated Alkamides , Pyrazoles/antagonists & inhibitors , Rats , Rats, Inbred Lew , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Rimonabant , Seizures/physiopathology , Seizures/virology
17.
Virus Res ; 111(2): 224-34, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15885838

ABSTRACT

Viruses able to infect the central nervous system (CNS) are increasingly being recognized as important factors that can cause mental diseases by interfering with neuronal plasticity. The mechanisms whereby such infections disturb brain functions are beginning to emerge. Borna disease virus (BDV), which causes a persistent infection of neurons without direct cytolysis in several mammalian hosts, has recently gained interest as a unique model to study the mechanisms of viral interference with neuronal plasticity. This review will summarize several hypotheses that have been put forward to explain possible levels of BDV interference with brain function.


Subject(s)
Borna disease virus/pathogenicity , Neuronal Plasticity , Neurons/virology , Animals , Animals, Newborn , Borna Disease/physiopathology , Borna Disease/virology , Cells, Cultured , Central Nervous System Viral Diseases/physiopathology , Central Nervous System Viral Diseases/virology , Disease Models, Animal , Gerbillinae , Humans , Mice , Mice, Transgenic , Rats , Tupaiidae/virology
18.
J Virol ; 79(1): 314-25, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15596826

ABSTRACT

The Parapoxvirus Orf virus represents a promising candidate for novel vector vaccines due to its immune modulating properties even in nonpermissive hosts such as mouse or rat. The highly attenuated Orf virus strain D1701 was used to generate a recombinant virus (D1701-VrVp40) expressing nucleoprotein p40 of Borna disease virus, which represents a major antigen for the induction of a Borna disease virus-specific humoral and cellular immune response. Infection with Borna disease virus leads to distinct neurological symptoms mediated by the invasion of activated specific CD8+ T cells into the infected brain. Usually, Borna disease virus is not cleared from the brain but rather persists in neural cells. In the present study we show for the first time that intramuscular application of the D1701-VrVp40 recombinant protected rats against Borna disease, and importantly, virus clearance from the infected brain was demonstrated in immunized animals. Even 4 and 8 months after the last immunization, all immunized animals were still protected against the disease. Initial characterization of the immune cells attracted to the infected brain areas suggested that D1701-VrVp40 mediated induction of B cells and antibody-producing plasma cells as well as T cells. These findings suggest the induction of various defense mechanisms against Borna disease virus. First studies on the role of antiviral cytokines indicated that D1701-VrVp40 immunization did not lead to an enhanced early response of gamma or alpha interferon or tumor necrosis factor alpha. Collectively, this study describes the potential of the Orf virus vector system in mediating long-lasting, protective antiviral immunity and eliminating this persistent virus infection without provoking massive neuronal damage.


Subject(s)
Borna Disease/prevention & control , Borna disease virus/immunology , Orf virus/genetics , Viral Proteins/immunology , Viral Vaccines/administration & dosage , Animals , Antibodies, Viral/blood , Borna Disease/immunology , Borna Disease/physiopathology , Borna Disease/virology , Borna disease virus/physiology , Brain/virology , Cell Line , Cytokines/metabolism , Guinea Pigs , Immunization , Orf virus/metabolism , RNA, Viral/analysis , Rats , Rats, Inbred Lew , Recombination, Genetic , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Vaccines/genetics , Viral Vaccines/immunology
19.
J Neurovirol ; 10(5): 267-77, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15385249

ABSTRACT

Neonatal Borna disease virus (BDV) infection of the rat brain serves as a valuable model for studying the pathogenesis of neurodevelopmental abnormalities following early brain injury. Previous experiments have demonstrated significant alterations in regional tissue content of serotonin (5-HT) in neonatally BDV-infected Lewis rats. The present study sought to provide more insights into postnatal virus-associated alterations in 5-HT neurotransmission by evaluating the density of 5-HT1a receptors in the hippocampus and 5-HT2a receptors in the cortex, regional 5-HT tissue concentrations, behavioral responses to a 5-HT agonist, quipazine, and numbers of neurons in specific subfields of the hippocampus on days 7, 14, and 30 after neonatal BDV infection in Lewis rats. Neonatal BDV infection was found to be associated with a gradual increase in the density of 5-HT2a and 5-HT1a postsynaptic receptors followed by an elevation of 5-HT contents at both the levels of synaptic terminals (i.e., cortex and hippocampus) and cell bodies (i.e., raphe nuclei). In addition, there was an enhanced behavioral response to quipazine. Virus-associated neurochemical and behavioral changes were accompanied by a decline in the number of neurons in the dentate gyrus and in the CA1 field of the hippocampus. No change in the number of neurons in the CA3/2 field of the hippocampus was observed. The present pattern of BDV-associated alterations in 5-HT brain system along with available data from other laboratories suggest that BDV might compromise axonal transport and/or release of 5-HT, resulting in decreased 5-HT neurotransmission.


Subject(s)
Borna Disease/physiopathology , Borna disease virus , Neurotransmitter Agents/metabolism , Receptors, Serotonin, 5-HT1/metabolism , Animals , Animals, Newborn , Borna Disease/metabolism , Borna Disease/virology , Borna disease virus/immunology , Brain Chemistry/drug effects , Brain Chemistry/genetics , Disease Models, Animal , Hippocampus/chemistry , Hippocampus/growth & development , Hippocampus/virology , Neurotransmitter Agents/genetics , Rats , Serotonin/metabolism
20.
Viral Immunol ; 17(1): 79-85, 2004.
Article in English | MEDLINE | ID: mdl-15018664

ABSTRACT

Borna disease virus (BDV) can induce severe neurological disorder in Lewis rats and MRL mice. Antiviral CD8 T cells have been shown to be the mediators of disease in these animals. To define molecules involved in the disease process, we performed infection studies in MRL mice lacking either interferon-gamma, a functional Fas/FasL system, chemokine receptor CXCR3, or inducible NO synthase. We further used transgenic MRL mice expressing interferon-gamma-inducible, T cell-attracting chemokine CXCL10 in brain astrocytes. After intracerebral infection with BDV, wild-type and mutant mice developed CD8 T cell responses and neurological disease at similar frequency and with similar kinetics, suggesting that these factors are not required for initiation and maintenance of the immunopathological process. Similarly, the course of disease could not be altered by treating infected MRL mice or Lewis rats with the drug L-N(6)-(1-iminoethyl)-lysine (L-NIL) that specifically blocks the activity of the inducible NO synthase. We therefore have excluded a number of important factors that have been demonstrated to be crucial in the pathogenesis of a broad number of pathologic conditions. Thus, BDV-induced disease may not result from the action of a single dominant T cell-dependent effector molecule. Disease rather reflects a combined influence of several as yet undefined factors from CD8 T cells.


Subject(s)
Borna Disease/immunology , Borna Disease/physiopathology , Borna disease virus/pathogenicity , Nervous System Diseases/immunology , Nervous System Diseases/physiopathology , Animals , Borna Disease/virology , CD8-Positive T-Lymphocytes/immunology , Enzyme Induction , Interferon-gamma/genetics , Interferon-gamma/metabolism , Mice , Mice, Knockout , Nervous System Diseases/virology , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Rats , Rats, Inbred Lew , Receptors, CXCR3 , Receptors, Chemokine/genetics , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...