Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Neuropathology ; 37(5): 398-406, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28493345

ABSTRACT

The apoptosis of pyramidal neurons in CA2 and CA3 subregions of the hippocampus is induced after infection with Mu-3 virus (Mu-3), a neuropathogenic strain of the JHM virus (JHMV), at 4-5 days post-inoculation (dpi). The viral antigens in the hippocampus are mainly found in the CD11b-positive cells distributed in the stratum oriens located outside the pyramidal layer, and only a few pyramidal neurons are infected. Furthermore, the apoptotic cells, indicated as showing caspase 3 (Cas3) activation, consist of a high number of uninfected cells. Therefore, it is considered that the apoptotic lesions occur through the indirect effects of infection, and not as a result of direct infection with Mu-3, similar to the reported neuronal apoptosis in the hippocampus after other types of infection. The apoptosis in the pyramidal neurons is accompanied by various types of proinflammatory cytokines depending on the causative agents. Thus, the local expression of proinflammatory cytokines was studied, revealing no correlation in the distribution of cytokine expression with the subregions showing apoptosis. However, the anti-inflammatory cytokine IL-10 was produced by pyramidal neurons of CA2 and CA3 at 3 dpi when there is no destructive change or viral invasion in the hippocampus.


Subject(s)
Apoptosis/immunology , Coronavirus Infections/immunology , Interleukin-10/biosynthesis , Pyramidal Cells/immunology , Pyramidal Cells/virology , Animals , Mice , Murine hepatitis virus , Pyramidal Cells/pathology
2.
Neuropathology ; 37(4): 311-320, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28317173

ABSTRACT

During the early phase of infection with an extremely neurovirulent murine coronavirus, cl-2, the ER-TR7 antigen (ERag)-positive fibers (ERfibs) associated with laminin and collagen III show a rapid increase in expression levels in the meninges, followed by an appearance of the antigens in the ventricle and brain parenchyma. Then, cl-2 invades the ventricle and ventricular wall along the newly assembled ERfibs after infection, using them as a pathway from the meninges, the initial site of infection. In the lymph nodes and spleen, ERag is mainly produced by fibroblastic reticular cells (FRCs), which play a key role in nursing the ERfibs to form a fibroblastic reticular network (FRN). The FRN functions as a conduit system to transfer antigens, cytokines or leukocytes in the lymphoid organs. In the brain parenchyma, astrocytes were found to produce the main components of mature ERfibs, such as collagen, laminin and ERag, which have been identified in the lymphoid organs. The producibility of these extracellular matrices (ECMs) by astrocytes was further confirmed by primary brain cultures, which disclosed the dissociation of laminin and ERag production, and the close association of ERag production with that of collagen, forming a fibrous structure. The pattern of ECM production in vitro indicated the process of forming mature ERfibs in the brain, that is, fibers made of collagen fibers and ERag are wrapped by laminin prepared as a sheet structure. In addition, the brain parenchymal cells that produce interferon ß after infection in spite of their residence away from the sites of viral invasion were surrounded by ERfibs, which were closely associated with astrocytic fibers. These findings indicate that astrocytes play a central role in forming the astrocytic reticular network (ARN) in the brain parenchyma, as FRCs do to form FRN in the lymphoid organs.


Subject(s)
Brain/pathology , Coronavirus Infections/pathology , Extracellular Matrix/pathology , Murine hepatitis virus/pathogenicity , Animals , Astrocytes/pathology , Mice , Mice, Inbred BALB C , Virulence
3.
Neuropathology ; 36(6): 513-526, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27121485

ABSTRACT

cl-2 virus is an extremely neurovirulent murine coronavirus. However, during the initial phase of infection between 12 and 24 h post-inoculation (hpi), the viral antigens are detected only in the meninges, followed by viral spread into the ventricular wall before invasion into the brain parenchyma, indicating that the viruses employ a passage between the meninges and ventricular wall as an entry route into the brain parenchyma. At 48 hpi, the passage was found to be constructed by ER-TR7 antigen (ERag)-positive fibers (ERfibs) associated with laminin and collagen III between the fourth ventricle and meninges at the cerebellopontine angle. The construct of the fibers mimics the reticular fibers of the fibroblastic reticular network, which comprises a conduit system in the lymphoid organs. In the meninges, ERfibs together with collagen fibers, lining in a striped pattern, made up a pile of thin sheets. In the brain parenchyma, mature ERfibs associated with laminin were found around blood vessels. Besides mature ERfibs, immature Erfibs without associations with other extracellular matrix components like laminin and collagen appeared after infection, suggesting that the CNS creates a unique conduit system for immune communication triggered by viral invasion.


Subject(s)
Brain/pathology , Brain/virology , Coronavirus Infections/pathology , Coronavirus Infections/virology , Fibroblasts/virology , Murine hepatitis virus/pathogenicity , Animals , Fibroblasts/pathology , Mice , Mice, Inbred BALB C , Virulence
4.
Jpn J Infect Dis ; 69(5): 405-13, 2016 Sep 21.
Article in English | MEDLINE | ID: mdl-26902214

ABSTRACT

Although Lewis X (Le(x)), a carbohydrate structure, is involved in innate immunity through cell-to-cell and pathogen recognition, its expression has not been observed in mouse monocytes/macrophages (Mo/Mas). The Mo/Mas that infiltrate the meninges after infection with the neuropathogenic murine coronavirus strain srr7 are an initial target of infection. Furthermore, higher inflammatory responses were observed in gene-manipulated mice lacking α1,3-fucosyltransferase 9, which determines the expression of the Le(x) structure, than in wild type mice after infection. We investigated Le(x) expression using CD11b-positive peritoneal exudate cells (PECs) and found that Le(x) is inducible in Mo/Mas after infection with srr7, especially in the syncytial cells during the late phase of infection. The number of syncytial cells was reduced after treatment of the infected PECs with anti-Le(x) antibody, during the late phase of infection. In addition, the antibody treatment induced a marked reduction in the number of the infected cells at 24 hours post inoculation, without changing the infected cell numbers during the initial phase of infection. These data indicate that the Le(x) structure could play a role in syncytial formation and cell-to-cell infection during the late phase of infection.


Subject(s)
Coronavirus/growth & development , Lewis X Antigen/metabolism , Macrophages/immunology , Macrophages/virology , Monocytes/immunology , Monocytes/virology , Animals , Cells, Cultured , Coronavirus/physiology , Macrophages/chemistry , Mice, Inbred BALB C , Monocytes/chemistry , Virus Internalization
5.
Pathol Int ; 64(5): 199-208, 2014 May.
Article in English | MEDLINE | ID: mdl-24888773

ABSTRACT

Carbohydrate structures, including Lewis X (Le(x)), which is not synthesized in mutant mice that lack α1,3-fucosyltransferase 9 (Fut9(-/-)), are involved in cell-cell recognition and inflammation. However, immunological alteration in Fut9(-/-) mice has not been studied. Thus, the inflammatory response of Fut9(-/-) mice was examined using the highly neurovirulent mouse hepatitis virus (MHV) JHMV srr7 strain. Pathological study revealed that inflammation induced in the brains of Fut9(-/-) mice after infection was more extensive compared with that of wild-type mice, although viral titers obtained from the brains of mutant mice were lower than those of wild-type mice. Furthermore, the reduction in cell numbers in the spleens of wild-type mice after infection was not observed in the infected Fut9(-/-) mice. Although there were no clear differences in the levels of cytokines examined in the brains between Fut9(-/-) and wild-type mice except for interferon-ß expression, some of those in the spleens, including interferon-γ, interleukin-6, and monocyte chemoattractant protein 1, showed higher levels in Fut9(-/-) than in wild-type mice. Furthermore, Fut9(-/-) mice were refractory to the in vivo inoculation of endotoxin (LPS) compared with wild-type mice. These results indicate that Le(x) structures are involved in host responses against viral or bacterial challenges.


Subject(s)
Fucosyltransferases/deficiency , Immunity, Innate/immunology , Lipopolysaccharides/immunology , Murine hepatitis virus/immunology , Animals , Brain/pathology , Brain/virology , Cell Count , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Coronavirus Infections/prevention & control , Cytokines/metabolism , Disease Models, Animal , Fucosyltransferases/genetics , Fucosyltransferases/physiology , Immunity, Innate/genetics , Immunity, Innate/physiology , Mice , Mice, Knockout , Mice, Mutant Strains , Murine hepatitis virus/isolation & purification , Spleen/pathology , Spleen/virology
6.
Jpn J Infect Dis ; 67(1): 9-16, 2014.
Article in English | MEDLINE | ID: mdl-24451095

ABSTRACT

The mutant virus Mu-3 was isolated from the soluble receptor-resistant mutant 7 virus (srr7), which is a neuropathogenic strain of the mouse hepatitis virus JHMV, and cloned as a soluble receptor-resistant mutant from the highly neuropathogenic JHMV strain cl-2 virus (cl-2). In order to identify specific characteristics of Mu-3, the pathology of Mu-3-infected mice was compared with that of srr7- and cl-2-infected mice. The neuropathology after Mu-3 infection exhibited a mixed pattern comparable to that induced by srr7 and cl-2 infections. In addition, Mu-3 infection caused marked apoptotic lesions in the hippocampal region, particularly in the CA2 and CA3 subregions, in the brains of all infected mice. In contrast, in cl-2 infection, 10-20% of the infected mice exhibited apoptosis in the hippocampus, which was primarily observed in the CA1 subregion. Apoptosis also occurred in the pyramidal neurons and CD11b-bearing cells. The apoptotic cells, indicated by caspase 3-activation, were a mixed population of infected and a higher number of uninfected cells. These data indicated that apoptosis observed in Mu-3 infection could be induced by the indirect effects of infection in addition to direct effects of the infected cells occurring in a cell-autonomous manner.


Subject(s)
Apoptosis , Coronavirus Infections/veterinary , Hepatitis, Viral, Animal/pathology , Hippocampus/pathology , Hippocampus/virology , Murine hepatitis virus/genetics , Rodent Diseases/pathology , Animals , Coronavirus Infections/pathology , Coronavirus Infections/virology , Hepatitis, Viral, Animal/virology , Mice , Mice, Inbred BALB C , Rodent Diseases/virology
7.
Jpn J Infect Dis ; 64(6): 499-505, 2011.
Article in English | MEDLINE | ID: mdl-22116329

ABSTRACT

The mouse hepatitis virus (MHV) has a high mutation rate, leading to various neuropathologies after infection. The srr7 mutant was isolated from the MHV strain cl-2, which induces characteristic spongiform degeneration in the brain. To investigate outcomes of srr7 infection, we re-cloned srr7(H2) from the viral stock srr7(Mix). During this re-cloning, we obtained the mutant viruses, Mu-1, Mu-2, and Br-1 which was isolated from the mice brain infected with srr7(Mix). We examined mutant viruses for infectivity independent of the major MHV receptor (MHVR), because these mutants exhibited high virulence similar to cl-2, which is MHVR-independent. To confirm MHVR-independence in vitro, we used a combination of spinoculation and ultraviolet radiation to detect distinct plaque formation (SpinoPlaque(UV+)) afrer infection of BHK cells, which do not express MHVR. Using this technique, we found that the unique neuropathologies caused by infection with the mutant viruses result from infecting neurons, which do not express MHVR. Infection with the mutant viruses was 100% correlated with SpinoPlaque(UV+) formation. This is in contrast to infection with srr7, which does not from SpinoPlaque(UV+).


Subject(s)
Central Nervous System Infections/pathology , Central Nervous System Infections/virology , Murine hepatitis virus/pathogenicity , Murine hepatitis virus/radiation effects , Mutation , Ultraviolet Rays , Animals , Brain/pathology , Brain/virology , Histocytochemistry , Immunohistochemistry , Mice , Mice, Inbred BALB C , Microscopy , Murine hepatitis virus/isolation & purification , Virulence
8.
Microbiol Immunol ; 55(10): 694-703, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21831205

ABSTRACT

Friend murine leukemia virus clone A8 causes spongiform neurodegeneration in the rat brain, and the env gene of A8 is a primary determinant of neuropathogenicity. In order to narrow down the critical region within the env gene that determines neuropathogenicity, we constructed chimeric viruses having chimeric env between A8 and non-neuropathogenic 57 on the background of A8 virus. After replacement of the BamHI (at nucleotide 5715)-AgeI (at nucleotide 6322) fragment of A8 virus with the corresponding fragment of 57, neuropathogenicity was lost. In contrast, the chimeric viruses that have the BamHI (5715)-AgeI (6322) fragment of A8 induced spongiosis in 100% of infected rats at the same or slightly lower intensity than A8 virus. These results indicate that the BamHI (5715)-AgeI (6322) fragment of A8, which contains the signal sequence and the N-terminal half of RBD, is crucial for the induction of spongiform neurodegeneration. In the BamHI (5715)-AgeI (6322) fragment, seven amino acids differed between A8 and 57, one in the signal sequence and six in RBD, which suggests that these amino acids significantly contribute to the neuropathogenicity of A8.


Subject(s)
Friend murine leukemia virus/physiology , Gene Products, env/chemistry , Gene Products, env/genetics , Retroviridae Infections/virology , Tumor Virus Infections/virology , 3T3 Cells , Amino Acid Motifs , Animals , Brain/pathology , Brain/virology , Friend murine leukemia virus/genetics , Friend murine leukemia virus/pathogenicity , Gene Products, env/metabolism , Mice , Rats , Rats, Inbred Lew , Retroviridae Infections/pathology , Tumor Virus Infections/pathology
9.
Jpn J Infect Dis ; 64(3): 256-9, 2011.
Article in English | MEDLINE | ID: mdl-21617315

ABSTRACT

The highly neuropathogenic cl-2 and less virulent srr7 viruses isolated from the neurotropic JHM strain of the mouse hepatitis virus exhibit super acute spread of virus (SAS), a term applied when rapid viral spread from an organ or part of the initially infected site to another non-adjacent organ or part is detected within 12 h after infection. Herein, we used a cytospin procedure to confirm SAS in splenic cells derived from mice whose brains were infected with these viruses. The cytospin procedure enabled effective preservation of the cells on glass slides. With this method, we could characterize extremely low populations of infected cells in the spleen (less than 0.1%) at 12 h post-inoculation with srr7. We observed that all kinds of splenic cells examined were infected, including B220(+)Ly-6C(+) plasmacytoid dendritic cells. The population of viral antigen-positive splenic cells was only slightly higher in cl-2 infection than in srr7 infection, but the cells showing viral production were present in numbers significantly higher in cl-2 infection compared with srr7 infection.


Subject(s)
Hepatitis, Viral, Animal/virology , Murine hepatitis virus/pathogenicity , Rodent Diseases/immunology , Rodent Diseases/virology , Spleen/virology , Animals , Disease Models, Animal , Hepatitis, Viral, Animal/pathology , Mice , Mice, Inbred BALB C , Murine hepatitis virus/isolation & purification , Spleen/pathology , Time Factors
10.
Pathol Int ; 61(4): 184-91, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21418390

ABSTRACT

Soluble receptor-resistant mutant 7 (ssr7) is isolated from a highly neurovirulent mouse hepatitis virus (MHV) JHMV cl-2 strain (cl-2). srr7 exhibits lower virulence than its maternal strain in infected mice, which is typically manifested in a longer lifespan. In this study, during the course of infection with srr7, small spongiotic lesions became apparent at 2 days post-inoculation (pi), they spread out to form spongiform encephalopathy by 8 to 10 days pi. We recently reported that the initial expressions of viral antigens in the brain are detected in the infiltrating monocyte lineage and in ependymal cells. Here, we demonstrate that the next viral spread was observed in glial fibrillary acidic protein-positive cells or nestin-positive progenitor cells which take up positions in the subventricular zone (SVZ). From this restricted site of infection in the SVZ, a large area of gliosis extended deep into the brain parenchyma where no viral antigens were detected but vacuolar degeneration started at 48 h pi of the virus. The extremely short incubation period compared with other experimental models of infectious spongiform degeneration in the brain would provide a superior experimental model to investigate the mechanism of spongiotic lesions formation.


Subject(s)
Coronavirus Infections/complications , Coronavirus Infections/pathology , Prion Diseases/pathology , Prion Diseases/virology , Animals , Antigens, Viral/analysis , Antigens, Viral/metabolism , Fluorescent Antibody Technique , Immunohistochemistry , Mice , Mice, Inbred BALB C , Prion Diseases/etiology
11.
Neuropathology ; 30(4): 361-71, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20051016

ABSTRACT

Viral spread during the early stages after infection was compared between a highly neurovirulent mouse hepatitis virus (MHV), JHMV cl-2 strain (cl-2), and its low-virulent mutant, soluble-receptor-resistant (srr)7. The infection of cells with srr7 (soluble-receptor-resistant mutant 7) is dependent on a known MHV receptor (MHVR), carcinoembryonic cell adhesion molecule 1a, whereas cl-2 shows MHVR-independent infection. Initial viral antigens were detected between 12 and 24 h post-inoculation (p.i) in the infiltrating cells that appeared in the subarachnoidal space of mouse brains infected with viruses. There were no significant differences in the intensity or spread of viral antigens in the inflammatory cells between the two viruses. However, 48 h after infection with cl-2, viral antigen-positive cells in the grey matter with the shape of neurons, which do not express MHVR, were detected, while srr7 infection was observed primarily in the white matter. Some of the viral antigen-positive inflammatory cells found in the subarachnoidal space during the early phase of infection reacted with anti-F4/80 or anti-CD11b monoclonal antibodies. Syncytial giant cells (SGCs) expressing viral and CD11b antigens were also detected among these inflammatory cells. These antigen-positive cells appeared in the subarachnoidal space prior to viral antigen spread into the brain parenchyma, indicating that viral encephalitis starts with the infection of infiltrating monocytes which express MHVR. Furthermore, the observation indicates that viral infection has cytopathic effects on the monocyte lineage, which plays a critical role in innate immunity, leading to the rapid spread of viruses during the early stage of infection.


Subject(s)
Brain/virology , Coronaviridae Infections/virology , Glycoproteins/immunology , Monocytes/virology , Murine hepatitis virus/pathogenicity , Animals , Antigens, Viral/immunology , Brain/immunology , Cell Adhesion Molecules , Cell Lineage , Coronaviridae Infections/immunology , Fluorescent Antibody Technique , Immunohistochemistry , Mice , Mice, Inbred BALB C , Monocytes/immunology , Murine hepatitis virus/immunology
12.
Neuropathology ; 26(3): 188-95, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16771173

ABSTRACT

A8-V is a neuropathogenic clone isolated from the Friend murine leukemia virus which causes spongiosis in the rat brain after infection at birth. Serial studies using chimeric viruses derived from the A8-V and the 57 virus (57-V), which is a non-neuropathogenic strain of Friend murine leukemia virus, proved that the long terminal repeat (LTR) and 5' leader (LTR-leader/A8) derived from A8-V, in addition to the env gene (env/A8) of A8-V, are necessary for the neuropathogenesis of A8-V. The enhancer element within the LTR of A8-V (LTR/A8) has been supposed to contribute to the severe manifestation of spongiosis by inducing high levels of viral production in the brain after A8-V infection. However, the recombinant viruses R7c and R7f, which lack the enhancer element of A8-V, induced spongiosis with high incidence rates, although the isolated viral titers of the infected brain display very low levels, which are even comparable to the 57-V infection. Immunohistochemical studies demonstrated that infection with neuropathogenic chimerae, R7c and R7f, induced increased expression of viral antigens than that produced by infection with non-neuropathogenic chimeric virus, Rec5, despite the fact that R7c, R7f and Rec5 all exhibited similar levels of viral proliferation in the brain postinfection. Thus, neuropathology induced by A8 infection is not dependent upon the viral proliferation rate but rather the level of viral antigen expression.


Subject(s)
Antigens, Viral/biosynthesis , Brain Diseases/pathology , Brain Diseases/virology , Retroviridae Infections/pathology , Tumor Virus Infections/pathology , Animals , Brain Diseases/immunology , Clone Cells , Enhancer Elements, Genetic , Friend murine leukemia virus/genetics , Immunohistochemistry , Neuroglia/virology , Rats , Rats, Inbred Lew , Retroviridae Infections/metabolism , Tumor Virus Infections/metabolism
13.
J Vet Med Sci ; 68(3): 259-65, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16598170

ABSTRACT

Friend murine leukemia virus A8 and PVC211 cause spongiform neurodegeneration in rat brains. Glutamate is an important neurotransmitter synthesized from alpha-ketoglutaric acid, an intermediate product of the citric acid cycle, and glutamine is synthesized from glutamate. To examine the brain metabolism of rats infected with neuropathogenic viruses, the amount of glutamate and glutamine in the brains of rats infected with A8, PVC211, and non-neuropathogenic 57 was measured using high performance liquid chromatography, and the (13)C-label incorporation into the C4 position of glutamate and glutamine from [1-(13)C] glucose was measured with (13)C nuclear magnetic resonance. In the cerebral hemisphere and region containing the brain stem and basal ganglia of rats infected with A8 and PVC211 at 8-9 weeks post-infection (wpi), the amount of glutamine was decreased compared with the 57-infected rats. The amount of glutamate was decreased in the cerebral hemisphere of the A8-infected rats and the region containing the brain stem and basal ganglia of PVC211-infected rats at 8-9 wpi. The amount of [4-(13)C] glutamine and [4-(13)C] glutamate in the cerebral hemisphere and region containing the brain stem and basal ganglia of rats infected with A8 and PVC211 at 8-9 wpi was equivalent to that of the 57-infected rats. These results suggest that in the brains of rats infected with neuropathogenic viruses, de novo synthesis of glutamate and glutamine is not decreased, but the ability to maintain quantitative levels of glutamate and glutamine is decreased compared with the brains of rats infected with non-neuropathogenic virus.


Subject(s)
Brain Diseases/veterinary , Friend murine leukemia virus/growth & development , Leukemia, Experimental/virology , Retroviridae Infections/veterinary , Rodent Diseases/virology , Tumor Virus Infections/veterinary , Animals , Animals, Newborn , Basal Ganglia/metabolism , Basal Ganglia/pathology , Basal Ganglia/virology , Brain Diseases/metabolism , Brain Diseases/pathology , Brain Diseases/virology , Brain Stem/metabolism , Brain Stem/pathology , Brain Stem/virology , Glucose/metabolism , Glutamic Acid/analysis , Glutamic Acid/metabolism , Glutamine/analysis , Glutamine/metabolism , Histocytochemistry/veterinary , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Nuclear Magnetic Resonance, Biomolecular , Rats , Rats, Inbred Lew , Retroviridae Infections/metabolism , Retroviridae Infections/pathology , Retroviridae Infections/virology , Rodent Diseases/metabolism , Rodent Diseases/pathology , Telencephalon/metabolism , Telencephalon/pathology , Telencephalon/virology , Tumor Virus Infections/metabolism , Tumor Virus Infections/pathology , Tumor Virus Infections/virology
14.
Microbiol Immunol ; 50(3): 197-201, 2006.
Article in English | MEDLINE | ID: mdl-16547417

ABSTRACT

Friend murine leukemia virus clone A8 causes spongiform neurodegeneration in the rat brain. A 0.3-kb fragment containing the R-U5-5' leader sequence of A8 is required in addition to the A8-env gene to induce spongiosis. The A8-env gene is a primary determinant of neuropathogenicity. Comparative studies of the neuropathogenic virus R7f, which carries the 0.3-kb fragment of A8 and A8-env on the background of the non-neuropathogenic clone 57, and the non-neuropathogenic virus Rec5, which carries A8-env on the background of 57, showed that the 0.3-kb fragment of A8 was responsible for increasing the ratio of Env/Gag expression in the brain, but not in the spleen.


Subject(s)
5' Untranslated Regions/genetics , Brain/metabolism , Friend murine leukemia virus/pathogenicity , Gene Expression Regulation, Viral , Gene Products, env/metabolism , Animals , Brain/virology , DNA, Viral/analysis , DNA, Viral/isolation & purification , Friend murine leukemia virus/genetics , Gene Products, env/genetics , Gene Products, gag/genetics , Gene Products, gag/metabolism , Rats , Rats, Inbred Lew , Retroviridae Infections/virology , Spleen/metabolism , Spleen/virology , Tumor Virus Infections/virology
15.
Microbiol Immunol ; 49(12): 1069-74, 2005.
Article in English | MEDLINE | ID: mdl-16365532

ABSTRACT

Infection of rats with Friend murine leukemia virus (Fr-MLV) clone A8 causes thymoma in all the animals within 7 weeks. The rapid induction of thymoma is associated with a unique enhancer structure in the U3 region of the A8-LTR. Our Southern blot analyses showed that the thymomas were oligo clonal. The A8-induced thymomas showed 3-to 11-fold overexpression of c-myc mRNA. These results suggest that provirus insertion into particular positions of the host genome is correlated with tumorigenesis after A8 infection and that up-regulation of c-myc plays an important role in the induction of thymoma.


Subject(s)
Leukemia Virus, Murine , Leukemia, Experimental/metabolism , Proto-Oncogene Proteins c-myc/genetics , Retroviridae Infections/metabolism , Thymoma/metabolism , Tumor Virus Infections/metabolism , Animals , Animals, Newborn , Blotting, Southern , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/genetics , Rats , Rats, Inbred Lew , Reverse Transcriptase Polymerase Chain Reaction , Thymus Gland/metabolism , Up-Regulation , Virus Integration
16.
Microbiol Immunol ; 49(12): 1075-81, 2005.
Article in English | MEDLINE | ID: mdl-16365533

ABSTRACT

A8-V and PVC211 are neuropathogenic strains of the Friend murine leukemia virus (Fr-MLV) that cause spongiosis in the rat brain after infection at birth. PVC211 exhibited stronger neuropathogenicity than A8-V, and induced more severe neurological symptoms such as hind-leg paralysis. These symptoms correlated with the neuropathological spread and intensity, which were more severe in the spinal cord of rats infected with PVC211 than in those infected with A8-V, without exhibiting neuropathological differences in other areas of the CNS. Interestingly, virus titers recovered from infected spinal cords were similar in PVC211 and A8-V infected animals. However, in the spinal cord infected with PVC211, glial cells attained higher immunohistochemical expression scores for the viral surface antigen, gp70 (Env) than in the A8-V infected spinal cord, although expression levels of viral antigens in blood vessel walls were similar in A8-V and PVC211 infections. Furthermore, many of those glial cells which carried viral antigens were found, by double immunostaining, to be microglia. The results suggested that the spread of viral antigen positive microglia plays an important role in forming the different neuro-pathogenicity observed in A8-V and PVC211 infections.


Subject(s)
Friend murine leukemia virus , Leukemia, Experimental/metabolism , Retroviridae Infections/metabolism , Retroviridae Proteins, Oncogenic/metabolism , Tumor Virus Infections/metabolism , Viral Envelope Proteins/metabolism , Animals , Animals, Newborn , Hindlimb/pathology , Immunohistochemistry , Leukemia, Experimental/pathology , Microglia/metabolism , Paralysis/pathology , Rats , Rats, Inbred Lew , Retroviridae Infections/pathology , Spinal Cord/metabolism , Tumor Virus Infections/pathology
17.
Jpn J Infect Dis ; 58(6): 380-2, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16377873

ABSTRACT

In order to invent a screening system to check in vivo gene function and the efficiency of gene transfer mediated by a retroviral vector system, we established a novel packaging cell, PacNIH/A8, based on the neuropathogenic retrovirus A8-V. To construct the expression vector, pA8(Psi-), which expresses the genes gag, pol and env derived from A8-V, the SV40 early region was used for the polyadenylation signal (polyA signal). When a 0.85 kbp fragment in the SV40 early region was employed for the expression vector (pA8(Psi-)beta), env expression was abolished. This abolition was rescued by shortening the SV40 early region to 0.14 kbp (pA8(Psi-)delta). The NHI3T3 cells transfected with pA8(Psi-)delta showed expressions of both env and gag genes.


Subject(s)
Friend murine leukemia virus/genetics , Gene Expression Regulation, Viral , Genes, env/genetics , Poly A/genetics , Regulatory Sequences, Nucleic Acid/genetics , Simian virus 40/genetics , Animals , Mice , NIH 3T3 Cells
18.
Virology ; 336(1): 1-10, 2005 May 25.
Article in English | MEDLINE | ID: mdl-15866066

ABSTRACT

Friend murine leukemia virus (Fr-MLV) clone A8 causes spongiform neurodegeneration in the rat brain. The A8-env gene is a primary determinant of neuropathogenicity, and the 1.5-kb ClaI-HindIII fragment containing the LTR and 5' leader from A8 are additionally required for spongiosis. After replacement of the A8 enhancer region of the neuropathogenic chimera with the enhancer region of non-neuropathogenic 57, viral titer in the brain was reduced by two orders of magnitude. However, the A8 enhancer region was not responsible for the induction of spongiosis. The region responsible for neuropathogenesis was located in the 0.3-kb KpnI-AatII fragment of A8 containing the R-U5-5' leader. The chimeric virus possessing this 0.3-kb fragment of A8 and the A8-env in the 57 background induced a high rate of spongiform neurodegeneration within 7 weeks (9/9 of infected rats). Studies using cultured cells suggest that the 0.3-kb fragment influences the expression of Env protein. Furthermore, these neuropathogenic chimerae, despite low viral replication in the brain, exhibited a stronger expression of Env protein compared with that of non-neuropathogenic viruses.


Subject(s)
5' Untranslated Regions/genetics , Friend murine leukemia virus/genetics , Friend murine leukemia virus/pathogenicity , Neurodegenerative Diseases/virology , RNA, Viral/genetics , Retroviridae Infections/pathology , Animals , Base Sequence , Brain/pathology , Brain/virology , Cell Line , Disease Models, Animal , Gene Expression Regulation, Viral , Genes, env , Molecular Sequence Data , RNA, Messenger/analysis , Rats , Retroviridae , Sequence Alignment , Terminal Repeat Sequences , Viral Plaque Assay
19.
Virology ; 326(1): 29-40, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15262492

ABSTRACT

Friend murine leukemia virus (Fr-MLV) clone A8 causes thymoma 7 weeks postinfection in rats with a more rapid progression than clone 57. The U3 region of A8-LTR contains a unique structure of enhancer motifs consisting of three repeats of a 38-bp sequence containing FVa, LVb/C4, and CORE motifs. Replacement or deletion of the 38-bp sequence in the A8-U3 resulted in a marked reduction in tumorigenicity. Furthermore, the virus with 57-U3 gained high tumorigenicity after construction of the three 38-bp repeats in the U3 region. These findings indicated that the repeats of the 38-bp sequence of A8-LTR are essential for the rapid induction of thymoma. Interestingly, the repeat of the 38-bp sequence did not accelerate the amount of integrated viral DNA in the thymus during the early phase of infection, although it contributed to higher production of infectious virus. Thus, it was demonstrated that the ability to induce thymoma, which correlates with virus titer in the thymus, is not determined by the rate of viral DNA integration into the host genome.


Subject(s)
Friend murine leukemia virus/pathogenicity , Retroviridae Infections/virology , Terminal Repeat Sequences/genetics , Thymoma/virology , Thymus Neoplasms/virology , Tumor Virus Infections/virology , Animals , Animals, Newborn , Base Sequence , DNA, Viral/analysis , Disease Models, Animal , Enhancer Elements, Genetic , Friend murine leukemia virus/genetics , Friend murine leukemia virus/isolation & purification , Mice , Molecular Sequence Data , Mutation , NIH 3T3 Cells , Rats , Rats, Inbred Lew , Sequence Alignment , Terminal Repeat Sequences/physiology , Virulence , Virus Integration
20.
Cell Transplant ; 11(5): 459-64, 2002.
Article in English | MEDLINE | ID: mdl-12382674

ABSTRACT

In order to invent a screening system to check in vivo gene function and the efficiency of gene transfer mediated by a retroviral vector system, we established a novel packaging cell, PacC6/A8, that is transplantable to rat brains. The packaging cell is based on the gene of the neuropatogenic retrovirus, A8-V. For expression in the brain, a vector that expresses brain-derived neurotrophic factor (BDNF) tagged by c-Myc-His6 (LxA/bdmh) was constructed. After transfection of LxA/bdmh to PacC6/A8, a cloned cell line, PacC6/A8/bmh, was established. PacC6/A8/bmh cells stably produced pseudotyped retroviruses carrying LxA/bdmh. For a control, a retroviral vector that bears the gene that codes enhanced green fluorescent protein (EGFP) tagged by C-Mic-His6 was also created and used for the establishment of PacC6/A8/gfmh cells that produce pseudotyped retroviruses carrying LxA/gfmh. PacC6/A8/bmh and PacC6/A8/gfmh cells were injected to the brain of newborn rats. A tumor was formed in all the rats injected that did not exhibit any symptoms until 3-4 weeks after the injection. A histological study of the injected rats revealed that the transferred BDNF gene was expressed in the brain of rats injected with PacC6/A8/bmh cells, but not in rats with PacC6/A8/gfmh cells. Interestingly, many activated microglia had migrated into the tumor induced by PacC6/A8/bmh cells, and expressed a high amount of BDNF.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Brain/surgery , Cell Transplantation/methods , Genetic Vectors , Retroviridae/genetics , Animals , Brain/anatomy & histology , Brain-Derived Neurotrophic Factor/biosynthesis , COS Cells , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Green Fluorescent Proteins , Immunohistochemistry , Luminescent Proteins , Rats , Retroviridae/isolation & purification , Transduction, Genetic/methods , Tumor Cells, Cultured , Virus Assembly
SELECTION OF CITATIONS
SEARCH DETAIL
...