Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Immunity ; 47(2): 310-322.e7, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28813660

RESUMO

Select humans and animals control persistent viral infections via adaptive immune responses that include production of neutralizing antibodies. The precise genetic basis for the control remains enigmatic. Here, we report positional cloning of the gene responsible for production of retrovirus-neutralizing antibodies in mice of the I/LnJ strain. It encodes the beta subunit of the non-classical major histocompatibility complex class II (MHC-II)-like molecule H2-O, a negative regulator of antigen presentation. The recessive and functionally null I/LnJ H2-Ob allele supported the production of virus-neutralizing antibodies independently of the classical MHC haplotype. Subsequent bioinformatics and functional analyses of the human H2-Ob homolog, HLA-DOB, revealed both loss- and gain-of-function alleles, which could affect the ability of their carriers to control infections with human hepatitis B (HBV) and C (HCV) viruses. Thus, understanding of the previously unappreciated role of H2-O (HLA-DO) in immunity to infections may suggest new approaches in achieving neutralizing immunity to viruses.


Assuntos
Anticorpos Neutralizantes , Antígenos HLA-D/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Imunidade Humoral , Vírus do Tumor Mamário do Camundongo/imunologia , Vírus Rauscher/imunologia , Infecções por Retroviridae/imunologia , Animais , Anticorpos Neutralizantes/metabolismo , Anticorpos Antivirais/metabolismo , Apresentação de Antígeno/genética , Biologia Computacional , Feminino , Predisposição Genética para Doença , Antígenos HLA-D/genética , Células HeLa , Hepatite B/imunologia , Hepatite B/transmissão , Hepatite C/imunologia , Hepatite C/transmissão , Antígenos de Histocompatibilidade Classe II/genética , Humanos , Imunidade Humoral/genética , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Mutação/genética , Polimorfismo Genético , Infecções por Retroviridae/transmissão
2.
Elife ; 42015 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-25974216

RESUMO

IL-6 plays an important role in determining the fate of effector CD4 cells and the cytokines that these cells produce. Here we identify a novel molecular mechanism by which IL-6 regulates CD4 cell effector function. We show that IL-6-dependent signal facilitates the formation of mitochondrial respiratory chain supercomplexes to sustain high mitochondrial membrane potential late during activation of CD4 cells. Mitochondrial hyperpolarization caused by IL-6 is uncoupled from the production of ATP by oxidative phosphorylation. However, it is a mechanism to raise the levels of mitochondrial Ca(2+) late during activation of CD4 cells. Increased levels of mitochondrial Ca(2+) in the presence of IL-6 are used to prolong Il4 and Il21 expression in effector CD4 cells. Thus, the effect of IL-6 on mitochondrial membrane potential and mitochondrial Ca(2+) is an alternative pathway by which IL-6 regulates effector function of CD4 cells and it could contribute to the pathogenesis of inflammatory diseases.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Cálcio/metabolismo , Interleucina-6/metabolismo , Potenciais da Membrana/fisiologia , Mitocôndrias/metabolismo , Animais , Linfócitos T CD4-Positivos/citologia , Sinalização do Cálcio/fisiologia , Interleucina-6/deficiência , Interleucina-6/genética , Interleucinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/fisiologia , Membranas Mitocondriais/fisiologia , Transdução de Sinais/fisiologia , Proteínas de Transporte Vesicular
3.
Biol Sex Differ ; 6: 6, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25866616

RESUMO

Sexually dimorphic traits arise through the combined effects of sex hormones and sex chromosomes on sex-biased gene expression, and experimental mouse models have been instrumental in determining their relative contribution in modulating sex differences. A role for the Y chromosome (ChrY) in mediating sex differences outside of development and reproduction has historically been overlooked due to its unusual genetic composition and the predominant testes-specific expression of ChrY-encoded genes. However, ample evidence now exists supporting ChrY as a mediator of other physiological traits in males, and genetic variation in ChrY has been linked to several diseases, including heart disease, cancer, and autoimmune diseases in experimental animal models, as well as humans. The genetic and molecular mechanisms by which ChrY modulates phenotypic variation in males remain unknown but may be a function of copy number variation between homologous X-Y multicopy genes driving differential gene expression. Here, we review the literature identifying an association between ChrY polymorphism and phenotypic variation and present the current evidence depicting the mammalian ChrY as a member of the regulatory genome in males and as a factor influencing paternal parent-of-origin effects in female offspring.

4.
FASEB J ; 28(12): 5042-54, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25212221

RESUMO

The estrogens are female sex hormones that are involved in a variety of physiological processes, including reproductive development and function, wound healing, and bone growth. They are mainly known for their roles in reproductive tissues--specifically, 17ß-estradiol (E2), the primary estrogen, which is secreted by the ovaries and induces cellular proliferation and growth of the uterus and mammary glands. In addition to the role of estrogens in promoting tissue growth and development during normal physiological states, they have a well-established role in determining susceptibility to disease, particularly cancer, in reproductive tissues. The responsiveness of various tissues to estrogen is genetically controlled, with marked quantitative variation observed across multiple species, including humans. This variation presents both researchers and clinicians with a veritable physiological puzzle, the pieces of which--many of them unknown--are complex and difficult to fit together. Although genetics is known to play a major role in determining sensitivity to estrogens, there are other factors, including parent of origin and the maternal environment, that are intimately linked to heritable phenotypes but do not represent genotype, per se. The objectives of this review article were to summarize the current knowledge of the role of genotype, and uterine and neonatal environments, in phenotypic variation in the response to estrogens; to discuss recent findings and the potential mechanisms involved; and to highlight exciting research opportunities for the future.


Assuntos
Estrogênios/fisiologia , Animais , Feminino , Humanos , Glândulas Mamárias Humanas/fisiologia , Útero/fisiologia , Vagina/fisiologia
5.
Endocrinology ; 155(8): 3025-35, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24708240

RESUMO

The uterotropic response of the uterus to 17ß-estradiol (E2) is genetically controlled, with marked variation observed depending on the mouse strain studied. Previous genetic studies from our laboratory using inbred mice that are high (C57BL6/J; B6) or low (C3H/HeJ; C3H) responders to E2 led to the identification of quantitative trait loci (QTL) associated with phenotypic variation in uterine growth and leukocyte infiltration. Like the uterus, phenotypic variation in the responsiveness of the mammary gland to E2 during both normal and pathologic conditions has been reported. In the current experiment, we utilized an E2-specific model of mammary ductal growth combined with a microarray approach to determine the degree to which genotype influences the responsiveness of the mammary gland to E2, including the associated transcriptional programs, in B6 and C3H mice. Our results reveal that E2-induced mammary ductal growth and ductal morphology are genetically controlled. In addition, we observed a paradoxical effect of mammary ductal growth in response to E2 compared with what has been reported for the uterus; B6 is a high responder for the uterus and was a low responder for mammary ductal growth, whereas the reverse was observed for C3H. In contrast, B6 was a high responder for mammary ductal side branching. The B6 phenotype was associated with increased mammary epithelial cell proliferation and apoptosis, and a distinct E2-induced transcriptional program. These findings lay the groundwork for future experiments designed to investigate the genes and mechanisms underlying phenotypic variation in tissue-specific sensitivity to systemic and environmental estrogens during various physiological and disease states.


Assuntos
Estradiol/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Glândulas Mamárias Animais/crescimento & desenvolvimento , Maturidade Sexual/genética , Animais , Apoptose , Proliferação de Células , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Células Epiteliais/fisiologia , Feminino , Genótipo , Camundongos , Camundongos Endogâmicos C3H , Útero/fisiologia
6.
Genome Res ; 23(9): 1474-85, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23800453

RESUMO

Understanding the DNA elements that constitute and control the regulatory genome is critical for the appropriate therapeutic management of complex diseases. Here, using chromosome Y (ChrY) consomic mouse strains on the C57BL/6J (B6) background, we show that susceptibility to two diverse animal models of autoimmune disease, experimental allergic encephalomyelitis (EAE) and experimental myocarditis, correlates with the natural variation in copy number of Sly and Rbmy multicopy ChrY genes. On the B6 background, ChrY possesses gene regulatory properties that impact genome-wide gene expression in pathogenic CD4(+) T cells. Using a ChrY consomic strain on the SJL background, we discovered a preference for ChrY-mediated gene regulation in macrophages, the immune cell subset underlying the EAE sexual dimorphism in SJL mice, rather than CD4(+) T cells. Importantly, in both genetic backgrounds, an inverse correlation exists between the number of Sly and Rbmy ChrY gene copies and the number of significantly up-regulated genes in immune cells, thereby supporting a link between copy number variation of Sly and Rbmy with the ChrY genetic element exerting regulatory properties. Additionally, we show that ChrY polymorphism can determine the sexual dimorphism in EAE and myocarditis. In humans, an analysis of the CD4(+) T cell transcriptome from male multiple sclerosis patients versus healthy controls provides further evidence for an evolutionarily conserved mechanism of gene regulation by ChrY. Thus, as in Drosophila, these data establish the mammalian ChrY as a member of the regulatory genome due to its ability to epigenetically regulate genome-wide gene expression in immune cells.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Encefalomielite Autoimune Experimental/genética , Predisposição Genética para Doença , Macrófagos/metabolismo , Miocardite/genética , Transcriptoma , Cromossomo Y/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transporte Vesicular , Animais , Variações do Número de Cópias de DNA , Feminino , Dosagem de Genes , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Caracteres Sexuais
7.
FASEB J ; 27(5): 1874-86, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23371066

RESUMO

The uterotropic response of the uterus to 17ß-estradiol (E2) is genetically controlled, with marked variation observed depending on the mouse strain studied. Previous genetic studies from our laboratory using inbred mice that are high [C57BL/6J (B6)] or low [C3H/HeJ (C3H)] responders to E2 led to the identification of quantitative trait (QT) loci associated with phenotypic variation in uterine growth and leukocyte infiltration. The mechanisms underlying differential responsiveness to E2, and the genes involved, are unknown. Therefore, we used a microarray approach to show association of distinct E2-regulated transcriptional signatures with genetically controlled high and low responses to E2 and their segregation in (C57BL/6J×C3H/HeJ) F1 hybrids. Among the 6664 E2-regulated transcripts, analysis of cellular functions of those that were strain specific indicated C3H-selective enrichment of apoptosis, consistent with a 7-fold increase in the apoptosis indicator CASP3, and a 2.4-fold decrease in the apoptosis inhibitor Naip1 (Birc1a) in C3H vs. B6 following treatment with E2. In addition, several differentially expressed transcripts reside within our previously identified QT loci, including the ERα-tethering factor Runx1, demonstrated to enhance E2-mediated transcript regulation. The level of RUNX1 in uterine epithelial cells was shown to be 3.5-fold greater in B6 compared to C3H. Our novel insights into the mechanisms underlying the genetic control of tissue sensitivity to estrogen have great potential to advance understanding of individualized effects in physiological and disease states.


Assuntos
Caspase 3/genética , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Estradiol/farmacologia , Receptor alfa de Estrogênio/genética , Proteína Inibidora de Apoptose Neuronal/genética , Transcrição Gênica/genética , Útero/fisiologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Proliferação de Células/efeitos dos fármacos , Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Células Epiteliais/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Peroxidase/genética , Análise Serial de Proteínas , Locos de Características Quantitativas/fisiologia , Transcrição Gênica/efeitos dos fármacos , Transcriptoma , Útero/efeitos dos fármacos , Útero/crescimento & desenvolvimento
8.
Cell Immunol ; 278(1-2): 27-34, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23121973

RESUMO

The histamine H(1) receptor (Hrh1/H(1)R) was identified as an autoimmune disease gene in experimental allergic encephalomyelitis (EAE), the principal autoimmune model of multiple sclerosis (MS). Previously, we showed that selective re-expression of H(1)R by endothelial cells or T cells in H(1)RKO mice significantly reduced or complemented EAE severity and cytokine responses, respectively. H(1)R regulates innate immune cells, which in turn influences peripheral and central nervous system CD4(+) T cell effector responses. Therefore, we selectively re-expressed H(1)R in CD11b(+) cells of H(1)RKO mice to test the hypothesis that H(1)R signaling in these cells contributes to EAE susceptibility. We demonstrate that transgenic re-expression of H(1)R by H(1)RKO-CD11b(+) cells neither complements EAE susceptibility nor T cell cytokine responses highlighting the cell-specific effects of Hrh1 in the pathogenesis of EAE and MS, and the need for cell-specific targeting in optimizing therapeutic interventions based on such genes.


Assuntos
Antígenos CD1/imunologia , Encefalomielite Autoimune Experimental/imunologia , Receptores Histamínicos H1/imunologia , Animais , Antígenos CD1/genética , Citocinas/biossíntese , Citocinas/imunologia , Células Dendríticas/imunologia , Células Dendríticas/patologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/patologia , Expressão Gênica , Predisposição Genética para Doença , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Esclerose Múltipla/genética , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Especificidade de Órgãos , Receptores Histamínicos H1/deficiência , Receptores Histamínicos H1/genética , Índice de Gravidade de Doença , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Linfócitos T/patologia
9.
Science ; 334(6053): 245-9, 2011 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-21998394

RESUMO

To establish chronic infections, viruses must develop strategies to evade the host's immune responses. Many retroviruses, including mouse mammary tumor virus (MMTV), are transmitted most efficiently through mucosal surfaces rich in microbiota. We found that MMTV, when ingested by newborn mice, stimulates a state of unresponsiveness toward viral antigens. This process required the intestinal microbiota, as antibiotic-treated mice or germ-free mice did not transmit infectious virus to their offspring. MMTV-bound bacterial lipopolysaccharide triggered Toll-like receptor 4 and subsequent interleukin-6 (IL-6)-dependent induction of the inhibitory cytokine IL-10. Thus, MMTV has evolved to rely on the interaction with the microbiota to induce an immune evasion pathway. Together, these findings reveal the fundamental importance of commensal microbiota in viral infections.


Assuntos
Fenômenos Fisiológicos Bacterianos , Evasão da Resposta Imune , Mucosa Intestinal/virologia , Intestinos/microbiologia , Vírus do Tumor Mamário do Camundongo/imunologia , Vírus do Tumor Mamário do Camundongo/patogenicidade , Metagenoma , Infecções por Retroviridae/transmissão , Animais , Animais Recém-Nascidos , Antibacterianos/farmacologia , Anticorpos Antivirais/biossíntese , Antígenos Virais/imunologia , Feminino , Vida Livre de Germes , Interleucina-10/genética , Interleucina-10/metabolismo , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Gravidez , Complicações Infecciosas na Gravidez/virologia , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/virologia , Organismos Livres de Patógenos Específicos , Receptor 4 Toll-Like/imunologia , Receptor 4 Toll-Like/metabolismo , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/transmissão , Infecções Tumorais por Vírus/virologia , Replicação Viral
10.
Immunity ; 35(1): 135-45, 2011 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-21723157

RESUMO

Innate immune sensors are required for induction of pathogen-specific immune responses. Retroviruses are notorious for their ability to evade immune defenses and establish long-term persistence in susceptible hosts. However, some infected animals are able to develop efficient virus-specific immune responses, and thus can be employed for identification of critical innate virus-sensing mechanisms. With mice from two inbred strains that control retroviruses via adaptive immune mechanisms, we found that of all steps in viral replication, the ability to enter the host cell was sufficient to induce antivirus humoral immune responses. Virus sensing occurred in endosomes via a MyD88-Toll-like receptor 7-dependent mechanism and stimulated virus-neutralizing immunity independently of type I interferons. Thus, efficient adaptive immunity to retroviruses is induced in vivo by innate sensing of the early stages of retroviral infection.


Assuntos
Glicoproteínas de Membrana/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Infecções por Retroviridae/imunologia , Retroviridae/fisiologia , Receptor 7 Toll-Like/metabolismo , Internalização do Vírus , Imunidade Adaptativa , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Células Cultivadas , Suscetibilidade a Doenças , Endossomos/metabolismo , Interações Hospedeiro-Patógeno , Imunidade Inata , Interferon Tipo I/metabolismo , Camundongos , Camundongos Endogâmicos , Infecções por Retroviridae/virologia , Transdução de Sinais
11.
J Virol ; 85(7): 3415-23, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21248041

RESUMO

Antiviral adaptive immune defenses consist of humoral and cell-mediated responses, which together eliminate extracellular and intracellular virus. As most retrovirus-infected individuals do not raise efficient protective antivirus immune responses, the relative importance of humoral and cell-mediated responses in restraining retroviral infection is not well understood. We utilized retrovirus-resistant I/LnJ mice, which control infection with mouse mammary tumor virus (MMTV) and murine leukemia virus (MuLV) via an adaptive immune mechanism, to assess the contribution of cellular responses and virus-neutralizing antibodies (Abs) to the control of retroviral infection. We found that in retrovirus-infected CD8-deficient I/LnJ mice, viral titers exceed the neutralizing capability of antiviral Abs, resulting in augmented virus spread and disease induction. Thus, even in the presence of robust neutralizing Ab responses, CD8-mediated responses are essential for full protection against retroviral infection.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vírus da Leucemia Murina/imunologia , Vírus do Tumor Mamário do Camundongo/imunologia , Infecções por Retroviridae/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Camundongos
12.
Cell Immunol ; 260(2): 119-27, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19914609

RESUMO

The postnatal maternal environment is known to increase susceptibility to a number of autoimmune diseases. Here we asked whether the postnatal maternal environment could influence autoimmune disease development to day 3 thymectomy (d3tx)-induced autoimmune ovarian disease (AOD) and experimental allergic encephalomyelitis (EAE) in cross-fostered A/J and B6 mice. A/J pups foster-nursed by B6 mothers exhibit an increase in autoimmune disease development while cross-fostering B6 pups on A/J mothers did not alter their susceptibility. The increase in AOD incidence seen in foster-nursed d3tx A/J mice correlated with a decrease in the total number of CD4(+) T cells in the lymph nodes of these animals. Analysis of the cellular composition in the milk revealed that B6 mice shed significantly more maternally derived lymphocytes into their milk compared to A/J mothers. These data suggest that there are maternally derived postnatal factors that influence the development of autoimmune disease in A/J mice.


Assuntos
Animais Recém-Nascidos/imunologia , Animais Lactentes/imunologia , Doenças Autoimunes/imunologia , Suscetibilidade a Doenças/imunologia , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Quimiocina CXCL1/metabolismo , Citocinas/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Feminino , Imunidade Materno-Adquirida/imunologia , Interleucina-13/metabolismo , Interleucina-9/metabolismo , Lactação/imunologia , Lactação/metabolismo , Linfonodos/citologia , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Doenças Ovarianas/imunologia , Timectomia
13.
J Virol ; 82(3): 1438-47, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18057254

RESUMO

Mice of the I/LnJ inbred strain are unique in their ability to mount a robust and sustained humoral immune response capable of neutralizing infection with a betaretrovirus, mouse mammary tumor virus (MMTV). Virus-neutralizing antibodies (Abs) coat MMTV virions secreted by infected cells, preventing virus spread and hence the formation of mammary tumors. To investigate whether I/LnJ mice resist infection with other retroviruses besides MMTV, the animals were infected with murine leukemia virus (MuLV), a gammaretrovirus. MuLV-infected I/LnJ mice produced virus-neutralizing Abs that block virus transmission and virally induced disease. Generation of virus-neutralizing Abs required gamma interferon but was independent of interleukin-12. This unique mechanism of retrovirus resistance is governed by a single recessive gene, virus infectivity controller 1 (vic1), mapped to chromosome 17. In addition to controlling the antivirus humoral immune response, vic1 is also required for an antiviral cytotoxic response. Both types of responses were maintained in mice of the susceptible genetic background but congenic for the I/LnJ vic1 locus. Although the vic1-mediated resistance to MuLV resembles the mechanism of retroviral recovery controlled by the resistance to Friend virus 3 (rfv3) gene, the rfv3 gene has been mapped to chromosome 15 and confers resistance to MuLV but not to MMTV. Thus, we have identified a unique virus resistance mechanism that controls immunity against two distinct retroviruses.


Assuntos
Imunidade Inata/genética , Vírus da Leucemia Murina/imunologia , Vírus do Tumor Mamário do Camundongo/imunologia , Camundongos Endogâmicos/virologia , Infecções por Retroviridae/genética , Infecções por Retroviridae/imunologia , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/imunologia , Animais , Anticorpos Antivirais/sangue , Mapeamento Cromossômico , Feminino , Interferons/imunologia , Interleucina-12/imunologia , Masculino , Camundongos , Testes de Neutralização
14.
J Virol ; 80(5): 2206-15, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16474128

RESUMO

Mouse mammary tumor virus (MMTV), a well-characterized retrovirus that causes mammary tumors in susceptible mice, is commonly used to investigate virus-host interactions. We have shown that YBR/Ei mice demonstrate a novel, dominant mechanism of resistance to MMTV infection and MMTV-induced mammary tumors. MMTV can both establish infection in YBR/Ei mice and be transmitted by YBR/Ei mice as an infectious virus. However, virus production is severely attenuated, resulting in gradual clearance of infection in successive generations. Our transfer experiments showed that T cells generated in MMTV-infected resistant mice were required to restrict MMTV replication in susceptible mice. These results emphasize the importance of inducing T-cell responses for effective protection against retroviral infections.


Assuntos
Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/virologia , Vírus do Tumor Mamário do Camundongo/fisiologia , Infecções por Retroviridae/imunologia , Linfócitos T/imunologia , Infecções Tumorais por Vírus/imunologia , Animais , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/imunologia , DNA Viral/análise , Modelos Animais de Doenças , Suscetibilidade a Doenças , Retrovirus Endógenos/fisiologia , Feminino , Imunidade Inata , Tecido Linfoide/virologia , Glândulas Mamárias Animais/virologia , Camundongos , Provírus/genética , Infecções por Retroviridae/genética , Infecções Tumorais por Vírus/genética , Integração Viral/genética
15.
J Immunol ; 175(11): 7543-9, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16301663

RESUMO

Previously, we showed that IFN-gamma elicited by mouse mammary tumor virus (MMTV) infection in I/LnJ mice stimulated production of virus-neutralizing Abs, mostly of the IgG2a isotype. These Abs coated virions secreted by infected I/LnJ cells, and thus completely prevented virus transmission to offspring. However, the mechanism of virus neutralization by isotype-specific Abs remained unknown. Ab coating is capable of blocking virus infection by interfering with receptor-virus binding, by virus opsonization, by complement activation, and via FcgammaR-mediated effector mechanisms. The aim of the studies described in this work was to uncover the cellular basis of anti-virus Ab production, to evaluate the importance of the IgG2a subclass of IgGs in virus neutralization, and to investigate which of the blocking mechanisms plays a role in virus neutralization. We showed that I/LnJ-derived bone marrow cells, specifically IFN-gamma-producing CD4+ T cells, were key cells conferring resistance to MMTV infection in susceptible mice upon transfer. We also established that a unique bias in the subclass selection toward the IgG2a isotype in infected I/LnJ mice was not due to their potent neutralizing ability, as anti-virus Abs of other isotypes were also able to neutralize the virus, but were a product of virally induced IFN-gamma. Finally, we demonstrated that F(ab')2 of anti-MMTV IgGs neutralized the virus as efficiently as total IgGs, suggesting that Ab-mediated interference with viral entry is the sole factor inhibiting virus replication in I/LnJ mice. We propose and discuss possible mechanisms by which infected I/LnJ mice eradicate retrovirus.


Assuntos
Anticorpos Antivirais/imunologia , Linfócitos T CD4-Positivos/virologia , Imunidade Inata/imunologia , Vírus do Tumor Mamário do Camundongo/imunologia , Vírion/imunologia , Transferência Adotiva , Animais , Células da Medula Óssea/imunologia , Linfócitos T CD4-Positivos/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Fragmentos Fab das Imunoglobulinas/imunologia , Imunoglobulina G/imunologia , Interferon gama/imunologia , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Infecções Tumorais por Vírus/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA