Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogenesis ; 5(6): e231, 2016 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-27270613

RESUMO

A growing body of evidence implicates the noncanonical NF-κB pathway as a key driver of glioma invasiveness and a major factor underlying poor patient prognoses. Here, we show that NF-κB-inducing kinase (NIK/MAP3K14), a critical upstream regulator of the noncanonical NF-κB pathway, is both necessary and sufficient for cell-intrinsic invasion, as well as invasion induced by the cytokine TWEAK, which is strongly associated with tumor pathogenicity. NIK promotes dramatic alterations in glioma cell morphology that are characterized by extensive membrane branching and elongated pseudopodial protrusions. Correspondingly, NIK increases the phosphorylation, enzymatic activity and pseudopodial localization of membrane type-1 matrix metalloproteinase (MT1-MMP/MMP14), which is associated with enhanced tumor cell invasion of three-dimensional collagen matrices. Moreover, NIK regulates MT1-MMP activity in cells lacking the canonical NF-κB p65 and cRel proteins. Finally, increased expression of NIK is associated with elevated MT1-MMP phosphorylation in orthotopic xenografts and co-expression of NIK and MT1-MMP in human tumors is associated with poor glioma patient survival. These data reveal a novel role of NIK to enhance pseudopodia formation, MT1-MMP enzymatic activity and tumor cell invasion independently of p65. Collectively, our findings underscore the therapeutic potential of approaches targeting NIK in highly invasive tumors.

2.
Gene Ther ; 19(8): 818-27, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22052242

RESUMO

Epstein-Barr virus (EBV) infection leads to Hodgkin's disease (HD) in some immunocompetent hosts. The malignant Reed-Sternberg cells of HD only express a limited array of subdominant EBV antigens to evade pre-existing immune responses to EBV. The EBV-encoded latent membrane proteins (LMP1 and LMP2), which are expressed by HD and various EBV-associated malignancies, have been proposed as a potential target for cytotoxic T lymphocyte (CTL)-based therapy. However, the precursor frequency for LMP-specific CTL is generally low in healthy EBV-infected hosts, and immunotherapy based on these antigens is often compromised by the poor immunogenicity and the oncogenic potential. In the present study, we report that transiently expressing an inhibitor of A20, a key negative regulator of inflammatory signaling pathways, together with the LMP antigens (truncated LMP1 and full-length LMP2) greatly enhances maturation and cytokine production of human (h) monocyte-derived dendritic cells (DCs). As a consequence, LMP1/2-expressed, A20-silenced hDCs have an enhanced potency to prime LMP-specific T-cell response. When the in vitro primed T cells are adoptively transferred into tumor-xenografted, severe-combined immunodeficient mice, some of the xenografted tumors approach complete regression. Thus, the study may provide an available resource of LMP-specific T cells for T-cell immunotherapy.


Assuntos
Proteínas de Ligação a DNA/antagonistas & inibidores , Células Dendríticas/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Proteínas Nucleares/antagonistas & inibidores , Linfócitos T Citotóxicos/imunologia , Proteínas da Matriz Viral/imunologia , Animais , Células Dendríticas/metabolismo , Imunoterapia Adotiva/métodos , Ativação Linfocitária , Linfoma não Hodgkin/terapia , Camundongos , Camundongos SCID , Linfócitos T Citotóxicos/metabolismo , Transfecção , Proteína 3 Induzida por Fator de Necrose Tumoral alfa , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Cell Death Differ ; 18(3): 452-64, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20885445

RESUMO

The anti-apoptotic Bcl-2 protein, which confers oncogenic transformation and drug resistance in most human cancers, including breast cancer, has recently been shown to effectively counteract autophagy by directly targeting Beclin1, an essential autophagy mediator and tumor suppressor. However, it remains unknown whether autophagy inhibition contributes to Bcl-2-mediated oncogenesis. Here, by using a loss-of-function mutagenesis study, we show that Bcl-2-mediated antagonism of autophagy has a critical role in enhancing the tumorigenic properties of MCF7 breast cancer cells independent of its anti-apoptosis activity. A Bcl-2 mutant defective in apoptosis inhibition but competent for autophagy suppression promotes MCF7 breast cancer cell growth in vitro and in vivo as efficiently as wild-type Bcl-2. The growth-promoting activity of this Bcl-2 mutant is strongly correlated with its suppression of Beclin1-dependent autophagy, leading to sustained p62 expression and increased DNA damage in xenograft tumors, which may directly contribute to tumorigenesis. Thus, the anti-autophagic property of Bcl-2 is a key feature of Bcl-2-mediated oncogenesis and may in some contexts, serve as an attractive target for breast and other cancer therapies.


Assuntos
Apoptose , Autofagia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Regulação para Baixo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Beclina-1 , Neoplasias da Mama/ultraestrutura , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Dano ao DNA , Feminino , Proteínas de Choque Térmico/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Nus , Proteínas Mutantes/metabolismo , Células NIH 3T3 , Ligação Proteica , Estrutura Secundária de Proteína , Proteínas Proto-Oncogênicas c-bcl-2/química , Proteína Sequestossoma-1 , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Curr Top Microbiol Immunol ; 269: 187-201, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12224509

RESUMO

To establish lifelong infection in the presence of an active host immune system, herpesviruses have acquired an impressive array of immune modulatory mechanisms that contribute to their success as long-term parasites. Kaposi's sarcoma-associated herpesvirus (KSHV) is the most recently discovered human tumor virus and is associated with the pathogenesis of Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. KSHV has acquired a battery of genes to assist in viral survival against the host immune response. These viral gene products target a variety of host immune surveillance mechanisms, including the cytokine-mediated immune response, apoptosis, natural killer (NK) cell killing and T cell-mediated responses. This review summarizes our understanding of the role of these viral proteins in the escape from host immune surveillance, which ultimately contributes to lifelong infection and pathogenesis of KSHV.


Assuntos
Infecções por Herpesviridae/imunologia , Herpesvirus Humano 8/imunologia , Animais , Apoptose , Citocinas , Genes Virais , Herpesvirus Humano 8/genética , Humanos , Imunidade Ativa/genética , Imunidade Inata/genética , Interferons , Células Matadoras Naturais , Linfócitos T Citotóxicos , Proteínas Virais/imunologia , Replicação Viral
6.
J Immunol ; 167(5): 2657-65, 2001 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-11509608

RESUMO

The molecular basis of X-linked lymphoproliferative (XLP) disease has been attributed to mutations in the signaling lymphocytic activation molecule-associated protein (SAP), an src homology 2 domain-containing intracellular signaling molecule known to interact with the lymphocyte-activating surface receptors signaling lymphocytic activation molecule and 2B4. To investigate the effect of SAP defects on TCR signal transduction, herpesvirus saimiri-immortalized CD4 Th cells from XLP patients and normal healthy individuals were examined for their response to TCR stimulation. CD4 T cells of XLP patients displayed elevated levels of tyrosine phosphorylation compared with CD4 T cells from healthy individuals. In addition, downstream serine/threonine kinases are constitutively active in CD4 T cells of XLP patients. In contrast, TCR-mediated activation of Akt, c-Jun-NH(2)-terminal kinases, and extracellular signal-regulated kinases in XLP CD4 T cells was transient and rapidly diminished when compared with that in control CD4 T cells. Consequently, XLP CD4 T cells exhibited severe defects in up-regulation of IL-2 and IFN-gamma cytokine production upon TCR stimulation and in MLRs. Finally, SAP specifically interacted with a 75-kDa tyrosine-phosphorylated protein upon TCR stimulation. These results demonstrate that CD4 T cells from XLP patients exhibit aberrant TCR signal transduction and that the defect in SAP function is likely responsible for this phenotype.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular , Transtornos Linfoproliferativos/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Sequência de Bases , Complexo CD3/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Transformação Celular Viral , Células Cultivadas , Citocinas/biossíntese , Primers do DNA/genética , Herpesvirus Saimiriíneo 2 , Herpesvirus Humano 4 , Humanos , Transtornos Linfoproliferativos/genética , Transtornos Linfoproliferativos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutação , Proteína Oncogênica v-cbl , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Proteínas Oncogênicas de Retroviridae/metabolismo , Transdução de Sinais , Proteína Associada à Molécula de Sinalização da Ativação Linfocitária , Linfócitos T Auxiliares-Indutores/metabolismo , Tirosina/metabolismo , Proteína-Tirosina Quinase ZAP-70
7.
J Virol ; 75(16): 7572-82, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11462029

RESUMO

The irreversible cell cycle arrest and apoptosis induced by p53 are part of the host surveillance mechanisms for viral infection and tumor induction. Kaposi's sarcoma-associated herpesvirus (KSHV), the most recently discovered human tumor virus, is associated with the pathogenesis of Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease. The K9 open reading frame of KSHV encodes a viral interferon (IFN) regulatory factor (vIRF) which functions as a repressor for cellular IFN-mediated signal transduction and as an oncoprotein to induce cell growth transformation. Here, we demonstrate that KSHV vIRF interacts with the cellular p53 tumor suppressor through the putative DNA binding region of vIRF and the central region of p53. This interaction suppresses the level of phosphorylation and acetylation of p53 and inhibits transcriptional activation of p53. As a consequence, vIRF efficiently prevents p53-mediated apoptosis. These results suggest that KSHV vIRF interacts with and inhibits the p53 tumor suppressor to circumvent host growth surveillance and to facilitate uncontrolled cell proliferation.


Assuntos
Regulação Viral da Expressão Gênica , Genes p53 , Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/fisiologia , Interferons/genética , Animais , Linhagem Celular , Genes Supressores de Tumor , Infecções por Herpesviridae/genética , Humanos , Replicação Viral/genética
8.
Cytokine Growth Factor Rev ; 12(2-3): 245-57, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11325605

RESUMO

Kaposi's Sarcoma associated Herpesvirus (KSHV) is the most recently discovered human tumor virus and is associated with the pathogenesis of Kaposi's sarcoma, primary effusion lymphoma, and Multicentric Casttleman's disease. KSHV contains numerous open reading frames with striking homology to cellular genes. These viral gene products play a variety of roles in KSHV-associated pathogenesis by disrupting cellular signal transduction pathways, which include interferon-mediated anti-viral responses, cytokine-regulated cell growth, apoptosis, and cell cycle control. In this review, we will attempt to cover our understanding of how viral proteins deregulate cellular signaling pathways, which ultimately contribute to the conversion of normal cells to cancerous cells.


Assuntos
Herpesvirus Humano 8/metabolismo , Mimetismo Molecular , Animais , Apoptose , Transformação Celular Neoplásica/metabolismo , Evolução Molecular , Genes Virais/genética , Haplorrinos/virologia , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/imunologia , Herpesvirus Humano 8/isolamento & purificação , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virologia , Transdução de Sinais , Proteínas Virais/genética , Proteínas Virais/imunologia , Proteínas Virais/metabolismo
9.
Adv Cancer Res ; 80: 51-82, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11034540

RESUMO

Members of the gamma herpesvirus family include the lymphocryptoviruses (gamma-1 herpesviruses) and the rhadinoviruses (gamma-2 herpesviruses). Gammaherpesvirinae uniformly establish long-term, latent, reactivatable infection of lymphocytes, and several members of the gamma herpesviruses are associated with lymphoproliferative diseases. Epstein-Barr virus is a lymphocryptovirus, whereas Kaposi sarcoma-associated herpesvirus and Herpesvirus saimiri are members of the rhadinovirus family. Genes encoded by these viruses are involved in a diverse array of cellular signaling pathways. This review attempts to cover our understanding of how viral proteins deregulate cellular signaling pathways that ultimately contribute to the conversion of normal cells to cancerous cells.


Assuntos
Herpesvirus Saimiriíneo 2/metabolismo , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 8/metabolismo , Transformação Genética , Herpesvirus Saimiriíneo 2/genética , Herpesvirus Humano 4/genética , Herpesvirus Humano 8/genética , Modelos Biológicos , Filogenia
10.
Mol Cell Biol ; 20(21): 8254-63, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11027294

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) has been consistently identified in Kaposi's sarcomas, body cavity-based lymphomas, and some forms of Castleman's disease. The K9 open reading frame of KSHV encodes a viral interferon regulatory factor (vIRF) which functions as a repressor for cellular interferon-mediated signal transduction and as an oncogene to induce cell growth transformation. We demonstrate that KSHV vIRF directly interacts with cellular transcriptional coactivator p300 and displaces p300/CBP-associated factor from p300 complexes. This interaction inhibits the histone acetyltransferase activity of p300, resulting in drastic reduction of nucleosomal histone acetylation and alteration of chromatin structure. As a consequence, vIRF expression markedly alters cellular cytokine expression, which is regulated by acetylation of nucleosomal histones. These results demonstrate that KSHV vIRF interacts with and inhibits the p300 transcriptional coactivator to circumvent the host antiviral immune response and to induce a global alteration of cellular gene expression. These studies also illustrate how a cellular gene captured by a herpesvirus has evolved several functions that suit the needs of the virus.


Assuntos
Acetiltransferases/antagonistas & inibidores , Proteínas de Ciclo Celular/antagonistas & inibidores , Herpesvirus Humano 8/metabolismo , Proteínas de Saccharomyces cerevisiae , Células 3T3 , Acetilação , Acetiltransferases/metabolismo , Animais , Células COS , Ciclo Celular , Linhagem Celular , Separação Celular , Cromatina/metabolismo , Citocinas/metabolismo , DNA Complementar/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Citometria de Fluxo , Imunofluorescência , Genes Reporter , Histona Acetiltransferases , Histonas/metabolismo , Insetos , Fatores Reguladores de Interferon , Camundongos , Microscopia Confocal , Modelos Genéticos , Mutagênese Sítio-Dirigida , Nucleossomos/metabolismo , Fases de Leitura Aberta , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Transdução de Sinais , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Ativação Transcricional , Transfecção , Proteínas Virais , Fatores de Transcrição de p300-CBP
11.
Immunity ; 13(3): 365-74, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11021534

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) K3 and K5 proteins dramatically downregulate MHC class I molecules. However, although MHC class I downregulation may protect KSHV-infected cells from cytotoxic T lymphocyte recognition, these cells become potential targets for natural killer (NK) cell-mediated lysis. We now show that K5 also downregulates ICAM-1 and B7-2, which are ligands for NK cell-mediated cytotoxicity receptors. As a consequence, K5 expression drastically inhibits NK cell-mediated cytotoxicity. Conversely, de novo expression of B7-2 and ICAM-1 resensitizes the K5-expressing cells to NK cell-mediated cytotoxicity. This is a novel viral immune evasion strategy where KSHV K5 achieves immune avoidance by downregulation of cellular ligands for NK cell-mediated cytotoxicity receptors.


Assuntos
Citotoxicidade Imunológica/imunologia , Herpesvirus Humano 8/imunologia , Proteínas Imediatamente Precoces/fisiologia , Imunossupressores/farmacologia , Células Matadoras Naturais/imunologia , Antígenos CD/biossíntese , Antígenos CD/fisiologia , Antígeno B7-2 , Membrana Celular/imunologia , Membrana Celular/virologia , Citoplasma/imunologia , Citoplasma/virologia , Regulação para Baixo/imunologia , Sinergismo Farmacológico , Humanos , Proteínas Imediatamente Precoces/biossíntese , Imunidade Inata , Molécula 1 de Adesão Intercelular/biossíntese , Molécula 1 de Adesão Intercelular/fisiologia , Células Matadoras Naturais/virologia , Ativação Linfocitária/imunologia , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/fisiologia , Células Tumorais Cultivadas/imunologia , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/virologia
12.
J Virol ; 74(17): 8102-10, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10933720

RESUMO

Herpesvirus saimiri (HVS) is divided into three subgroups, A, B, and C, based on sequence divergence at the left end of genomic DNA in which the saimiri transforming protein (STP) resides. Subgroup A and C strains transform primary common marmoset lymphocytes to interleukin-2-independent growth, whereas subgroup B strains do not. To investigate the nononcogenic phenotype of the subgroup B viruses, STP genes from seven subgroup B virus isolates were cloned and sequenced. Consistent with the lack of oncogenic activity of HVS subgroup B viruses, STP-B was deficient for transforming activity in rodent fibroblast cells. Sequence comparison reveals that STP-B lacks the signal-transducing modules found in STP proteins of the other subgroups, collagen repeats and an authentic SH2 binding motif. Substitution mutations demonstrated that the lack of collagen repeats but not an SH2 binding motif contributed to the nontransforming phenotype of STP-B. Introduction of the collagen repeat sequence induced oligomerization of STP-B, resulting in activation of NF-kappaB activity and deregulation of cell growth control. These results demonstrate that the collagen repeat sequence is a determinant of the degree of HVS STP transforming activity.


Assuntos
Transformação Celular Neoplásica , Colágeno/genética , Herpesvirus Saimiriíneo 2/genética , Proteínas Oncogênicas Virais/genética , Motivos de Aminoácidos , Sequência de Aminoácidos , Divisão Celular , Linhagem Celular , Colágeno/metabolismo , Herpesvirus Saimiriíneo 2/isolamento & purificação , Herpesvirus Saimiriíneo 2/metabolismo , Immunoblotting , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , NF-kappa B/metabolismo , Proteínas Oncogênicas Virais/metabolismo , Fenótipo , Sequências Repetitivas de Ácido Nucleico , Análise de Sequência de Proteína , Quinases da Família src/metabolismo
13.
J Exp Med ; 192(1): 11-21, 2000 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-10880522

RESUMO

The B cell antigen receptor (BCR) is a large complex that consists of a disulfide-linked tetramer of two transmembrane heavy (mu) chains and two light (lambda or kappa) chains in association with a heterodimer of Igalpha and Igbeta. Kaposi's sarcoma-associated herpesvirus (KSHV) encodes a transforming protein called K1, which has structural and functional similarity to Igalpha and Igbeta. We demonstrate that K1 downregulates the expression of BCR complexes on the surface. The NH(2)-terminal region of K1 specifically interacts with the mu chains of BCR complexes, and this interaction retains BCR complexes in the endoplasmic reticulum, preventing their intracellular transport to the cell surface. Thus, KSHV K1 resembles Igalpha and Igbeta in its ability to induce signaling and to interact with mu chains of the BCR. However, unlike Igalpha and Igbeta, which interact with mu chains to direct BCR complexes to the cell surface, K1 interacts with mu chains to block the intracellular transport of BCR complexes to the cell surface. These results demonstrate a unique feature of the K1 transforming protein, which may confer virus-infected cells with a long-term survival advantage.


Assuntos
Herpesvirus Humano 8/fisiologia , Proteínas de Membrana/fisiologia , Receptores de Antígenos de Linfócitos B/fisiologia , Proteínas do Envelope Viral/fisiologia , Proteínas Virais/fisiologia , Sítios de Ligação , Linhagem Celular , Regulação para Baixo , Herpesvirus Humano 8/genética , Humanos , Cadeias kappa de Imunoglobulina/química , Cadeias kappa de Imunoglobulina/fisiologia , Cadeias lambda de Imunoglobulina/química , Cadeias lambda de Imunoglobulina/fisiologia , Cadeias mu de Imunoglobulina/química , Cadeias mu de Imunoglobulina/fisiologia , Proteínas de Membrana/genética , Fases de Leitura Aberta , Receptores de Antígenos de Linfócitos B/química , Receptores de Antígenos de Linfócitos B/genética , Proteínas do Envelope Viral/genética , Proteínas Virais/genética
14.
J Virol ; 74(11): 5300-9, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10799607

RESUMO

The T-cell-mediated immune response plays a central role in the defense against intracellular pathogens. To avoid this immune response, viruses have evolved elaborate mechanisms that target and modulate many different aspects of the host's immune system. A target common to many of these viruses is the major histocompatibility complex (MHC) class I molecules. Kaposi's sarcoma-associated herpesvirus (KSHV) encodes K3 and K5 zinc finger membrane proteins which remove MHC class I molecules from the cell surface. K3 and K5 exhibit 40% amino acid identity to each other and localize primarily near the plasma membrane. While K3 and K5 dramatically downregulated class I molecules, they displayed different specificities in downregulation of HLA allotypes. K5 significantly downregulated HLA-A and -B and downregulated HLA-C only weakly, but not HLA-E, whereas K3 downregulated all four HLA allotypes. This selective downregulation of HLA allotypes by K5 was partly due to differences in amino acid sequences in their transmembrane regions. Biochemical analyses demonstrated that while K3 and K5 did not affect expression and intracellular transport of class I molecules, their expression induced rapid endocytosis of the molecules. These results demonstrate that KSHV has evolved a novel immune evasion mechanism by harboring similar but distinct genes, K3 and K5, which target MHC class I molecules in different ways.


Assuntos
Regulação para Baixo , Antígenos de Histocompatibilidade Classe I/imunologia , Proteínas Imediatamente Precoces/imunologia , Sarcoma de Kaposi/imunologia , Proteínas Virais/imunologia , Dedos de Zinco , Animais , Sítios de Ligação , Células COS , Endocitose/imunologia , Hexosaminidases , Proteínas Imediatamente Precoces/metabolismo , Frações Subcelulares , Proteínas Virais/metabolismo
15.
J Virol ; 74(6): 2721-30, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10684288

RESUMO

Rhesus monkey rhadinovirus (RRV) is a gamma-2 herpesvirus that exhibits a considerable degree of similarity to the human Kaposi's sarcoma-associated herpesvirus (KSHV). The R1 protein of RRV is distantly related to the K1 protein of KSHV, and R1, like K1, can contribute to cell growth transformation. In this study we analyzed the ability of the cytoplasmic tail of R1 to function as a signal transducer. The cytoplasmic domain of the R1 protein contains several tyrosine residues whose phosphorylation is induced in cells expressing Syk kinase. Expression of a CD8 chimera protein containing the extracellular and transmembrane domains of CD8 fused to the cytoplasmic domain of R1 mobilized intracellular calcium and induced cellular tyrosine phosphorylation in B cells upon stimulation with anti-CD8 antibody. None of the CD8-R1 cytoplasmic deletion mutants tested were able to mobilize intracellular calcium or to induce tyrosine phosphorylation to a significant extent upon addition of anti-CD8 antibody. Expression of wild-type R1 protein activated nuclear factor of activated T lymphocytes (NFAT) eightfold in B cells in the absence of antibody stimulation; expression of the CD8-R1C chimera strongly induced NFAT activity (60-fold) but only upon the addition of anti-CD8 antibody. We conclude that the cytoplasmic domain of R1 is capable of transducing signals that elicit B-lymphocyte activation events. The signal-inducing properties of R1 appear to be similar to those of K1 but differ in that the required sequences are distributed over a much longer stretch of the cytoplasmic domain (>150 amino acids). In addition, the induction of calcium mobilization was considerably longer in duration and stronger with R1 than with K1.


Assuntos
Linfócitos B/imunologia , Proteínas de Ligação a DNA/metabolismo , Glicoproteínas/metabolismo , Ativação Linfocitária , Proteínas Nucleares , Proteínas Oncogênicas Virais/metabolismo , Rhadinovirus/imunologia , Transdução de Sinais , Linfócitos T/imunologia , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Linfócitos B/metabolismo , Sítios de Ligação , Antígenos CD8/genética , Antígenos CD8/metabolismo , Células COS , Cálcio/metabolismo , Linhagem Celular , Citoplasma/metabolismo , Precursores Enzimáticos/metabolismo , Engenharia Genética , Glicoproteínas/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Macaca mulatta/virologia , Dados de Sequência Molecular , Fatores de Transcrição NFATC , Proteínas Oncogênicas Virais/genética , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Rhadinovirus/metabolismo , Quinase Syk , Linfócitos T/metabolismo
17.
J Virol ; 74(1): 436-46, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10590133

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) encodes a distinct open reading frame called K15 at a position equivalent to the gene encoding LMP2A of Epstein-Barr virus (EBV). K15 isolates from body cavity-based lymphoma (BCBL) cells exhibited a dramatic sequence variation and a complex splicing pattern. However, all K15 alleles are organized similarly with the potential SH2 and SH3 binding motifs in their cytoplasmic regions. Northern blot analysis showed that K15 was weakly expressed in latently infected BCBL-1 cells, and the level of its expression was significantly induced by tetradecanoyl phorbol acetate stimulation. K15 encoded 40- to 55-kDa proteins, as determined by sodium dodecyl sulfate-polyacrylamide gel electrophoresis, and was localized at the cytoplasm and plasma membrane. To demonstrate the signal-transducing activity of the K15 protein, we constructed a chimeric protein in which the cytoplasmic tail of the human CD8alpha polypeptide was replaced with that of KSHV K15. While the CD8-K15 chimera was not capable of eliciting cellular signal transduction upon stimulation with an anti-CD8 antibody, it significantly inhibited B-cell receptor signaling, as evidenced by a suppression of tyrosine phosphorylation and intracellular calcium mobilization. This inhibition required the putative SH2 or SH3 binding motif in the cytoplasmic region of K15. Biochemical study of CD8-K15 chimeras showed that the cytoplasmic region of K15 was constitutively tyrosine phosphorylated and that the tyrosine residue within the putative SH2 binding motif of K15 was a primary site of phosphorylation. These results demonstrate that KSHV K15 resembles LMP2A in genomic location, splicing pattern, and protein structure and by the presence of functional signal-transducing motifs in the cytoplasmic region. Thus, KSHV K15 is likely a distant evolutionary relative of EBV LMP2A.


Assuntos
Genoma Viral , Herpesvirus Humano 8/genética , Proteínas Virais/genética , Alelos , Sequência de Aminoácidos , Animais , Linhagem Celular , Quimera , Clonagem Molecular , Citoplasma/metabolismo , Imunofluorescência , Regulação Viral da Expressão Gênica , Variação Genética , Dados de Sequência Molecular , Fases de Leitura Aberta , Fosforilação , Receptores de Antígenos de Linfócitos B/metabolismo , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Tirosina/metabolismo , Proteínas Virais/química
18.
Mol Cells ; 9(4): 345-9, 1999 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-10515596

RESUMO

Gammaherpesviruses are the most rapidly growing members of the herpesviridae family. Gamma herpesviruses share similarity in their genome organizations and in early and late lytic genes that are required for viral replication. A distinct characteristic of gamma herpesviruses is their ability to establish latent infection in lymphoid cells, and some of these viruses are closely associated with abnormal proliferation and cancer in primates. The first open reading frame of the primate gamma herpesviruses has been shown to directly contribute to virus-associated pathogenesis. This open reading frame encodes latent membrane protein-1 (LMP1) in Epstein-Barr virus, Saimiri transformation protein (STP) in Herpesvirus Saimiri, K1 in Kaposi's sarcoma-associated herpesvirus, and R1 in Rhesus monkey Rhadinovirus. All of these gene products are capable of eliciting cellular signal transduction events, resulting in cell growth transformation. This review briefly summarizes the current view on the transforming mechanisms utilized by primate herpesviral oncogenes.


Assuntos
Gammaherpesvirinae/genética , Proteínas Oncogênicas Virais/genética , Animais , Transformação Celular Neoplásica , Herpesvirus Humano 8/genética , Macaca mulatta/virologia , Modelos Genéticos , Rhadinovirus/genética , Transdução de Sinais , Proteínas da Matriz Viral/genética , Proteínas Virais/genética
19.
Structure ; 7(3): 245-54, 1999 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-10368294

RESUMO

BACKGROUND: Cyclin-dependent kinases (CDKs) have a central role in cell-cycle control and are activated by complex formation with positive regulatory proteins called cyclins and by phosphorylation. The overexpression and mutation of cyclins and CDKs has been associated with tumorigenesis and oncogenesis. A virus-encoded cyclin (v-cyclin) from herpesvirus saimiri has been shown to exhibit highest sequence homology to type D cyclins and specifically activates CDK6 of host cells to a very high degree. RESULTS: We have determined the first X-ray structure of a v-cyclin to 3.0 A resolution. The structure of the core domains is very similar to those of cyclin A and cyclin H from human cells. To understand the structural basis for the v-cyclin specificity for CDK6 and the insensitivity of the complex to inhibitors of the p21 and INK4 families, a v-cyclin-CDK2 model was built on the basis of the known structures of human cyclin A in complex with CDK2 and the CDK inhibitor p27(Kip1). CONCLUSIONS: Although many critical interactions between cyclin A and CDK2 would be conserved in a v-cyclin-CDK2 complex, some appear sterically or electrostatically unfavorable due to shifts in the backbone conformation or sidechain differences and may contribute to v-cyclin selectivity for CDK6. The insensitivity of v-cyclin-CDK6 complexes to inhibitors of the p21 family is probably due to structural changes in v-cyclin that lead to a flatter surface area offering fewer potential contacts with the protein inhibitor. In addition, sequence changes in v-cyclin eliminate hydrogen-bonding partners for atoms of the p27(Kip1) inhibitor. This structure provides the first model for interactions between v-cyclins and host cell-cycle proteins; these interactions may be important for virus survival as well as oncogenic transformation of host cells.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Proteínas de Ciclo Celular , Ciclinas/química , Herpesvirus Saimiriíneo 2/química , Conformação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas , Proteínas Supressoras de Tumor , Proteínas Virais/química , Sequência de Aminoácidos , Ciclo Celular , Cristalografia por Raios X , Ciclina A/química , Ciclina H , Quinase 2 Dependente de Ciclina , Quinase 4 Dependente de Ciclina , Quinase 6 Dependente de Ciclina , Inibidor p16 de Quinase Dependente de Ciclina/química , Inibidor p16 de Quinase Dependente de Ciclina/farmacologia , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/química , Ciclinas/genética , Ciclinas/fisiologia , Ativação Enzimática , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Substâncias Macromoleculares , Proteínas Associadas aos Microtúbulos/química , Proteínas Associadas aos Microtúbulos/farmacologia , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/química , Proteínas Recombinantes de Fusão/química , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
20.
Semin Cancer Biol ; 9(3): 231-9, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10343074

RESUMO

Herpesvirus saimiri (HVS) causes T-lymphoproliferative dis-$borders in several New World and Old World primate species and in certain rabbits.In vitro infection leads to permanent growth of primary T cells of primate and human origins. The transformation-relevant proteins of HVS interact with cellular proto-oncoproteins which results in cell growth transformation. In addition, virus-encoded cellular homologues may contribute to transformation or persistence of HVS by altering cellular signal transduction and deregulating cell growth control. Because of the presence of a permissive cell culture system and in vitro Land in vivo transformation assays, HVS provides a unique opportunity to investigate the mechanisms of cancer induction by oncogenic herpesviruses.


Assuntos
Herpesvirus Saimiriíneo 2/genética , Neoplasias/etiologia , Animais , Transformação Celular Neoplásica , Modelos Animais de Doenças , Humanos , Ativação Linfocitária , Oncogenes , Fosfoproteínas/fisiologia , Primatas , Coelhos , Proteínas Virais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...