Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
J Immunol ; 205(4): 923-935, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32690655

RESUMO

HLA molecules of the MHC class II (MHCII) bind and present pathogen-derived peptides for CD4 T cell activation. Peptide loading of MHCII in the endosomes of cells is controlled by the interplay of the nonclassical MHCII molecules, HLA-DM (DM) and HLA-DO (DO). DM catalyzes peptide loading, whereas DO, an MHCII substrate mimic, prevents DM from interacting with MHCII, resulting in an altered MHCII-peptide repertoire and increased MHCII-CLIP. Although the two genes encoding DO (DOA and DOB) are considered nonpolymorphic, there are rare natural variants. Our previous work identified DOB variants that altered DO function. In this study, we show that natural variation in the DOA gene also impacts DO function. Using the 1000 Genomes Project database, we show that ∼98% of individuals express the canonical DOA*0101 allele, and the remaining individuals mostly express DOA*0102, which we found was a gain-of-function allele. Analysis of 25 natural occurring DOα variants, which included the common alleles, identified three null variants and one variant with reduced and nine with increased ability to modulate DM activity. Unexpectedly, several of the variants produced reduced DO protein levels yet efficiently inhibited DM activity. Finally, analysis of associated single-nucleotide polymorphisms genetically linked the DOA*0102 common allele, a gain-of-function variant, with human hepatitis B viral persistence. In contrast, we found that the DOα F114L null allele was linked with viral clearance. Collectively, these studies show that natural variation occurring in the human DOA gene impacts DO function and can be linked to specific outcomes of viral infections.


Assuntos
Antígenos HLA-D/genética , Hepatite B/genética , Antígenos de Histocompatibilidade Classe II/genética , Polimorfismo de Nucleotídeo Único/genética , Alelos , Apresentação de Antígeno/genética , Linhagem Celular Tumoral , Células HeLa , Hepatite B/virologia , Humanos , Peptídeos/genética
2.
Cell ; 181(6): 1276-1290.e13, 2020 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-32402238

RESUMO

At the species level, immunity depends on the selection and transmission of protective components of the immune system. A microbe-induced population of RORγ-expressing regulatory T cells (Tregs) is essential in controlling gut inflammation. We uncovered a non-genetic, non-epigenetic, non-microbial mode of transmission of their homeostatic setpoint. RORγ+ Treg proportions varied between inbred mouse strains, a trait transmitted by the mother during a tight age window after birth but stable for life, resistant to many microbial or cellular perturbations, then further transferred by females for multiple generations. RORγ+ Treg proportions negatively correlated with IgA production and coating of gut commensals, traits also subject to maternal transmission, in an immunoglobulin- and RORγ+ Treg-dependent manner. We propose a model based on a double-negative feedback loop, vertically transmitted via the entero-mammary axis. This immunologic mode of multi-generational transmission may provide adaptability and modulate the genetic tuning of gut immune responses and inflammatory disease susceptibility.


Assuntos
Sistema Digestório/imunologia , Linfócitos T Reguladores/imunologia , Animais , Suscetibilidade a Doenças/imunologia , Feminino , Microbioma Gastrointestinal/imunologia , Homeostase/imunologia , Imunoglobulina A/imunologia , Inflamação/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Endogâmicos NOD , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia
3.
Annu Rev Virol ; 6(1): 525-546, 2019 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-31567067

RESUMO

Genetic alleles that contribute to enhanced susceptibility or resistance to viral infections and virally induced diseases have often been first identified in mice before humans due to the significant advantages of the murine system for genetic studies. Herein we review multiple discoveries that have revealed significant insights into virus-host interactions, all made using genetic mapping tools in mice. Factors that have been identified include innate and adaptive immunity genes that contribute to host defense against pathogenic viruses such as herpes viruses, flaviviruses, retroviruses, and coronaviruses. Understanding the genetic mechanisms that affect infectious disease outcomes will aid the development of personalized treatment and preventive strategies for pathogenic infections.


Assuntos
Camundongos/genética , Viroses/genética , Fenômenos Fisiológicos Virais , Imunidade Adaptativa , Animais , Predisposição Genética para Doença , Humanos , Imunidade Inata , Camundongos/imunologia , Camundongos/virologia , Viroses/imunologia , Viroses/virologia , Vírus/genética
4.
J Virol ; 92(16)2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-29875252

RESUMO

An essential step in the development of effective antiviral humoral responses is cytokine-triggered class switch recombination resulting in the production of antibodies of a specific isotype. Most viral and parasitic infections in mice induce predominantly IgG2a-specific antibody responses that are stimulated by interferon gamma (IFN-γ). However, in some mice deficient in IFN-γ, class switching to IgG2a antibodies is relatively unaffected, indicating that another signal(s) can be generated upon viral or parasitic infections that trigger this response. Here, we found that a single recessive locus, provisionally called IFN-γ-independent IgG2a (Igii), confers the ability to produce IFN-γ-independent production of IgG2a antibodies upon retroviral infection. The Igii locus was mapped to chromosome 9 and was found to function in the radiation-resistant compartment. Thus, our data implicate nonhematopoietic cells in activation of antiviral antibody responses in the absence of IFN-γ.IMPORTANCE Understanding the signals that stimulate antibody production and class switch recombination to specific antibody isotypes is crucial for the development of novel vaccines and adjuvants. While an interferon gamma-mediated switch to the IgG2a isotype upon viral infection in mice has been well established, this investigation reveals a noncanonical, interferon gamma-independent pathway for antiretroviral antibody production and IgG2a class switch recombination that is controlled by a single recessive locus. Furthermore, this study indicates that the radiation-resistant compartment can direct antiviral antibody responses, suggesting that detection of infection by nonhematopoietic cells is involved is stimulating adaptive immunity.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Imunoglobulina G/sangue , Vírus/imunologia , Animais , Mapeamento Cromossômico , Interferon gama/deficiência , Camundongos , Camundongos Knockout
5.
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
6.
ILAR J ; 57(1): 12-23, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27034391

RESUMO

Mouse mammary tumor virus (MMTV), which was discovered as a milk-transmitted, infectious, cancer-inducing agent in the 1930s, has been used as an animal model for the study of retroviral infection and transmission, antiviral immune responses, and breast cancer and lymphoma biology. The main target cells for MMTV infection in vivo are cells of the immune system and mammary epithelial cells. Although the host mounts an immune response to the virus, MMTV has evolved multiple means of evading this response. MMTV causes mammary tumors when the provirus integrates into the mammary epithelial and lymphoid cell genome during viral replication and thereby activates cellular oncogene expression. Thus, tumor induction is a by-product of the infection cycle. A number of important oncogenes have been discovered by carrying out MMTV integration site analysis, some of which may play a role in human breast cancer.


Assuntos
Vírus do Tumor Mamário do Camundongo/fisiologia , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/virologia , Modelos Animais de Doenças , Feminino , Humanos , Camundongos
7.
Cell Host Microbe ; 18(4): 456-62, 2015 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-26468748

RESUMO

The orally transmitted retrovirus mouse mammary tumor virus (MMTV) requires the intestinal microbiota for persistence. Virion-associated lipopolysaccharide (LPS) activates Toll-like receptor 4 (TLR4), stimulating production of the immunosuppressive cytokine IL-10 and MMTV evasion of host immunity. However, the mechanisms by which MMTV associates with LPS remain unknown. We find that the viral envelope contains the mammalian LPS-binding factors CD14, TLR4, and MD-2, which, in conjunction with LPS-binding protein (LBP), bind LPS to the virus and augment transmission. MMTV isolated from infected mice lacking these LBPs cannot engage LPS or stimulate TLR4 and have a transmission defect. Furthermore, MMTV incorporation of a weak agonist LPS from Bacteroides, a prevalent LPS source in the gut, significantly enhances the ability of this LPS to stimulate TLR4, suggesting that MMTV intensifies these immunostimulatory properties. Thus, an orally transmitted retrovirus can capture, modify, and exploit mammalian receptors for bacterial ligands to ensure successful transmission.


Assuntos
Interações Hospedeiro-Patógeno , Receptores de Lipopolissacarídeos/metabolismo , Vírus do Tumor Mamário do Camundongo/fisiologia , Proteínas do Envelope Viral/metabolismo , Animais , Evasão da Resposta Imune , Imunossupressores/metabolismo , Interleucina-10/metabolismo , Lipopolissacarídeos/metabolismo , Camundongos , Ligação Proteica , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo
8.
Nature ; 514(7524): 638-41, 2014 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-25274297

RESUMO

Systemic infection induces conserved physiological responses that include both resistance and 'tolerance of infection' mechanisms. Temporary anorexia associated with an infection is often beneficial, reallocating energy from food foraging towards resistance to infection or depriving pathogens of nutrients. However, it imposes a stress on intestinal commensals, as they also experience reduced substrate availability; this affects host fitness owing to the loss of caloric intake and colonization resistance (protection from additional infections). We hypothesized that the host might utilize internal resources to support the gut microbiota during the acute phase of the disease. Here we show that systemic exposure to Toll-like receptor (TLR) ligands causes rapid α(1,2)-fucosylation of small intestine epithelial cells (IECs) in mice, which requires the sensing of TLR agonists, as well as the production of interleukin (IL)-23 by dendritic cells, activation of innate lymphoid cells and expression of fucosyltransferase 2 (Fut2) by IL-22-stimulated IECs. Fucosylated proteins are shed into the lumen and fucose is liberated and metabolized by the gut microbiota, as shown by reporter bacteria and community-wide analysis of microbial gene expression. Fucose affects the expression of microbial metabolic pathways and reduces the expression of bacterial virulence genes. It also improves host tolerance of the mild pathogen Citrobacter rodentium. Thus, rapid IEC fucosylation appears to be a protective mechanism that utilizes the host's resources to maintain host-microbial interactions during pathogen-induced stress.


Assuntos
Doença , Epitélio/metabolismo , Epitélio/microbiologia , Fucose/metabolismo , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Simbiose , Animais , Anorexia/complicações , Anorexia/microbiologia , Bactérias/genética , Bactérias/metabolismo , Bactérias/patogenicidade , Citrobacter rodentium/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Ingestão de Alimentos , Ácidos Graxos/química , Ácidos Graxos/metabolismo , Feminino , Fucosiltransferases/metabolismo , Regulação Bacteriana da Expressão Gênica , Glicosilação , Tolerância Imunológica , Imunidade Inata , Interleucinas/biossíntese , Interleucinas/imunologia , Ligantes , Masculino , Redes e Vias Metabólicas/genética , Camundongos , Microbiota/fisiologia , Fatores de Proteção , Receptores Toll-Like/agonistas , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo , Fatores de Virulência/genética , Interleucina 22 , Galactosídeo 2-alfa-L-Fucosiltransferase
9.
Immunol Rev ; 255(1): 222-9, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23947358

RESUMO

With our abilities to culture and sequence the commensal bacteria that dwell on and within a host, we can now study the host in its entirety, as a supraorganism that must be navigated by the pathogen invader. At present, the majority of studies have focused on the interaction between the host's microbiota and bacterial pathogens. This is not unwarranted, given that bacterial pathogens must compete with commensal organisms for the limited territory afforded by the host. However, viral pathogens also enter the host through surfaces coated with microbial life and encounter an immune system shaped by this symbiotic community. Therefore, we believe that the microbiota cannot be ignored when examining the interplay between the host and viral pathogens. Here, we review work that details mechanisms by which the microbiota either promotes or inhibits viral replication and virally induced pathogenesis. The impact of the microbitota on viral infection promises to be a new and exciting avenue of investigation, which will ultimately lead to better treatments and preventions of virally induced disease.


Assuntos
Bactérias , Fenômenos Fisiológicos Bacterianos , Simbiose , Viroses/imunologia , Viroses/virologia , Vírus/imunologia , Animais , Interações Hospedeiro-Patógeno/imunologia , Humanos , Metagenoma , Replicação Viral
10.
J Virol ; 87(2): 1069-82, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23135726

RESUMO

The mouse mammary tumor virus (MMTV) Gag protein directs the assembly in the cytoplasm of immature viral capsids, which subsequently bud from the plasma membranes of infected cells. MMTV Gag localizes to discrete cytoplasmic foci in mouse mammary epithelial cells, consistent with the formation of cytosolic capsids. Unexpectedly, we also observed an accumulation of Gag in the nucleoli of infected cells derived from mammary gland tumors. To detect Gag-interacting proteins that might influence its subcellular localization, a yeast two-hybrid screen was performed. Ribosomal protein L9 (RPL9 or L9), an essential component of the large ribosomal subunit and a putative tumor suppressor, was identified as a Gag binding partner. Overexpression of L9 in cells expressing the MMTV(C3H) provirus resulted in specific, robust accumulation of Gag in nucleoli. Förster resonance energy transfer (FRET) and coimmunoprecipitation analyses demonstrated that Gag and L9 interact within the nucleolus, and the CA domain was the major site of interaction. In addition, the isolated NC domain of Gag localized to the nucleolus, suggesting that it contains a nucleolar localization signal (NoLS). To determine whether L9 plays a role in virus assembly, small interfering RNA (siRNA)-mediated knockdown was performed. Although Gag expression was not reduced with L9 knockdown, virus production was significantly impaired. Thus, our data support the hypothesis that efficient MMTV particle assembly is dependent upon the interaction of Gag and L9 in the nucleoli of infected cells.


Assuntos
Nucléolo Celular/metabolismo , Produtos do Gene gag/metabolismo , Interações Hospedeiro-Patógeno , Vírus do Tumor Mamário do Camundongo/fisiologia , Proteínas Ribossômicas/metabolismo , Montagem de Vírus , Animais , Linhagem Celular , Células Epiteliais/virologia , Transferência Ressonante de Energia de Fluorescência , Imunoprecipitação , Camundongos , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Sinais Direcionadores de Proteínas , Transporte Proteico , Técnicas do Sistema de Duplo-Híbrido
11.
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
12.
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
13.
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
14.
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
15.
J Virol ; 80(7): 3215-24, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16537589

RESUMO

Previously, we identified a group of replication-competent exogenous mouse mammary tumor viruses that failed to induce mammary tumors in susceptible mice. Sequence comparison of tumorigenic and tumor-attenuated virus variants has linked the ability of virus to cause high-frequency mammary tumors to the gag gene. To determine the specific sequences within the gag gene that contribute to tumor induction, we constructed five distinct chimeric viruses that have various amino acid coding sequences of gag derived from a tumor-attenuated virus replaced by those of highly tumorigenic virus and tested these viruses for tumorigenic capacities in virus-susceptible C3H/HeN mice. Comparing the tumorigenic potentials of these viruses has allowed us to map the region responsible for tumorigenesis to a 253-amino-acid region within the CA and NC regions of the Gag protein. Unlike C3H/HeN mice, BALB/cJ mice develop tumors when infected with all viral variants, irrespective of the gag gene sequences. Using genetic crosses between BALB/cJ and C3H/HeN mice, we were able to determine that the mechanism that confers susceptibility to Gag-independent mammary tumors in BALB/cJ mice is inherited as a dominant trait and is controlled by a single gene, called mammary tumor susceptibility (mts), that maps to chromosome 14.


Assuntos
Transformação Celular Neoplásica , Genes gag , Neoplasias Mamárias Experimentais/virologia , Vírus do Tumor Mamário do Camundongo/genética , Sequência de Aminoácidos , Animais , Western Blotting , Mapeamento Cromossômico , Cromossomos , Clonagem Molecular , Sequência Conservada , Cruzamentos Genéticos , Feminino , Regulação Neoplásica da Expressão Gênica , Regulação Viral da Expressão Gênica , Engenharia Genética , Haplótipos , Neoplasias Mamárias Experimentais/etiologia , Vírus do Tumor Mamário do Camundongo/patogenicidade , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Transgênicos , Modelos Genéticos , Dados de Sequência Molecular , Mutagênese
16.
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
17.
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
18.
J Immunol ; 172(9): 5582-7, 2004 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15100301

RESUMO

Exogenous mouse mammary tumor virus (MMTV) is transmitted via the milk from infected mothers to newborn pups. Efficient MMTV transmission is dependent on proliferation of T cells with particular TCR beta-chains, which occurs upon recognition of virally encoded superantigen (SAg) bound to MHC class II molecules. It is assumed that infection of these dividing cells favors MMTV amplification. SAg is important for MMTV infection, as mice that lack SAg-cognate T cells due to expression of endogenous Mtv loci or mice that express inappropriate MHC haplotypes unable to present viral SAg efficiently were shown to be resistant to MMTV infection. However, this resistance was not absolute, as these mice developed late onset MMTV-induced mammary tumors. In this study, we show that the success of initial MMTV infection in neonates is independent of SAg function but depends on the developmentally regulated proliferation of target cells. However, SAg was absolutely required for virus spread following completion of this proliferative stage.


Assuntos
Vírus do Tumor Mamário do Camundongo/imunologia , Infecções por Retroviridae/imunologia , Superantígenos/fisiologia , Infecções Tumorais por Vírus/imunologia , Animais , Animais Recém-Nascidos , Apresentação de Antígeno/genética , Apresentação de Antígeno/imunologia , Predisposição Genética para Doença , Imunidade Inata/genética , Vírus do Tumor Mamário do Camundongo/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Fenótipo , Período Pós-Parto/genética , Período Pós-Parto/imunologia , Infecções por Retroviridae/genética , Infecções por Retroviridae/virologia , Superantígenos/imunologia , Superantígenos/metabolismo , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/virologia , Replicação Viral/imunologia
19.
Cancer Biol Ther ; 2(6): 687-93, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14688478

RESUMO

We had previously demonstrated that in mice acute toxoplasmosis leads to systemic inhibition of angiogenesis and, consequently, strong suppression of neoplastic growth. Here we investigated the role of Th1 cytokines, in particular interferon gamma (IFN-gamma), in this phenomenon. Besides toxoplasma, neoplastic growth was readily blocked during acute infection with other Th1 response-inducing pathogens such as Listeria monocytogenes and lymphocytic choriomeningitis virus (LCMV). In contrast, chronic infection with LCMV (when Th1 responses were strongly suppressed) and acute infection with Schistosoma mansoni (when Th2 responses predominated) afforded no anti-tumor protection. To corroborate the involvement of Th1 cytokines in infection-mediated suppression of neoplastic growth, we utilized mice deficient in interleukin-10 (IL10), a suppressor of Th1 responses. When challenged with B16 cells concomitantly with toxoplasma infection, both IL10-null and wild type mice exhibited resistance to neoplastic growth. However, tumors borne by IL10-null animals were even smaller than those borne by their wild type counterparts. This enhanced resistance correlated with dramatically elevated levels of circulating IFN-gamma, a principal Th1 cytokine. Furthermore, while interleukin-12 and tumor necrosis factor a were dispensable for tumor suppression, in animals deficient in IFN-gamma production or signaling, tumor growth and neovascularization were markedly enhanced. Interestingly, the enhancement was also apparent in uninfected animals suggesting that IFN-gamma and its anti-angiogenic effects underlie both infection-dependent and -independent tumor surveillance.


Assuntos
Citocinas/imunologia , Infecções/imunologia , Interferon gama/imunologia , Melanoma Experimental/imunologia , Células Th1/imunologia , Doença Aguda , Animais , Linhagem Celular Tumoral , Transplante de Células , Células Clonais , Colágeno/metabolismo , Meios de Cultivo Condicionados/análise , Combinação de Medicamentos , Infecções/sangue , Interferon gama/análise , Interleucina-10/deficiência , Laminina/metabolismo , Listeria monocytogenes/patogenicidade , Vírus da Coriomeningite Linfocítica/patogenicidade , Melanoma Experimental/sangue , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Neovascularização Patológica , Proteoglicanas/metabolismo , Fatores de Tempo , Toxoplasma/patogenicidade , Toxoplasmose , Fator A de Crescimento do Endotélio Vascular/análise , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
20.
Nat Immunol ; 4(6): 573-8, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12730691

RESUMO

Retroviruses evolve rapidly to avoid the immune response of the infected host. We show here that the wild-type mouse mammary tumor virus MMTV(C3H) persisted indefinitely in C3H/HeN mice. However, it was rapidly lost in mice of the closely related C3H/HeJ strain and was replaced by a virus recombinant with an endogenous Mtv provirus. Maintenance of the wild-type virus was dependent on Toll-like receptor-4 (TLR4) signaling, which triggered production of the immunosuppressive cytokine interleukin-10. In the presence of mutant TLR4 in C3H/HeJ mice, wild-type virus was eliminated by the cytotoxic immune response, promoting selection of the immune escape recombinant MMTV variants. Thus, subversion of the innate immune system is yet another survival strategy used by retroviruses.


Assuntos
Vírus do Tumor Mamário do Camundongo/imunologia , Infecções por Retroviridae/imunologia , Infecções Tumorais por Vírus/imunologia , Animais , Cruzamentos Genéticos , Testes Imunológicos de Citotoxicidade , Interleucina-10/imunologia , Interleucina-10/metabolismo , Vírus do Tumor Mamário do Camundongo/genética , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Receptores de Superfície Celular/imunologia , Proteínas Recombinantes , Infecções por Retroviridae/genética , Seleção Genética , Transdução de Sinais/imunologia , Receptor 4 Toll-Like , Receptores Toll-Like , Infecções Tumorais por Vírus/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...