Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Exp Med ; 186(1): 139-46, 1997 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-9207008

RESUMO

Ligation of CCR5 by the CC chemokines RANTES, MIP-1alpha or MIP-1beta, and of CXCR4 by the CXC chemokine SDF-1alpha, profoundly inhibits the replication of HIV strains that use these coreceptors for entry into CD4(+) T lymphocytes. The mechanism of entry inhibition is not known. We found a rapid and extensive downregulation of CXCR4 by SDF-1alpha and of CCR5 by RANTES or the antagonist RANTES(9-68). Confocal laser scanning microscopy showed that CCR5 and CXCR4, after binding to their ligands, are internalized into vesicles that qualify as early endosomes as indicated by colocalization with transferrin receptors. Internalization was not affected by treatment with Bordetella pertussis toxin, showing that it is independent of signaling via Gi-proteins. Removal of SDF-1alpha led to rapid, but incomplete surface reexpression of CXCR4, a process that was not inhibited by cycloheximide, suggesting that the coreceptor is recycling from the internalization pool. Deletion of the COOH-terminal, cytoplasmic domain of CXCR4 did not affect HIV entry, but prevented SDF-1alpha-induced receptor downregulation and decreased the potency of SDF-1alpha as inhibitor of HIV replication. Our results indicate that the ability of the coreceptor to internalize is not required for HIV entry, but contributes to the HIV suppressive effect of CXC and CC chemokines.


Assuntos
Quimiocinas CXC , Citocinas/fisiologia , Infecções por HIV/virologia , HIV-1/fisiologia , Proteínas de Membrana/fisiologia , Receptores de HIV/fisiologia , Replicação Viral , Animais , Células CHO , Quimiocina CXCL12 , Cricetinae , Regulação para Baixo , Citometria de Fluxo , Células HeLa , Humanos , Receptores CXCR4
2.
J Exp Med ; 188(5): 855-66, 1998 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-9730887

RESUMO

Human cytomegalovirus (HCMV), a betaherpesvirus, has developed several ways to evade the immune system, notably downregulation of cell surface expression of major histocompatibility complex class I heavy chains. Here we report that HCMV has devised another means to compromise immune surveillance mechanisms. Extracellular accumulation of both constitutively produced monocyte chemoattractant protein (MCP)-1 and tumor necrosis factor-superinduced RANTES (regulated on activation, normal T cell expressed and secreted) was downregulated in HCMV-infected fibroblasts in the absence of transcriptional repression or the expression of polyadenylated RNA for the cellular chemokine receptors CCR-1, CCR-3, and CCR-5. Competitive binding experiments demonstrated that HCMV-infected cells bind RANTES, MCP-1, macrophage inflammatory protein (MIP)-1beta, and MCP-3, but not MCP-2, to the same receptor as does MIP-1alpha, which is not expressed in uninfected cells. HCMV encodes three proteins with homology to CC chemokine receptors: US27, US28, and UL33. Cells infected with HCMV mutants deleted of US28, or both US27 and US28 genes, failed to downregulate extracellular accumulation of either RANTES or MCP-1. In contrast, cells infected with a mutant deleted of US27 continues to bind and downregulate those chemokines. Depletion of chemokines from the culture medium was at least partially due to continuous internalization of extracellular chemokine, since exogenously added, biotinylated RANTES accumulated in HCMV-infected cells. Thus, HCMV can modify the chemokine environment of infected cells through intense sequestering of CC chemokines, mediated principally by expression of the US28-encoded chemokine receptor.


Assuntos
Quimiocinas CC/metabolismo , Citomegalovirus/fisiologia , Receptores de Quimiocinas/metabolismo , Receptores Virais/metabolismo , Replicação Viral/imunologia , Sítios de Ligação , Células Cultivadas , Quimiocina CCL2/biossíntese , Quimiocina CCL2/metabolismo , Quimiocina CCL5/biossíntese , Quimiocina CCL5/metabolismo , Quimiocinas CC/genética , Meios de Cultura/metabolismo , Citomegalovirus/genética , Fibroblastos/metabolismo , Fibroblastos/virologia , Deleção de Genes , Humanos , Líquido Intracelular/metabolismo , Receptores CCR2 , Receptores de Quimiocinas/genética , Fator de Necrose Tumoral alfa/farmacologia
3.
J Exp Med ; 192(4): 495-506, 2000 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-10952719

RESUMO

Leukocyte recruitment to target tissue is initiated by weak rolling attachments to vessel wall ligands followed by firm integrin-dependent arrest triggered by endothelial chemokines. We show here that immobilized chemokines can augment not only arrest but also earlier integrin-mediated capture (tethering) of lymphocytes on inflamed endothelium. Furthermore, when presented in juxtaposition to vascular cell adhesion molecule 1 (VCAM-1), the endothelial ligand for the integrin very late antigen 4 (VLA-4, alpha4beta1), chemokines rapidly augment reversible lymphocyte tethering and rolling adhesions on VCAM-1. Chemokines potentiate VLA-4 tethering within <0.1 s of contact through Gi protein signaling, the fastest inside-out integrin signaling events reported to date. Although VLA-4 affinity is not altered upon chemokine signaling, subsecond VLA-4 clustering at the leukocyte-substrate contact zone results in enhanced leukocyte avidity to VCAM-1. Endothelial chemokines thus regulate all steps in adhesive cascades that control leukocyte recruitment at specific vascular beds.


Assuntos
Adesão Celular/fisiologia , Endotélio Vascular/metabolismo , Integrinas/metabolismo , Leucócitos/metabolismo , Receptores de Retorno de Linfócitos/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo , Anticorpos Monoclonais , Movimento Celular/fisiologia , Células Cultivadas , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Endotélio Vascular/citologia , Citometria de Fluxo , Humanos , Integrina alfa4beta1 , Microscopia Confocal , Microscopia de Vídeo , Transdução de Sinais , Linfócitos T/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Molécula 1 de Adesão de Célula Vascular/genética
4.
Curr Biol ; 7(9): 652-60, 1997 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9285716

RESUMO

BACKGROUND: Chemokines bind to specific receptors and mediate leukocyte migration to sites of inflammation. Recently, some chemokine receptors, notably CXCR4 and CCR5, have been shown to be essential fusion factors on target cells for infection by human immunodeficiency virus (HIV); the chemokines bound by these receptors have also been shown to act as potent inhibitors of HIV infection. Here, we describe the isolation of a novel, putative chemokine receptor. RESULTS: We have isolated the cDNA for a putative human chemokine receptor, which we have termed TYMSTR (T-lymphocyte-expressed seven-transmembrane domain receptor). The TYMSTR gene is localized to human chromosome 3 and encodes a protein that has a high level of identity with chemokine receptors. TYMSTR mRNA was selectively expressed in interleukin-2-stimulated T lymphocytes but not in freshly isolated lymphocytes and leukocytes or related cell lines. The natural ligand for TYMSTR was not identified among 32 human chemokines and other potential ligands. Cells co-expressing TYMSTR and human CD4 fused with cells expressing envelope glycoproteins of macrophage (M)-tropic HIV-1 as well as T-cell line (T)-tropic HIV-1 isolates. Addition of infectious, T-tropic HIV-1 particles to TYMSTR/CD4-expressing cells resulted in viral entry and proviral DNA formation. CONCLUSIONS: Our findings demonstrate that TYMSTR, in combination with CD4, mediates HIV-1 fusion and entry. The high-level expression of TYMSTR in CD4(+) T lymphocytes and the selectivity of this receptor for T-tropic and M-tropic HIV-1 strains indicates that TYMSTR might function as HIV coreceptor at both early and late stages of infection.


Assuntos
HIV-1 , Receptores de Quimiocinas/biossíntese , Receptores de HIV/biossíntese , Linfócitos T/metabolismo , Sequência de Aminoácidos , Mapeamento Cromossômico , Clonagem Molecular , DNA Complementar/química , Humanos , Ligantes , Ativação Linfocitária , Dados de Sequência Molecular , Fases de Leitura Aberta , Reação em Cadeia da Polimerase , Receptores CCR1 , Receptores de Quimiocinas/química , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo , Receptores de HIV/genética , Receptores de Interleucina/química , Receptores de Interleucina/metabolismo , Receptores de Interleucina-8B , Alinhamento de Sequência , Linfócitos T/virologia
5.
Curr Biol ; 10(6): 325-8, 2000 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-10744978

RESUMO

HIV particles that use the chemokine receptor CXCR4 as a coreceptor for entry into cells (X4-HIV) inefficiently transmit infection across mucosal surfaces [1], despite their presence in seminal fluid and mucosal secretions from infected individuals [2] [3] [4]. In addition, although intestinal lymphocytes are susceptible to infection with either X4-HIV particles or particles that use the chemokine receptor CCR5 for viral entry (R5-HIV) during ex vivo culture [5], only systemic inoculation of R5-chimeric simian-HIV (S-HIV) results in a rapid loss of CD4(+) intestinal lymphocytes in macaques [6]. The mechanisms underlying the inefficient capacity of X4-HIV to transmit infection across mucosal surfaces and to infect intestinal lymphocytes in vivo have remained elusive. The CCR5 ligands RANTES, MIP-1alpha and MIP-1beta suppress infection by R5-HIV-1 particles via induction of CCR5 internalization, and individuals whose peripheral blood lymphocytes produce high levels of these chemokines are relatively resistant to infection [7] [8] [9]. Here, we show that the CXCR4 ligand stromal derived factor-1 (SDF-1) is constitutively expressed by mucosal epithelial cells at sites of HIV transmission and propagation. Furthermore, CXCR4 is selectively downmodulated on intestinal lymphocytes within the setting of prominent SDF-1 expression. We postulate that mucosally derived SDF-1 continuously downmodulates CXCR4 on resident HIV target cells, thereby reducing the transmission and propagation of X4-HIV at mucosal sites. Moreover, such a mechanism could contribute to the delayed emergence of X4 isolates, which predominantly occurs during the later stages of the HIV infection.


Assuntos
Quimiocinas CXC/fisiologia , HIV/crescimento & desenvolvimento , Mucosa Intestinal/metabolismo , Quimiocina CXCL12 , Quimiocinas CXC/biossíntese , Humanos , Receptores CCR5/metabolismo , Receptores CXCR4/biossíntese , Receptores CXCR4/genética
6.
J Clin Invest ; 77(1): 326-9, 1986 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-2418061

RESUMO

We have investigated the secretory function of cell suspensions from bone eosinophilic granulomas surgically collected in two patients with histiocytosis X. Unseparated cell preparations spontaneously produced interleukin 1 (IL-1) and prostaglandin E2 (PGE2). In order to ascertain that this secretion was due to the characteristic Langerhans cell-like histiocytosis X cells predominantly found in the bone lesions, we have purified T6+ cells by the use of a fluorescence-activated cell sorter. Such highly purified cell preparations were found to secrete IL-1 and PGE2 spontaneously in culture. Stimulation with endotoxins and treatment with interferon gamma (IFN gamma) revealed an intense IL-1 secretory function of histiocytosis X cells. Since both IL-1 and PGE2 are able to induce bone resorption in vitro, our findings are compatible with the hypothesis that histiocytosis X cells are responsible for the typical osteolytic lesion observed in histiocytosis X through the local secretion of these two mediators.


Assuntos
Doenças Ósseas/imunologia , Granuloma Eosinófilo/imunologia , Interleucina-1/biossíntese , Prostaglandinas E/biossíntese , Linfócitos T/metabolismo , Antígenos de Diferenciação de Linfócitos T , Antígenos de Superfície/análise , Antígenos de Superfície/imunologia , Doenças Ósseas/metabolismo , Separação Celular , Células Cultivadas , Criança , Pré-Escolar , Dinoprostona , Granuloma Eosinófilo/metabolismo , Citometria de Fluxo , Humanos , Interferons/metabolismo , Interleucina-2/metabolismo , Masculino , Crânio
7.
J Clin Invest ; 99(2): 257-66, 1997 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9005994

RESUMO

The study of the pathology of HIV-1 infection in chimpanzees supports the idea of the crucial role of HIV-infected monocytes in the pathogenesis of AIDS, although viral mechanisms that lead to T cell dysfunction and deletion during HIV infection are still unclear. We show here that HIV-1-infected antigen-presenting monocytes (APCs) are able to prime in vitro non-HIV-infected antigen-specific CD4+ T cell lines or peripheral blood CD4+ T cells to undergo apoptosis after antigen-specific restimulation. The priming of T cells for apoptosis occurs in the absence of HIV replication in the T cells. Priming for apoptosis required two concomitant signals present on the same APC, an antigenic stimulus and a second signal provided by the HIV gp120 protein as demonstrated by the use as APCs of EBV-LCLs infected with different recombinant deleted proviruses or transfected with different HIV proteins. These results provide a mechanism for the priming for apoptosis of T cells in HIV-infected patients, implicating a role for HIV-infected APCs in the induction of T cell dysfunction and depletion in AIDS.


Assuntos
Apoptose , Linfócitos T CD4-Positivos/imunologia , HIV-1/imunologia , Monócitos/imunologia , Monócitos/virologia , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/virologia , Genes env , Genes tat , Proteína gp120 do Envelope de HIV/imunologia , HIV-1/genética , Humanos , Ativação Linfocitária , Provírus/genética , Provírus/imunologia , Transcrição Gênica
8.
J Clin Invest ; 104(9): 1199-211, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10545519

RESUMO

The chemokine SDF-1 plays a central role in the repopulation of the bone marrow (BM) by circulating CD34(+) progenitors, but the mechanisms of its action remain obscure. To extravasate to target tissue, a blood-borne cell must arrest firmly on vascular endothelium. Murine hematopoietic progenitors were recently shown in vivo to roll along BM microvessels that display selectins and integrins. We now show that SDF-1 is constitutively expressed by human BM endothelium. In vitro, human CD34(+) cells establish efficient rolling on P-selectin, E-selectin, and the CD44 ligand hyaluronic acid under physiological shear flow. ICAM-1 alone did not tether CD34(+) cells under flow, but, in the presence of surface-bound SDF-1, CD34(+) progenitors rolling on endothelial selectin rapidly developed firm adhesion to the endothelial surface, mediated by an interaction between ICAM-1 and its integrin ligand, which coimmobilized with SDF-1. Human CD34(+) cells accumulated efficiently on TNF-activated human umbilical cord endothelial cells in the absence of SDF-1, but they required immobilized SDF-1 to develop firm integrin-mediated adhesion and spreading. In the absence of selectins, SDF-1 also promoted VLA-4-mediated, Gi protein-dependent tethering and firm adhesion to VCAM-1 under shear flow. To our knowledge, this is the first demonstration that SDF-1 expressed on vascular endothelium is crucial for translating rolling adhesion of CD34(+) progenitors into firm adhesion by increasing the adhesiveness of the integrins VLA-4 and LFA-1 to their respective endothelial ligands, VCAM-1 and ICAM-1.


Assuntos
Antígenos CD34/metabolismo , Medula Óssea/metabolismo , Quimiocinas CXC/fisiologia , Endotélio Vascular/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Integrinas/metabolismo , Adesão Celular , Quimiocina CXCL12 , Selectina E/metabolismo , Sangue Fetal/metabolismo , Citometria de Fluxo , Humanos , Receptores de Hialuronatos/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Selectina-P/metabolismo , Estresse Mecânico , Linfócitos T/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo
9.
J Clin Invest ; 106(11): 1331-9, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11104786

RESUMO

The chemokine stromal-derived factor-1 (SDF-1) controls many aspects of stem cell function. Details of its regulation and sites of production are currently unknown. We report that in the bone marrow, SDF-1 is produced mainly by immature osteoblasts and endothelial cells. Conditioning with DNA-damaging agents (ionizing irradiation, cyclophosphamide, and 5-fluorouracil) caused an increase in SDF-1 expression and in CXCR4-dependent homing and repopulation by human stem cells transplanted into NOD/SCID mice. Our findings suggest that immature osteoblasts and endothelial cells control stem cell homing, retention, and repopulation by secreting SDF-1, which also participates in host defense responses to DNA damage.


Assuntos
Quimiocinas CXC/genética , Dano ao DNA , Células-Tronco/metabolismo , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Linhagem Celular , Células Cultivadas , Quimiocina CXCL12 , Ciclofosfamida/farmacologia , Relação Dose-Resposta à Radiação , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Citometria de Fluxo , Fluoruracila/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos SCID , Osteoblastos/citologia , Osteoblastos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células-Tronco/citologia , Células Tumorais Cultivadas
10.
Mol Cell Biol ; 15(5): 2413-9, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7739525

RESUMO

After exposure of cells to tumor necrosis factor (TNF), I kappa B alpha is rapidly degraded by a proteolytic activity that is required for nuclear localization and activation of transcription factor NF-kappa B. To investigate this problem, we have developed a cell-free system to study the degradation of I kappa B alpha initiated in vivo. In this in vitro system, characteristics of endogenous I kappa B alpha degradation were comparable to those observed in vivo. Recombinant I kappa B alpha, when added to lysates from cells exposed to TNF, was specifically degraded by a cellular proteolytic activity; however, it was stable in extracts from unstimulated cells. Inhibition characteristics of the proteolytic activity responsible for I kappa B alpha degradation suggest the involvement of a serine protease. Analysis of mutated forms of I kappa B alpha in the in vitro system demonstrated that an I kappa B alpha species which was unable to interact with NF-kappa B was still efficiently degraded. In contrast, deletion of the C-terminal 61 amino acids from I kappa B alpha rendered the protein resistant to proteolytic degradation. Expression of I kappa B alpha mutated forms in COS-7 cells confirmed the importance of the C-terminal domain for the degradation of the protein in vivo following cell activation. Thus, it is likely that the acidic, negatively charged region represented by the C-terminal 61 amino acids of the protein contains residues critical for TNF-inducible degradation of I kappa B alpha.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas I-kappa B , Sequência de Aminoácidos , Animais , Linhagem Celular , Sistema Livre de Células , Proteínas de Ligação a DNA/genética , Células HeLa , Humanos , Dados de Sequência Molecular , Mutação , Inibidor de NF-kappaB alfa , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Inibidores de Serina Proteinase/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
11.
Mol Cell Biol ; 15(5): 2689-96, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7739549

RESUMO

The transcription factor NF-kappa B is exploited by many viruses, including the human immunodeficiency virus, for expression of viral genes, but its primary role appears to be in the rapid induction of cellular genes during immune and inflammatory responses. The inhibitor protein I kappa B alpha maintains NF-kappa B in an inactive form in the cytoplasms of unstimulated cells, but upon cell activation, I kappa B alpha is rapidly degraded, leading to nuclear translocation of free NF-kappa B. However, NF-kappa B-dependent transcription of the I kappa B alpha gene leads to rapid resynthesis of the I kappa B alpha protein and inhibition of NF-kappa B-dependent transcription. Here we demonstrate a new regulatory function of I kappa B alpha exerted on NF-kappa B in the nuclear compartment. Although normally found in the cytoplasm, I kappa B alpha, newly synthesized in response to tumor necrosis factor or interleukin I, is transported to the nucleus. In the nucleus I kappa B alpha associates with the p50 and p65 subunits of NF-kappa B, inhibiting DNA binding of the transcription factor. Furthermore, nuclear expression of I kappa B alpha correlates with transcription termination of transfected NF-kappa B-dependent luciferase genes. Following the appearance of I kappa B alpha in the nuclei of activated cells, a dramatic reduction in the amount of nuclear p50 occurs, suggesting that NF-kappa B-I kappa B alpha complexes are cleared from the nucleus.


Assuntos
Proteínas de Ligação a DNA/biossíntese , DNA/metabolismo , Proteínas I-kappa B , NF-kappa B/metabolismo , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/genética , Células HeLa , Humanos , Interleucina-1/farmacologia , Inibidor de NF-kappaB alfa , NF-kappa B/antagonistas & inibidores , Transcrição Gênica , Fator de Necrose Tumoral alfa/farmacologia
12.
Mol Cell Biol ; 13(2): 1290-5, 1993 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8423794

RESUMO

Nuclear factor kappa B (NF-kappa B) plays a critical role in the regulation of a large variety of cellular genes. However, the mechanism whereby this nuclear factor is activated remains to be determined. In this report, we present evidence that in oocytes from Xenopus laevis, (i) ras p21- and phospholipase C (PLC)-mediated phosphatidylcholine (PC) hydrolysis activates NF-kappa B and (ii) protein kinase C zeta subspecies is involved in the activation of NF-kappa B in response to insulin/ras p21/PC-PLC. Thus, the microinjection of either ras p21 or PC-PLC, or the exposure of oocytes to insulin, promotes a significant translocation to the nucleus of an NF-kappa B-like activity. This effect is not observed when oocytes are incubated with phorbol myristate acetate or progesterone, both of which utilize a ras p21-independent pathway for oocyte activation. These data strongly suggest a critical role of the insulin/ras p21/PC-PLC/protein kinase C zeta pathway in the control of NF-kappa B activation.


Assuntos
NF-kappa B/metabolismo , Proteína Quinase C/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Células Cultivadas , Citosol/metabolismo , Elementos Facilitadores Genéticos , HIV-1/genética , Insulina/farmacologia , Cinética , Dados de Sequência Molecular , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/farmacologia , Ativação Transcricional , Xenopus laevis
13.
Oncogene ; 4(11): 1359-62, 1989 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-2682462

RESUMO

Transient transfection of ras expression vectors into human fibroblasts and astrocytes has been used to test the hypothesis that p21 ras, a known membrane signal transductor, may participate in pathways linking cellular activation and human immunodeficiency virus (HIV) reactivation. Expression vectors carrying the chloramphenicol acetyl transferase coding sequence under the control of various fragments of the long terminal repeat (LTR) of HIV were co-transfected with expression vectors of the mutated (val 12) c-Ha-ras gene or of its normal counterpart. Both forms of the ras gene induced transactivation of the HIV-LTR via the two direct repeat sequences which constitute the HIV enhancer. This repeat sequence was shown to be sufficient for ras-induced LTR transactivation. Other LTR sequences tested were not found to be responsive to co-transfected ras expression vectors. Deletion of the TAR sequence impaired the response to tat, but not to ras co-transfection. The mutated ras gene was more efficient than the proto-oncogene in activating the HIV enhancer. Transfection of ras was shown to enhance transcription of a complete provirus DNA clone of HIV-1. Such findings may shed new light on the mechanisms through which cell membrane activation signals result in HIV reactivation.


Assuntos
Astrócitos/metabolismo , Elementos Facilitadores Genéticos , Genes ras , HIV/genética , Transcrição Gênica , Transfecção , Astrocitoma , Sequência de Bases , Linhagem Celular , Células Cultivadas , Fibroblastos , Vetores Genéticos , Humanos , Dados de Sequência Molecular , Mutação , Sondas de Oligonucleotídeos/síntese química , Proto-Oncogene Mas , Transdução de Sinais , Ativação Transcricional
14.
Oncogene ; 15(15): 1841-50, 1997 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-9362451

RESUMO

The Rel/NF-kappaB family of transcription factors controls the expression of a wide variety of genes that are implicated in immune and inflammatory responses and cellular proliferation. Disregulation of NF-kappaB is associated with cellular transformation and the maintenance of a high anti-apoptotic threshold in transformed cells. NF-kappaB activity is in turn regulated by its sequestration in the cytoplasm by the inhibitor I kappaB. I kappaB alpha, the most abundant and well-characterized member of the I kappaB multiprotein family, is rapidly degraded in response to multiple physiologic stimuli. In the present study we show that not only the amino-terminus, but also the carboxy-terminus of I kappaB alpha contain transferable signals that must be simultaneously present in an unrelated protein to render it susceptible to activation-induced, proteasome-mediated degradation. We show here that I kappaB alpha amino-terminal modifications occur independently of the carboxy-terminus. Moreover, we present evidence indicating a critical role for the carboxy-terminal region in facilitating proteolysis by the catalytic core of the proteasome. When incubated with 20S proteasome extracted from rat liver, I kappaB alpha was quickly degraded while a deletion mutant lacking the carboxy-terminus was resistant to proteolysis. Likewise, chimeric proteins of beta-galactosidase with the I kappaB alpha carboxy-terminus were degraded in vitro independently of the presence of the I kappaB alpha amino-terminus, whereas chimeric proteins lacking the I kappaB alpha carboxy-terminus were stable. Our results identify the carboxy-terminus of I kappaB alpha as a domain critical for degradation through interaction with an as yet unidentified component of the proteasome.


Assuntos
Cisteína Endopeptidases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas I-kappa B , Complexos Multienzimáticos/metabolismo , Animais , Células COS , Catálise , Linhagem Celular , Proteínas de Ligação a DNA/química , Células HeLa , Humanos , Hidrólise , Inibidor de NF-kappaB alfa , Fosforilação , Complexo de Endopeptidases do Proteassoma , Ratos , Fator de Necrose Tumoral alfa/farmacologia , Ubiquitinas/metabolismo
15.
Oncogene ; 12(11): 2425-35, 1996 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-8649784

RESUMO

Activation of transcription factor NF-kappaB involves signal-induced degradation of the protein inhibitor IkappaB-alpha and release of NF-kappaB which translocates to the nucleus where it influences transcription of responsive genes. Although multiple regions of IkappaB-alpha are involved in this process, the N-terminal region of the protein has been identified as a regulatory region that is required for signal induced phosphorylation and degradation. The sensitivity of IkappaB-alpha degradation to peptide aldehydes which inhibit components of the proteasome and the detection of ubiquitinated forms of IkappaB-alpha indicate that IkappaB-alpha is degraded by the ubiquitin-proteasome pathway. To identify lysine residues that represent the sites of ubiquitin addition, a series of lysine to arginine mutations were introduced into IkappaB-alpha and the mutant proteins tested for their ability to function in vivo. Exposure of COS7 cells, cotransfected with IkappaB-alpha and a TNF-responsive NF-kappaB reporter gene, resulted in stimulation of reporter activity as a consequence of IkappaB-alpha degradation. In contrast, this effect was drastically reduced when an IkappaKB-alpha mutant carrying serine to alanine changes at amino-acids, 32 and 36, which blocks both signal-induced phosphorylation and ubiquitin conjugation of the protein, was co-transfected with the reporter gene. Likewise, a mutant form of IkappaB-alpha containing lysine to arginine changes at positions 21 and 22 (K21R, K22R) severely reduces TNF-induced activation of the NF-kappaB-dependent reporter gene. Examination of the metabolism of mutant IkappaB-alpha molecules reveals that, while the K21R, K22R mutant inhibits the DNA-binding activity of NF-kappaB and undergoes signal induced phosphorylation, it is neither ubiquitinated nor degraded in response to TNF. Thus, it is likely that after signal-induced phosphorylation Of IkappaB-alpha on serine residues 32 and 36, lysine residues 21 and 22 are major sites of ubiquitin ligation which target the protein for rapid degradation by the proteasome.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas I-kappa B , Lisina/metabolismo , NF-kappa B/metabolismo , Transcrição Gênica , Ubiquitinas/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Éteres Cíclicos/farmacologia , Genes Reporter , Interleucina-1/farmacologia , Lisina/química , Lisina/genética , Dados de Sequência Molecular , Mutagênese , Inibidor de NF-kappaB alfa , NF-kappa B/antagonistas & inibidores , Ácido Okadáico , Fosforilação , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA , Fator de Necrose Tumoral alfa/farmacologia
16.
Chem Biol ; 6(10): 689-98, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10508680

RESUMO

BACKGROUND: The fungal epipolythiodioxopiperazine metabolite gliotoxin has a variety of toxic effects such as suppression of antigen processing, induction of macrophagocytic apoptosis and inhibition of transcription factor NF-kappaB activation. How gliotoxin acts remains poorly understood except that the molecule's characteristic disulfide bridge is important for immunomodulation. As this fungal metabolite stabilizes the NF-kappaB inhibitor IkappaBalpha in the cytoplasm, we decided to investigate its molecular mechanism of action. RESULTS: We show that gliotoxin is an efficient, noncompetitive inhibitor of the chymotrypsin-like activity of the 20S proteasome in vitro. Proteasome inhibition can be reversed by dithiothreitol, which reduces gliotoxin to the dithiol compound. In intact cells, gliotoxin inhibits NF-kappaB induction through inhibition of proteasome-mediated degradation of IkappaBalpha. CONCLUSIONS: Gliotoxin targets catalytic activities of the proteasome efficiently. Inhibition by gliotoxin may be countered by reducing agents, which are able to inactivate the disulfide bridge responsible for the inhibitory capacity of gliotoxin.


Assuntos
Cisteína Endopeptidases/metabolismo , Gliotoxina/metabolismo , Proteínas I-kappa B , Complexos Multienzimáticos/metabolismo , Células Cultivadas , Quimotripsina/metabolismo , Cumarínicos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células HeLa , Humanos , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , Oligopeptídeos/metabolismo , Complexo de Endopeptidases do Proteassoma , Fator de Necrose Tumoral alfa/farmacologia
17.
FEBS Lett ; 426(2): 271-8, 1998 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-9599023

RESUMO

CXCR4 is the receptor for the CXC chemokine SDF1 that has essential functions on embryo organogenesis, immunological functions and T lymphocyte trafficking. Recently, CXCR4 has drawn unexpected attention as it was recently identified as a co-factor required for entry of lymphotropic HIV isolates in CD4+ T lymphocytes. CXCR4 is the only SDF1 receptor identified so far. This suggests that CXCR4 expression is critical for the biological effects of SDF1. To investigate the mechanisms controlling both the constitutive and induced expression of CXCR4 receptors we have isolated and characterized the promoter region and determined the genomic structure of the human gene. The CXCR4 gene contains two exons separated by an intronic sequence. A 2.6 kb 5'-flanking region located upstream the CXCR4 open reading frame contains a TATA box and the transcription start site characteristic of a functional promoter. This region also contains putative consensus binding sequences for different transcription factors, some of them associated with the hemopoiesis and lymphocyte development.


Assuntos
Receptores CXCR4/genética , Sequência de Bases , Éxons , Expressão Gênica/efeitos dos fármacos , Genes , Células HeLa , Humanos , Íntrons , Ionomicina/farmacologia , Dados de Sequência Molecular , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Acetato de Tetradecanoilforbol/farmacologia , Transcrição Gênica , Transfecção
18.
Pediatrics ; 76(2): 263-8, 1985 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-3839578

RESUMO

Familial erythrophagocytic lymphohistiocytosis, a rare disorder affecting infants, is characterized by a visceral infiltration of histiocytes and lymphocytes resulting in rapid death. It has recently been reported that use of epipodophyllotoxin, VP 16-213, could induce a complete remission of the disease. Such treatment does not, however, prevent fatal CNS relapse. Four patients with the characteristic features of the disease--fever, hepatosplenomegaly, pancytopenia, low plasmatic fibrinogen level, hyperlipidemia, and histiocytic meningitis--are described. These patients were treated with a combination therapy including systemic administration of VP 16-213, steroids, and intrathecal methotrexate followed by cranial irradiation after the age of 12 months. The four patients achieved complete remission of the disease after clearing of the CNS localization. Two patients had secondary relapses, but all four patients have had a disease-free survival exceeding 12 months. All patients have been in remission of the disease for 27, 20, 16, and 13 months, respectively, after disease onset without major setbacks from the treatment. This combination therapy appears to be a promising approach toward long-term remission of the disease.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Eritrócitos , Doenças Linfáticas/tratamento farmacológico , Fagocitose , Terapia Combinada , Eritrócitos/efeitos dos fármacos , Etoposídeo/administração & dosagem , Feminino , Humanos , Lactente , Injeções Espinhais , Doenças Linfáticas/genética , Doenças Linfáticas/radioterapia , Masculino , Metotrexato/administração & dosagem , Metilprednisolona/administração & dosagem , Fagocitose/efeitos dos fármacos , Prednisona/administração & dosagem
19.
AIDS Res Hum Retroviruses ; 8(5): 545-51, 1992 May.
Artigo em Inglês | MEDLINE | ID: mdl-1355346

RESUMO

While recent studies in Rhesus monkeys have pointed out the importance of an intact nef gene for the development of acquired immunodeficiency syndrome (AIDS), no biological function has been so far unambiguously attributed to its product. Since Nef has been described to possess GTP-binding properties and to down-regulate CD4 cell surface expression, we looked for evidences of Nef interfering with the transduction of activating signals in human CD4+ T cells. We used a murine leukemia retroviral vector to express the HIV-1BRU nef gene in two permanent tumoral T-cell lines (CEM and Jurkat) and in two nonimmortalized, interleukin-2 (IL2)-dependent, T-cell clones. The single copy recombinant provirus integrated in the genome of these cells directed the synthesis of a 27-kD protein with a half-life greater than 5 h. The levels of expression of cell surface molecules involved in T-cell functions (CD4, CD3, CD28, CD29, IL-2 receptor) were not modified in cell populations expressing Nef. In immunocompetent T-cell clones, cell proliferation and lymphokine production in response to activating stimuli (IL-2, alloantigens, phorbol esters, or antibodies directed against CD2, CD3, CD4, CD28) remained unmodified. Moreover, the presence of Nef did not change the kinetics of human immunodeficiency virus (HIV) infection.


Assuntos
Linfócitos T CD4-Positivos/microbiologia , Produtos do Gene nef/fisiologia , HIV-1/fisiologia , Ativação Linfocitária , Células 3T3 , Animais , Antígenos de Superfície/biossíntese , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular , Produtos do Gene nef/genética , Genes nef , Humanos , Camundongos , Transfecção , Replicação Viral , Produtos do Gene nef do Vírus da Imunodeficiência Humana
20.
AIDS Res Hum Retroviruses ; 5(2): 217-24, 1989 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-2540782

RESUMO

Susceptibility of a human astrocytoma cell line to human cytomegalovirus (HCMV) infection was investigated. Infection of U-373MG astrocytoma cells with two strains of HCMV resulted in both production of extracellular, infectious virus and expression of immediate early and early antigens within 18 hours and late antigens after 72 hours of infection. The kinetics of infection in U-373MG cells were the same as in human diploid fibroblasts (MRC-5). Since HCMV and human immunodeficiency virus (HIV) have reportedly been found in astrocytic cells in vivo, we studied the possible interaction between HCMV and HIV long terminal repeat (LTR) elements in this cellular environment. HCMV infection transactivated the LTR of HIV-1 and HIV-2 to similar levels. Interestingly, transfection of these cells with infectious HIV-1 provirus did not result in expression of gag, env, or F proteins detectable by immunofluorescence. However, provirus gene expression was not completely silent, since it transactivated HIV-1 LTR. The level of this transactivation was similar to that seen following cotransfection with a tat expression vector. These results suggest that opportunistic infection with HCMV may reactivate latent HIV genomes in glial cells.


Assuntos
Infecções por Citomegalovirus/fisiopatologia , Regulação da Expressão Gênica , HIV-1/genética , HIV-2/genética , Animais , Cloranfenicol O-Acetiltransferase/genética , Imunofluorescência , Humanos , Provírus/genética , Sequências Repetitivas de Ácido Nucleico , Transfecção , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA