Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Genes Immun ; 12(6): 399-414, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21490621

RESUMO

Interferon regulatory factor 7 (IRF7) was originally identified in the context of Epstein-Barr virus (EBV) infection, and has since emerged as the crucial regulator of type I interferons (IFNs) against pathogenic infections, which activate IRF7 by triggering signaling cascades from pathogen recognition receptors (PRRs) that recognize pathogenic nucleic acids. Moreover, IRF7 is a multifunctional transcription factor, underscored by the fact that it is associated with EBV latency, in which IRF7 is induced as well as activated by the EBV principal oncoprotein latent membrane protein-1 (LMP1). Aberrant production of type I IFNs is associated with many types of diseases such as cancers and autoimmune disorders. Thus, tight regulation of IRF7 expression and activity is imperative in dictating appropriate type I IFN production for normal IFN-mediated physiological functions. Posttranslational modifications have important roles in regulation of IRF7 activity, exemplified by phosphorylation, which is indicative of its activation. Furthermore, mounting evidence has shed light on the importance of regulatory ubiquitination in activation of IRF7. Albeit these exciting findings have been made in the past decade since its discovery, many questions related to IRF7 remain to be addressed.


Assuntos
Infecções por Vírus Epstein-Barr/genética , Infecções por Vírus Epstein-Barr/metabolismo , Fator Regulador 7 de Interferon/metabolismo , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/fisiologia , Humanos , Fator Regulador 7 de Interferon/genética , Interferon Tipo I/metabolismo , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Latência Viral
2.
Science ; 257(5077): 1685-9, 1992 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-1382315

RESUMO

The double-stranded RNA-dependent protein kinase (dsRNA-PK) is thought to be a key mediator of the antiviral and antiproliferative effects of interferons (IFNs). Studies examining the physiological function of the kinase suggest that it participates in cell growth and differentiation by regulating protein synthesis. Autophosphorylation and consequent activation of dsRNA-PK in vitro and in vivo result in phosphorylation of the alpha subunit of eukaryotic initiation factor-2 (eIF-2) and inhibition of protein synthesis. Expression of a functionally defective mutant of human dsRNA-PK in NIH 3T3 cells resulted in malignant transformation, suggesting that dsRNA-PK may function as a suppressor of cell proliferation and tumorigenesis.


Assuntos
Transformação Celular Neoplásica , Interferons/farmacologia , Mutação , Proteínas Quinases/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Divisão Celular , Linhagem Celular , Clonagem Molecular , DNA/genética , Indução Enzimática , Expressão Gênica , Humanos , Immunoblotting , Camundongos , Dados de Sequência Molecular , Fosforilação , Proteínas Quinases/química , Proteínas Quinases/fisiologia , Transfecção , eIF-2 Quinase
3.
Science ; 285(5424): 107-10, 1999 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-10390359

RESUMO

Most isolates of hepatitis C virus (HCV) infections are resistant to interferon, the only available therapy, but the mechanism underlying this resistance has not been defined. Here it is shown that the HCV envelope protein E2 contains a sequence identical with phosphorylation sites of the interferon-inducible protein kinase PKR and the translation initiation factor eIF2alpha, a target of PKR. E2 inhibited the kinase activity of PKR and blocked its inhibitory effect on protein synthesis and cell growth. This interaction of E2 and PKR may be one mechanism by which HCV circumvents the antiviral effect of interferon.


Assuntos
Hepacivirus , Interferon-alfa/farmacologia , Proteínas do Envelope Viral/fisiologia , eIF-2 Quinase/antagonistas & inibidores , Linhagem Celular , Cloranfenicol O-Acetiltransferase/biossíntese , Resistência Microbiana a Medicamentos , Retículo Endoplasmático/metabolismo , Indução Enzimática , Fator de Iniciação 2 em Eucariotos/química , Fator de Iniciação 2 em Eucariotos/metabolismo , Células HeLa , Hepacivirus/efeitos dos fármacos , Humanos , Fosforilação , Biossíntese de Proteínas , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento , Saccharomyces cerevisiae/metabolismo , Transfecção , Transformação Genética , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/farmacologia , eIF-2 Quinase/química , eIF-2 Quinase/metabolismo
4.
Science ; 255(5043): 456-9, 1992 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-1531159

RESUMO

Simian immunodeficiency virus (SIV) is a primate lentivirus related to human immunodeficiency viruses and is an etiologic agent for acquired immunodeficiency syndrome (AIDS)-like diseases in macaques. To date, only inactivated whole virus vaccines have been shown to protect macaques against SIV infection. Protective immunity was elicited by recombinant subunit vaccines. Four Macaca fascicularis were immunized with recombinant vaccinia virus expressing SIVmne gp160 and were boosted with gp160 produced in baculovirus-infected cells. All four animals were protected against an intravenous challenge of the homologous virus at one to nine animal-infectious doses. These results indicate that immunization with viral envelope antigens alone is sufficient to elicit protective immunity against a primate immunodeficiency virus. The combination immunization regimen, similar to one now being evaluated in humans as candidate human immunodeficiency virus (HIV)-1 vaccines, appears to be an effective way to elicit such immune responses.


Assuntos
Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/imunologia , Vacinas Sintéticas/imunologia , Vacinas Virais/imunologia , Animais , Sequência de Bases , DNA Viral/genética , Produtos do Gene env , Vetores Genéticos , Ativação Linfocitária , Macaca fascicularis , Dados de Sequência Molecular , Testes de Neutralização , Oligonucleotídeos/química , Reação em Cadeia da Polimerase , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Fatores de Tempo , Vacinação
5.
Mucosal Immunol ; 11(3): 820-834, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29346345

RESUMO

STING (stimulator of interferon genes) is a cytosolic sensor for cyclic dinucleotides and also an adaptor molecule for intracellular DNA receptors. Although STING has important functions in the host defense against pathogens and in autoimmune diseases, its physiological relevance in intestinal homeostasis is largely unknown. In this study, we show that STING-/- mice presented defective protective mechanisms of intestinal mucosa, including decreased number of goblet cells, diminished mucus production, and lower levels of secretory IgA, when compared with wild-type (WT) mice. Fecal content and microbiota DNA could activate STING, indicating a role of this molecule in gut. Microbiota composition was altered in STING-/- mice toward a more inflammatory profile, evidencing a reduction in the Allobacolum and Bifidobacterium groups along with increase in Disulfovibrio bacteria. Absence of STING lead to decrease in induced intraepithelial lymphocytes (IEL) and to increase in group 1 innate lymphoid cell (ILC1) as well as ILC3 frequencies and decrease in ILC2 in the colon. Development and function of Foxp3+ and LAP+ regulatory T cells were also compromised in STING-/- mice. Moreover, these mice were highly susceptible to dextran sodium sulfate-induced colitis, T-cell-induced colitis, and enteric Salmonella typhimurium infection when compared with WT animals. Therefore, our results identify an important role of STING in maintaining gut homeostasis and also a protective effect in controlling gut inflammation.


Assuntos
Colite/imunologia , Microbioma Gastrointestinal/fisiologia , Mucosa Intestinal/imunologia , Intestinos/fisiologia , Linfócitos/imunologia , Proteínas de Membrana/metabolismo , Infecções por Salmonella/imunologia , Salmonella typhimurium/imunologia , Linfócitos T Reguladores/imunologia , Animais , Colite/induzido quimicamente , Colite/genética , Sulfato de Dextrana , Feminino , Fatores de Transcrição Forkhead/metabolismo , Homeostase , Imunidade Inata , Imunoglobulina A Secretora/sangue , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Salmonella/genética , Células Th1/imunologia
6.
Cell Death Differ ; 13(11): 1982-93, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16575407

RESUMO

Melanoma differentiation-associated gene-5 (mda-5) was the first molecule identified in nature whose encoded protein embodied the unique structural combination of an N-terminal caspase recruitment domain and a C-terminal DExD/H RNA helicase domain. As suggested by its structure, cumulative evidences documented that ectopic expression of mda-5 leads to growth inhibition and/or apoptosis in various cell lines. However, the signaling pathways involved in mda-5-mediated killing have not been elucidated. In this study, we utilized either genetically modified cloned rat embryo fibroblast cells overexpressing different functionally and structurally distinct oncogenes or human pancreatic and colorectal carcinoma cells containing mutant active ras to resolve the role of the Ras/Raf signaling pathway in mda-5-mediated growth inhibition/apoptosis induction. Rodent and human tumor cells containing constitutively activated Raf/Raf/MEK/ERK pathways were resistant to mda-5-induced killing and this protection was antagonized by intervening in this signal transduction cascade either by directly inhibiting ras activity using an antisense strategy or by targeting ras-downstream factors, such as MEK1/2, with the pharmacological inhibitor PD98059. The present findings provide a further example of potential cross-talk between growth-inhibitory and growth-promoting pathways in which the ultimate balance of these factors defines cellular homeostasis, leading to survival or induction of programmed cell death.


Assuntos
Apoptose , Diferenciação Celular/fisiologia , RNA Helicases DEAD-box/metabolismo , Melanoma/patologia , Proteínas Proto-Oncogênicas c-raf/metabolismo , Proteínas ras/metabolismo , Adenoviridae/metabolismo , Animais , Linhagem Celular Transformada , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , RNA Helicases DEAD-box/genética , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Humanos , Helicase IFIH1 Induzida por Interferon , Proteínas Mutantes/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos
7.
Mol Cell Biol ; 15(6): 3138-46, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7539103

RESUMO

Recently we reported that introduction of catalytically inactive PKR molecules into NIH 3T3 cells causes malignant transformation and the development of tumors in nude mice. We have proposed that PKR may be a tumor suppressor gene possibly because of its translational inhibitory properties. We have now designed and characterized a number of PKR mutants encoding proteins that retain their catalytic competence but are mutated in their regulatory double-stranded RNA (dsRNA) binding domains (RBDs). RNA binding analysis revealed that PKR proteins either lacking or with point mutations in the first RBD (RBD-1) bound negligible amounts of dsRNA activator or adenovirus VAI RNA inhibitor. Despite the lack of binding, such variants remained functionally competent but were much less active than wild-type PKR. PKR variants completely lacking RBD-1 were largely unresponsive to dsRNA in activation assays but could be activated by heparin. To complement these studies, we evaluated the effects of point mutations in RBD-1 or the removal of either RBD-1 or RBD-2 on the proliferation rate of mouse 3T3 cells. We were unsuccessful at isolating stably transformed cells expressing RBD-1 point mutants or RBD-2-minus mutants. In contrast, NIH 3T3 cells, which constitutively expressed PKR proteins that lacked RBD-1, were selected. These cells displayed a transformed phenotype and caused tumors after inoculation in nude mice. Further, levels of endogenous eIF-2 alpha phosphorylation in RBD-1-minus cell lines were reduced, suggesting that such mutants act in a dominant negative manner to inhibit the function of endogenous PKR. These results emphasize the importance of RBD-1 in PKR control of cell growth and provide additional evidence for the critical role played by PKR in the regulation of malignant transformation.


Assuntos
Transformação Celular Neoplásica/metabolismo , Proteínas Serina-Treonina Quinases/genética , Células 3T3 , Animais , Sequência de Bases , Sítios de Ligação/genética , Transformação Celular Neoplásica/genética , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas Serina-Treonina Quinases/metabolismo , RNA/antagonistas & inibidores , RNA/metabolismo , eIF-2 Quinase
8.
Mol Cell Biol ; 15(1): 365-78, 1995 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-7799945

RESUMO

The protein kinase DAI is activated upon viral infection of mammalian cells and inhibits protein synthesis by phosphorylation of the alpha subunit of translation initiation factor 2 (eIF-2 alpha). DAI is activated in vitro by double-stranded RNAs (dsRNAs), and binding of dsRNA is dependent on two copies of a conserved sequence motif located N terminal to the kinase domain in DAI. High-level expression of DAI in Saccharomyces cerevisiae cells is lethal because of hyperphosphorylation of eIF-2 alpha; at lower levels, DAI can functionally replace the protein kinase GCN2 and stimulate translation of GCN4 mRNA. These two phenotypes were used to characterize structural requirements for DAI function in vivo, by examining the effects of amino acid substitutions at matching positions in the two dsRNA-binding motifs and of replacing one copy of the motif with the other. We found that both copies of the dsRNA-binding motif are required for high-level kinase function and that the N-terminal copy is more important than the C-terminal copy for activation of DAI in S. cerevisiae. On the basis of these findings, we conclude that the requirements for dsRNA binding in vitro and for activation of DAI kinase function in vivo closely coincide. Two mutant alleles containing deletions of the first or second binding motif functionally complemented when coexpressed in yeast cells, strongly suggesting that the active form of DAI is a dimer. In accord with this conclusion, overexpression of four catalytically inactive alleles containing different deletions in the protein kinase domain interfered with wild-type DAI produced in the same cells. Interestingly, three inactivating point mutations in the kinase domain were all recessive, suggesting that dominant interference involves the formation of defective heterodimers rather than sequestration of dsRNA activators by mutant enzymes. We suggest that large structural alterations in the kinase domain impair an interaction between the two protomers in a DAI dimer that is necessary for activation by dsRNA or for catalysis of eIF-2 alpha phosphorylation.


Assuntos
Proteínas Serina-Treonina Quinases/metabolismo , Sequência de Aminoácidos , Ativação Enzimática , Fator de Iniciação 2 em Eucariotos/metabolismo , Regulação Enzimológica da Expressão Gênica , Genes Dominantes , Teste de Complementação Genética , Humanos , Substâncias Macromoleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fosforilação , Ligação Proteica , Biossíntese de Proteínas , Proteínas Serina-Treonina Quinases/química , RNA de Cadeia Dupla/metabolismo , Proteínas Recombinantes , Saccharomyces cerevisiae , Relação Estrutura-Atividade , eIF-2 Quinase
9.
Mol Cell Biol ; 11(11): 5497-505, 1991 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-1717830

RESUMO

Eukaryotic viruses have devised numerous strategies to downregulate activity of the interferon-induced, double-stranded (dsRNA)-activated protein kinase (referred to as p68 on the basis of its Mr of 68,000 in human cells). Viruses must exert this control to avoid extensive phosphorylation of the alpha subunit of eukaryotic initiation factor 2 (eIF-2) by p68 and the resultant negative effects on protein synthesis initiation. To begin to define the molecular mechanisms underlying this regulation, we optimized expression of p68 in an in vitro transcription-translation system utilizing the full-length cDNA clone. The in vitro-expressed kinase was autophosphorylated in response to dsRNAs and heparin in a manner similar to that for the native p68 provided that the kinase inhibitor, 2-aminopurine, was present during the in vitro translation reaction. Further, the activated kinase efficiently phosphorylated its natural substrate, the alpha subunit of eIF-2. Binding experiments revealed that the expressed kinase complexed with the dsRNA activator, reovirus dsRNA, as well as the adenovirus-encoded inhibitor, VAI RNA. Interestingly, both the reovirus RNAs and VAI RNA also complexed with protein kinase molecules that lacked the carboxyl terminus and all catalytic domains. Deletion analysis confirmed that the p68 amino terminus contained critical determinants for reovirus dsRNA and VAI RNA binding. Further, reovirus dsRNA efficiently bound to, but failed to activate, p68 kinase molecules containing a single amino acid substitution in the invariant lysine 295 present in catalytic domain II. Taken together, these data demonstrate that this expression system permits a detailed mutagenic analysis of the regions of p68 required for interaction with virus-encoded activators and repressors.


Assuntos
DNA Viral/metabolismo , Interferons/farmacologia , Mutagênese Sítio-Dirigida , Biossíntese de Proteínas , Proteínas Quinases/genética , RNA de Cadeia Dupla/metabolismo , Adenoviridae/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Sistema Livre de Células , Deleção Cromossômica , Ativação Enzimática , Indução Enzimática , Fator de Iniciação 2 em Eucariotos/metabolismo , Humanos , Cinética , Substâncias Macromoleculares , Dados de Sequência Molecular , Peso Molecular , Oligonucleotídeos , Fosforilação , Proteínas Quinases/biossíntese , Proteínas Quinases/metabolismo , Coelhos , Reoviridae/genética , Reticulócitos/metabolismo , Transcrição Gênica
10.
Nucleic Acids Res ; 31(5): 1470-80, 2003 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-12595555

RESUMO

The RNA helicases p68 and p72 are highly related members of the DEAD box family of proteins, sharing 90% identity across the conserved core, and have been shown to be involved in both transcription and mRNA processing. We previously showed that these proteins co-localise in the nucleus of interphase cells. In this study we show that p68 and p72 can interact with each other and self-associate in the yeast two-hybrid system. Co-immunoprecipitation experiments confirmed that p68 and p72 can interact in the cell and indicated that these proteins preferentially exist as hetero-dimers. In addition, we show that p68 can interact with NFAR-2, a protein that is also thought to function in mRNA processing. Moreover, gel filtration analysis suggests that p68 and p72 can exist in a variety of complexes in the cell (ranging from approximately 150 to approximately 400 kDa in size), with a subset of p68 molecules being in very large complexes (>2 MDa). The potential to exist in different complexes that may contain p68 and/or p72, together with a range of other factors, would provide the potential for these proteins to interact with different RNA substrates and would be consistent with recent reports implying a wide range of functions for p68/p72.


Assuntos
Adenosina Trifosfatases/metabolismo , Fosfoproteínas , Proteínas Quinases/metabolismo , RNA Helicases/metabolismo , Adenosina Trifosfatases/química , Adenosina Trifosfatases/genética , Ligação Competitiva , Linhagem Celular , Proteínas Cromossômicas não Histona/metabolismo , RNA Helicases DEAD-box , Dimerização , Células HeLa , Humanos , Microscopia de Fluorescência , Proteínas do Fator Nuclear 90 , Testes de Precipitina , Ligação Proteica , Proteínas Quinases/química , Proteínas Quinases/genética , RNA Helicases/química , RNA Helicases/genética , Proteínas de Ligação a RNA/metabolismo , Saccharomyces cerevisiae/genética , Técnicas do Sistema de Duplo-Híbrido
11.
Oncogene ; 20(7): 800-11, 2001 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11314014

RESUMO

Human herpes virus 8 (HHV-8) has developed unique mechanisms for altering cellular proliferative and apoptotic control pathways by incorporating viral homologs to several cellular regulatory genes into its genome. One of the important pirated genes encoded by the ORF K9 reading frame is a viral homolog of the interferon regulatory factors (IRF), a family of cellular transcription proteins that regulates expression of genes involved in pathogen response, immune modulation and cell proliferation. vIRF-1 has been shown to downregulate the interferon- and IRF-mediated transcriptional activation of ISG and murine IFNA4 gene promoters. In this study we demonstrate that vIRF-1 efficiently inhibited virus-induced expression of endogenous interferon B, CC chemokine RANTES and CXC chemokine IP-10 genes. Co-expression analysis revealed that vIRF-1 selectively blocked IRF-3 but not IRF-7-mediated transactivation. vIRF-1 was able to bind to both IRF-3 and IRF-7 in vivo as detected by coimmunoprecipitation analysis, but did not affect IRF-3 dimerization, nuclear translocation and DNA binding activity. Rather, vIRF-1 interacted with the CBP/p300 coactivators and efficiently inhibited the formation of transcriptionally competent IRF-3-CBP/p300 complexes. These results illustrate that vIRF-1 is able to block the early stages of the IFN response to virus infection by interfering with the activation of IRF-3 responsive, immediate early IFN genes.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Herpesvirus Humano 8/imunologia , Interferons/metabolismo , Proteínas Nucleares/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Virais/metabolismo , Antivirais/metabolismo , Células Cultivadas , Proteínas de Ligação a DNA/genética , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/patogenicidade , Humanos , Fator Regulador 3 de Interferon , Fator Regulador 7 de Interferon , Fatores Reguladores de Interferon , Ligação Proteica , Fatores de Transcrição/genética , Ativação Transcricional , Proteínas Virais/genética
12.
Oncogene ; 20(48): 7029-40, 2001 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-11704827

RESUMO

Gammaherpes viruses are often detected in lymphomas arising in immunocompromised patients. We have found that Azidothymidine (AZT) alone induces apoptosis in Epstein Barr Virus (EBV) positive Burkitt's lymphoma (BL) cells but requires interferon alpha (IFN-alpha) to induce apoptosis in Human Herpes Virus Type 8 (HHV-8) positive Primary Effusion Lymphomas (PEL). Our analysis of a series of AIDS lymphomas revealed that IFN-alpha selectively induced very high levels of the Death Receptor (DR) tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in HHV-8 positive PEL lines and primary tumor cells whereas little or no induction was observed in primary EBV+ AIDS lymphomas and EBV-Burkitt's lines. AZT and IFN-alpha mediated apoptosis in PEL was blocked by stable overexpression of dominant negative Fas Associated Death Domain (FADD), decoy receptor 2 (DcR2), soluble TRAIL receptor fusion proteins (DR-4 and DR-5) and thymidine. Trimeric TRAIL (in place of IFN-alpha) similarly synergized with AZT to induce apoptosis in HHV-8 positive PEL cells. This is the first demonstration that IFN-alpha induces functional TRAIL in a malignancy that can be exploited to effect a suicide program. This novel antiviral approach to Primary Effusion lymphomas is targeted and may represent a highly effective and relatively non-toxic therapy.


Assuntos
Antivirais/farmacologia , Apoptose/efeitos dos fármacos , Proteínas de Arabidopsis , Fatores Imunológicos/farmacologia , Interferon-alfa/farmacologia , Linfoma Relacionado a AIDS/terapia , Linfoma de Células B/terapia , Glicoproteínas de Membrana/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Antimetabólitos Antineoplásicos/farmacologia , Antimetabólitos Antineoplásicos/uso terapêutico , Antivirais/uso terapêutico , Proteínas Reguladoras de Apoptose , Biopolímeros , Cisteína Endopeptidases/metabolismo , Sinergismo Farmacológico , Ativação Enzimática/efeitos dos fármacos , Infecções por Vírus Epstein-Barr/complicações , Etoposídeo/farmacologia , Ácidos Graxos Dessaturases/biossíntese , Ácidos Graxos Dessaturases/genética , Ácidos Graxos Dessaturases/fisiologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes bcl-2 , Infecções por HIV/complicações , Infecções por Herpesviridae/complicações , Herpesvirus Humano 4/isolamento & purificação , Herpesvirus Humano 8/isolamento & purificação , Humanos , Hospedeiro Imunocomprometido , Fatores Imunológicos/uso terapêutico , Interferon-alfa/uso terapêutico , Linfoma Relacionado a AIDS/etiologia , Linfoma Relacionado a AIDS/imunologia , Linfoma Relacionado a AIDS/patologia , Linfoma de Células B/etiologia , Linfoma de Células B/imunologia , Linfoma de Células B/patologia , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/farmacologia , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/biossíntese , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/fisiologia , Ligante Indutor de Apoptose Relacionado a TNF , Timidina/farmacologia , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/química , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/farmacologia
13.
Cell Death Differ ; 8(2): 113-26, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11313713

RESUMO

To thwart viral infection, the host has developed a formidable and integrated defense network that comprises our innate and adaptive immune response. In recent years, it has become clear that in an attempt to prevent viral replication, viral dissemination or persistent viral infection of the cell, many of these protective measures actually involve the induction of programmed cell death, or apoptosis. An initial response to viral infection primarily involves the innate arm of immunity and the killing of infected cells with cytotoxic lymphocytes such as natural killer (NK) cells through mechanisms that include the employment of perforin and granzymes. Once the virus has invaded the cell, however, a second host defense-mediated response is also triggered which involves the induction of a family of cytokines known as the interferons (IFNs). The IFNs, which are essential for initiating and coordinating a successful antiviral response, function by stimulating the adaptive arm of immunity involving cytotoxic T cells (CTLs), and by inducing a number of intracellular genes that directly prevent virus replication/cytolysis or that facilitate apoptosis. The IFN-induced gene family is now known to comprise the death ligand TRAIL, the dsRNA-dependent protein kinase (PKR), interferon regulatory factors (IRFs) and the promyelocytic leukemia gene (PML), all of which have been reported to be mediators of cell death. That DNA array analyses indicate that numerous cellular genes, many as yet uncharacterized, may similarly be induced by IFN, further emphasizes the likely importance that these cytokines have in the modulation of apoptosis. This likelihood is additionally underlined by the elaborate strategies developed by viruses to inhibit IFN-antiviral function and the mechanisms of cell death.


Assuntos
Apoptose/fisiologia , Interferons/metabolismo , Proteínas Nucleares , Viroses/imunologia , Vírus/metabolismo , Nucleotídeos de Adenina/metabolismo , Animais , Antígenos CD/metabolismo , Proteínas Reguladoras de Apoptose , Endorribonucleases/metabolismo , Genes Reguladores/genética , Genes Reguladores/fisiologia , Humanos , Interferons/imunologia , Glicoproteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias/terapia , Oligorribonucleotídeos/metabolismo , Proteína da Leucemia Promielocítica , RNA de Cadeia Dupla/fisiologia , Receptores do Fator de Necrose Tumoral/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral , Transdução de Sinais , Ligante Indutor de Apoptose Relacionado a TNF , Fatores de Transcrição/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteína Supressora de Tumor p53 , Proteínas Supressoras de Tumor , Vírus/patogenicidade , eIF-2 Quinase/metabolismo , Receptor fas/metabolismo
14.
Oncogene ; 34(41): 5302-8, 2015 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-25639870

RESUMO

Stimulator of interferon genes (STING) is a cellular sensor that controls cytosolic DNA-activated innate immune signaling. We have previously demonstrated that STING-deficient mice are resistant to carcinogen-induced skin cancer, similar to myeloid differentiation primary response gene 88 (MyD88) deficient mice, since the production of STING-dependent DNA-damage-induced proinflammatory cytokines, that likely require MyD88 signaling to exert their growth-promoting activity, are prevented. In contrast, MyD88-deficient mice are sensitive to colitis-associated cancer (CAC), since selected cytokines generated following DNA-damage also activate repair pathways, which can help prevent tumor development. Here, we demonstrate that STING signaling facilitates wound repair processes and that analogous to MyD88-deficient mice, STING-deficient mice (SKO) are prone to CAC induced by DNA-damaging agents. SKO mice harboring tumors exhibited low levels of tumor-suppressive interleukin-22 binding protein (IL-22BP) compared to normal mice, a cytokine considered critical for preventing colon-related cancer. Our data indicate that STING constitutes a critical component of the host early response to intestinal damage and is essential for invigorating tissue repair pathways that may help prevent tumorigenesis.


Assuntos
Carcinogênese/metabolismo , Neoplasias do Colo/metabolismo , Proteínas de Membrana/fisiologia , Animais , Azoximetano/farmacologia , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Humanos , Imunidade Inata , Camundongos , Camundongos Knockout , Transdução de Sinais , Ativação Transcricional/efeitos dos fármacos
15.
Int J Biochem Cell Biol ; 31(1): 123-38, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10216948

RESUMO

The double-stranded (ds) RNA-regulated serine/threonine protein kinase, PKR, is an interferon-inducible enzyme of widespread occurrence in mammalian cells. PKR is activated by dsRNA via a mechanism involving autophosphorylation. Once activated, the enzyme phosphorylates the alpha-subunit of protein synthesis initiation factor eIF2, thereby inhibiting translation. Accumulating data suggest that PKR has additional substrates, and that the kinase may also regulate gene transcription and signal transduction pathways. Although PKR plays an important role in mediating the antiviral effects of interferons, PKR is also implicated in regulating cell proliferation in uninfected cells and may have a tumor suppressor function under normal conditions. Studies of human malignancies and tumor cell lines suggest that, in general, patients bearing tumors with a higher PKR content have a more favorable prognosis. However, in human breast carcinoma cells, dysregulation of PKR may be associated with the establishment or maintenance of the transformed state.


Assuntos
Apoptose/fisiologia , Neoplasias/enzimologia , Proteínas Proto-Oncogênicas c-bcl-2 , Proteínas de Ligação a RNA/metabolismo , Proteínas Repressoras/metabolismo , eIF-2 Quinase/metabolismo , Animais , Sequência de Bases , Neoplasias da Mama/enzimologia , Carcinoma/enzimologia , Diferenciação Celular/fisiologia , Divisão Celular , Transformação Celular Neoplásica , Humanos , Dados de Sequência Molecular , Proteínas de Ligação a Poli(A) , Biossíntese de Proteínas , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas Repressoras/química , Proteínas Repressoras/genética , Proteína X Associada a bcl-2 , eIF-2 Quinase/química , eIF-2 Quinase/genética
16.
Gene ; 56(1): 137-44, 1987.
Artigo em Inglês | MEDLINE | ID: mdl-3315857

RESUMO

The Lassa virus nucleocapsid protein gene and segments from it were expressed in Escherichia coli under the control of the lac promoter in pUC-based plasmids. Expression of the near full-length protein [amino acid (aa) residues 12-570] fused to an N-terminal sequence of vector-derived 6 aa was not particularly efficient, and neither was that of a smaller N-terminal segment (aa 6-201) which was also fused at its C terminus to the remainder of the lacZ gene product. By contrast, the C-terminal 370 aa could be expressed at levels approaching 10% of total cellular protein. All the recombinant proteins were associated with the insoluble fraction after sonication of the bacteria. The inefficiently expressed products did not appear to be any more susceptible to proteolytic degradation. The distribution of codons rarely used in E. coli genes was relatively uniform along the nucleocapsid gene sequence. These results are consistent with the regulation of transcriptional or translational efficiency by features of the sequence downstream from the promoter and ribosome-binding site. The C-terminal segment (aa 201-570 representing 65% of the authentic protein) was purified by ion exchange chromatography and shown to be active when used as antigen in enzyme-linked immunoassays for virus-specific antibodies.


Assuntos
Arenaviridae/genética , Capsídeo/genética , Vírus Lassa/genética , Proteínas do Core Viral/genética , Anticorpos Antivirais/análise , Capsídeo/imunologia , Capsídeo/isolamento & purificação , Clonagem Molecular , Ensaio de Imunoadsorção Enzimática , Escherichia coli/genética , Regulação da Expressão Gênica , Genes Virais , Plasmídeos , Proteínas Recombinantes/genética , Proteínas do Core Viral/imunologia , Proteínas do Core Viral/isolamento & purificação
17.
Cancer Gene Ther ; 19(7): 443-50, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22522623

RESUMO

Multiple myeloma (MM) is an incurable malignancy of plasma secreting B cells disseminated in the bone marrow. Successful utilization of oncolytic virotherapy for myeloma treatment requires a systemically administered virus that selectively destroys disseminated myeloma cells in an immune-competent host. Vesicular stomatitis virus (VSV)-expressing interferon-ß (IFNß) is a promising new oncolytic agent that exploits tumor-associated defects in innate immune signaling pathways to destroy cancer cells specifically. We demonstrate here that a single, intravenous dose of VSV coding for IFNß (VSV-IFNß) specifically destroys subcutaneous and disseminated 5TGM1 myeloma in an immune-competent myeloma model. VSV-IFN treatment significantly prolonged survival in mice bearing orthotopic myeloma. Viral murine IFNß expression further delayed myeloma progression and significantly enhanced survival compared with VSV-expressing human IFNß. Evaluation of VSV-IFNß oncolytic activity in human myeloma cell lines and primary patient samples confirmed myeloma-specific oncolytic activity, but revealed variable susceptibility to VSV-IFNß oncolysis. The results indicate that VSV-IFNß is a potent, safe oncolytic agent that can be systemically administered to target and destroy disseminated myeloma effectively in immune-competent mice. IFNß expression improves cancer specificity and enhances VSV therapeutic efficacy against disseminated myeloma. These data show VSV-IFNß to be a promising vector for further development as a potential therapy for the treatment of MM.


Assuntos
Interferon beta/genética , Mieloma Múltiplo/terapia , Vírus Oncolíticos/genética , Vírus da Estomatite Vesicular Indiana/genética , Animais , Linhagem Celular Tumoral , Feminino , Vetores Genéticos , Humanos , Interferon beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Terapia Viral Oncolítica
18.
Leukemia ; 26(8): 1870-8, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22425894

RESUMO

Current therapy for multiple myeloma is complex and prolonged. Antimyeloma drugs are combined in induction, consolidation and/or maintenance protocols to destroy bulky disease, then suppress or eradicate residual disease. Oncolytic viruses have the potential to mediate both tumor debulking and residual disease elimination, but this curative paradigm remains unproven. Here, we engineered an oncolytic vesicular stomatitis virus to minimize its neurotoxicity, enhance induction of antimyeloma immunity and facilitate noninvasive monitoring of its intratumoral spread. Using high-resolution imaging, autoradiography and immunohistochemistry, we demonstrate that the intravenously administered virus extravasates from tumor blood vessels in immunocompetent myeloma-bearing mice, nucleating multiple intratumoral infectious centers that expand rapidly and necrose at their centers, ultimately coalescing to cause extensive tumor destruction. This oncolytic tumor debulking phase lasts only for 72 h after virus administration, and is completed before antiviral antibodies become detectable in the bloodstream. Antimyeloma T cells, cross-primed as the virus-infected cells provoke an antiviral immune response, then eliminate residual uninfected myeloma cells. The study establishes a curative oncolytic paradigm for multiple myeloma where direct tumor debulking and immune eradication of minimal disease are mediated by a single intravenous dose of a single therapeutic agent. Clinical translation is underway.


Assuntos
Vetores Genéticos/administração & dosagem , Mieloma Múltiplo/terapia , Terapia Viral Oncolítica , Animais , Linhagem Celular Tumoral , Cricetinae , Terapia Genética , Vetores Genéticos/genética , Interferon beta/genética , Interferon beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mieloma Múltiplo/genética , Mieloma Múltiplo/imunologia , Vírus Oncolíticos/genética , Simportadores/genética , Simportadores/metabolismo , Transplante Isogênico , Vírus da Estomatite Vesicular Indiana/genética
20.
Semin Cancer Biol ; 10(2): 103-11, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10936061

RESUMO

The interferons (IFNs) play an integral role in cellular host defense against virus infection and conceivably tumorigenesis. Despite over 50 years of research, however, the molecular mechanisms underlining IFN action remain to be fully elucidated, in part because of the large number of genes, with an uncharacterized function that appears to be induced by these cytokines. Although the majority of in vitro studies indicate that IFNs antiviral properties involve inhibiting viral replication while maintaining the integrity of the cell, numerous reports have now implicated that a number of IFN-induced genes, IFN transcriptional regulatory factors and IFN signaling molecules can also mediate apoptosis. Here, we review some of what is known about IFN's ability to invoke programmed cell death as part of an intricate arsenal intended to prevent viral infection and malignant disease.


Assuntos
Apoptose/fisiologia , Interferon Tipo I/fisiologia , Interferon gama/fisiologia , Animais , Humanos , Fatores de Transcrição/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA