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










Base de dados
Intervalo de ano de publicação
2.
J Virol ; 73(9): 7899-902, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10438888

RESUMO

Mouse mammary tumor virus (MMTV) has been shown to preferentially infect B lymphocytes in vivo. We have used recombinant envelope-coated fluospheres and highly purified MMTV particles to study the distribution of the viral receptors on fresh mouse lymphocytes. A preferential dose-dependent binding to B lymphocytes was observed which could be competed with neutralizing antibodies. In contrast, T-lymphocyte binding remained at background levels. These results strongly suggest a higher density of viral receptor molecules on B lymphocytes than on T lymphocytes and correlate with the preferential initial infection of B lymphocytes observed in vivo.


Assuntos
Antígenos Virais de Tumores/metabolismo , Linfócitos B/virologia , Vírus do Tumor Mamário do Camundongo/metabolismo , Animais , Antígenos Virais de Tumores/genética , Antígenos Virais de Tumores/isolamento & purificação , Linfócitos B/citologia , Linfócitos B/metabolismo , Células Cultivadas , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Baço/citologia , Linfócitos T/citologia , Linfócitos T/metabolismo
3.
Immunol Rev ; 168: 287-303, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10399081

RESUMO

Mouse mammary tumor virus has developed strategies to exploit the immune response. It requires vigorous immune stimulation to achieve efficient infection. The infected antigen-presenting cells present a viral superantigen on the cell surface which stimulates strong CD4-mediated T-cell help but CD8 T-cell responses are undetectable. Despite the high frequency of superantigen-reactive T cells, the superantigen-induced immune response is comparable to classical antigen responses in terms of T-cell priming, T-cell-B-cell collaboration as well as follicular and extra-follicular B-cell differentiation. Induction of systemic anergy is observed, similar to classical antigen responses where antigen is administered systemically but does not influence the role of the superantigen-reactive T cells in the maintenance of the chronic germinal center reaction. So far we have been unable to detect a cytotoxic T-cell response to mouse mammary tumor virus peptide antigens or to the superantigen. This might yet represent another step in the viral infection strategy.


Assuntos
Formação de Anticorpos , Imunidade Celular , Vírus do Tumor Mamário do Camundongo/imunologia , Infecções por Retroviridae/imunologia , Infecções Tumorais por Vírus/imunologia , Sequência de Aminoácidos , Animais , Células Apresentadoras de Antígenos/imunologia , Antígenos Virais/imunologia , Humanos , Camundongos , Dados de Sequência Molecular , Superantígenos/imunologia , Linfócitos T/imunologia , Linfócitos T Citotóxicos/imunologia , Viroses/imunologia
4.
J Virol ; 71(10): 7295-9, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9311805

RESUMO

Mouse mammary tumor virus (MMTV) infects B lymphocytes and expresses a superantigen on the cell surface after integration of its reverse-transcribed genome. Superantigen-dependent B- and T-cell activation becomes detectable 2 to 3 days after infection. We show here that before this event, B cells undergo a polyclonal activation which does not involve massive proliferation. This first phase of B-cell activation is T cell independent. Moreover, during the first phase of activation, when only a small fraction of B cells is infected by MMTV(SW), viral DNA is detected only in activated B cells. Such a B-cell activation is also seen after injection of murine leukemia virus but not after injection of vaccinia virus, despite the very similar kinetics and intensity of the immune response. Since retroviruses require activated target cells to induce efficient infection, these data suggest that the early polyclonal retrovirus-induced target cell activation might play an important role in the establishment of retroviral infections.


Assuntos
Linfócitos B/virologia , DNA Viral/biossíntese , Ativação Linfocitária , Neoplasias Mamárias Experimentais/imunologia , Vírus do Tumor Mamário do Camundongo/imunologia , Infecções por Retroviridae/imunologia , Linfócitos T/virologia , Infecções Tumorais por Vírus/imunologia , Animais , Linfócitos B/imunologia , Replicação do DNA , DNA Viral/análise , Feminino , Genoma Viral , Cinética , Linfonodos/imunologia , Linfonodos/virologia , Neoplasias Mamárias Experimentais/virologia , Vírus do Tumor Mamário do Camundongo/genética , Vírus do Tumor Mamário do Camundongo/patogenicidade , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos , Camundongos Nus , Leite/virologia , Linfócitos T/imunologia , Fatores de Tempo , Integração Viral , Replicação Viral
5.
J Immunol ; 159(6): 2807-14, 1997 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-9300703

RESUMO

Infectious mouse mammary tumor virus (MMTV) is a retrovirus that expresses a superantigen shortly after infection of B cells. The superantigen first drives the polyclonal activation and proliferation of superantigen-reactive CD4+ T cells, which then induce the infected B cells to proliferate and differentiate. Part of the MMTV-induced B cell response leads to the production of Abs that are specific for the viral envelope protein gp52. Here we show that this Ab response has virus-neutralizing activity and confers protection against superinfection by other MMTV strains in vivo as soon as 4 to 7 days after infection. A protective Ab titer is maintained lifelong. Viral infection as well as the superantigen-induced T-B collaboration are required to generate this rapid and long lasting neutralizing Ab response. Polyclonal or superantigen-independent B cell activation, on the contrary, does not lead to detectable virus neutralization. The early onset of this superantigen-dependent neutralizing response suggests that viral envelope-specific B cells are selectively recruited to form part of the extrafollicular B cell response and are subsequently amplified and maintained by superantigen-reactive Th cells.


Assuntos
Anticorpos Antivirais/imunologia , Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Vírus do Tumor Mamário do Camundongo/imunologia , Infecções por Retroviridae/imunologia , Superantígenos/imunologia , Infecções Tumorais por Vírus/imunologia , Animais , Linfócitos T CD4-Positivos/virologia , Imunidade , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C
6.
Eur J Immunol ; 26(7): 1595-602, 1996 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8766566

RESUMO

B cells are the primary targets of infection for mouse mammary tumor virus (MMTV). However, for productive retroviral infection, T cell stimulation through the virally-encoded superantigen (SAG) is necessary. It activates B cells and leads to cell division and differentiation. To characterize the role of B cell differentiation for the MMTV life cycle, we studied the course of infection in transgenic mice deficient for CD28/CTLA4-B7 interactions (mCTLA4-H gamma 1 transgenic mice). B cell infection occurred in CTLA4-H gamma 1 transgenic mice as integrated proviral DNA could be detected in draining lymph node cells early after infection by polymerase chain reaction analysis. In mice expressing I-E, B cells were able to present the viral SAG efficiently to V beta 6+ T cells. These cells expanded specifically and were triggered to express the activation marker CD69. Further stages of progression of infection appeared to be defective. Kinetics experiments indicated that T and B cell stimulation stopped more rapidly than in control mice. B cells acquired an activated CD69+ phenotype, were induced to produce IgM but only partially switched to IgG secretion. Finally, the dissemination of infected cells to other lymph nodes and spleen was reduced and the peripheral deletion of V beta 6+ T cells was minimal. In contrast, in mice lacking I-E, T cell stimulation was also impaired and B cell activation undetectable. These data implicate B7-dependent cellular interactions for superantigenic T cell stimulation by low-affinity TCR ligands and suggest a role of B cell differentiation in viral dissemination and peripheral T cell deletion.


Assuntos
Antígenos de Diferenciação/metabolismo , Antígenos Virais/imunologia , Linfócitos B/imunologia , Antígeno B7-1/metabolismo , Antígenos CD28/metabolismo , Imunoconjugados , Ativação Linfocitária , Vírus do Tumor Mamário do Camundongo/imunologia , Superantígenos/farmacologia , Linfócitos T/imunologia , Abatacepte , Animais , Antígenos CD , Antígenos de Diferenciação/genética , Sequência de Bases , Antígeno CTLA-4 , Diferenciação Celular/imunologia , Deleção Clonal , Antígenos de Histocompatibilidade Classe II/imunologia , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Infecções por Retroviridae/imunologia , Infecções Tumorais por Vírus/imunologia
7.
Cell ; 74(3): 529-40, 1993 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-8394220

RESUMO

Endogenous and infectious mouse mammary tumor viruses (MMTVs) encode in their 3' long terminal repeat a protein that exerts superantigen activity; that is, it is able to interact with T cells via the variable domain of the T cell receptor (TCR) beta chain. We show here that transmission of an infectious MMTV is prevented when superantigen-reactive cells are absent through either clonal deletion due to the expression of an endogenous MTV with identical superantigen specificity or exclusion due to expression of a transgenic TCR beta chain that does not interact with the viral superantigen. A strict requirement for superantigen-reactive T cells is also seen for a local immune response following MMTV infection. This immune response locally amplifies the number of MMTV-infected B cells, most likely owing to their clonal expansion. Collectively, our data indicate that a superantigen-induced immune response is critical for the MMTV life cycle.


Assuntos
Antígenos Virais/imunologia , Neoplasias Mamárias Experimentais/microbiologia , Vírus do Tumor Mamário do Camundongo/fisiologia , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Animais , Antígenos Virais/biossíntese , Antígenos Virais/metabolismo , Linfócitos B/imunologia , Sequência de Bases , DNA Viral/genética , DNA Viral/isolamento & purificação , Citometria de Fluxo , Linfonodos/microbiologia , Neoplasias Mamárias Experimentais/imunologia , Vírus do Tumor Mamário do Camundongo/genética , Vírus do Tumor Mamário do Camundongo/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos , Reação em Cadeia da Polimerase , Sequências Repetitivas de Ácido Nucleico , Linfócitos T/imunologia
8.
Nucleic Acids Res ; 21(6): 1457-61, 1993 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-8464737

RESUMO

Iron regulatory factor (IRF) is a cytoplasmic mRNA-binding protein that coordinates post-transcriptionally the expression of several important proteins in iron metabolism. Binding of IRF to iron-responsive elements (IRE) in the 5' untranslated region (UTR) of ferritin and erythroid 5-aminolevulinic acid-synthase mRNAs inhibits their translation, whereas binding to IREs in the 3' UTR of transferrin receptor (TfR) mRNA prevents the degradation of this mRNA. IRF binds RNA strongly after iron deprivation, but is inactive, yet present, under conditions of high cellular iron supply. Recently, IRF was also shown to have aconitase activity indicating the existence of an Fe-S cluster in the protein. In the current study we expressed human IRF in insect cells from recombinant baculovirus and analysed IRE-binding and aconitase activities under various culture conditions. Newly made apoprotein, synthesized in the absence of iron, was fully active in IRE-binding, but showed no aconitase activity. In contrast, IRF made by cells grown in high iron medium bound RNA poorly, but exhibited high aconitase activity with a Km of 9.2 microM for cis-aconitate. Apo-IRF was converted in vitro to active aconitase by Fe-S cluster-generating conditions, and under the same conditions lost its RNA-binding capacity. These results indicate that the two activities are mutually exclusive and controlled through formation of the Fe-S cluster.


Assuntos
Aconitato Hidratase/química , Proteínas Ferro-Enxofre/metabolismo , Ferro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Aconitato Hidratase/metabolismo , Animais , Apoproteínas/metabolismo , Baculoviridae , Linhagem Celular , Clonagem Molecular , Humanos , Técnicas In Vitro , Insetos , Proteínas Reguladoras de Ferro , Proteínas Ferro-Enxofre/química , Cinética , Proteínas de Ligação a RNA/genética , Proteínas Recombinantes/metabolismo
9.
J Exp Med ; 177(2): 359-66, 1993 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-8093892

RESUMO

Superantigens are defined by their ability to stimulate a large fraction of T cells via interaction with the T cell receptor (TCR) V beta domain. Endogenous superantigens, classically termed minor lymphocyte-stimulating (Mls) antigens, were recently identified as products of open reading frames (ORF) in integrated proviral copies of mouse mammary tumor virus (MMTV). We have described an infectious MMTV homologue of the classical endogenous superantigen Mls-1a (Mtv-7). The ORF molecules of both the endogenous Mtv-7 and the infectious MMTV(SW) interact with T cells expressing the TCR V beta 6, 7, 8.1, and 9 domains. Furthermore, the COOH termini of their ORF molecules, thought to confer TCR specificity, are very similar. Since successful transport of MMTV from the site of infection in the gut to the mammary gland depends on a functional immune system, we were interested in determining the early events after and requirements for MMTV infection. We show that MMTV(SW) infection induces a massive response of V beta 6+ CDC4+ T cells, which interact with the viral ORF. Concomitantly, we observed a B cell response and differentiation that depends on both the presence and stimulation of the superantigen-reactive T cells. Furthermore, we show that B cells are the main target of the initial MMTV infection as judged by the presence of the reverse-transcribed viral genome and ORF transcripts. Thus, we suggest that MMTV infection of B cells leads to ORF-mediated B-T cell interaction, which maintains and possibly amplifies viral infection.


Assuntos
Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Ativação Linfocitária , Vírus do Tumor Mamário do Camundongo/imunologia , Antígenos Secundários de Estimulação de Linfócitos/imunologia , Infecções Tumorais por Vírus/imunologia , Animais , Formação de Anticorpos , Antígenos Virais/imunologia , Linfócitos B/microbiologia , Sequência de Bases , Regulação Viral da Expressão Gênica , Genes Virais , Vírus do Tumor Mamário do Camundongo/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos/química , RNA Mensageiro/genética , RNA Viral/genética , Infecções Tumorais por Vírus/microbiologia , Proteínas Estruturais Virais/genética
11.
J Exp Med ; 176(6): 1769-72, 1992 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-1334118

RESUMO

Minor lymphocyte stimulating (Mls) antigens specifically stimulate T cell responses that are restricted to particular T cell receptor (TCR) beta chain variable domains. The Mls phenotype is genetically controlled by an open reading frame (orf) located in the 3' long terminal repeat of mouse mammary tumor virus (MMTV); however, the mechanism of action of the orf gene product is unknown. Whereas predicted orf amino acid sequences show strong overall homology, the 20-30 COOH-terminal residues are strikingly polymorphic. This polymorphic region correlates with TCR V beta specificity. We have generated monoclonal antibodies to a synthetic peptide encompassing the 19 COOH-terminal amino acid residues of Mtv-7 orf, which encodes the Mls-1a determinant. We show here that these antibodies block Mls responses in vitro and can interfere specifically with thymic clonal deletion of Mls-1a reactive V beta 6+ T cells in neonatal mice. Furthermore, the antibodies can inhibit V beta 6+ T cell responses in vivo to an infectious MMTV that shares orf sequence homology and TCR specificity with Mtv-7. These results confirm the predicted extracellular localization of the orf COOH terminus and imply that the orf proteins of both endogenous and exogenous MMTV interact directly with TCR V beta.


Assuntos
Anticorpos Monoclonais/imunologia , Vírus do Tumor Mamário do Camundongo/genética , Antígenos Secundários de Estimulação de Linfócitos/imunologia , Fases de Leitura Aberta , Linfócitos T/imunologia , Sequência de Aminoácidos , Animais , Baculoviridae/genética , Sequência de Bases , Linhagem Celular , DNA/genética , DNA/isolamento & purificação , Imunidade Celular , Vírus do Tumor Mamário do Camundongo/imunologia , Camundongos , Camundongos Endogâmicos BALB C/imunologia , Camundongos Endogâmicos , Antígenos Secundários de Estimulação de Linfócitos/genética , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos , Peptídeos/síntese química , Peptídeos/imunologia , Reação em Cadeia da Polimerase , Receptores de Antígenos de Linfócitos T/genética , Transfecção
12.
J Exp Med ; 175(6): 1623-33, 1992 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-1316932

RESUMO

The classical minor lymphocyte stimulating (Mls) antigens, which induce a strong primary T cell response in vitro, are closely linked to endogenous copies of mouse mammary tumor viruses (MMTV). Expression of Mls genes leads to clonal deletion of T cell subsets expressing specific T cell receptor (TCR) V beta chains. We describe the isolation and characterization of a new exogenous (infectious) MMTV with biological properties similar to the Mls antigen Mls-1a. In vivo administration of either Mls-1a-expressing B cells or the infectious MMTV (SW) led to an increase of T cells expressing V beta 6 followed by their deletion. Surprisingly, different kinetics of deletion were observed with the exogenous virus depending upon the route of infection. Infection through the mucosa led to a slow deletion of V beta 6+ T cells, whereas deletion was rapid after subcutaneous infection. Sequence analysis of the open reading frames in the 3' long terminal repeat of both this exogenous MMTV (SW) and of Mtv-7 (which is closely linked to Mls-1a) revealed striking similarities, particularly in the COOH terminus, which has been implicated in TCR V beta recognition. The identification of an infectious MMTV with the properties of a strong Mls antigen provides a new, powerful tool to study immunity and tolerance in vivo.


Assuntos
Genes Virais , Vírus do Tumor Mamário do Camundongo/genética , Antígenos Secundários de Estimulação de Linfócitos/genética , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais , Sequência de Bases , Clonagem Molecular , Feminino , Linfonodos/imunologia , Vírus do Tumor Mamário do Camundongo/isolamento & purificação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos , Leite/microbiologia , Antígenos Secundários de Estimulação de Linfócitos/análise , Dados de Sequência Molecular , Oligodesoxirribonucleotídeos , Reação em Cadeia da Polimerase/métodos , Homologia de Sequência do Ácido Nucleico , Especificidade da Espécie , Timo/imunologia
13.
Int Rev Immunol ; 8(4): 327-36, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-1351535

RESUMO

The nature of the mysterious minor lymphocyte stimulating (Mls) antigens has recently been clarified. These molecules which were key elements for our current understanding of immune tolerance, have a strong influence on the mouse immune system and are encoded by the open reading frame (orf) of endogenous and exogenous mouse mammary tumor viruses (MMTV's). The knowledge that these antigens are encoded by cancerogenic retroviruses opens an interdisciplinary approach for understanding the mechanisms of immune responses and immune tolerance, retroviral carcinogenesis, and retroviral strategies for infection.


Assuntos
Antígenos Virais/imunologia , Vírus do Tumor Mamário do Camundongo/imunologia , Antígenos Secundários de Estimulação de Linfócitos/imunologia , Sequência de Aminoácidos , Animais , Linfócitos T CD4-Positivos/imunologia , Vírus do Tumor Mamário do Camundongo/genética , Antígenos Secundários de Estimulação de Linfócitos/genética , Dados de Sequência Molecular , Fases de Leitura Aberta , Provírus/genética , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Linfócitos T Reguladores/imunologia
14.
Nature ; 350(6315): 207-11, 1991 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-1848685

RESUMO

Autoreactive T lymphocytes are clonally deleted during maturation in the thymus. Deletion of T cells expressing particular receptor V beta elements is controlled by poorly defined autosomal dominant genes. A gene has now been identified by expression of transgenes in mice which causes deletion of V beta 14+ T cells. The gene lies in the open reading frame of the long terminal repeat of the mouse mammary tumour virus.


Assuntos
Genes Virais , Vírus do Tumor Mamário do Camundongo/genética , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T/imunologia , Sequência de Aminoácidos , Animais , Deleção Cromossômica , Eletroforese em Gel de Poliacrilamida , Feminino , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Fases de Leitura Aberta , Fenótipo , Provírus/genética
16.
EMBO J ; 9(12): 3815-9, 1990 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-2249651

RESUMO

The granule/perforin exocytosis model of CTL mediated cytolysis proposes that CTL, upon recognition of the specific targets, release the cytolytic, pore-forming protein perforin into the intercellular space which then mediates the cytotoxic effect. However, direct evidence for the involvement of perforin is still lacking, and indeed, recent results even seem incompatible with the model. To determine directly the role of perforin in CTL cytotoxicity, perforin antisense oligonucleotides were exogenously added during the stimulation of mouse spleen derived T cells and human peripheral blood lymphocytes (PBL), respectively. Perforin protein expression in lymphocytes was reduced by up to 65%, and cytotoxicity of stimulated T cells by as much as 69% (5.7-fold). These results provide the first experimental evidence for a crucial role of perforin in lymphocyte mediated cytotoxicity.


Assuntos
Citotoxicidade Imunológica/efeitos dos fármacos , Glicoproteínas de Membrana , Proteínas de Membrana/genética , Oligonucleotídeos Antissenso/farmacologia , Linfócitos T Citotóxicos/imunologia , Animais , Sequência de Bases , Linhagem Celular , Células Cultivadas , Humanos , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Oligonucleotídeos Antissenso/síntese química , Oligonucleotídeos Antissenso/metabolismo , Perforina , Proteínas Citotóxicas Formadoras de Poros , Baço/imunologia , Linfócitos T Citotóxicos/efeitos dos fármacos
17.
J Immunol ; 145(6): 1960-7, 1990 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-2391425

RESUMO

B cells express MHC class II Ag in a constitutive fashion, whereas macrophages can do so only after induction by a variety of exogenous stimuli. In this study we describe interspecies somatic cell hybrids between the human B cell Raji and the murine macrophage cell P388 D1. This murine cell line does not express detectable levels of class II mRNA. Phenotypic, molecular, and karyotype analysis of a series of hybrids showed that murine macrophage class II genes can be expressed in a constitutive fashion under the control of the human B cell genome. This event is the consequence of de novo accumulation of class II specific mRNA and thus probably reflects activation of transcription. In certain cases the amount of murine class II Ag expressed on the surface of the hybrid cell was significantly higher than the one observed in the parental macrophage cells after induction with IFN-gamma and was not further modified by treatment with the murine lymphokine. Reversion from a murine class II-positive to class II-negative cell surface phenotype in the hybrids correlated with reduced expression of human markers and more important with segregation of human chromosomes. Interestingly, in this case certain hybrids still expressed detectable levels of murine class II mRNA and increased levels of murine invariant chain mRNA when compared with parental P388 D1 murine macrophage cells. These results indicate that constitutive class II gene expression behaves as a dominant trait in B cell x macrophage somatic cell hybrids. Possible mechanisms responsible of the different control of class II gene expression during cell type differentiation are discussed.


Assuntos
Regulação da Expressão Gênica , Antígenos de Histocompatibilidade Classe II/genética , Complexo Principal de Histocompatibilidade , Animais , Linfócitos B/fisiologia , Northern Blotting , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Células Híbridas , Técnicas In Vitro , Cariotipagem , Macrófagos/fisiologia , Camundongos , RNA Mensageiro/genética
19.
J Immunol ; 142(8): 2902-10, 1989 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-2495328

RESUMO

In this study we investigated the molecular mechanisms responsible for the extinction of the constitutive MHC class II gene expression of human B cells on somatic cell hybridization with murine plasmocytoma cells. We found that this event is due to trans-acting suppressor functions of mouse origin pre-existing in the plasmocytoma cells and acting at transcriptional level. Transcription of the entire family of human class II genes is suppressed, including genes as DO beta for which a distinct regulation of expression in B cells had been previously demonstrated. Suppression appears specific for class II genes because in the hybrids expression of MHC class I genes of mouse is unaffected and of human only partially reduced. Interestingly, also murine invariant chain gene is expressed in both parental plasmocytoma and hybrid cells although at reduced amounts as compared to a murine class II positive B cell line. The class II negative phenotype of hybrid cells and parental plasmocytoma cells is highly stable and unaffected by treatment with protein synthesis inhibitors, suggesting that the transcriptional suppressor function is not mediated by rapid, labile turning-over proteins. Possible mechanisms responsible for transcriptional regulation of MHC class II gene expression during terminal differentiation of B cells to plasma cells are discussed.


Assuntos
Linfócitos B/fisiologia , Genes MHC da Classe II , Plasmócitos/fisiologia , Transcrição Gênica , Linfócitos B/imunologia , Diferenciação Celular , Cicloeximida/farmacologia , Humanos , Células Híbridas , Fenótipo , Plasmócitos/imunologia , RNA Mensageiro/análise , Supressão Genética , Células Tumorais Cultivadas
20.
Proc Natl Acad Sci U S A ; 85(7): 2229-33, 1988 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-3127829

RESUMO

Constitutive expression of major histocompatibility complex class II genes is acquired very early in B-cell ontogeny and is maintained up to the B-cell blast stage. Terminal differentiation in plasma cells is, however, accompanied by a loss of class II gene expression. In B cells this gene system is under the control of several loci encoding transacting factors with activator function, one of which, the aIr-1 gene product, operates across species barriers. In this report human class II gene expression is shown to be extinguished in somatic cell hybrids between the human class II-positive B-cell line Raji and the mouse class II-negative plasmacytoma cell line P3-U1. Since all murine chromosomes are retained in these hybrids and no preferential segregation of a specific human chromosome is observed, the results are compatible with the presence of suppressor factors of mouse origin, operating across species barriers and inhibiting class II gene expression. Suppression seems to act at the level of transcription or accumulation of class II-specific mRNA, since no human, and very few murine, class II transcripts are detectable in the hybrids.


Assuntos
Linfócitos B/citologia , Diferenciação Celular , Regulação da Expressão Gênica , Genes MHC da Classe II , Antígenos de Histocompatibilidade Classe II/biossíntese , Plasmócitos/citologia , Animais , Linfoma de Burkitt/patologia , Antígenos de Histocompatibilidade Classe II/genética , Humanos , Células Híbridas/metabolismo , Camundongos , Fenótipo , Plasmocitoma/patologia , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...