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










Base de dados
Intervalo de ano de publicação
1.
Viruses ; 14(4)2022 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-35458563

RESUMO

Tissue and subcellular localization and its changes upon cell activation of virus-restricting APOBEC3 at protein levels are important to understanding physiological functions of this cytidine deaminase, but have not been thoroughly analyzed in vivo. To precisely follow the possible activation-induced changes in expression levels of APOBEC3 protein in different mouse tissues and cell populations, genome editing was utilized to establish knock-in mice that express APOBEC3 protein with an in-frame FLAG tag. Flow cytometry and immunohistochemical analyses were performed prior to and after an immunological stimulation. Cultured B cells expressed higher levels of APOBEC3 protein than T cells. All differentiation and activation stages of freshly prepared B cells expressed significant levels of APOBEC3 protein, but germinal center cells possessed the highest levels of APOBEC3 protein localized in their cytoplasm. Upon immunological stimulation with sheep red blood cells in vivo, germinal center cells with high levels of APOBEC3 protein expression increased in their number, but FLAG-specific fluorescence intensity in each cell did not change. T cells, even those in germinal centers, did not express significant levels of APOBEC3 protein. Thus, mouse APOBEC3 protein is expressed at distinctively high levels in germinal center B cells. Antigenic stimulation did not affect expression levels of cellular APOBEC3 protein despite increased numbers of germinal center cells.


Assuntos
Linfócitos B , Citidina Desaminase , Centro Germinativo , Infecções por HIV , Animais , Antirretrovirais , Citidina Desaminase/genética , Camundongos , Proteínas , Ovinos
2.
Int Immunopharmacol ; 107: 108672, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35279511

RESUMO

The anti-prion activity of cellulose ether (CE) has been reported in rodents, but the mechanism of action is not well understood. As defects in early T-cell development have been reported in Tga20 mice which show only a slight effect of CE administration, we investigated the involvement of immune functions in the CE action. We confirmed an insertion of the prion protein transgene into the pre T-cell antigen receptor α gene of Tga20 mice, and its impaired expression in the thymus and other tissues. The influence of immune suppression on the CE effect was then examined in high CE-responder mice treated with immunosuppressive agents or neonatal thymectomy. As neonatal thymectomy significantly reduced the CE effect, we compared the influence of various T-cell defects in mice with similar genetic backgrounds. The CE effect was increased or unchanged in mice with defects in the αß T-cell lineage, whereas it was abolished in T-cell receptor δ deficient mice. Further, when other immune defects were examined, the CE effect was reduced in mice with lysosomal trafficking dysfunction, but was unchanged in mice deficient in B-cell differentiation or toll-like receptor 4 signaling. These findings collectively suggest that the mechanism of CE action may involve γδ T cells and lytic granule function, as well as immune factors like natural killer T cells which are lacking in pre T-cell antigen receptor α deficient mice and neonatally thymectomized mice.


Assuntos
Príons , Animais , Celulose , Éter , Éteres , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T gama-delta
4.
PLoS One ; 10(4): e0122458, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25874769

RESUMO

The aim of this study is to explore a cause-oriented therapy for patients with uterine cervical cancer that expresses erythropoietin (Epo) and its receptor (EpoR). Epo, by binding to EpoR, stimulates the proliferation and differentiation of erythroid progenitor cells into hemoglobin-containing red blood cells. In this study, we report that the HeLa cells in the xenografts expressed ε, γ, and α globins as well as myoglobin (Mb) to produce tetrameric α2ε2 and α2γ2 and monomeric Mb, most of which were significantly suppressed with an EpoR antagonist EMP9. Western blotting revealed that the EMP9 treatment inhibited the AKT-pAKT, MAPKs-pMAPKs, and STAT5-pSTAT5 signaling pathways. Moreover, the treatment induced apoptosis and suppression of the growth and inhibited the survival through disruption of the harmonized hemoprotein syntheses in the tumor cells concomitant with destruction of vascular nets in the xenografts. Furthermore, macrophages and natural killer (NK) cells with intense HIF-1α expression recruited significantly more in the degenerating foci of the xenografts. These findings were associated with the enhanced expressions of nNOS in the tumor cells and iNOS in macrophages and NK cells in the tumor sites. The treated tumor cells exhibited a substantial number of perforations on the cell surface, which indicates that the tumors were damaged by both the nNOS-induced nitric oxide (NO) production in the tumor cells as well as the iNOS-induced NO production in the innate immune cells. Taken together, these data suggest that HeLa cells constitutively acquire ε, γ and Mb synthetic capacity for their survival. Therefore, EMP9 treatment might be a cause-oriented and effective therapy for patients with squamous cell carcinoma of the uterine cervix.


Assuntos
Hemoglobinas/biossíntese , Xenoenxertos/efeitos dos fármacos , Neoplasias Experimentais/metabolismo , Peptídeos/farmacologia , Receptores da Eritropoetina/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Proliferação de Células/efeitos dos fármacos , Eritropoetina/química , Eritropoetina/farmacologia , Expressão Gênica/efeitos dos fármacos , Células HeLa , Hemoglobinas/genética , Xenoenxertos/metabolismo , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Peptídeos/síntese química , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores da Eritropoetina/genética , Receptores da Eritropoetina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transplante Heterólogo
5.
J Virol ; 89(2): 1468-73, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25378499

RESUMO

Toll-like receptor 7 and Myd88 are required for antiretroviral antibody and germinal center responses, but whether somatic hypermutation and class-switch recombination are required for antiretroviral immunity has not been examined. Mice deficient in activation-induced cytidine deaminase (AID) resisted Friend virus infection, produced virus-neutralizing antibodies, and controlled viremia. Passive transfer demonstrated that immune IgM from AID-deficient mice contributes to Friend virus control in the presence of virus-specific CD4+ T cells.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Vírus da Leucemia Murina de Friend/imunologia , Leucemia Experimental/imunologia , Infecções por Retroviridae/imunologia , Hipermutação Somática de Imunoglobulina , Infecções Tumorais por Vírus/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Citidina Desaminase/deficiência , Imunização Passiva , Switching de Imunoglobulina , Imunoglobulina M/imunologia , Leucemia Experimental/virologia , Camundongos , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia
7.
PLoS Pathog ; 10(3): e1003937, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24651250

RESUMO

In chronic viral infections, persistent antigen presentation causes progressive exhaustion of virus-specific CD8+ T cells. It has become clear, however, that virus-specific naïve CD8+ T cells newly generated from the thymus can be primed with persisting antigens. In the setting of low antigen density and resolved inflammation, newly primed CD8+ T cells are preferentially recruited into the functional memory pool. Thus, continual recruitment of naïve CD8+ T cells from the thymus is important for preserving the population of functional memory CD8+ T cells in chronically infected animals. Friend virus (FV) is the pathogenic murine retrovirus that establishes chronic infection in adult mice, which is bolstered by the profound exhaustion of virus-specific CD8+ T cells induced during the early phase of infection. Here we show an additional evasion strategy in which FV disseminates efficiently into the thymus, ultimately leading to clonal deletion of thymocytes that are reactive to FV antigens. Owing to the resultant lack of virus-specific recent thymic emigrants, along with the above exhaustion of antigen-experienced peripheral CD8+ T cells, mice chronically infected with FV fail to establish a functional virus-specific CD8+ T cell pool, and are highly susceptible to challenge with tumor cells expressing FV-encoded antigen. However, FV-specific naïve CD8+ T cells generated in uninfected mice can be primed and differentiate into functional memory CD8+ T cells upon their transfer into chronically infected animals. These findings indicate that virus-induced central tolerance that develops during the chronic phase of infection accelerates the accumulation of dysfunctional memory CD8+ T cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Tolerância Imunológica/imunologia , Memória Imunológica/imunologia , Infecções por Retroviridae/imunologia , Timo/virologia , Envelhecimento , Animais , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular/imunologia , Doença Crônica , Feminino , Citometria de Fluxo , Vírus da Leucemia Murina de Friend/imunologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Timo/imunologia
9.
J Virol ; 87(24): 13760-74, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24109240

RESUMO

To assess the possible contribution of host immune responses to the exertion of Fv2-associated resistance to Friend virus (FV)-induced disease development, we inoculated C57BL/6 (B6) mice that lacked various subsets of lymphocytes with FV containing no lactate dehydrogenase-elevating virus. Fv2(r) B6 mice lacking CD4(+) T cells developed early polycythemia and fatal erythroleukemia, while B6 mice lacking CD8(+) T cells remained resistant. Erythroid progenitor cells infected with spleen focus-forming virus (SFFV) were eliminated, and no polycythemia was observed in B cell-deficient B6 mice, but they later developed myeloid leukemia associated with oligoclonal integration of ecotropic Friend murine leukemia virus. Additional depletion of natural killer and/or CD8(+) T cells from B cell-deficient B6 mice resulted in the expansion of SFFV proviruses and the development of polycythemia, indicating that SFFV-infected erythroid cells are not only restricted in their growth but are actively eliminated in Fv2(r) mice through cellular immune responses.


Assuntos
Vírus da Leucemia Murina de Friend/imunologia , Imunidade Celular , Imunidade Humoral , Leucemia Eritroblástica Aguda/veterinária , Doenças dos Roedores/imunologia , Animais , Linfócitos B/imunologia , Progressão da Doença , Resistência à Doença , Feminino , Vírus da Leucemia Murina de Friend/genética , Humanos , Leucemia Eritroblástica Aguda/imunologia , Leucemia Eritroblástica Aguda/virologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Doenças dos Roedores/virologia , Vírus Formadores de Foco no Baço/genética , Vírus Formadores de Foco no Baço/imunologia , Linfócitos T/imunologia
10.
J Immunol ; 191(1): 91-6, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23720810

RESUMO

We investigated the role of effector CD8 T cells in the pathogenesis of immune glomerular injury. BALB/c mice are not prone to autoimmune disease, but after 12 immunizations with OVA they developed a variety of autoantibodies and glomerulonephritis accompanied by immune complex (IC) deposition. In these mice, IFN-γ-producing effector CD8 T cells were significantly increased concomitantly with glomerulonephritis. In contrast, after 12 immunizations with keyhole limpet hemocyanin, although autoantibodies appeared, IFN-γ-producing effector CD8 T cells did not develop, and glomerular injury was not induced. In ß2-microglobulin-deficient mice lacking CD8 T cells, glomerular injury was not induced after 12 immunizations with OVA, despite massive deposition of IC in the glomeruli. In mice containing a targeted disruption of the exon encoding the membrane-spanning region of the Ig µ-chain (µMT mice), 12 immunizations with OVA induced IFN-γ-producing effector CD8 T cells but not IC deposition or glomerular injury. When CD8 T cells from mice immunized 12 times with OVA were transferred into naive recipients, glomerular injury could be induced, but only when a single injection of OVA was also given simultaneously. Importantly, injection of OVA could be replaced by one injection of the sera from mice that had been fully immunized with OVA. This indicates that deposition of IC is required for effector CD8 T cells to cause immune tissue injury. Thus, in a mouse model of systemic lupus erythematosus, glomerular injury is caused by effector CD8 T cells that recognize Ag presented as IC on the target renal tissue.


Assuntos
Apresentação de Antígeno/imunologia , Complexo Antígeno-Anticorpo/imunologia , Linfócitos T CD8-Positivos/imunologia , Glomerulonefrite/imunologia , Interferon gama/biossíntese , Animais , Apresentação de Antígeno/genética , Complexo Antígeno-Anticorpo/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Feminino , Glomerulonefrite/metabolismo , Glomerulonefrite/patologia , Cadeias mu de Imunoglobulina/genética , Interferon gama/fisiologia , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/patologia , Microglobulina beta-2/deficiência
11.
J Virol ; 85(11): 5423-35, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21411527

RESUMO

Natural killer (NK) cells function as early effector cells in the innate immune defense against viral infections and also participate in the regulation of normal and malignant hematopoiesis. NK cell activities have been associated with early clearance of viremia in experimental simian immunodeficiency virus and clinical human immunodeficiency virus type 1 (HIV-1) infections. We have previously shown that NK cells function as major cytotoxic effector cells in vaccine-induced immune protection against Friend virus (FV)-induced leukemia, and NK cell depletion totally abrogates the above protective immunity. However, how NK cells recognize retrovirus-infected cells remains largely unclear. The present study demonstrates a correlation between the expression of the products of retinoic acid early transcript-1 (RAE-1) genes in target cells and their susceptibility to killing by NK cells isolated from FV-infected animals. This killing was abrogated by antibodies blocking the NKG2D receptor in vitro. Further, the expression of RAE-1 proteins on erythroblast surfaces increased early after FV inoculation, and administration of an RAE-1-blocking antibody resulted in increased spleen infectious centers and exaggerated pathology, indicating that FV-infected erythroid cells are recognized by NK cells mainly through the NKG2D-RAE-1 interactions in vivo. Enhanced retroviral replication due to host gene-targeting resulted in markedly increased RAE-1 expression in the absence of massive erythroid cell proliferation, indicating a direct role of retroviral replication in RAE-1 upregulation.


Assuntos
Células Precursoras Eritroides/imunologia , Células Precursoras Eritroides/virologia , Vírus da Leucemia Murina de Friend/imunologia , Células Matadoras Naturais/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Proteínas Associadas à Matriz Nuclear/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Animais , Células Cultivadas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
12.
PLoS One ; 6(3): e17655, 2011 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-21408016

RESUMO

Regulators of G protein signaling (RGS) are a multi-functional protein family, which functions in part as GTPase-activating proteins (GAPs) of G protein α-subunits to terminate G protein signaling. Previous studies have demonstrated that the Rgs16 transcripts exhibit robust circadian rhythms both in the suprachiasmatic nucleus (SCN), the master circadian light-entrainable oscillator (LEO) of the hypothalamus, and in the liver. To investigate the role of RGS16 in the circadian clock in vivo, we generated two independent transgenic mouse lines using lentiviral vectors expressing short hairpin RNA (shRNA) targeting the Rgs16 mRNA. The knockdown mice demonstrated significantly shorter free-running period of locomotor activity rhythms and reduced total activity as compared to the wild-type siblings. In addition, when feeding was restricted during the daytime, food-entrainable oscillator (FEO)-driven elevated food-anticipatory activity (FAA) observed prior to the scheduled feeding time was significantly attenuated in the knockdown mice. Whereas the restricted feeding phase-advanced the rhythmic expression of the Per2 clock gene in liver and thalamus in the wild-type animals, the above phase shift was not observed in the knockdown mice. This is the first in vivo demonstration that a common regulator of G protein signaling is involved in the two separate, but interactive circadian timing systems, LEO and FEO. The present study also suggests that liver and/or thalamus regulate the food-entrained circadian behavior through G protein-mediated signal transduction pathway(s).


Assuntos
Antecipação Psicológica , Ritmo Circadiano/genética , Comportamento Alimentar/fisiologia , Técnicas de Silenciamento de Genes , Atividade Motora/genética , Proteínas RGS/genética , Animais , Encéfalo/metabolismo , Regulação da Expressão Gênica , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Proteínas RGS/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Tálamo/metabolismo , Fatores de Tempo
13.
J Virol ; 84(12): 6082-95, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20375169

RESUMO

Several host genes control retroviral replication and pathogenesis through the regulation of immune responses to viral antigens. The Rfv3 gene influences the persistence of viremia and production of virus-neutralizing antibodies in mice infected with Friend mouse retrovirus complex (FV). This locus has been mapped within a narrow segment of mouse chromosome 15 harboring the APOBEC3 and BAFF-R loci, both of which show functional polymorphisms among different strains of mice. The exon 5-lacking product of the APOBEC3 allele expressed in FV-resistant C57BL/6 (B6) mice directly restricts viral replication, and mice lacking the B6-derived APOBEC3 exhibit exaggerated pathology and reduced production of neutralizing antibodies. However, the mechanisms by which the polymorphisms at the APOBEC3 locus affect the production of neutralizing antibodies remain unclear. Here we show that the APOBEC3 genotypes do not directly affect the B-cell repertoire, and mice lacking B6-derived APOBEC3 still produce FV-neutralizing antibodies in the presence of primed T helper cells. Instead, higher viral loads at a very early stage of FV infection caused by either a lack of the B6-derived APOBEC3 or a lack of the wild-type BAFF-R resulted in slower production of neutralizing antibodies. Indeed, B cells were hyperactivated soon after infection in the APOBEC3- or BAFF-R-deficient mice. In contrast to mice deficient in the B6-derived APOBEC3, which cleared viremia by 4 weeks after FV infection, mice lacking the functional BAFF-R allele exhibited sustained viremia, indicating that the polymorphisms at the BAFF-R locus may better explain the Rfv3-defining phenotype of persistent viremia.


Assuntos
Anticorpos Neutralizantes/imunologia , Citidina Desaminase/genética , Vírus da Leucemia Murina de Friend/imunologia , Polimorfismo Genético , Infecções por Retroviridae/veterinária , Doenças dos Roedores/genética , Doenças dos Roedores/imunologia , Viremia/genética , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Receptor do Fator Ativador de Células B/genética , Receptor do Fator Ativador de Células B/imunologia , Citidina Desaminase/imunologia , Vírus da Leucemia Murina de Friend/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Retroviridae/genética , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/virologia , Doenças dos Roedores/virologia , Viremia/imunologia , Viremia/virologia
14.
J Immunol ; 184(9): 4696-707, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20351188

RESUMO

During chronic viral infection, persistent exposure to viral Ags leads to the overexpression of multiple inhibitory cell-surface receptors that cause CD8(+) T cell exhaustion. The severity of exhaustion correlates directly with the level of infection and the number and intensity of inhibitory receptors expressed, and it correlates inversely with the ability to respond to the blockade of inhibitory pathways. Friend virus (FV) is a murine retrovirus complex that induces acute high-level viremia, followed by persistent infection and leukemia development, when inoculated into immunocompetent adult mice. In this article, we provide conclusive evidence that FV infection results in the generation of virus-specific effector CD8(+) T cells that are terminally exhausted. Acute FV-induced disease is characterized by a rapid increase in the number of virus-infected erythroblasts, leading to massive splenomegaly. Most of the expanded erythroblasts strongly express programmed death ligand-1 and MHC class I, thereby creating a highly tolerogenic environment. Consequently, FV-specific effector CD8(+) T cells uniformly express multiple inhibitory receptors, such as programmed cell death 1 (PD-1), T cell Ig domain and mucin domain 3 (Tim-3), lymphocyte activation gene-3, and CTLA-4, rapidly become nonresponsive to restimulation and are no longer reinvigorated by combined in vivo blockade of PD-1 and Tim-3 during the memory phase. However, combined blockade of PD-1 and Tim-3 during the priming/differentiation phase rescued FV-specific CD8(+) T cells from becoming terminally exhausted, resulting in improved CD8(+) T cell functionality and virus control. These results highlight FV's unique ability to evade virus-specific CD8(+) T cell responses and the importance of an early prophylactic approach for preventing terminal exhaustion of CD8(+) T cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Vírus da Leucemia Murina de Friend/imunologia , Ativação Linfocitária/imunologia , Receptores KIR/biossíntese , Animais , Antígeno B7-1/fisiologia , Antígeno B7-H1 , Linfócitos T CD8-Positivos/virologia , Células Cultivadas , Epitopos de Linfócito T/imunologia , Eritroblastos/imunologia , Eritroblastos/patologia , Eritroblastos/virologia , Feminino , Receptor Celular 1 do Vírus da Hepatite A , Receptor Celular 2 do Vírus da Hepatite A , Evasão da Resposta Imune/imunologia , Masculino , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/fisiologia , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Peptídeos/antagonistas & inibidores , Peptídeos/fisiologia , Receptores KIR/fisiologia , Receptores Virais/antagonistas & inibidores , Receptores Virais/fisiologia , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/patologia , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/patologia , Infecções Tumorais por Vírus/virologia
15.
J Virol ; 82(22): 10998-1008, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18786991

RESUMO

Several members of the apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like complex 3 (APOBEC3) family in primates act as potent inhibitors of retroviral replication. However, lentiviruses have evolved mechanisms to specifically evade host APOBEC3. Likewise, murine leukemia viruses (MuLV) exclude mouse APOBEC3 from the virions and cleave virion-incorporated APOBEC3. Although the betaretrovirus mouse mammary tumor virus has been shown to be susceptible to mouse APOBEC3, it is not known if APOBEC3 has a physiological role in restricting more widely distributed and long-coevolved mouse gammaretroviruses. The pathogenicity of Friend MuLV (F-MuLV) is influenced by several host genes: some directly restrict the cell entry or integration of the virus, while others influence the host immune responses. Among the latter, the Rfv3 gene has been mapped to chromosome 15 in the vicinity of the APOBEC3 locus. Here we have shown that polymorphisms at the mouse APOBEC3 locus indeed influence F-MuLV replication and pathogenesis: the APOBEC3 alleles of F-MuLV-resistant C57BL/6 and -susceptible BALB/c mice differ in their sequences and expression levels in the hematopoietic tissues and in their abilities to restrict F-MuLV replication both in vitro and in vivo. Furthermore, upon infection with the pathogenic Friend virus complex, (BALB/c x C57BL/6)F(1) mice displayed an exacerbated erythroid cell proliferation when the mice carried a targeted disruption of the C57BL/6-derived APOBEC3 allele. These results indicate, for the first time, that mouse APOBEC3 is a physiologically functioning restriction factor to mouse gammaretroviruses.


Assuntos
Citidina Desaminase/genética , Citidina Desaminase/imunologia , Vírus da Leucemia Murina de Friend/imunologia , Vírus da Leucemia Murina de Friend/patogenicidade , Sequência de Aminoácidos , Animais , Linhagem Celular , Suscetibilidade a Doenças , Feminino , Perfilação da Expressão Gênica , Leucemia Experimental/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Mutagênese Insercional , Polimorfismo Genético , Infecções por Retroviridae/imunologia , Alinhamento de Sequência , Infecções Tumorais por Vírus/imunologia , Replicação Viral/imunologia
16.
Vaccine ; 26(24): 2981-96, 2008 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-18255203

RESUMO

Several host genes control retroviral replication and pathogenesis. These include genes that directly affect the replication of retroviruses in target cells and those that control the host immune responses to the viral antigens. Host genetic factors that affect retroviral replication and immune responses to the viral antigens have been best studied in mouse models of Friend leukemia virus (FV) infection. Several genes located within the major histocompatibility complex (MHC), along with a separate gene not linked to the MHC, influence the host immune responses to FV antigens. The latter, the Rfv3, regulates the production of virus-neutralizing antibodies, and thus affects the duration of viremia. T-cell responses to the viral epitopes are controlled by MHC class I and class II genotypes, and both CD8(+) and CD4(+) T-cells are required for spontaneous immune resistance to FV infection. When CD4(+) T-helper cells are efficiently primed with a viral epitope, however, CD8(+) T-cells are not required for immune protection against FV infection, while B cells are absolutely required. There are individuals who possess human immunodeficiency virus type 1 (HIV-1)-reactive IgA antibodies in their mucosal secretions and show strong T-cell responses to HIV-1 antigens, even though they are negative for HIV-1 genome and HIV-1-reactive serum IgG. These HIV-1-exposed but uninfected individuals rarely possess resistance-associated alleles at known AIDS-restricting loci such as CCR5Delta32. Recent genetic analyses have indicated that a large proportion of such exposed but uninfected individuals may share a common genetic background.


Assuntos
Vírus da Leucemia Murina de Friend/imunologia , Vírus da Leucemia Murina de Friend/fisiologia , Leucemia Experimental/imunologia , Infecções por Retroviridae/imunologia , Infecções Tumorais por Vírus/imunologia , Animais , Cromossomos Humanos Par 22/genética , Anticorpos Anti-HIV/imunologia , Infecções por HIV/genética , Infecções por HIV/imunologia , Soronegatividade para HIV , HIV-1/imunologia , HIV-1/fisiologia , Humanos , Leucemia Experimental/genética , Leucemia Experimental/virologia , Camundongos , Fenótipo , Infecções por Retroviridae/genética , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/virologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/virologia , Infecções Tumorais por Vírus/genética , Replicação Viral
17.
J Immunol ; 177(5): 3108-15, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16920948

RESUMO

MILL (MHC class I-like located near the leukocyte receptor complex) is a family of MHC class I-like molecules encoded outside the MHC, which displays the highest sequence similarity to human MICA/B molecules among known class I molecules. In the present study, we show that the two members of the mouse MILL family, MILL1 and MILL2, are GPI-anchored glycoproteins associated with beta2-microglobulin (beta2m) and that cell surface expression of MILL1 or MILL2 does not require functional TAP molecules. MILL1 and MILL2 molecules expressed in bacteria could be refolded in the presence of beta2m, without adding any peptides. Hence, neither MILL1 nor MILL2 is likely to be involved in the presentation of peptides. Immunohistochemical analysis revealed that MILL1 is expressed in a subpopulation of thymic medullary epithelial cells and a restricted region of inner root sheaths in hair follicles. The present study provides additional evidence that MILL is a class I family distinct from MICA/B.


Assuntos
Membrana Celular/metabolismo , Glicoproteínas/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Microglobulina beta-2/metabolismo , Animais , Antígenos Ly/metabolismo , Linhagem Celular Tumoral , Separação Celular , Células Epiteliais/metabolismo , Folículo Piloso/metabolismo , Antígenos de Histocompatibilidade Classe I/genética , Proteínas de Membrana/metabolismo , Camundongos , Ligação Proteica , Dobramento de Proteína , Coelhos , Timo/metabolismo
18.
Cancer Immunol Immunother ; 55(12): 1459-69, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16491400

RESUMO

Hyperthermia (HT), in combination with other conventional therapeutic modalities, has become a promising approach in cancer therapy. In addition to heat-induced apoptosis, an augmented immunological effect is considered to be a benefit of hyperthermic treatment over chemo- or radiotherapy. Here, we investigated the effect of regional HT targeting the liver on immune cells, especially T cells and antigen-presenting cells, which are important in recognizing and eliminating tumor cells and pathogens such as viruses. In healthy volunteers exposed to such regional HT, both CD4(+) and CD8(+) T cells that express an activation marker CD69 increased transiently at 1 h post-treatment, with a subsequent decrease to base levels at 6 h after the treatment. At 24 h post-treatment, the percentage of CD69-positive cells significantly increased again but only among CD8(+) T cells. IFN-gamma production from PHA-stimulated peripheral blood mononuclear cells was gradually and significantly increased in the 2 days following the heating procedure, peaking at 36 h post-treatment. Furthermore, we found marked increases in plasma levels of IL-1beta and IL-6 starting at 24 h post-treatment. With regard to the number of each leukocyte subpopulation, a transient and dramatic decrease in the number of a subset of monocytes, CD14(+) CD16(-) cells, was observed at 1 h after the hyperthermic treatment, suggesting that the regional HT aimed at the liver may have influenced the extravasation of blood monocytes. No significant changes in T-cell activities or monocyte counts were observed in the volunteers exposed to heating of the lungs or the legs. These results suggest that heating of the liver may efficiently induce cellular immune responses to liver cancers.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Hipertermia Induzida , Fígado/imunologia , Monócitos/imunologia , Adulto , Idoso , Formação de Anticorpos , Antígenos CD/análise , Antígenos de Diferenciação de Linfócitos T/análise , Contagem de Células , Citocinas/sangue , Feminino , Humanos , Hipertermia Induzida/instrumentação , Interferon gama/metabolismo , Lectinas Tipo C , Neoplasias Hepáticas/terapia , Ativação Linfocitária , Masculino , Pessoa de Meia-Idade , Temperatura
19.
Int Immunol ; 18(1): 183-98, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16352628

RESUMO

CD8+ CTLs and virus-neutralizing antibodies have been associated with spontaneous and vaccine-induced immune control of retroviral infections. We previously showed that a single immunization with an env gene-encoded CD4+ T cell epitope protected mice against fatal Friend retrovirus infection. Here, we analyzed immune cell components required for the peptide-induced anti-retroviral protection. Mice lacking CD8+ T cells were nevertheless protected against Friend virus infection, while mice lacking B cells were not. Virus-producing cells both in the spleen and bone marrow decreased rapidly in their number and became undetectable by 4 weeks after infection in the majority of the peptide-immunized animals even in the absence of CD8+ T cells. In the vaccinated animals the production and class switching of virus-neutralizing and anti-leukemia cell antibodies were facilitated; however, virus-induced erythroid cell expansion was suppressed before neutralizing antibodies became detectable in the serum. Further, the numbers of virus-producing cells in the spleen and bone marrow in the early stage of the infection were smaller in the peptide-immunized than in unimmunized control mice in the absence of B cells. Thus, peptide immunization facilitates both early cellular and late humoral immune responses that lead to the effective control of the retrovirus-induced disease, but CD8+ T cells are not crucial for the elimination of virus-infected cells in the peptide-primed animals.


Assuntos
Epitopos de Linfócito T/administração & dosagem , Vírus da Leucemia Murina de Friend/imunologia , Produtos do Gene env/administração & dosagem , Leucemia Experimental/prevenção & controle , Infecções por Retroviridae/prevenção & controle , Infecções Tumorais por Vírus/prevenção & controle , Vacinação , Vacinas Virais/administração & dosagem , Animais , Anticorpos Antivirais/imunologia , Linfócitos B , Medula Óssea/imunologia , Medula Óssea/virologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos , Epitopos de Linfócito T/imunologia , Produtos do Gene env/imunologia , Leucemia Experimental/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Infecções por Retroviridae/imunologia , Baço/imunologia , Baço/virologia , Infecções Tumorais por Vírus/imunologia , Vacinação/métodos , Vacinas Virais/imunologia
20.
J Virol ; 78(12): 6322-34, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15163726

RESUMO

Recent studies have demonstrated an essential role of Gag-specific CD4+ T-cell responses for viral control in individuals infected with human immunodeficiency virus type 1. However, little is known about epitope specificities and functional roles of the Gag-specific helper T-cell responses in terms of vaccine-induced protection against a pathogenic retroviral challenge. We have previously demonstrated that immunization with Friend murine leukemia virus (F-MuLV) Gag proteins protects mice against the fatal Friend retrovirus (FV) infection. We report here the structure of a protective T helper cell (Th) epitope, (I)VTWEAIAVDPPP, identified in the p15 (MA) region of F-MuLV Gag. In mice immunized with the Th epitope-harboring peptide or a vaccinia virus-expressed native full-length MA protein, FV-induced early splenomegaly regressed rapidly. In these mice, FV-infected cells were eliminated within 4 weeks and the production of virus-neutralizing antibodies was induced rapidly after FV challenge, resulting in strong protection against the virus infection. Interestingly, mice immunized with the whole MA mounted strong CD4+ T-cell responses to the identified Th epitope, whereas mice immunized with mutant MA proteins that were not bound to the plasma membrane failed to mount efficient CD4+ T-cell responses, despite the presence of the Th epitope. These mutant MA proteins also failed to induce strong protection against FV challenge. These data indicate the importance of the properly processible MA molecule for CD4+ T-cell priming and for the resultant induction of an effective immune response against retrovirus infections.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Produtos do Gene gag/imunologia , Animais , Anticorpos Antivirais/sangue , Feminino , Vírus da Leucemia Murina de Friend/imunologia , Vírus da Leucemia Murina de Friend/fisiologia , Produtos do Gene gag/química , Produtos do Gene gag/genética , Leucemia Experimental/prevenção & controle , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infecções por Retroviridae/prevenção & controle , Infecções Tumorais por Vírus/prevenção & controle , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...