Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
J Virol ; 83(12): 6079-86, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19339354

ABSTRACT

A small group of ecotropic murine retroviruses cause a spongiform neurodegenerative disease manifested by tremor, paralysis, and wasting. The neurovirulence of these viruses has long been known to be determined by the sequence of the viral envelope protein, although the nature of the neurotoxicity remains to be clarified. Studies on the neurovirulent viruses FrCas(NC) and Moloney murine leukemia virus ts1 indicate that the nascent envelope protein misfolds, is retained in the endoplasmic reticulum (ER), and induces an unfolded protein response. In the present study we constructed a series of viruses with chimeric envelope genes containing segments from virulent and avirulent retroviruses. Each of the viruses studied was highly neuroinvasive but differed in the severity of the neurological disease they induced. Only viruses that contained the receptor-binding domain (RBD) of the neurovirulent virus induced neurological disease. Likewise, only viruses containing the RBD of the neurovirulent virus exhibited increased binding of the ER chaperone BiP to the envelope precursor protein and induced the unfolded protein response. Thus, the RBD determined both neurovirulence and folding instability. Among viruses carrying the neurovirulent RBD, the severity of the disease was increased when envelope sequences from the neurovirulent virus outside the RBD were also present. Interestingly, these sequences appeared to further increase the degree of folding instability (BiP binding) of the viral envelope protein. These results provide strong support for the hypothesis that this spongiform neurodegenerative disease represents a virus-induced protein folding disorder.


Subject(s)
Neurodegenerative Diseases/virology , Protein Folding , Retroviridae Infections/virology , Retroviridae/pathogenicity , Viral Envelope Proteins/metabolism , Animals , Endoplasmic Reticulum/metabolism , Mice , Mice, Inbred Strains , NIH 3T3 Cells , RNA, Viral/metabolism , Retroviridae/genetics , Viral Envelope Proteins/genetics , Virulence
2.
J Virol ; 75(22): 11239-43, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11602765

ABSTRACT

Glycosylated Gag (Glycogag) is a transmembrane protein encoded by murine and feline oncornaviruses. While the protein is dispensible for virus replication, Glycogag-null mutants of a neurovirulent murine oncornavirus are slow to spread in vivo and exhibit a loss of pathogenicity. The function of this protein in the virus life cycle, however, is not understood. Glycogag is expressed at the plasma membrane of infected cells but has not been detected in virions. In the present study we have reexamined this issue and have found an N-terminal cleavage fragment of Glycogag which was pelleted by high-speed centrifugation and sedimented in sucrose density gradients at the same bouyant density as virus particles. Its association with virions was confirmed by velocity sedimentation through iodixanol, which effectively separated membrane microvesicles from virus particles. Furthermore, the apparent molecular weight of the virion-associated protein was different from that of the protein extracted from the plasma membrane, suggesting some level of specificity or selectivity of incorporation.


Subject(s)
Gene Products, gag/chemistry , Retroviridae/chemistry , Virion/chemistry , Animals , Glycosylation
4.
Infect Immun ; 69(3): 1832-40, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11179361

ABSTRACT

Immunization(s) fostering the induction of genital mucosa-targeted immune effectors is the goal of vaccines against sexually transmitted diseases. However, it is uncertain whether vaccine administration should be based on the current assumptions about the common mucosal immune system. We investigated the relationship between mucosal sites of infection, infection-induced inflammation, and immune-mediated bacterial clearance in mice using the epitheliotropic pathogen Chlamydia trachomatis. Chlamydial infection of the conjunctival, pulmonary, or genital mucosae stimulated significant changes in tissue architecture with dramatic up-regulation of the vascular addressin, VCAM, a vigorous mixed-cell inflammatory response with an influx of alpha4beta1+ T cells, and clearance of bacteria within 30 days. Conversely, intestinal mucosa infection was physiologically inapparent, with no change in expression of the local MAdCAM addressin, no VCAM induction, no histologically detectable inflammation, and no tissue pathology. Microbial clearance was complete within 60 days in the small intestine but bacterial titers remained at high levels for at least 8 months in the large intestine. These findings are compatible with the notion that VCAM plays a functional role in recruiting cells to inflammatory foci, and its absence from the intestinal mucosa contributes to immunologic homeostasis at that site. Also, expression of type 1 T cell-mediated immunity to intracellular Chlamydia may exhibit tissue-specific variation, with the rate and possibly the mechanism(s) of clearance differing between enteric and nonenteric mucosae. The implications of these data for the common mucosal immune system and the delivery of vaccines against mucosal pathogens are discussed.


Subject(s)
Chlamydia Infections/immunology , Chlamydia trachomatis/immunology , Mucous Membrane/immunology , Animals , Antigens, Surface/isolation & purification , B-Lymphocytes/immunology , Cell Adhesion Molecules/isolation & purification , Conjunctiva/immunology , Epithelial Cells/immunology , Female , Inflammation , Integrins/isolation & purification , Intestinal Mucosa/immunology , Membrane Proteins , Mice , Mice, Inbred C57BL , Respiratory Mucosa/immunology , T-Lymphocytes/immunology , Urogenital System/immunology
5.
J Virol ; 75(6): 2665-74, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11222690

ABSTRACT

The chimeric murine oncornavirus FrCas(E) causes a rapidly progressive paralytic disease associated with spongiform neurodegeneration throughout the neuroaxis. Neurovirulence is determined by the sequence of the viral envelope gene and by the capacity of the virus to infect microglia. The neurocytopathic effect of this virus appears to be indirect, since the cells which degenerate are not infected. In the present study we have examined the possible role of inflammatory responses in this disease and have used as a control the virus F43. F43 is an highly neuroinvasive but avirulent virus which differs from FrCas(E) only in 3' pol and env sequences. Like FrCas(E), F43 infects large numbers of microglial cells, but it does not induce spongiform neurodegeneration. RNAase protection assays were used to detect differential expression of genes encoding a variety of cytokines, chemokines, and inflammatory cell-specific markers. Tumor necrosis factor alpha (TNF-alpha) and TNF-beta mRNAs were upregulated in advanced stages of disease but not early, even in regions with prominent spongiosis. Surprisingly there was no evidence for upregulation of the cytokines interleukin-1 alpha (IL-1 alpha), IL-1 beta, and IL-6 or of the microglial marker F4/80 at any stage of this disease. In contrast, increased levels of the beta-chemokines MIP-1 alpha and -beta were seen early in the disease and were concentrated in regions of the brain rich in spongiosis, and the magnitude of responses was similar to that observed in the brains of mice injected with the glutamatergic neurotoxin ibotenic acid. MIP-1alpha and MIP-1beta mRNAs were also upregulated in F43-inoculated mice, but the responses were three- to fivefold lower and occurred later in the course of infection than was observed in FrCas(E)-inoculated mice. These results suggest that the robust increase in expression of MIP-1 alpha and MIP-1 beta in the brain represents a correlate of neurovirulence in this disease, whereas the TNF responses are likely secondary events.


Subject(s)
Brain/pathology , Macrophage Inflammatory Proteins/metabolism , Neurodegenerative Diseases/virology , Retroviridae Infections/virology , Retroviridae/pathogenicity , Animals , Brain/immunology , Brain/virology , Cell Death/drug effects , Chemokine CCL3 , Chemokine CCL4 , Chemokines, CC/metabolism , Ibotenic Acid/pharmacology , Immunohistochemistry , Inflammation , Macrophage Inflammatory Proteins/genetics , Mice , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/pathology , Neurons/drug effects , Neurons/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retroviridae Infections/immunology , Retroviridae Infections/pathology , Virulence
6.
J Virol ; 75(6): 2848-56, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11222710

ABSTRACT

Infection of the central nervous system (CNS) by several viruses can lead to upregulation of proinflammatory cytokines and chemokines. In immunocompetent adults, these molecules induce prominent inflammatory infiltrates. However, with immunosuppressive retroviruses, such as human immunodeficiency virus (HIV), little CNS inflammation is observed yet proinflammatory cytokines and chemokines are still upregulated in some patients and may mediate pathogenesis. The present study examined expression of cytokines and chemokines in brain tissue of neonatal mice infected with virulent (Fr98) and avirulent (Fr54) polytropic murine retroviruses. While both viruses infect microglia and endothelia primarily in the white matter areas of the CNS, only Fr98 induces clinical CNS disease. The pathology consists of gliosis with minimal morphological changes and no inflammation, similar to HIV. In the present experiments, mice infected with Fr98 had increased cerebellar mRNA levels of proinflammatory cytokines tumor necrosis factor alpha (TNF-alpha), TNF-beta, and interleukin-1 alpha and chemokines macrophage inflammatory protein-1 alpha (MIP-1 alpha), MIP-1 beta, monocyte chemoattractant protein 1 (MCP-1), gamma-interferon-inducible protein 10 (IP-10), and RANTES compared to mice infected with Fr54 or mock-infected controls. The increased expression of these genes occurred prior to the development of clinical symptoms, suggesting that these cytokines and chemokines might be involved in induction of neuropathogenesis. Two separate regions of the Fr98 envelope gene are associated with neurovirulence. CNS disease associated with the N-terminal portion of the Fr98 env gene was preceded by upregulation of cytokines and chemokines. In contrast, disease associated with the central region of the Fr98 env gene showed no upregulation of cytokines or chemokines and thus did not require increased expression of these genes for disease induction.


Subject(s)
Central Nervous System Viral Diseases/virology , Chemokines/metabolism , Cytokines/metabolism , Genes, env , Leukemia Virus, Murine/pathogenicity , Retroviridae Infections/virology , Animals , Brain/metabolism , Brain/pathology , Brain/virology , Central Nervous System Viral Diseases/immunology , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/immunology , Leukemia, Experimental/immunology , Leukemia, Experimental/virology , Mice , RNA, Messenger/metabolism , Retroviridae Infections/immunology , Tumor Virus Infections/immunology , Tumor Virus Infections/virology , Viral Envelope Proteins/genetics
7.
Virology ; 271(2): 227-33, 2000 Jun 05.
Article in English | MEDLINE | ID: mdl-10860875

ABSTRACT

The discovery within the past decade that neural stem cells (NSCs) from the developing and adult mammalian brain can be propagated, cloned, and genetically manipulated ex vivo for ultimate transfer back into the CNS has opened the door to a novel means for modifying the CNS environment for experimental and therapeutic purposes. While a great deal of interest has been focused on the properties and promise of this new technology, especially in regard to cellular replacement and gene therapy, this minireview will focus on the recent use of NSCs to study the neuropathogenesis of the murine oncornaviruses. In brief, the use of this NSC-based approach has provided a means for selective reconstitution within the brain, of specific retroviral life cycle events, in order to consider their contribution to the induction of neurodegeneration. Furthermore, by virtue of their ability to disseminate virus within the brain, NSCs have provided a reliable means for assessing the true neurovirulence potential of murine oncornaviruses by directly circumventing a restriction to virus entry into the CNS. Importantly, these experiments have demonstrated that the neurovirulence of oncornaviruses requires late virus life cycle events occurring specifically within microglia, the resident macrophages of the brain. This initial application of NSC biology to the analysis of oncornavirus-CNS interactions may serve as an example for how other questions in viral neuropathogenesis might be addressed in the future.


Subject(s)
Neurons/virology , Retroviridae/pathogenicity , Stem Cells/virology , Animals , Humans , Membrane Fusion/physiology , Microglia/virology , Retroviridae Infections/virology , Retroviridae Proteins/metabolism , Viral Envelope Proteins/metabolism , Virulence
8.
J Virol ; 74(1): 465-73, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10590136

ABSTRACT

The chimeric murine oncornavirus FrCas(E) causes a rapidly progressive noninflammatory spongiform encephalomyelopathy after neonatal inoculation. The virus was constructed by the introduction of pol-env sequences from the wild mouse virus CasBrE into the genome of a neuroinvasive but nonneurovirulent strain of Friend murine leukemia virus (FMuLV), FB29. Although the brain infection by FrCas(E) as well as that by other neurovirulent murine retroviruses has been described in detail, little attention has been paid to the neuroinvasive but nonneurovirulent viruses. The purpose of the present study was to compare brain infection by FrCas(E) with that by FB29 and another nonneurovirulent virus, F43, which contains pol-env sequences from FMuLV 57. Both FB29 and F43 infected the same spectrum of cell types in the brain as that infected by FrCas(E), including endothelial cells, microglia, and populations of neurons which divide postnatally. Viral burdens achieved by the two nonneurovirulent viruses in the brain were actually higher than that of FrCas(E). The widespread infection of microglia by the two nonneurovirulent viruses is notable because it is infection of these cells by FrCas(E) which is thought to be a critical determinant of its neuropathogenicity. These results indicate that although the sequence of the envelope gene determines neurovirulence, this effect appears to operate through a mechanism which does not influence either viral tropism or viral burden in the brain. Although all three viruses exhibited similar tropism for granule neurons in the cerebellar cortex, there was a striking difference in the distribution of envelope proteins in those cells in vivo. The FrCas(E) envelope protein accumulated in terminal axons, whereas those of FB29 and F43 remained predominantly in the cell bodies. These observations suggest that differences in the intracellular sorting of these proteins may exist and that these differences appear to correlate with neurovirulence.


Subject(s)
Encephalitis, Viral/virology , Retroviridae Infections/virology , Retroviridae/pathogenicity , Animals , Astrocytes/pathology , Astrocytes/virology , Mice , Mice, Inbred Strains , Microglia/pathology , Microglia/virology , Phenotype , Virulence
10.
Virology ; 248(2): 199-207, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9721229

ABSTRACT

Murine leukemia virus (MuLV) clone Fr98 is a recombinant polytropic virus that causes neurological disease characterized by ataxia in susceptible mouse strains. The envelope gene of Fr98 has been previously shown to encode at least two separate neurovirulence determinants. In the present study, the determinant encoded within the EcoRI/AvrII fragment of the envelope gene was further defined. In these experiments, neurovirulence was associated with a change from a serine to an arginine at position 195 and a glycine to an alanine at position 198 within the envelope protein. Neurovirulent and nonvirulent virus clones, which differed only at these two amino acid residues, showed no difference in the type or location of cells infected. Furthermore, equivalent levels of viral p30 capsid protein were detected in the brains of mice infected with either the neurovirulent or nonvirulent virus clones. These results were consistent with the interpretation that the envelope protein of the neurovirulent virus differed from that of the nonvirulent virus by having a greater toxic effect on central nervous system function.


Subject(s)
Central Nervous System Diseases/virology , Leukemia Virus, Murine/pathogenicity , Viral Envelope Proteins/physiology , Animals , Astrocytes/physiology , Capsid/metabolism , Immunohistochemistry , Leukemia Virus, Murine/chemistry , Leukemia Virus, Murine/genetics , Leukemia, Experimental/pathology , Mice , Protein Conformation , Retroviridae Infections/pathology , Tumor Virus Infections/pathology , Viral Envelope Proteins/analysis , Viral Envelope Proteins/genetics , Virulence/genetics
11.
J Virol ; 72(7): 5619-25, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9621020

ABSTRACT

The tempo and intensity of retroviral neuropathogenesis are dependent on the capacity of the virus to invade the central nervous system. For murine leukemia viruses, an important determinant of neuroinvasiveness is the virus-encoded protein glycosylated Gag, the function of which in the virus life cycle is not known. While this protein is dispensable for virus replication, mutations which prevent its expression slow the spread of virus in vivo and restrict virus dissemination to the brain. To further explore the function of this protein, we compared two viruses, CasFrKP (KP) and CasFrKP41 (KP41), which differ dramatically in neurovirulence. KP expresses high early viremia titers, is neuroinvasive, and induces clinical neurologic disease in 100% of neonatally inoculated mice, with an incubation period of 18 to 23 days. In contrast, KP41 expresses early viremia titers 100- fold lower than those of KP, exhibits attenuated neuroinvasiveness, and induces clinical neurologic disease infrequently, with a relatively long incubation period. The genomes of these two viruses differ by only 10 nucleotides, resulting in differences at five residues, all located within the N-terminal cytoplasmic tail of glycosylated Gag. In this study, using KP as the parental virus, we systematically mutated each of the five amino acid residues to those of KP41 and found that substitution mutation of two membrane-proximal residues, E53 and L56, to K and P, respectively produced the greatest effect on early viremia kinetics and neurovirulence. These mutations disrupted the KP sequence E53FLL56, the leucine dipeptide of which suggests the possibility that it may represent a sorting signal for glycosylated Gag. Supporting this idea was the finding that alteration of this sequence motif increased the level of cell surface expression of the protein, which suggests that analysis of the intracellular trafficking of glycosylated Gag may provide further clues to its function.


Subject(s)
Brain/virology , Gene Products, gag/chemistry , Leukemia Virus, Murine/pathogenicity , Amino Acid Sequence , Animals , Cytoplasm/chemistry , Glycosylation , Leucine , Leukemia Virus, Murine/chemistry , Mice , Molecular Sequence Data , Spleen/virology , Viremia/virology , Virulence
12.
J Immunol ; 160(6): 2905-14, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9510194

ABSTRACT

Immunity to genital tract infection with Chlamydia trachomatis is mediated by type 1 CD4+ T lymphocytes. To define the signals that govern lymphocyte trafficking to the genital mucosa, integrins expressed by infiltrating T cells and endothelial addressins displayed on local vasculature were characterized during the course of infection. All T cells expressed the alphaLbeta2 heterodimer that binds vascular ICAM-1, and most displayed enhanced levels of the alpha4beta1 integrin that interacts with VCAM-1. AlphaE and beta7(low) integrin chains were detected on approximately 15 and 30% of infiltrating T cells, respectively. Lymphocytes derived from the spleen or draining lymph nodes expressed this same integrin profile, suggesting that cells are recruited to the genital mucosa from the systemic circulation without significant selection pressure for these markers. Immunofluorescent staining for the corresponding vascular addressins revealed intense expression of VCAM-1 on small vessels within Chlamydia-infected genital tracts and up-regulation of ICAM-1 on endothelial, stromal, and epithelial cells. Mucosal addressin cell adhesion molecule-1 was not detected within genital tissues. These results indicate that T lymphocyte homing to the genital mucosa requires the interaction of alphaLbeta2 and alpha4beta1 with endothelial ICAM-1 and VCAM-1, respectively, which is the same pathway that directs lymphocytes to systemic sites of inflammation. Homing pathways defined for the intestinal mucosa and assumed to be relevant to all mucosal sites are not well represented in the genital tract. The identification of T lymphocyte trafficking pathways shared between systemic and mucosal tissues should facilitate vaccine strategies aimed at maximizing immune responses against Chlamydia and other pathogens of the urogenital tract.


Subject(s)
Chlamydia Infections/immunology , Chlamydia trachomatis/immunology , Genitalia, Female/immunology , Intestinal Mucosa/immunology , T-Lymphocytes/physiology , Animals , Cell Adhesion Molecules , Female , Immunoglobulins/analysis , Immunologic Memory , Immunophenotyping , Integrins/analysis , Intercellular Adhesion Molecule-1/analysis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mucoproteins/analysis , Receptors, Antigen, T-Cell/analysis , Vascular Cell Adhesion Molecule-1/analysis
13.
J Virol ; 71(7): 5287-94, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9188597

ABSTRACT

Several murine leukemia viruses (MuLV) induce neurologic disease in susceptible mice. To identify features of central nervous system (CNS) infection that correlate with neurovirulence, we compared two neurovirulent MuLV, Fr98 and Fr98/SE, with a nonneurovirulent MuLV, Fr54. All three viruses utilize the polytropic receptor and are coisogenic, each containing a different envelope gene within a common genetic background. Both Fr98 and Fr98/SE induce a clinical neurologic disease characterized by hyperexcitability and ataxia yet differ in incubation period, 16 to 30 and 30 to 60 days, respectively. Fr54 infects the CNS but fails to induce clinical signs of neurologic disease. In this study, we compared the histopathology, regional virus distribution, and cell tropism in the brain, as well as the relative CNS viral burdens. All three viruses induced similar histopathologic effects, characterized by intense reactive astrogliosis and microglial activation associated with minimal vacuolar degeneration. The infected target cells for each virus consisted primarily of endothelial and microglial cells, with rare oligodendrocytes. Infection localized predominantly in white matter tracts of the cerebellum, internal capsule, and corpus callosum. The only feature that correlated with relative neurovirulence was viral burden as measured by both viral CA protein expression in cerebellar homogenates and quantification of infected cells. Interestingly, Fr54 (nonneurovirulent) and Fr98/SE (slow disease) had similar viral burdens at 3 weeks postinoculation, suggesting that they entered the brain with comparable efficiencies. However, spread of Fr98/SE within the brain thereafter exceeded that of Fr54, reaching levels of viral burden comparable to that seen for Fr98 (rapid disease) at 3 weeks. These results suggest that the determinants of neurovirulence in the envelope gene may influence the efficiency of virus spread within the brain and that a critical number of infected cells may be required for induction of clinical neurologic disease.


Subject(s)
Brain/virology , Leukemia, Experimental/virology , Microglia/virology , Mink Cell Focus-Inducing Viruses/isolation & purification , Retroviridae Infections/virology , Tumor Virus Infections/virology , Animals , Brain/cytology , Leukemia, Experimental/pathology , Mice , Mice, Inbred Strains , Mink Cell Focus-Inducing Viruses/pathogenicity , Retroviridae Infections/pathology , Tumor Virus Infections/pathology , Viral Load , Virulence
14.
J Virol ; 71(7): 5355-60, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9188605

ABSTRACT

The neuroinvasiveness of a chimeric murine retrovirus, CasFrKP (KP), is dependent on the expression of glycosylated Gag (gp85gag). This viral protein is the product of alternate translation initiation 88 codons upstream of and in frame with the initiation codon of pr65gag, the precursor of the viral core proteins. Although expression of glycosylated Gag affects virus spread in the spleen, it appears not to affect virus spread in vitro in fibroblast cell lines (J. L. Portis et al., J. Virol. 68:3879-3887, 1994). The differential effects of this protein in vitro and in vivo have not been explained, and its function is unknown. We have here compared the in vitro processing of this molecule with that expressed in spleens of infected mice. In vitro, gp85gag was cleaved near the middle of the molecule, releasing the C-terminal half (containing capsid and nucleocapsid domains of pr65gag) as a secreted glycoprotein. The N-terminal half of the protein was associated with the plasma membrane as a approximately 55-kDa glycoprotein bearing the matrix domain of pr65gag as well as the N-terminal 88 residue L domain. This processing scheme was also observed in vivo, although two differences were seen. There were differences in N-linked glycosylation of the secreted form of the protein expressed in the spleen. In addition, whereas the membrane-associated species assumed the orientation of a type II integral membrane protein (N(cyto) C(exo)) in fibroblasts in vitro, a subpopulation of spleen cells was detected in which the N terminus of the protein was exposed at the cell surface. These results suggest that the differential effects of glycosylated Gag expression in vivo and in vitro may be related to differences in posttranslational processing of the protein.


Subject(s)
Friend murine leukemia virus/metabolism , Gene Products, gag/metabolism , Protein Processing, Post-Translational , Animals , Cell Line , Fibroblasts/cytology , Friend murine leukemia virus/pathogenicity , Gene Expression , Gene Products, gag/genetics , Glycosylation , Mice , Mice, Inbred Strains , Spleen/cytology , Spleen/metabolism , Virulence
15.
Virology ; 226(2): 384-92, 1996 Dec 15.
Article in English | MEDLINE | ID: mdl-8955058

ABSTRACT

Neuroinvasiveness is a property of all neurovirulent murine retroviruses, although the factors which facilitate infection of the CNS are not understood. We previously showed that mutant MuLV which lack expression of an accessory protein, glycosylated gag, had lost neurovirulence, indicating that this protein may be involved in promoting CNS infection. The mutations were located in the "Kozak" consensus sequence of the initiation codon for this protein. Here it is shown that shortly after inoculation of mice with one of these mutant viruses, revertants emerged which had regained expression of glycosylated gag and had also regained the neuroinvasiveness and neurovirulence exhibited by the wild-type virus. The phenotypic revertants retained the mutations in the "Kozak" consensus sequence but exhibited a G-->A mutation 12 codons downstream from the mutated start site, creating a new initiation codon and a glycosylated gag protein, which was truncated at its N-terminus. Using antibodies specific for glycosylated gag it is shown that the frequency of splenic infectious centers expressing revertant virus increased progressively during the 2 months following inoculation of mutant virus until > or = 50% of the virus-producing cells in the spleen expressed revertant virus. In contrast, although phenotypic revertants were detectable at low frequency after cell-free passage in vitro in M. dunni fibroblasts, there was no evidence for selection. These results indicate that glycosylated gag facilitates virus spread within the spleen and to extra-splenic sites, such as the CNS, and suggest that the protein may function through its interaction with the host.


Subject(s)
Gene Products, gag/physiology , Leukemia Virus, Murine/pathogenicity , Nervous System Diseases/virology , Retroviridae Infections/virology , Tumor Virus Infections/virology , Amino Acid Sequence , Animals , Animals, Newborn , DNA, Viral , Gene Products, gag/genetics , Glycosylation , Immunoblotting , Leukemia Virus, Murine/genetics , Mice , Molecular Sequence Data , Mutagenesis , Phenotype , Recombination, Genetic , Spleen/virology , Virulence/genetics
16.
J Virol ; 70(12): 8896-907, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8971019

ABSTRACT

CasBrE is a neurovirulent murine retrovirus which induces a spongiform myeloencephalopathy in susceptible mice. Genetic mapping studies have indicated that sequences responsible for neurovirulence reside within the env gene. To address the question of direct envelope protein neuroxicity in the central nervous system (CNS), we have generated chimeric mice expressing the CasBrE envelope protein in cells of neuroectodermal origin. Specifically, the multipotent neural progenitor cell line C17.2 was engineered to express the CasBrE env gene as either gp70/p15E (CasE) or gp70 alone (CasES). CasE expression in these cells resulted in complete (>10(5)) interference of superinfection with Friend murine leukemia virus clone FB29, whereas CasES expression resulted in a 1.8-log-unit decrease in FB29 titer. Introduction of these envelope-expressing C17.2 cells into the brains of highly susceptible IRW mice resulted in significant engraftment as integral cytoarchitecturally correct components of the CNS. Despite high-level envelope protein expression from the engrafted cells, no evidence of spongiform neurodegeneration was observed. To examine whether early virus replication events were necessary for pathogenesis, C17.2 cells expressing whole virus were transplanted into mice in which virus replication in the host was specifically restricted by Fv-1 to preintegration events. Again, significant C17.2 cell engraftment and infectious virus expression failed to precipitate spongiform lesions. In contrast, transplantation of virus-expressing C17.2 progenitor cells in the absence of the Fv-1 restriction resulted in extensive spongiform neurodegeneration by 2 weeks postengraftment. Cytological examination indicated that infection had spread beyond the engrafted cells, and in particular to host microglia. Spongiform neuropathology in these animals was directly correlated with CasBrE env expression in microglia rather than expression from neural progenitor cells. These results suggest that the envelope protein of CasBrE is not itself neurotoxic but that virus infectious events beyond binding and fusion in microglia are necessary for the induction of CNS disease.


Subject(s)
Brain/metabolism , Gene Products, gag/metabolism , Microglia/metabolism , Prion Diseases/metabolism , Retroviridae Proteins, Oncogenic/metabolism , Retroviridae/metabolism , Viral Envelope Proteins/metabolism , Animals , Base Sequence , Brain/virology , Cell Line , Central Nervous System/virology , DNA, Viral , Gene Expression , Gene Products, gag/genetics , Mice , Molecular Sequence Data , Prion Diseases/virology , Retroviridae/genetics , Retroviridae/pathogenicity , Retroviridae/physiology , Retroviridae Proteins, Oncogenic/genetics , Viral Envelope Proteins/genetics , Virulence , Virus Replication
17.
J Virol ; 69(12): 8070-5, 1995 Dec.
Article in English | MEDLINE | ID: mdl-7494324

ABSTRACT

A variety of ecotropic murine leukemia viruses cause neurodegenerative disease. We describe here the clinical and histopathological features of a neurologic disease induced by a polytropic murine leukemia virus, FMCF98. Clinical disease was dominated by hyperexcitability and ataxia, and the histopathology was characterized primarily by astrocytosis and astrocytic degeneration. The viral envelope gene harbored the determinants of neurovirulence, since the chimeric virus Fr98E, which contained the envelope gene of FMCF98 on a background of the nonneurovirulent virus FB29, caused a similar disease. The disease caused by Fr98E differed from that induced by the coisogenic neurovirulent ecotropic virus FrCasE in clinical presentation, histopathology, and distribution of virus in the central nervous system. Since Fr98E contains a polytropic envelope gene and FrCasE contains an ecotropic envelope gene, these phenotypic differences appeared to be determined by envelope sequences and may reflect differences in virus receptor usage in the central nervous system.


Subject(s)
Brain Diseases/pathology , Brain Diseases/virology , Brain/virology , Leukemia Virus, Murine , Retroviridae Infections/pathology , Tumor Virus Infections/pathology , 3T3 Cells , Animals , Animals, Newborn , Antigens, Viral/analysis , Astrocytes/pathology , Chimera , Friend murine leukemia virus/isolation & purification , Gliosis , Leukemia Virus, Murine/isolation & purification , Leukemia Virus, Murine/pathogenicity , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Nerve Degeneration , Virulence
18.
J Virol ; 69(11): 7300-3, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7474158

ABSTRACT

The neurovirulent chimeric mouse ecotropic retrovirus FrCasE causes a rapid neurodegenerative disease of the central nervous system (CNS) characterized by the appearance of spongiform lesions in motor areas 10 days after neonatal inoculation. To study the details of the pathogenic process, we examined the ability of an ex vivo spinal cord model to recapitulate disease. Organotypic spinal cord slice cultures were established from IRW mice 7 days after neonatal inoculation. This corresponds to a time when virus expression in the CNS is first detectable but spongiform changes have yet to evolve. Infectivity associated with these cultures peaked at 7 days in vitro and persisted at this level for 6 weeks. FrCasE infection of the spinal cord slices was primarily found associated with microglial cells. Infection of neurons, astrocytes, oligodendroglia, and endothelial cells was not observed; however, significant astrogliosis was found. Despite the presence of extensive microglial infection in close association with spinal motor neurons in organotypic cultures, no virus-specific spongiform degenerative changes were observed. These results suggest that removal of motor neurons from the developing CNS, despite maintaining the local cytoarchitectural relationships, prevents the virus from eliciting its pathological effects. Possible reasons for the interruption of lesion development are discussed.


Subject(s)
Central Nervous System Diseases/virology , Retroviridae/pathogenicity , Spinal Cord/pathology , Spinal Cord/virology , Animals , Antibodies, Monoclonal , Central Nervous System Diseases/pathology , Chimera , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/analysis , Mice , Organ Culture Techniques , Retroviridae/isolation & purification , Retroviridae/physiology , Viral Envelope Proteins/analysis
19.
Virology ; 212(2): 367-82, 1995 Oct 01.
Article in English | MEDLINE | ID: mdl-7571406

ABSTRACT

Characterization of the SRS murine retrovirus complex, derived from the TSZ system of murine leukemia developed in China, was carried out. The initial stock contained XC+, NB-tropic virus (and possibly other viruses), and induced several neoplastic diseases in neonatally inoculated NIH Swiss mice: erythroid leukemia, myeloid leukemia (acute myeloblastic leukemia), and lymphoblastic lymphoma (both B- and T-lymphoid). In addition, approximately 30% of inoculated animals developed central nervous system disease--hindlimb paralysis or semilateral paralysis. Rescue of virus from the spleen of an animal with combined erythroid/myeloid leukemia, followed by endpoint dilution gave two stocks: 19-6 (XC+, NB-tropic) and 19-7 (XC-, NB-tropic). Both stocks induced erythroid and myeloid leukemia, and 19-6 induced CNS symptoms as well. Southern blot analysis indicated that the predominant viruses from the 19-6 and the 19-7 cultures were related, but different in the env region. An infectious virus molecular clone of provirus from 19-6 cells was obtained. The resulting cloned virus [SRS 19-6 murine leukemia virus (MuLV)] induced four kinds of leukemia: erythroid, myeloid, B-lymphoma, and T-lymphoma; in many cases, more than one tumor type was identified in the same animal. Such a broad spectrum of leukemias induced by a cloned MuLV is unusual. Flaccid hindlimb paralysis induced by SRS 19-6 MuLV could be attributed to meningeal B-lymphoma. Immunofluorescent staining with a panel of env-specific monoclonal antibodies confirmed that the 19-6 and 19-7 viral stocks contained different viruses, which differed from previously characterized MuLVs. The viruses of the SRS complex may provide interesting reagents for investigations of MuLV-induced disease.


Subject(s)
Leukemia Virus, Murine/genetics , Leukemia, Experimental/virology , Retroviridae Infections/virology , Tumor Virus Infections/virology , Animals , Animals, Newborn , Base Sequence , China , Cloning, Molecular , DNA, Viral/genetics , Genes, env/genetics , Leukemia Virus, Murine/pathogenicity , Leukemia, Experimental/pathology , Mice , Molecular Sequence Data , Paralysis/virology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/virology , Proviruses/genetics , Restriction Mapping , Retroviridae Infections/pathology , Sequence Analysis, DNA , Spleen/virology , Tumor Virus Infections/pathology
20.
J Virol ; 69(3): 1408-19, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7853473

ABSTRACT

FrCasE is a highly neurovirulent murine leukemia virus which causes a noninflammatory spongiform neurodegenerative disease after neonatal inoculation. The central nervous system (CNS) infection is wide-spread, involving several different cell types, whereas the lesions are localized to motor areas of the brain and spinal cord. Inoculation of FrCasE at 10 days of age (P10) results in viremia, but infection of the CNS is restricted and neurological disease is not observed (M. Czub, S. Czub, F. McAtee, and J. Portis, J. Virol. 65:2539-2544, 1991). In this study, we used this developmental resistance to restrict the extent and the distribution of FrCasE in the brain to examine whether the spongiform degeneration is a consequence of infection of cells in proximity to the lesions. Two approaches were used to infect the brain on or after P10. First, mice were inoculated with FrCasE at P10 to induce viremia and then at P17 were subjected to focal CNS injury within brain regions known to be susceptible to virus-induced spongiform degeneration. The injury resulted in local inflammation, glial activation, migration of inflammatory cells into the wound site, and high-level parenchymal infection about the wound site. However, no evidence of spongiform neurodegeneration was observed over a period of 3 months. The second approach involved the implantation of FrCasE-infected microglia into the CNS at > or = P10. This resulted in microglial engraftment and focal CNS infection unilaterally at the implantation sites and bilaterally along white matter tracts of the corpus callosum and pons and in cells of the subventricular layers of the lateral cerebral ventricles. Strikingly, focal spongiform degeneration colocalized with the sites of infection. In contrast to the wounding experiments, the implantation model was not associated with an inflammatory response or significant glial activation. Results of these studies suggest that (i) the developmental resistance of the CNS to infection lies at the blood-brain barrier and can be bypassed by direct introduction into the brain of virus-infected cells, (ii) the neuropathology induced by this virus is a consequence of local effects of the infection and does not appear to require endothelial or neuronal infection, and (iii) elements of the inflammatory response and/or glial activation may modulate the expression of neuropathology induced by neurovirulent retroviruses.


Subject(s)
Leukemia Virus, Murine/pathogenicity , Microglia/microbiology , Nerve Degeneration , Nervous System Diseases/microbiology , Age Factors , Animals , Blood-Brain Barrier , Mice , Mice, Inbred Strains , Microglia/transplantation , Nervous System Diseases/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...