Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
J Virol ; 75(13): 6007-15, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11390602

ABSTRACT

Induction of apoptosis by different types of pathogenic retroviruses is an important step in disease development. We have observed that infection of thymic lymphocytes by the mink cell focus-forming murine leukemia virus (MCF MLV) during the preleukemic period resulted in an enhancement of apoptosis of these cells. To further study the ability of MCF MLVs to induce apoptosis and the role of this process in viral pathogenesis, we have developed an in vitro system of virus-induced apoptosis. MCF13 MLV infection of mink epithelial cells resulted in the production of cytopathic foci. In contrast, infection of mink cells with the 4070A amphotropic MLV did not produce any cytopathic effects. Staining of MCF13 MLV-infected cells with propidium iodide and annexin V-fluorescein isothiocyanate indicated that virus-induced cell death was due to apoptosis. At 6 days postinfection, the percentage of apoptotic MCF13 MLV-infected cells was 27% compared with 2 to 3% for mock- or amphotropic MLV-infected cells, representing a 9- to 14-fold difference. Assays for caspase-3 activation confirmed the detection by flow cytometry of apoptosis of MCF13 MLV-infected cells. Large amounts of unintegrated linear viral DNA were detectable by Southern blot analysis during the acute phase of infection, which indicated that MCF13 MLV is able to superinfect mink cells. Unintegrated viral DNA of only the linear form was detectable in thymic lymphocytes isolated from MCF13 MLV-inoculated mice during the preleukemic period. These results indicated that the ability of MCF13 MLV to induce apoptosis is correlated with its ability to superinfect cells and that this occurs as an early step in thymic lymphoma development.


Subject(s)
Apoptosis , Mink Cell Focus-Inducing Viruses/physiology , Mink/virology , Animals , DNA, Viral/analysis , DNA, Viral/physiology , Epithelial Cells/virology , Glycoproteins/physiology , Receptors, Virus/physiology
2.
Dev Biol (Basel) ; 106: 163-7; discussion 167-8, 253-63, 2001.
Article in English | MEDLINE | ID: mdl-11761228

ABSTRACT

Using neoplastic cell lines as substrates for vaccine development could inadvertently result in viral-viral or viral-cellular interactions whose biological consequences are unclear. In this review, the generation of mink cell focus-inducing (MCF) retroviruses in the mouse is discussed as a model for understanding how viral-viral and viral-cellular interactions can result in the generation of new retroviruses with pathological consequences.


Subject(s)
Leukemia, Experimental/virology , Membrane Fusion , Mink Cell Focus-Inducing Viruses/physiology , Models, Biological , Animals , Mice , Mink Cell Focus-Inducing Viruses/pathogenicity , Tumor Cells, Cultured
3.
J Virol ; 73(7): 5671-80, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10364317

ABSTRACT

One hallmark of murine leukemia virus (MuLV) leukemogenesis in mice is the appearance of env gene recombinants known as mink cell focus-inducing (MCF) viruses. The site(s) of MCF recombinant generation in the animal during Moloney MuLV (M-MuLV) infection is unknown, and the exact roles of MCF viruses in disease induction remain unclear. Previous comparative studies between M-MuLV and an enhancer variant, Mo+PyF101 MuLV, suggested that MCF generation or early propagation might take place in the bone marrow under conditions of efficient leukemogenesis. Moreover, M-MuLV induces disease efficiently following both intraperitoneal (i.p.) and subcutaneous (s.c.) inoculation but leukemogenicity by Mo+PyF101 M-MuLV is efficient following i.p. inoculation but attenuated upon s. c. inoculation. Time course studies of MCF recombinant appearance in the bone marrow, spleen, and thymus of wild-type and Mo+PyF101 M-MuLV i.p.- and s.c.-inoculated mice were carried out by performing focal immunofluorescence assays. Both the route of inoculation and the presence of the PyF101 enhancer sequences affected the patterns of MCF generation or early propagation. The bone marrow was a likely site of MCF recombinant generation and/or early propagation following i.p. inoculation of M-MuLV. On the other hand, when the same virus was inoculated s.c., the primary site of MCF generation appeared to be the thymus. Also, when Mo+PyF101 M-MuLV was inoculated i.p., MCF generation appeared to occur primarily in the thymus. The time course studies indicated that MCF recombinants are not involved in preleukemic changes such as splenic hyperplasia. On the other hand, MCFs were detected in tumors from Mo+PyF101 M-MuLV s. c.-inoculated mice even though they were largely undetectable at preleukemic times. These results support a role for MCF recombinants late in disease induction.


Subject(s)
Enhancer Elements, Genetic , Genetic Variation , Leukemia, Experimental/virology , Mink Cell Focus-Inducing Viruses/physiology , Moloney murine leukemia virus/physiology , Retroviridae Infections/virology , Tumor Virus Infections/virology , Injections, Intraperitoneal , Injections, Subcutaneous , Moloney murine leukemia virus/genetics , Proviruses
4.
J Virol Methods ; 77(2): 217-24, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10092145

ABSTRACT

Murine acquired immunodeficiency syndrome is a lymphoproliferative disease with a concurrently developing immunodeficiency. The disease is induced after injection of supernatant of a chronically infected cell line that releases a mixture of two replication competent virus classes and a replication incompetent virus species responsible for pathogenicity. An immunochemical detection assay for virus infected foci on cell monolayers has been developed. This assay allows quantification of all three types of virus.


Subject(s)
Defective Viruses/physiology , Immunoenzyme Techniques , Murine Acquired Immunodeficiency Syndrome/virology , Virus Replication , Animals , Cell Line , Female , Mice , Mice, Inbred C57BL , Mink Cell Focus-Inducing Viruses/physiology
5.
J Virol ; 73(3): 2434-41, 1999 Mar.
Article in English | MEDLINE | ID: mdl-9971828

ABSTRACT

Moloney murine leukemia virus (M-MuLV) is a replication-competent, simple retrovirus that induces T-cell lymphoma with a mean latency of 3 to 4 months. During the preleukemic period (4 to 10 weeks postinoculation) a marked decrease in thymic size is apparent for M-MuLV-inoculated mice in comparison to age-matched uninoculated mice. We were interested in studying whether the thymic regression was due to an increased rate of thymocyte apoptosis in the thymi of M-MuLV-inoculated mice. Neonatal NIH/Swiss mice were inoculated subcutaneously (s.c.) with wild-type M-MuLV (approximately 10(5) XC PFU). Mice were sacrificed at 4 to 11 weeks postinoculation. Thymic single-cell suspensions were prepared and tested for apoptosis by two-parameter flow cytometry. Indications of apoptosis included changes in cell size and staining with 7-aminoactinomycin D or annexin V. The levels of thymocyte apoptosis were significantly higher in M-MuLV-inoculated mice than in uninoculated control animals, and the levels of apoptosis were correlated with thymic atrophy. To test the relevance of enhanced thymocyte apoptosis to leukemogenesis, mice were inoculated with the Mo+PyF101 enhancer variant of M-MuLV. When inoculated intraperitoneally, a route that results in wild-type M-MuLV leukemogenesis, mice displayed levels of enhanced thymocyte apoptosis comparable to those seen with wild-type M-MuLV. However, in mice inoculated s.c., a route that results in attenuated leukemogenesis, significantly lower levels of apoptosis were observed. This supported a role for higher levels of thymocyte apoptosis in M-MuLV leukemogenesis. To examine the possible role of mink cell focus-forming (MCF) recombinant virus in raising levels of thymocyte apoptosis, MCF-specific focal immunofluorescence assays were performed on thymocytes from preleukemic mice inoculated with M-MuLV and Mo+PyF101 M-MuLV. The results indicated that infection of thymocytes by MCF virus recombinants is not required for the increased level of apoptosis and thymic atrophy.


Subject(s)
Apoptosis , Moloney murine leukemia virus , Preleukemia/pathology , Retroviridae Infections/pathology , T-Lymphocytes/pathology , Thymus Gland/pathology , Tumor Virus Infections/pathology , Animals , Atrophy , Mice , Mink Cell Focus-Inducing Viruses/physiology
6.
Exp Lung Res ; 25(8): 671-87, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10643564

ABSTRACT

Idiopathic interstitial pneumonitis (IP), characterized by lymphocytic infiltration of the lung and pulmonary dysfunction, is a major noninfectious complication of human immunodeficiency virus (HIV) infection. The role of the CD4+ and CD8+ T cell populations and INF-gamma in the development of IP were analyzed using a murine model of retroviral-associated IP. Infected mice depleted of CD8+ T cells developed IP similarly to untreated infected mice, suggesting that the CD8+ T cell population does not play a role in IP. Furthermore, depletion of CD8+ T cells did not alter the level of viral RNA in lungs, suggesting that cytotoxic T cells may not serve a role in controlling virus burden in lungs. In contrast, depletion of CD4+ T cells in infected mice prevented the development of IP and inhibited inflammatory cytokine expression, suggesting that CD4+ T cells are important for the development of IP. IFN-gamma -/- mice infected with virus for 10 weeks developed IP, although the severity of lymphocytic infiltration was substantially reduced compared to infected wild-type mice. The data suggest that persistent viral antigen in the lung may drive a CD4+ T cell-mediated immune response, resulting in the chronic production of IFN-gamma which amplifies a chronic inflammatory response in the lung resulting in tissue injury.


Subject(s)
AIDS-Related Opportunistic Infections/immunology , Lung Diseases, Interstitial/immunology , Murine Acquired Immunodeficiency Syndrome/immunology , T-Lymphocyte Subsets/immunology , AIDS-Related Opportunistic Infections/pathology , AIDS-Related Opportunistic Infections/virology , Animals , Cytokines/biosynthesis , Female , Helper Viruses/physiology , Interferon-gamma/pharmacology , Leukemia Virus, Murine/physiology , Lung/immunology , Lung/metabolism , Lung/virology , Lung Diseases, Interstitial/pathology , Lung Diseases, Interstitial/virology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mink Cell Focus-Inducing Viruses/physiology , Murine Acquired Immunodeficiency Syndrome/pathology , Murine Acquired Immunodeficiency Syndrome/virology , Virus Replication
7.
J Virol ; 70(2): 830-3, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8551621

ABSTRACT

Cultured cells derived from the wild mouse species Mus castaneus were found to be uniquely resistant to exogenous infection by polytropic mink cell focus-forming (MCF) murine leukemia viruses (MuLVs). This MCF MuLV resistance is inherited as a genetically recessive trait in the progeny of F1 crosses between M. castaneus and MCF MuLV-susceptible laboratory mice. Examination of the progeny of backcrosses demonstrated that susceptibility is inherited as a single gene which maps to chromosome 1. The map location of this gene places it at or near the locus Rmc1, the gene encoding the receptor for MCF/xenotropic MuLVs, suggesting that resistance is mediated by the M. castaneus allele of this receptor.


Subject(s)
Mink Cell Focus-Inducing Viruses/physiology , Muridae/genetics , Animals , Animals, Wild , Cell Line , Cells, Cultured , Chromosome Mapping , Genes, Recessive , Immunity, Innate/genetics , Mice , Mice, Inbred AKR , Mice, Inbred DBA , Mink Cell Focus-Inducing Viruses/genetics , Muridae/virology , Receptors, Virus/genetics , Xenotropic and Polytropic Retrovirus Receptor
8.
Exp Hematol ; 24(2): 364-70, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8641367

ABSTRACT

Friend mink cell focus-forming (FMCF) viruses are recombinants between the Friend murine leukemia virus (F-MuLV) and endogenous polytropic retroviruses involved in a number of retrovirus-induced malignancies of the myelo-erythroid compartment. To analyze the contribution of the viral cis regulatory elements to the host range determinants within the hematopoietic system, we performed a series of marker gene experiments using both transient transfection and retroviral-mediated stable transduction of indicator cell lines representing distinct developmental stages. According to our data, the U3 region in the long terminal repeat (LTR) of FMCF viruses possesses an enhancer assembly that allows efficient transcription in both early and late myelo-erythroid stem and progenitor cells. Retroviral gene expression, however, is subjected to stage-dependent transcriptional controls during blood cell maturation. We obtained evidence that a repressor element overlapping with the primer binding site in the viral leader region compromises U3-mediated gene expression in a stage-dependent manner, with the strongest restriction observed in the most primitive cells analyzed, FDCP-mix. In addition, our data indicate a second hurdle for retroviral gene expression in early hematopoietic cells that is independent of the primer binding site and most likely related to inefficient utilization of U3-located enhancers. These data shed light on the mechanisms of host range restriction within the hematopoietic system and define a basis for the design of retroviral vectors aimed to overcome transcriptional inefficiency in early hematopoietic cells. Thus, we developed novel retroviral vectors combining FMCF-type U3 regions with a permissive leader from the murine embryonic stem cell virus. These vectors are highly efficient for gene transfer and expression in both early and late myelo-erythroid cells, indicating that they will be of great use for a variety of experimental and therapeutic applications.


Subject(s)
Gene Expression Regulation, Viral , Hematopoiesis , Mink Cell Focus-Inducing Viruses/physiology , 3T3 Cells , Animals , Cell Differentiation , Cell Line , Enhancer Elements, Genetic , Fibroblasts , Friend murine leukemia virus/genetics , Genes, Reporter , Genetic Vectors , Hematopoietic Stem Cells , Humans , Mice , Mink Cell Focus-Inducing Viruses/genetics , Moloney murine leukemia virus/genetics , Repetitive Sequences, Nucleic Acid , Transcription, Genetic , Transfection , Tumor Cells, Cultured
9.
Virology ; 199(2): 497-9, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8122379

ABSTRACT

Whether the foci on S+L- mink cells transformed by the superinfection of MuLV were induced by the secondary infection of M-MSV pseudotype generated in the cells or by the secondary infection of MuLV has remained unresolved since the requirement of secondary infection was first reported (A. Ishimoto, J. Virol. 36, 18-21, 1980). Here, we show that infection with ecotropic MuLV of S+L- mink cells transfected with the receptor gene for ecotropic MuLV induced transformed foci resembling those induced by xenotropic and amphotropic viruses. This observation and our previous results (A. Ishimoto, J. Virol. 36, 18-21, 1980) that clonal lines of S+L- mink cells chronically infected with ecotropic MuLV are morphologically indistinguishable from normal S+L- mink cells suggest that the focus formation of S+L- mink cells by superinfection with MuLV is not due to secondary spread of helper virus which transactivates the expression of the v-mos oncogene by the MuLV, but is due to the secondary infection of the defective M-MSV genome. A new S+L- mink cell line with a receptor gene for ecotropic MuLV, designated ID cells, provided a new method for titrating the ecotropic MuLV that develop few XC foci and to simultaneously detect viruses in various host ranges.


Subject(s)
Cell Transformation, Viral/physiology , Mink Cell Focus-Inducing Viruses/physiology , Mink/microbiology , Moloney murine sarcoma virus/physiology , Animals , Cells, Cultured
10.
Genes Chromosomes Cancer ; 4(1): 46-57, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1377009

ABSTRACT

The karyotypic and phenotypic stability of cultured rat fibrosarcoma cells was challenged by infection with Moloney murine sarcoma virus (MoMuSV). After transformation, the spindle-like morphology of the parental HH-16 cl.2/1 cells had altered to a rounded phenotype, which was maintained in tumors produced by inoculating transformed cells into congenic animals. In contrast to the parental cells, transformed cells lacked cables of cytokeratins 14-16 and 19 and showed reduction of the mesenchymal marker protein vimentin. Additionally, the morphologically altered cell clones tf-1 to tf-3 had lost growth arrest in the presence of dexamethasone. The DNA of the transformed cells contained between four and six randomly integrated proviral copies. Karyotypic alterations were manifested by reduction of morphologically intact chromosomes in the MoMuSV-transformed cells together with increase of structural aberrations. Three additional markers were identified in the virus-transformed cell clones. Karyotypic instability induced by MoMuSV infection appeared closely related to reduction of the cellular differentiation status, although only cells of clone tf-1 had increased metastatic potential.


Subject(s)
Cell Transformation, Viral , Chromosome Aberrations , Moloney murine sarcoma virus/physiology , Sarcoma, Experimental/pathology , Age Factors , Animals , Animals, Newborn , Biomarkers, Tumor/analysis , Cell Differentiation , Cells, Cultured , DNA, Neoplasm/genetics , DNA, Viral/analysis , Extracellular Matrix Proteins/analysis , Female , Fibroblasts/microbiology , Fibroblasts/pathology , Fibrosarcoma/genetics , Fibrosarcoma/pathology , Karyotyping , Kidney , Mink Cell Focus-Inducing Viruses/physiology , Neoplasm Proteins/analysis , Neoplasm Transplantation , Proviruses/isolation & purification , Rats , Rats, Inbred Strains , Sarcoma, Experimental/genetics , Tumor Cells, Cultured/microbiology , Tumor Cells, Cultured/pathology , Tumor Cells, Cultured/transplantation , Virus Integration
11.
J Virol ; 65(5): 2408-14, 1991 May.
Article in English | MEDLINE | ID: mdl-1850020

ABSTRACT

The Friend or Moloney mink cell focus-forming (MCF) virus encodes a recombinant-type envelope glycoprotein, gp70, that is closely related to the membrane glycoprotein, gp55, of Friend spleen focus-forming virus (SFFV). We have shown previously that gp55 has the ability to activate cell growth by binding to the cellular receptor for erythropoietin. Here we show that gp70 encoded by either the Friend or Moloney MCF virus also binds to the erythropoietin receptor and that coexpression of the receptor and gp70 in an interleukin-3 (IL-3)-dependent cell line can activate IL-3-independent growth. Furthermore, when the cDNA for the human IL-2 receptor beta chain, which is related by sequence to the erythropoietin receptor, was introduced into this cell line, it became growth factor independent after infection either with SFFV or with one of the two MCF viruses but not with an ecotropic virus. Based on these observations, we propose a mechanism for the early stage of leukemogenesis induced by the MCF-type murine leukemia viruses.


Subject(s)
Cell Transformation, Neoplastic , Cell Transformation, Viral , Erythropoietin/metabolism , Leukemia/microbiology , Mink Cell Focus-Inducing Viruses/physiology , Retroviridae Proteins, Oncogenic/metabolism , Viral Envelope Proteins/metabolism , Animals , Cell Division , Cell Line , Electrophoresis, Polyacrylamide Gel , Friend murine leukemia virus/metabolism , Friend murine leukemia virus/physiology , Humans , Interleukin-3/pharmacology , Mice , Mink Cell Focus-Inducing Viruses/metabolism , Moloney murine leukemia virus/metabolism , Moloney murine leukemia virus/physiology , Receptors, Cell Surface/metabolism , Receptors, Erythropoietin , Receptors, Interleukin-2/metabolism
12.
J Virol ; 64(2): 493-8, 1990 Feb.
Article in English | MEDLINE | ID: mdl-2153219

ABSTRACT

Infection of certain strains of mice with the ecotropic Friend murine leukemia virus results in the generation of recombinant polytropic mink cell focus-inducing viruses and the development of erythroleukemia. We isolated a Friend mink cell focus-inducing virus (F-MCF-98D) from a Friend murine leukemia virus-infected BALB/c mouse which caused primarily a neurological disease as well as a low incidence of leukemia in susceptible IRW mice. Through genetic studies with the resistant C57BL/10 strain, we identified two genes which correlated with restricted viral replication and resistance to the development of disease caused by F-MCF-98D. One gene correlated with the expression of an endogenous gp70 linked to the Rmcf gene and might act by viral interference. The mechanism of action of the second gene was less clear, but it appeared to be associated with development of an antiviral antibody response.


Subject(s)
Central Nervous System Diseases/microbiology , Friend murine leukemia virus/genetics , Immunity, Innate , Leukemia Virus, Murine/genetics , Leukemia, Experimental/microbiology , Mink Cell Focus-Inducing Viruses/genetics , Animals , Antibodies, Viral/analysis , Antibody Formation , Cells, Cultured , Central Nervous System Diseases/immunology , Crosses, Genetic , Friend murine leukemia virus/immunology , Friend murine leukemia virus/physiology , Leukemia, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Mink Cell Focus-Inducing Viruses/immunology , Mink Cell Focus-Inducing Viruses/physiology , Species Specificity , Virus Replication
13.
Immunobiology ; 180(2-3): 195-207, 1990 Feb.
Article in English | MEDLINE | ID: mdl-2160912

ABSTRACT

Injection of the RadLV-Rs, a viral complex originally obtained from radio-induced thymic lymphosarcomas, into C57BL/6 mice induces a massive enlargement of spleen and lymph nodes. T- and B cell-proliferating populations display severe deterioration of their immune functions, resulting in death of the animals within three months. In contrast, the experiments reported here indicate that in such animals the natural killer (NK) activity is increased for about 2 months after viral injection. Interleukin 2 production is dramatically decreased, whereas interferon-gamma production is increased to twice the control value and thereafter decreases concomitantly with NK activity. This suggests that in RadLV-Rs-injected mice, the high NK activity is related to interferon-gamma production.


Subject(s)
Immunologic Deficiency Syndromes/immunology , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Killer Cells, Natural/metabolism , Leukemia Virus, Murine/physiology , Mink Cell Focus-Inducing Viruses/physiology , Retroviridae Infections/immunology , Acquired Immunodeficiency Syndrome , Animals , B-Lymphocytes/immunology , Disease Models, Animal , Female , Immunologic Deficiency Syndromes/etiology , Immunologic Deficiency Syndromes/pathology , Killer Cells, Natural/immunology , Male , Mice , Mice, Inbred C57BL/immunology , Retroviridae Infections/complications , T-Lymphocytes/immunology
14.
J Virol ; 62(9): 3156-66, 1988 Sep.
Article in English | MEDLINE | ID: mdl-2841468

ABSTRACT

We studied the relative importance of class I and class II major histocompatibility complex (MHC) immunoregulation in the control of T- and B-cell lymphomas induced by murine leukemia virus. Previously, we have described a mink cell focus-inducing (MCF) murine leukemia virus, MCF 1233, which induces not only lymphoblastic T-cell lymphomas but also follicle center cell or lymphoblastic B-cell lymphomas. We now report that the outcome of neonatal infection with MCF 1233 in H-2-congenic C57BL/10 and C57BL/6 mice is decisively influenced by the H-2 I-A locus. A total of 64% of H-2 I-Ak, d mice [B10.BR, B10.D2, B10.A(2R), B10.A(4R), and B10.MBR] developed T-cell lymphomas after MCF 1233 infection (mean latency, 37 weeks). In contrast, H-2 I-Ab [B10, B10.A(5R), B6], H-2 I-Ab/k [(B10.A x B10)F1 and (B10 x B10.A)F1], and H-2 I-Abm12 (bm12) mice were resistant against T-cell lymphomagenesis, but 65% of these H-2 I-Ab, b/k, bm12 animals developed B-cell lymphomas (mean latency, 71 weeks). Animals of T-cell lymphoma-susceptible strains that escaped from T-cell lymphomagenesis developed B-cell lymphomas with similar frequency as animals of T-cell lymphoma-resistant strains, but with a shorter latency. H-2 class II-determined regulation of antiviral immunity was reflected in the presence of high titers of antiviral envelope antibodies in T-cell lymphoma-resistant B-cell lymphoma-susceptible H-2 I-Ab, b/k, bm12 mice, whereas in T-cell lymphoma-susceptible H-2 I-Ak,d mice no antiviral antibodies were found. At week 4 after neonatal MCF 1233 infection, a high percentage of thymocytes were virally infected in both T-cell lymphoma-susceptible and -resistant mice. However, T-cell lymphoma-resistant animals cleared the thymic infection between weeks 4 and 10 of age, coinciding with a sharp rise in serum levels of antiviral antibodies. We conclude that the pleiotropic effects of MCF 1233 infection in H-2-congenic mice result from MHC class II I-A-determined T-cell response differences.


Subject(s)
Genes, MHC Class II , Leukemia Virus, Murine/immunology , Lymphoma/immunology , Mink Cell Focus-Inducing Viruses/immunology , Animals , Antibodies, Viral/biosynthesis , Antigens, Viral/analysis , B-Lymphocytes , Bone Marrow/microbiology , Disease Susceptibility , Fluorescent Antibody Technique , Immunity, Innate , Leukemia, Experimental/immunology , Lymphoma/genetics , Lymphoma/pathology , Mice , Mice, Mutant Strains , Mink Cell Focus-Inducing Viruses/physiology , Spleen/microbiology , T-Lymphocytes , Thymus Gland/microbiology
15.
J Exp Med ; 167(5): 1535-46, 1988 May 01.
Article in English | MEDLINE | ID: mdl-2835418

ABSTRACT

The Rmcf locus restricts the in vitro replication of mink cell focus-forming (MCF) viruses in cell cultures derived from mice carrying the resistance allele. Previously we reported that in cell cultures from first backcross progeny, this Rmcf-linked restriction segregates with the expression of an endogenous retroviral gp70 serologically related to that of MCF viruses. The current report details the results of genetic studies designed to examine the possible association of this endogenous gp70 with resistance of mice to Friend murine leukemia virus (F-MuLV)-induced erythroleukemia. This env gene segregates as a single dominant trait in (DBA/2 X IRW) X IRW progeny, in which the expression of the gene can be detected by serological techniques. Results indicated that the gp70- progeny developed leukemia at the same rate as the susceptible IRW parent, whereas the tempo of disease among the gp70+ progeny was significantly slower. However, the resistance mediated by this gene was only partial, since most of the gp70+ offspring eventually developed erythroleukemia when followed for 6 mo. This endogenous gp70 also segregated with a restriction to the expression of recombinant MCF viruses after infection with F-MuLV. Since in this study all unlinked genes segregated independently, this is direct evidence that MCF viruses participate in the induction of erythroleukemia.


Subject(s)
Genes, Viral , Leukemia Virus, Murine/physiology , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Experimental/genetics , Mice, Inbred DBA/genetics , Mice, Inbred Strains/genetics , Mink Cell Focus-Inducing Viruses/physiology , Retroviridae Proteins, Oncogenic , Retroviridae Proteins/physiology , Viral Envelope Proteins/physiology , Viral Interference , Animals , Cells, Cultured , Crosses, Genetic , Fibroblasts/microbiology , Friend murine leukemia virus/physiology , Genes, Dominant , Immunity, Innate , Leukemia, Erythroblastic, Acute/microbiology , Leukemia, Experimental/microbiology , Mice , Mice, Inbred DBA/microbiology , Mice, Inbred Strains/microbiology , Mink Cell Focus-Inducing Viruses/genetics , Mink Cell Focus-Inducing Viruses/isolation & purification , Retroviridae Proteins/genetics , Viral Envelope Proteins/genetics , Virus Replication
16.
Virology ; 154(2): 420-4, 1986 Oct 30.
Article in English | MEDLINE | ID: mdl-3020789

ABSTRACT

Two viruses which do not give rise to XC plaques in the standard XC assay (XC-negative) have been isolated from the Rauscher virus (RV) complex. These viruses differ in their host range. One, R-MCF-1, is dualtropic and will therefore infect both murine and non-murine cells. However, unlike other mink cell focus-inducing (MCF) viruses, it cannot infect NIH 3T3 cells. The other, R-XC-, is ecotropic. It will infect murine cells, including NIH 3T3 cells, but does not infect mink lung cells. Analysis of hybrid viruses, in which homologous regions of the genomes of R-MCF-1 and R-XC- virus were exchanged, indicated that the NH2-terminal portion of the gp70 is responsible for the particular host ranges of these viruses. The nucleotide sequence of the env gene of R-XC- virus was therefore determined and compared with the known env sequences of ecotropic MLVs and dualtropic MCF viruses of the Rauscher and Friend virus complexes. R-XC- virus was found to be a recombinant virus. Its env gene contained sequences derived from an endogenous env gene which were closely related to those of the MCF viruses but differed from any previously described sequences. The particular properties of R-MCF-1 and R-XC- virus suggest that the two viruses arose by recombination between R-MLV and two endogenous env sequences which differ from those of the known MCF viruses. If so, this suggests that the mouse genome contains at least five env sequences which can give rise to MCF-like viruses. In addition, since the host range and interference properties of R-XC- virus are very similar to those of the previously described ecotropic recombinant viruses, it may be that the ecotropic recombinant viruses arose by recombination with the same endogenous env sequences as did R-XC- virus.


Subject(s)
Genes, Viral , Leukemia Virus, Murine/genetics , Mink Cell Focus-Inducing Viruses/genetics , Rauscher Virus/genetics , Viral Envelope Proteins/genetics , Animals , Base Sequence , Cell Line , Mice , Mink Cell Focus-Inducing Viruses/physiology , Rauscher Virus/physiology
17.
Virology ; 151(1): 31-40, 1986 May.
Article in English | MEDLINE | ID: mdl-3008431

ABSTRACT

A new class of MuLV has been detected and isolated from normal and leukemic AKR, C58, SJL, and NFS.AKV mice as well as from NFS mice inoculated with Friend or Moloney ecotropic viruses. These new viruses are XC negative and serologically cross-react with MCF env antigens but are ecotropic in host range, being able to only infect mouse cells to varying degrees and unable to infect mink or other cells infectable by MCF or xenotropic viruses. Viruses of this type from AKR mice cross-interfere with Moloney ecotropic and MCF viruses in SC-1 cells and appear to have properties similar to those of the SL3-2, GPA-V2, and R-XC- isolates. Analysis of their genomes by restriction endonuclease mapping of proviral DNA indicates structures similar to class II MCFs with the 5' half of the genome being like ecotropic viruses and the env region exhibiting restriction sites characteristic of MCF viruses. In normal AKR mice, these ecotropic recombinant-like viruses are found in spleen and bone marrow as early as 1 week of age, but first appear in the thymus at 3-4 months of age. These viruses have not been detected in mice with no or low expression of ecotropic viruses (NFS, NZB, DBA/2, BALB/c, C57BL/6). Because of their apparent recombinant structure and ecotropic host range we have provisionally designated them ecotropic recombinant virus (ERV) to distinguish them from the MCF class of recombinant MuLV.


Subject(s)
Leukemia Virus, Murine/genetics , Leukemia, Experimental/microbiology , Recombination, Genetic , Animals , Bone Marrow/microbiology , Cell Line , DNA Restriction Enzymes , Genes, Viral , Leukemia Virus, Murine/growth & development , Leukemia Virus, Murine/physiology , Mice , Mice, Inbred AKR , Mice, Inbred Strains , Mink Cell Focus-Inducing Viruses/physiology , Moloney murine leukemia virus/physiology , Spleen/microbiology , Thymus Gland/microbiology , Viral Interference
SELECTION OF CITATIONS
SEARCH DETAIL
...