Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Genes Immun ; 14(6): 365-72, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23698707

RESUMO

Using an experimental evolution approach, we recently demonstrated that the mouse-specific pathogen Friend virus (FV) complex adapted to specific major histocompatibility complex (MHC) genotypes, which resulted in fitness tradeoffs when viruses were exposed to hosts possessing novel MHC polymorphisms. Here we report the analysis of patterns of pathogen adaptation and virulence evolution from viruses adapting to one of three hosts that differ across the entire genome (A/WySn, DBA/2J and BALB/c). We found that serial passage of FV complex through these mouse genotypes resulted in significant increases in pathogen fitness (156-fold) and virulence (11-fold). Adaptive responses by post-passage viruses also resulted in host-genotype-specific patterns of adaptation. To evaluate the relative importance of MHC versus non-MHC polymorphisms as factors influencing pathogen adaptation and virulence, we compared the magnitude of fitness tradeoffs incurred by post-passage viruses when infecting hosts possessing either novel MHC polymorphisms alone or hosts possessing novel MHC and non-MHC polymorphisms. MHC polymorphisms alone accounted for 71% and 83% of the total observed reductions in viral fitness and virulence in unfamiliar host genotypes, respectively. Strikingly, these data suggest that genetic polymorphisms within the MHC, a gene region representing only -0.1% of the genome, are major host factors influencing pathogen adaptation and virulence evolution.


Assuntos
Adaptação Fisiológica/genética , Evolução Molecular , Vírus da Leucemia Murina de Friend/patogenicidade , Interações Hospedeiro-Parasita , Complexo Principal de Histocompatibilidade/genética , Polimorfismo Genético , Animais , Vírus da Leucemia Murina de Friend/genética , Aptidão Genética , Genótipo , Especificidade de Hospedeiro , Camundongos , Camundongos Endogâmicos , Virulência/genética
2.
Proc Natl Acad Sci U S A ; 98(16): 9226-30, 2001 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-11459933

RESUMO

Normal levels of CD4(+) regulatory T cells are critical for the maintenance of immunological homeostasis and the prevention of autoimmune diseases. However, we now show that the expansion of CD4(+) regulatory T cells in response to a chronic viral infection can lead to immunosuppression. Mice persistently infected with Friend retrovirus develop approximately twice the normal percentage of splenic CD4(+) regulatory T cells and lose their ability to reject certain tumor transplants. The role of CD4(+) regulatory T cells was demonstrated by the transmission of immunosuppression to uninfected mice by adoptive transfers of CD4(+) T cells. CD4(+) T cells from chronically infected mice were also immunosuppressive in vitro, inhibiting the generation of cytolytic T lymphocytes in mixed lymphocyte cultures. Inhibition occurred at the level of blast-cell formation through a mechanism or mechanisms involving transforming growth factor-beta and the cell surface molecule CTLA-4 (CD152). These results suggest a possible explanation for HIV- and human T cell leukemia virus-I-induced immunosuppression in the absence of T cell depletion.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Vírus da Leucemia Murina de Friend/fisiologia , Tolerância Imunológica , Infecções por Retroviridae/imunologia , Transferência Adotiva , Animais , Camundongos , Camundongos Endogâmicos C57BL
3.
J Virol ; 75(11): 5018-26, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11333881

RESUMO

Infectious entry of enveloped viruses is thought to proceed by one of two mechanisms. pH-dependent viruses enter the cells by receptor-mediated endocytosis and are inhibited by transient treatment with agents that prevent acidification of vesicles in the endocytic pathway, while pH-independent viruses are not inhibited by such agents and are thought to enter the cell by direct fusion with the plasma membrane. Nearly all retroviruses, including amphotropic murine leukemia virus (MuLV) and human immunodeficiency virus type 1, are classified as pH independent. However, ecotropic MuLV is considered to be a pH-dependent virus. We have examined the infectious entry of ecotropic and amphotropic MuLVs and found that they were equally inhibited by NH4Cl and bafilomycin A. These agents inhibited both viruses only partially over the course of the experiments. Agents that block the acidification of endocytic vesicles also arrest vesicular trafficking. Thus, partial inhibition of the MuLVs could be the result of virus inactivation during arrest in this pathway. In support of this contention, we found that that the loss of infectivity of the MuLVs during treatment of target cells with the drugs closely corresponded to the loss of activity due to spontaneous inactivation at 37 degrees C in the same period of time. Furthermore, the drugs had no effect on the efficiency of infection under conditions in which the duration of infection was held to a very short period to minimize the effects of spontaneous inactivation. These results indicate that the infectious processes of both ecotropic and amphotropic MuLVs were arrested rather than aborted by transient treatment of the cells with the drugs. We also found that infectious viruses were efficiently internalized during treatment. This indicated that the arrest occurred in an intracellular compartment and that the infectious process of both the amphotropic and ecotropic MuLVs very likely involved endocytosis. An important aspect of this study pertains to the interpretation of experiments in which agents that block endocytic acidification inhibit infectivity. As we have found with the MuLVs, inhibition of infectivity may be secondary to the block of endocytic acidification. While this strongly suggests the involvement of an endocytic pathway, it does not necessarily indicate a requirement for an acidic compartment during the infectious process. Likewise, a lack of inhibition during transient treatment with the drugs would not preclude an endocytic pathway for viruses that are stable during the course of the treatment.


Assuntos
Células Eucarióticas/virologia , Vírus da Leucemia Murina/patogenicidade , Macrolídeos , Células 3T3 , Cloreto de Amônio/farmacologia , Animais , Antibacterianos/farmacologia , Transporte Biológico/efeitos dos fármacos , Membrana Celular/virologia , Endocitose/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Células Eucarióticas/efeitos dos fármacos , Concentração de Íons de Hidrogênio , Vírus da Leucemia Murina/metabolismo , Camundongos , ATPases Translocadoras de Prótons/antagonistas & inibidores , Temperatura , Fatores de Tempo
4.
J Virol ; 75(2): 654-60, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11134279

RESUMO

The immunological resistance of a host to viral infections may be strongly influenced by cytokines such as interleukin-12 (IL-12) and gamma interferon (IFN-gamma), which promote T helper type 1 responses, and IL-4, which promotes T helper type 2 responses. We studied the role of these cytokines during primary and secondary immune responses against Friend retrovirus infections in mice. IL-4- and IL-12-deficient mice were comparable to wild-type B6 mice in the ability to control acute and persistent Friend virus infections. In contrast, more than one-third of the IFN-gamma-deficient mice were unable to maintain long-term control of Friend virus and developed gross splenomegaly with high virus loads. Immunization with a live attenuated vaccine virus prior to challenge protected all three types of cytokine-deficient mice from viremia and high levels of spleen virus despite the finding that the vaccinated IFN-gamma-deficient mice were unable to class switch from immunoglobulin M (IgM) to IgG virus-neutralizing antibodies. The results indicate that IFN-gamma plays an important role during primary immune responses against Friend virus but is dispensable during vaccine-primed secondary responses.


Assuntos
Citocinas/fisiologia , Vírus da Leucemia Murina de Friend/imunologia , Infecções por Retroviridae/imunologia , Infecções Tumorais por Vírus/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Citocinas/genética , Feminino , Vírus da Leucemia Murina de Friend/isolamento & purificação , Vírus da Leucemia Murina de Friend/fisiologia , Interferon gama/genética , Interferon gama/fisiologia , Interleucina-12/genética , Interleucina-12/fisiologia , Interleucina-4/genética , Interleucina-4/fisiologia , Leucemia Experimental/imunologia , Leucemia Experimental/virologia , Camundongos , Camundongos Endogâmicos C57BL , Testes de Neutralização , Infecções por Retroviridae/virologia , Baço/virologia , Infecções Tumorais por Vírus/virologia , Vacinação/métodos , Carga Viral , Viremia/virologia
5.
J Virol ; 75(1): 52-60, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11119573

RESUMO

We have used the Friend virus model to determine the basic mechanisms by which the immune system can control persistent retroviral infections. Previously we showed that CD4(+) T cells play an essential role in keeping persistent retrovirus in check. The present in vitro experiments with a Friend virus-specific CD4(+) T-cell clone revealed that these cells produce gamma interferon (IFN-gamma), which acts with two distinct mechanisms of antiviral activity. First, IFN-gamma had a direct inhibitory effect on virus production. This inhibitory effect was noncytolytic and, interestingly, was not associated with decreased cell surface expression of viral antigens. The second mechanism of IFN-gamma-mediated antiviral activity was an enhancement of CD4(+) T-cell-mediated cytolytic activity. We also found an in vivo role for IFN-gamma in the control of persistent Friend virus infections. Neutralization of IFN-gamma in persistently infected mice resulted in significantly increased levels of virus in the spleen, and a significant percentage of IFN-gamma-deficient mice were unable to maintain long-term control over Friend virus infections.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Vírus da Leucemia Murina de Friend , Interferon gama/fisiologia , Infecções por Retroviridae/imunologia , Infecções Tumorais por Vírus/imunologia , Animais , Comunicação Celular , Citotoxicidade Imunológica , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Tumorais Cultivadas
6.
Curr Mol Med ; 1(4): 431-6, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11899087

RESUMO

More than 15 years after the discovery of human immunodeficiency virus (HIV), researchers are still struggling to design a protective AIDS vaccine. A remaining problem is a lack of basic knowledge about the immunological requirements for protection against retroviruses. Infection of macaque monkeys with simian immunodeficiency virus is still the best model for HIV vaccine research. However, in this model it remains difficult to determine protective immunological mechanisms because of limited numbers of experimental animals and their genetic heterogeneity. Thus, fundamental concepts in retroviral immunology have to be defined in other ways such as mouse models. This minireview summarizes new findings on cellular and molecular mechanisms in protection of mice against Friend murine retrovirus infection. It has been shown that complex immune responses, including B and T cell responses, are required for efficient protection in this model. Multiple viral antigens are necessary to elicit such broad immune reactivity. Efficacious vaccines must protect not only against acute disease, but also against the establishment of persistent infections or the host is at serious risk of virus reactivation. The minireview closes with a discussion on the relevance of findings from the mouse model on the design of a protective vaccine against HIV.


Assuntos
Infecções por Retroviridae/imunologia , Infecções por Retroviridae/prevenção & controle , Vacinas Virais/farmacologia , Vacinas contra a AIDS/farmacologia , Animais , Linfócitos B/imunologia , Vírus da Leucemia Murina de Friend/imunologia , Infecções por HIV/imunologia , Infecções por HIV/prevenção & controle , Humanos , Leucemia Experimental/imunologia , Leucemia Experimental/prevenção & controle , Camundongos , Modelos Biológicos , Vacinas contra a SAIDS/farmacologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Linfócitos T/imunologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/prevenção & controle
7.
Virology ; 272(1): 177-82, 2000 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-10873760

RESUMO

The propensity of retroviruses to rapidly establish persistent infections poses a formidable problem in vaccination strategies. In the current study, we use a live attenuated vaccine to study protection against acute and persistent Friend virus infections in mice. Adoptive transfers of immune CD8(+) T cells combined with passive immunizations with virus-neutralizing antibodies increased protection against acute disease compared with either treatment alone, but there was no protection against the establishment of persistent infection. In addition, the protection against acute disease elicited by the combination treatment was dependent on endogenous CD4(+) T cells as no protection was achieved in CD4(+) T-cell-depleted mice. Quantitative studies showed that doubling the numbers of immune lymphocytes used in adoptive transfer experiments increased protection against acute disease depending on the type of lymphocyte subset used in the transfer. CD8(+) T cells were the most potent subset for the transfer of such protection. However, even high numbers of immune CD8(+) T cells gave no protection against the establishment of persistent infections. The data indicate that strengthening the numbers of specific immune cell subsets may have a beneficial effect on protection against acute disease, but protection from establishment of persistence requires complex immune responses involving multiple lymphocyte subsets.


Assuntos
Vírus da Leucemia Murina de Friend/imunologia , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/prevenção & controle , Doença Aguda , Transferência Adotiva , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/administração & dosagem , Anticorpos Antivirais/imunologia , Linfócitos B/imunologia , Linfócitos B/transplante , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/transplante , Doença Crônica , Feminino , Vírus da Leucemia Murina de Friend/fisiologia , Camundongos , Testes de Neutralização , Infecções por Retroviridae/patologia , Infecções por Retroviridae/virologia , Esplenomegalia/imunologia , Esplenomegalia/patologia , Esplenomegalia/prevenção & controle , Esplenomegalia/virologia , Subpopulações de Linfócitos T/imunologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/patologia , Infecções Tumorais por Vírus/prevenção & controle , Infecções Tumorais por Vírus/virologia , Vacinas Atenuadas/imunologia , Vacinas Virais/imunologia , Viremia/imunologia , Viremia/prevenção & controle , Viremia/virologia
9.
J Virol ; 74(11): 5363-7, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10799615

RESUMO

Recovery from leukemia induced by Friend virus complex (FV) requires strong CD4(+) helper, CD8(+) cytotoxic T-lymphocyte, and B-cell responses. The development of these immune responses is dependent on the major histocompatibility complex (MHC) (H-2) genotype of the mouse. In H-2(b/b) mice, which spontaneously recover from FV-induced erythroleukemia, neutralization of gamma interferon (IFN-gamma) in vivo inhibited recovery, which indicated that IFN-gamma was a necessary component of the immune response to FV. Furthermore, in H-2(b/b) mice, high numbers of IFN-gamma-producing cells were detected after FV infection, whereas in H-2(a/b) mice, which have a low-recovery phenotype, only low numbers of IFN-gamma-producing cells were detected. Similarly, H-2(bm14/b) mice, which cannot recover from FV infection due to a point mutation in one allele of the H-2D(b) gene, also had low numbers of IFN-gamma-producing T cells. Surprisingly, this effect was observed for both CD8(+) and CD4(+) T cells. These findings reveal a novel influence of MHC class I genes on CD4(+) T-cell responses to viral infection. Furthermore, the influence of MHC class I genotype on the generation of both IFN-gamma-producing CD4(+) and CD8(+) T cells helps explain the major impact of the H-2D gene on recovery from FV disease.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Vírus da Leucemia Murina de Friend/imunologia , Antígenos H-2/genética , Interferon gama/biossíntese , Leucemia Experimental/imunologia , Infecções por Retroviridae/imunologia , Infecções Tumorais por Vírus/imunologia , Animais , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/virologia , Antígeno de Histocompatibilidade H-2D , Ativação Linfocitária , Camundongos , Mutação Puntual , Baço/citologia , Baço/imunologia , Esplenomegalia/imunologia
10.
J Virol ; 73(10): 8435-40, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10482595

RESUMO

Vaccination of mice with a live attenuated vaccine virus induces potent protection against subsequent challenge with pathogenic Friend retroviral complex. The kinetic studies presented here demonstrate protection from acute splenomegaly as early as 1 week postvaccination. At this time point virus-specific cytotoxic T lymphocytes (CTL) were demonstrable in direct chromium release assays. However, during the first 2 weeks after vaccination protection was incomplete since the mice were not protected against establishment of low-level persistent infections in the spleen. By 3 weeks postvaccination the animals were protected against the establishment of persistent virus as well as acute splenomegaly. The timing of this complete protection correlated with the presence of both virus-neutralizing antibodies and primed CTL in the immunized mice. Within 3 days of virus challenge, vaccinated mice showed high levels of activated B cells and CD4(+) and CD8(+) T cells, indicating an efficient priming of all lymphocyte subsets. Despite very limited replication of the vaccine virus, the protective effect was long lived and was still present 6 months after immunization.


Assuntos
Imunidade , Infecções por Retroviridae/imunologia , Retroviridae/imunologia , Vacinas Virais/imunologia , Animais , Linfócitos B/imunologia , Feminino , Camundongos , Infecções por Retroviridae/prevenção & controle , Linfócitos T Citotóxicos/imunologia , Vacinas Virais/administração & dosagem
11.
J Virol ; 73(9): 7848-52, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10438878

RESUMO

Rfv3 is a host resistance gene that operates through an unknown mechanism to control the development of the virus-neutralizing antibody response required for recovery from infection with Friend retrovirus. The Rfv3 gene was previously mapped to an approximately 20-centimorgan (cM) region of chromosome 15. More refined mapping was not possible, due to a lack of microsatellite markers and leakiness in the Rfv3 phenotype, which prevented definitive phenotyping of individual recombinant mice. In the present study, we overcame these difficulties by taking advantage of seven new microsatellite markers in the Rfv3 region and by using progeny tests to accurately determine the Rfv3 phenotype of recombinant mice. Detailed linkage analysis of relevant crossovers narrowed the location of Rfv3 to a 0.83-cM region. Mapping of closely linked genes in an interspecific backcross panel allowed us to exclude two previous candidate genes, Ly6 and Wnt7b. These studies also showed for the first time that the Hsf1 gene maps to the Rfv3-linked cluster of genes including Il2rb, Il3rb, and Pdgfb. This localization of Rfv3 to a region of less than 1 cM now makes it feasible to attempt the cloning of Rfv3 by physical methods.


Assuntos
Mapeamento Cromossômico , Vírus da Leucemia Murina de Friend/imunologia , Infecções por Retroviridae/genética , Infecções Tumorais por Vírus/genética , Animais , Proteínas de Ligação a DNA/genética , Feminino , Fatores de Transcrição de Choque Térmico , Imunidade Inata/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Retroviridae/imunologia , Fatores de Transcrição , Infecções Tumorais por Vírus/imunologia
12.
J Virol ; 73(8): 6468-73, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10400741

RESUMO

The study of genetic resistance to retroviral diseases provides insights into the mechanisms by which organisms overcome potentially lethal infections. Fv-2 resistance to Friend virus-induced erythroleukemia acts through nonimmunological mechanisms to prevent early virus spread, but it does not completely block infection. The current experiments were done to determine whether Fv-2 alone could provide resistance or whether immunological mechanisms were also required to bring infection under control. Fv-2-resistant mice that were CD4(+) T-cell deficient were able to restrict early virus replication and spread as well as normal Fv-2-resistant mice, but they could not maintain control and developed severe Friend virus-induced splenomegaly and erythroleukemia by 6 to 8 weeks postinfection. Mice deficient in CD8(+) T cells and, to a lesser extent, B cells were also susceptible to late Friend virus-induced disease. Thus, Fv-2 resistance does not independently prevent FV-induced erythroleukemia but works in concert with the immune system by limiting early infection long enough to allow virus-specific immunity time to develop and facilitate recovery.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Vírus da Leucemia Murina de Friend/imunologia , Leucemia Eritroblástica Aguda/imunologia , Leucemia Experimental/imunologia , Infecções por Retroviridae/imunologia , Infecções Tumorais por Vírus/imunologia , Animais , Suscetibilidade a Doenças/imunologia , Feminino , Imunidade Inata , Masculino , Camundongos , Camundongos Endogâmicos C57BL
14.
J Virol ; 73(5): 3753-7, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10196269

RESUMO

Many human viruses not only cause acute diseases but also establish persistent infections. Such persistent viruses can cause chronic diseases or can reactivate to cause acute diseases in AIDS patients or patients receiving immunosuppressive therapies. While the prevention of persistent infections is an important consideration in the design of modern vaccines, surprisingly little is known about this aspect of protection. In the current study, we tested the feasibility of vaccine prevention of retroviral persistence by using a Friend virus model that we recently developed. In this model, persistent virus can be detected at very low levels by immunosuppressing the host to reactivate virus or by transferring persistently infected spleen cells into highly susceptible mice. Two vaccines were analyzed, a recombinant vaccinia virus vector expressing Friend virus envelope protein and a live attenuated Friend virus. Both vaccines reduced pathogenic virus loads to levels undetectable by infectious center assays. However, only the live, attenuated vaccine prevented immunosuppression-induced reactivation of persistent virus. Thus, even very low levels of persistent Friend virus posed a significant threat during immunosuppression. Our results demonstrate that vaccine protection against establishment of retroviral persistence is attainable.


Assuntos
Vírus da Leucemia Murina de Friend/imunologia , Infecções por Retroviridae/prevenção & controle , Infecções Tumorais por Vírus/prevenção & controle , Vacinas Virais/imunologia , Latência Viral , Animais , Anticorpos Antivirais/imunologia , Feminino , Vírus da Leucemia Murina de Friend/fisiologia , Imunização Passiva , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Testes de Neutralização , Vacinas Atenuadas/imunologia
15.
Nat Med ; 5(2): 189-93, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9930867

RESUMO

Infection by live attenuated retroviruses provides excellent protection from challenge with pathogenic viruses in several animal models, but little is known about which immune effectors are necessary for protection. We examined this using adoptive transfer experiments in the Friend virus mouse model. Transfers of immune spleen cells into naive mice conferred complete protection, and transfers of purified lymphocyte subsets demonstrated that this effect required complex immune responses involving CD4+ and CD8+ T cells and also B cells. In addition, passive immunization experiments demonstrated that antibodies alone reduced virus loads but did not prevent infection. These findings may have implications for retroviral vaccine design in general.


Assuntos
Subpopulações de Linfócitos/imunologia , Infecções por Retroviridae/prevenção & controle , Vacinas Atenuadas/imunologia , Vacinas Virais/imunologia , Transferência Adotiva/métodos , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Antivirais , Feminino , Vírus da Leucemia Murina de Friend , Camundongos , Testes de Neutralização , Infecções por Retroviridae/imunologia
16.
Virology ; 248(1): 66-73, 1998 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-9705256

RESUMO

Several immunological epitopes are known to be located within the Friend murine leukemia virus (F-MuLV) envelope protein, but their relative contributions to protection from Friend virus-induced disease are not known. To determine how expression of various immunological determinants affected protection, mice were immunized with recombinant vaccinia viruses expressing different portions of the F-MuLV envelope protein, and they were then challenged with a lethal dose of Friend virus complex. The disease parameters that were followed in the mice were early viremia, early splenomegaly, and late splenomegaly. Both the N-terminal and C-terminal portions of the F-MuLV gp70 were found to protect against late splenomegaly, the primary clinical sign associated with virus-induced erythroleukemia. However, neither region alone protected against early splenomegaly and early viremia, indicating poor immunological control over early virus replication and spread through the spleen and blood. In contrast, mice immunized with a vaccine expressing the entire F-MuLV envelope protein were protected against all three disease parameters. The results indicated that expression of multiple immunological determinants including both T-helper and B cell epitopes was necessary for full protection.


Assuntos
Epitopos/análise , Vírus da Leucemia Murina de Friend/imunologia , Leucemia Experimental/imunologia , Infecções por Retroviridae/imunologia , Infecções Tumorais por Vírus/imunologia , Vacinas Sintéticas , Proteínas do Envelope Viral/imunologia , Vacinas Virais , Animais , Leucemia Experimental/patologia , Leucemia Experimental/prevenção & controle , Camundongos , Camundongos Endogâmicos , Proteínas Recombinantes de Fusão/imunologia , Infecções por Retroviridae/patologia , Infecções por Retroviridae/prevenção & controle , Esplenomegalia , Infecções Tumorais por Vírus/patologia , Infecções Tumorais por Vírus/prevenção & controle
17.
J Virol ; 72(8): 6554-8, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9658099

RESUMO

Live-attenuated retroviruses have been shown to be effective retroviral vaccines, but currently little is known regarding the mechanisms of protection. In the present studies, we used Friend virus as a model to analyze characteristics of a live-attenuated vaccine in protection against virus-induced disease. Highly susceptible mice were immunized with nonpathogenic Friend murine leukemia helper virus (F-MuLV), which replicates poorly in adult mice. Further attenuation of the vaccine virus was achieved by crossing the Fv-1 genetic resistance barrier. The minimum dose of vaccine virus required to protect 100% of the mice against challenge with pathogenic Friend virus complex was determined to be 10(3) focus-forming units of attenuated virus. Live vaccine virus was necessary for induction of immunity, since inactivated F-MuLV did not induce protection. To determine whether immune cells mediated protection, spleen cells from vaccinated donor mice were adoptively transferred into syngeneic recipients. The results indicated that immune mechanisms rather than viral interference mediated protection.


Assuntos
Vírus da Leucemia Murina de Friend/imunologia , Leucemia Experimental/prevenção & controle , Infecções por Retroviridae/prevenção & controle , Infecções Tumorais por Vírus/prevenção & controle , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/imunologia , Leucemia Experimental/imunologia , Camundongos , Testes de Neutralização , Infecções por Retroviridae/imunologia , Baço/citologia , Baço/imunologia , Infecções Tumorais por Vírus/imunologia , Vacinação , Vacinas Atenuadas/imunologia , Viremia/imunologia , Viremia/prevenção & controle
18.
J Virol ; 72(8): 6559-64, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9658100

RESUMO

Reactivations of persistent viral infections pose a significant medical problem in immunocompromised cancer, transplant, and AIDS patients, yet little is known about how persistent viral infections are immunologically controlled. Here we describe a mouse model for investigating the role of the immune response in controlling a persistent retroviral infection. We demonstrate that, following recovery from acute Friend virus infection, a small number of B cells evade immunological destruction and harbor persistent virus. In vivo depletions of T-cell subsets in persistently infected mice revealed a critical role for CD4(+) T cells in controlling virus replication, spread to the erythroid lineage, and induction of erythroleukemia. The CD4(+) T-cell effect was independent of CD8(+) T cells and in some cases was also independent of virus-neutralizing antibody responses. Thus, the CD4(+) T cells may have had a direct antiviral effect. These results may have relevance for human immunodeficiency virus (HIV) infections where loss of CD4(+) T cells is associated with an increase in HIV replication, reactivation of persistent viruses, and a high incidence of virus-associated cancers.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Vírus da Leucemia Murina de Friend/imunologia , Leucemia Experimental/imunologia , Infecções por Retroviridae/imunologia , Infecções Tumorais por Vírus/imunologia , Animais , Linfócitos B/virologia , Linfócitos T CD8-Positivos/imunologia , Feminino , Vírus da Leucemia Murina de Friend/fisiologia , Leucemia Eritroblástica Aguda , Leucemia Experimental/virologia , Depleção Linfocítica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Retroviridae/virologia , Esplenomegalia , Infecções Tumorais por Vírus/virologia , Latência Viral , Replicação Viral
19.
J Virol ; 71(7): 5287-94, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9188597

RESUMO

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.


Assuntos
Encéfalo/virologia , Leucemia Experimental/virologia , Microglia/virologia , Vírus Indutores de Focos em Células do Vison/isolamento & purificação , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia , Animais , Encéfalo/citologia , Leucemia Experimental/patologia , Camundongos , Camundongos Endogâmicos , Vírus Indutores de Focos em Células do Vison/patogenicidade , Infecções por Retroviridae/patologia , Infecções Tumorais por Vírus/patologia , Carga Viral , Virulência
20.
Proc Natl Acad Sci U S A ; 94(15): 7811-6, 1997 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-9223268

RESUMO

Friend virus infection of adult immunocompetent mice is a well established model for studying genetic resistance to infection by an immunosuppressive retrovirus. This paper reviews both the genetics of immune resistance and the types of immune responses required for recovery from infection. Specific major histocompatibility complex (MHC) class I and II alleles are necessary for recovery, as is a non-MHC gene, Rfv-3, which controls virus-specific antibody responses. In concordance with these genetic requirements are immunological requirements for cytotoxic T lymphocyte, T helper, and antibody responses, each of which provides essential nonoverlapping functions. The complexity of responses necessary for recovery from Friend virus infection has implications for both immunotherapies and vaccines. For example, it is shown that successful passive antibody therapy is dependent on MHC type because of the requirement for T cell responses. For vaccines, successful immunization requires priming of both T cell and B cell responses. In vivo depletion experiments demonstrate different requirements for CD8(+) T cells depending on the vaccine used. The implications of these studies for human retroviral diseases are discussed.


Assuntos
Vírus da Leucemia Murina de Friend/isolamento & purificação , Infecções por Retroviridae/imunologia , Animais , Genes MHC Classe I , Genes MHC da Classe II , Predisposição Genética para Doença , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Modelos Biológicos , Infecções por Retroviridae/genética , Infecções por Retroviridae/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA