Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
PLoS Pathog ; 13(4): e1006296, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28399146

RESUMO

Hepatitis B virus (HBV) replicates its DNA genome through reverse transcription of a viral RNA pregenome. We report herein that the interferon (IFN) stimulated exoribonuclease gene of 20 KD (ISG20) inhibits HBV replication through degradation of HBV RNA. ISG20 expression was observed at basal level and was highly upregulated upon IFN treatment in hepatocytes, and knock down of ISG20 resulted in elevation of HBV replication and attenuation of IFN-mediated antiviral effect. The sequence element conferring the susceptibility of HBV RNA to ISG20-mediated RNA degradation was mapped at the HBV RNA terminal redundant region containing epsilon (ε) stem-loop. Furthermore, ISG20-induced HBV RNA degradation relies on its ribonuclease activity, as the enzymatic inactive form ISG20D94G was unable to promote HBV RNA decay. Interestingly, ISG20D94G retained antiviral activity against HBV DNA replication by preventing pgRNA encapsidation, resulting from a consequence of ISG20-ε interaction. This interaction was further characterized by in vitro electrophoretic mobility shift assay (EMSA) and ISG20 was able to bind HBV ε directly in absence of any other cellular proteins, indicating a direct ε RNA binding capability of ISG20; however, cofactor(s) may be required for ISG20 to efficiently degrade ε. In addition, the lower stem portion of ε is the major ISG20 binding site, and the removal of 4 base pairs from the bottom portion of ε abrogated the sensitivity of HBV RNA to ISG20, suggesting that the specificity of ISG20-ε interaction relies on both RNA structure and sequence. Furthermore, the C-terminal Exonuclease III (ExoIII) domain of ISG20 was determined to be responsible for interacting with ε, as the deletion of ExoIII abolished in vitro ISG20-ε binding and intracellular HBV RNA degradation. Taken together, our study sheds light on the underlying mechanisms of IFN-mediated HBV inhibition and the antiviral mechanism of ISG20 in general.


Assuntos
Exonucleases/metabolismo , Exonucleases/farmacologia , Vírus da Hepatite B/metabolismo , RNA Viral/efeitos dos fármacos , Ribonucleases/metabolismo , Replicação Viral/efeitos dos fármacos , Antivirais/farmacologia , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/fisiologia , Exorribonucleases , Vírus da Hepatite B/isolamento & purificação , Hepatócitos/efeitos dos fármacos , Hepatócitos/virologia , Humanos , Estabilidade de RNA/efeitos dos fármacos , RNA Viral/metabolismo , Transcrição Reversa/efeitos dos fármacos , Replicação Viral/fisiologia
2.
J Gen Virol ; 98(7): 1864-1878, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28699853

RESUMO

The connection between the repression of human immunodeficiency virus type 1(HIV-1) transcription and the resting CD4+ T cell state suggests that the host transcription factors involved in the active maintenance of lymphocyte quiescence are likely to repress the viral transactivator, Tat, thereby restricting HIV-1 transcription. In this study, we analysed the interplay between Tat and the forkhead box transcription factors, FoxO1 and FoxO4. We show that FoxO1 and FoxO4 antagonize Tat-mediated transactivation of HIV-1 promoter through the repression of Tat protein expression. No effect was observed on the expression of two HIV-1 accessory proteins, Vif and Vpr. Unexpectedly, we found that FoxO1 and FoxO4 expression causes a strong dose-dependent post-transcriptional suppression of Tat mRNA, indicating that FoxO should effectively inhibit HIV-1 replication by destabilizing Tat mRNA and suppressing Tat-mediated HIV-1 transcription. In accordance with this, we observed that the Tat mRNA half-life is reduced by FoxO4 expression. The physiological relevance of our findings was validated using the J-Lat 10.6 model of latently infected cells. We demonstrated that the overexpression of a constitutively active FoxO4-TM mutant antagonized HIV-1 transcription reactivation in response to T cell activators, such as TNF-α or PMA. Altogether, our findings demonstrate that FoxO factors can control HIV-1 transcription and provide new insights into their potential role during the establishment of HIV-1 latency.


Assuntos
Regulação Viral da Expressão Gênica , Infecções por HIV/metabolismo , HIV-1/genética , Fatores de Transcrição/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Proteínas de Ciclo Celular , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Fatores de Transcrição Forkhead , Infecções por HIV/genética , Infecções por HIV/virologia , HIV-1/metabolismo , Humanos , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição/genética , Ativação Transcricional , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética
3.
J Virol ; 87(8): 4523-33, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23408607

RESUMO

Tripartite motif (TRIM) protein superfamily members are emerging as important effectors of the innate immune response against viral infections. In particular, TRIM22 was reported to exert antiviral activity against RNA viruses, such as hepatitis B virus (HBV), encephalomyocarditis virus (ECMV), and human immunodeficiency virus type 1 (HIV-1). We demonstrate here, for the first time, that TRIM22 is upregulated by influenza A virus (IAV) infection at both mRNA and protein levels in human alveolar epithelial A549 cells. Conversely, TRIM22 potently restricted IAV replication, in that prevention of TRIM22 expression by means of short hairpin RNA led to a 10-fold enhancement of IAV replication in these cells. Depletion of TRIM22 also reduced the anti-IAV activity of alpha interferon (IFN-α), suggesting that TRIM22 is an important IFN-stimulated gene that is required for maximal suppression of IAV by type I IFN. Furthermore, the IAV infectious titer decreased up to 100-fold in MDCK cells expressing exogenous human TRIM22. Restriction of IAV replication was accounted for by the interaction between TRIM22 and the viral nucleoprotein (NP), resulting in its polyubiquitination and degradation in a proteasome-dependent manner. Thus, TRIM22 represents a novel restriction factor upregulated upon IAV infection that curtails its replicative capacity in epithelial cells.


Assuntos
Interações Hospedeiro-Patógeno , Vírus da Influenza A/imunologia , Proteínas de Ligação a RNA/metabolismo , Proteínas Repressoras/metabolismo , Proteínas do Core Viral/metabolismo , Replicação Viral , Animais , Linhagem Celular , Cães , Células Epiteliais/imunologia , Células Epiteliais/virologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Vírus da Influenza A/fisiologia , Antígenos de Histocompatibilidade Menor , Proteínas do Nucleocapsídeo , Proteólise , Proteínas com Motivo Tripartido , Regulação para Cima
4.
J Virol ; 85(13): 6480-91, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21525355

RESUMO

Activation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway by the viral Tax oncoprotein plays a pivotal role in clonal expansion of human T-cell leukemia virus type 1 (HTLV-1)-infected cells. As the Forkhead box O (FoxO) tumor suppressors act as downstream effectors of PI3K/Akt, they represent good candidate targets whose dysregulation by Tax might be involved in HTLV-1-mediated activation and transformation of infected cells. In this report, we provide evidence showing that Tax induces a dose-dependent degradation of FoxO4 by the ubiquitin-proteasome pathway. Consistent with that, we demonstrate that Tax expression increases the interaction between FoxO4 and Mdm2 E3 ligase, leading to a strong FoxO4 polyubiquitination. These processes require the phosphorylation of FoxO4 by Akt, since a mutant of FoxO4 with mutations on its three Akt phosphorylation sites appears to be resistant to Tax-mediated degradation and ubiquitination. In addition, we show that Tax expression is associated with degradation and phosphorylation of endogenous FoxO4 in Jurkat T cells. Finally, we demonstrate that Tax represses FoxO4 transcriptional activity. Our study demonstrates that Tax can control FoxO4 protein stability and transcriptional activity and provides new insight into the subversion of cell signaling pathways during HTLV-1 infection.


Assuntos
Regulação da Expressão Gênica , Produtos do Gene tax/metabolismo , Vírus Linfotrópico T Tipo 1 Humano/patogenicidade , Complexo de Endopeptidases do Proteassoma/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular , Linhagem Celular Transformada , Fatores de Transcrição Forkhead , Produtos do Gene tax/genética , Células HeLa , Vírus Linfotrópico T Tipo 1 Humano/genética , Vírus Linfotrópico T Tipo 1 Humano/metabolismo , Humanos , Células Jurkat , Fosfatidilinositol 3-Quinase/genética , Fosfatidilinositol 3-Quinase/metabolismo , Fosforilação , Complexo de Endopeptidases do Proteassoma/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Transdução de Sinais , Linfócitos T , Fatores de Transcrição/genética , Ubiquitinação
5.
J Virol ; 85(10): 5183-96, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21345949

RESUMO

Previous studies identified clones of the U937 promonocytic cell line that were either permissive or nonpermissive for human immunodeficiency virus type 1 (HIV-1) replication. These clones were investigated further in the search for host restriction factors that could explain their differential capacity to support HIV-1 replication. Among known HIV-1 restriction factors screened, tripartite motif-containing protein 22 (TRIM22) was the only factor constitutively expressed in nonpermissive and absent in permissive U937 cells. Stable TRIM22 knockdown (KD) rescued HIV-1 long-terminal-repeat (LTR)-driven transcription in KD-nonpermissive cells to the levels observed in permissive cells. Conversely, transduction-mediated expression of TRIM22 in permissive cells reduced LTR-driven luciferase expression by ∼7-fold, supporting a negative role of TRIM22 in HIV-1 transcription. This finding was further confirmed in the human T cell line A3.01 expressing TRIM22. Moreover, overexpression of TRIM22 in 293T cells significantly impaired basal and phorbol myristate acetate-ionomycin-induced HIV-1 LTR-driven gene expression, whereas inhibition of tumor necrosis factor alpha-induced viral transcription was a consequence of lower basal expression. In agreement, TRIM22 equally inhibited an LTR construct lacking the tandem NF-κB binding sites. In addition, TRIM22 did not affect Tat-mediated LTR transactivation. Finally, these effects were independent of TRIM22 E3 ubiquitin-ligase activity. In the context of replication-competent virus, significantly higher levels of HIV-1 production were observed in KD-nonpermissive versus control nonpermissive U937 cells after infection. In contrast, lower peak levels of HIV-1 replication characterized U937 and A3.01 cells expressing TRIM22 versus their control transduced counterpart. Thus, nuclear TRIM22 significantly impairs HIV-1 replication, likely by interfering with Tat- and NF-κB-independent LTR-driven transcription.


Assuntos
HIV-1/crescimento & desenvolvimento , HIV-1/imunologia , Proteínas Repressoras/metabolismo , Transcrição Gênica , Replicação Viral , Linhagem Celular , Humanos , Antígenos de Histocompatibilidade Menor , Monócitos/imunologia , Monócitos/virologia , Linfócitos T/imunologia , Linfócitos T/virologia , Proteínas com Motivo Tripartido
6.
Nephron Exp Nephrol ; 119(2): e40-8, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21832855

RESUMO

BACKGROUND/AIMS: Interferon (IFN)-stimulated gene 20 (ISG20) is a 3'-to-5' exonuclease specific for single-stranded RNA and involved in host defense reactions against RNA viruses. The expression and the role of ISG20 in mesangial cells have not been reported. METHODS: Normal human mesangial cells were cultured and treated with polyinosinic-polycytidylic acid (poly (I:C)), an authentic double-stranded RNA which mimics viral infection to cells. The effect of RNA interference of Toll-like receptor 3 (TLR3) or IFN-ß on the ISG20 expression was examined. The effect of a blocking antibody against the receptor for IFN-ß or anti-inflammatory steroid dexamethasone was also examined. RESULTS: Treatment of cells with poly (I:C) induced the expression of ISG20. The poly (I:C)-induced expression of ISG20 was inhibited by knockdown of TLR3, IFN regulatery factor 3 (IRF3) or IFN-ß. Blocking of the receptor for IFN-ß suppressed and overexpression of IFN-ß enhanced ISG20 expression. The poly (I:C)-induced expressions of IFN-ß and ISG20 were inhibited by dexamethasone. Transfection of mesangial cells with poly (I:C) or 5'-triphosphate single-stranded RNA as a complex with cationic lipid also induced the expression of ISG20, and this was inhibited by knockdown of retinoic acid-inducible gene-I (RIG-I). CONCLUSION: Poly (I:C) induces the expression of ISG20 in mesangial cells. ISG20 may be involved in anti-viral reactions in renal mesangial cells. TLR3, IRF3 and de novo synthesized IFN-ß may mediate the poly (I:C)-induced expression of ISG20, and RIG-I may mediate ISG20 expression induced by poly (I:C)/cationic lipid complex.


Assuntos
Exonucleases/biossíntese , Células Mesangiais/metabolismo , Poli I-C/farmacologia , Células Cultivadas , Proteína DEAD-box 58 , RNA Helicases DEAD-box/fisiologia , Dexametasona/farmacologia , Exorribonucleases , Humanos , Fator Regulador 3 de Interferon/genética , Interferon beta/biossíntese , Interferon beta/genética , Células Mesangiais/efeitos dos fármacos , Interferência de RNA , Receptores Imunológicos , Receptor 3 Toll-Like/genética
7.
Neuropsychopharmacology ; 33(7): 1584-602, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17805310

RESUMO

Chromatin remodeling by posttranslational modification of histones plays an important role in brain plasticity, including memory, response to stress and depression. The importance of H3/4 histones acetylation by CREB-binding protein (CBP) or related histone acetyltransferase, including p300, was specifically demonstrated using knockout (KO) mouse models. The physiological role of a related protein that also acts as a transcriptional coactivator with intrinsic histone acetylase activity, the p300/CBP-associated factor (PCAF), is poorly documented. We analyzed the behavioral phenotype of homozygous male and female PCAF KO mice and report a marked impact of PCAF deletion on memory processes and stress response. PCAF KO animals showed short-term memory deficits at 2 months of age, measured using spontaneous alternation, object recognition, or acquisition of a daily changing platform position in the water maze. Acquisition of a fixed platform location was delayed, but preserved, and no passive avoidance deficit was noted. No gender-related difference was observed. These deficits were associated with hippocampal alterations in pyramidal cell layer organization, basal levels of Fos immunoreactivity, and MAP kinase activation. PCAF KO mice also showed an exaggerated response to acute stress, forced swimming, and conditioned fear, associated with increased plasma corticosterone levels. Moreover, learning and memory impairments worsened at 6 and 12 months of age, when animals failed to acquire the fixed platform location in the water maze and showed passive avoidance deficits. These observations demonstrate that PCAF histone acetylase is involved lifelong in the chromatin remodeling necessary for memory formation and response to stress.


Assuntos
Memória/fisiologia , Estresse Psicológico/genética , Estresse Psicológico/fisiopatologia , Fatores de Transcrição de p300-CBP/deficiência , Fatores Etários , Análise de Variância , Animais , Aprendizagem da Esquiva/fisiologia , Comportamento Animal , Condicionamento Psicológico/fisiologia , Corticosterona/sangue , Modelos Animais de Doenças , Emoções/fisiologia , Comportamento Exploratório/fisiologia , Medo , Feminino , Hipocampo/patologia , Masculino , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/genética , Transtornos da Memória/fisiopatologia , Camundongos , Camundongos Knockout , Reconhecimento Visual de Modelos/fisiologia , Fatores Sexuais , Estresse Psicológico/patologia
8.
mSphere ; 3(2)2018 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-29624498

RESUMO

Influenza A viruses (IAVs) can cause zoonotic infections with pandemic potential when most of the human population is immunologically naive. After a pandemic, IAVs evolve to become seasonal in the human host by acquiring adaptive mutations. We have previously reported that the interferon (IFN)-inducible tripartite motif 22 (TRIM22) protein restricts the replication of seasonal IAVs by direct interaction with the viral nucleoprotein (NP), leading to its polyubiquitination and proteasomal degradation. Here we show that, in contrast to seasonal H1N1 IAVs, the 2009 pandemic H1N1 strain as well as H1N1 strains from the 1930s are resistant to TRIM22 restriction. We demonstrate that arginine-to-lysine substitutions conferring an increased sensitivity to TRIM22-dependent ubiquitination accumulated progressively in the NP of seasonal influenza A (H1N1) viruses between 1918 and 2009. Our findings suggest that during long-term circulation and evolution of IAVs in humans, adaptive mutations are favored at the expense of an increased sensitivity to some components of the innate immune response.IMPORTANCE We have uncovered that long-term circulation of seasonal influenza A viruses (IAV) in the human population resulted in the progressive acquisition of increased sensitivity to a component of the innate immune response: the type I interferon-inducible TRIM22 protein, which acts as a restriction factor by inducing the polyubiquitination of the IAV nucleoprotein (NP). We show that four arginine residues present in the NP of the 1918 H1N1 pandemic strain and early postpandemic strains were progressively substituted for by lysines between 1918 and 2009, rendering NP more susceptible to TRIM22-mediated ubiquitination. Our observations suggest that during long-term evolution of IAVs in humans, variants endowed with increased susceptibility to TRIM22 restriction emerge, highlighting the complexity of selection pressures acting on the NP.


Assuntos
Evolução Molecular , Vírus da Influenza A Subtipo H1N1/genética , Antígenos de Histocompatibilidade Menor/metabolismo , Mutação , Proteínas de Ligação a RNA/genética , Proteínas Repressoras/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Proteínas do Core Viral/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Arginina/genética , Cães , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Influenza Humana/virologia , Lisina/genética , Células Madin Darby de Rim Canino , Mutagênese Sítio-Dirigida , Proteínas do Nucleocapsídeo , Estrutura Terciária de Proteína , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Replicação Viral
9.
J Interferon Cytokine Res ; 27(10): 857-64, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17970695

RESUMO

TRIM22 (Staf50) is an interferon (IFN)-inducible protein with unknown function. Recently, we identified TRIM22 as a novel p53 target gene and showed that overexpression of TRIM22 inhibits the clonogenic growth of monoblastic U937 cells. Moreover, expression of TRIM22 is high in lymphoid tissue, and levels decrease during T lymphocyte activation with CD3/CD2/CD28, suggesting that TRIM22 could exert antiproliferative effects. Here, a prominent increase in TRIM22 levels is observed during activation with interleukin-2 (IL-2) or IL-15 in contrast to the decrease observed during CD3/CD2/CD28-induced activation. However, stimulation of cells in these experiments was performed on crude T lymphocytes, allowing indirect regulation between different lymphocyte subtypes to take place. Therefore, to prevent interaction between different lymphocyte subtypes, expression of TRIM22 was examined during activation of sorted T lymphocyte subpopulations. In contrast to the marked changes of TRIM22 during activation of crude T lymphocytes, in isolated subpopulations, TRIM22 expression was not significantly affected in spite of IL-2-induced or CD3/CD2/CD28-induced activation. In addition, in contrast to the TRIM22 mouse ortholog Rpt-1, TRIM22 did not affect levels of CD25 (IL-2Ralpha) mRNA. Our data suggest a more complex role for TRIM22 during T lymphocyte activation than merely as an antiproliferative factor. TRIM22 probably has an activation stage-specific role connected to the paracrine crosstalk during T lymphocyte activation.


Assuntos
Interferons/farmacologia , Ativação Linfocitária/imunologia , Proteínas Repressoras/metabolismo , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/genética , Animais , Anticorpos/farmacologia , Antígenos CD2/imunologia , Antígenos CD28/imunologia , Complexo CD3/imunologia , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-15/farmacologia , Interleucina-2/farmacologia , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Células Jurkat , Leucócitos Mononucleares/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Antígenos de Histocompatibilidade Menor , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Proteínas com Motivo Tripartido
10.
Biochimie ; 89(6-7): 831-5, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17445960

RESUMO

The interferon (IFN) system is a major effector of the innate immunity that allows time for the subsequent establishment of an adaptive immune response against wide-range pathogens. The effectiveness of IFN to control initial infection requires the cooperation between several pathways induced in the target cells. Recent studies that highlight the implication of the 3'-5' exonuclease ISG20 (IFN Stimulated Gene product of 20 kDa) in the host's defenses against pathogens are summarised in this review.


Assuntos
Exonucleases/fisiologia , Imunidade Inata , Sequência de Aminoácidos , Animais , Linhagem Celular , Exonucleases/metabolismo , Exorribonucleases , Humanos , Inflamação , Interferons/metabolismo , Modelos Biológicos , Dados de Sequência Molecular , Vírus de RNA/metabolismo , RNA Interferente Pequeno/metabolismo , Receptores Toll-Like/metabolismo
11.
Leuk Res ; 31(7): 995-1001, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17257675

RESUMO

The interferon inducible protein TRIM22 has been identified as a p53 target gene, with possible involvement in proliferation and differentiation of leukaemia cells. Here, the expression levels of TRIM22 during haematopoietic differentiation are characterised. Expression of TRIM22 correlates inversely to differentiation, as TRIM22 is highly expressed in CD34(+) human bone marrow progenitor cells, but declines in mature populations. The erythroid lineage appears as a special case, as TRIM22 expression shows an extreme decrease during late erythroid maturation and is completely undetectable in nucleated erythroid populations in contrast to other lineages. In conclusion, our data could suggest lineage-specific roles for TRIM22 during haematopoietic differentiation.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular , Células Eritroides/citologia , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Antígenos CD34/metabolismo , Western Blotting , Regulação para Baixo , Citometria de Fluxo , Expressão Gênica , Granulócitos/citologia , Granulócitos/metabolismo , Humanos , Interferons/farmacologia , Antígenos de Histocompatibilidade Menor , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas com Motivo Tripartido
12.
Oncogene ; 23(26): 4636-40, 2004 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-15064705

RESUMO

Many interferon (IFN)-stimulated genes are also induced by double-stranded RNA (dsRNA), a component closely associated with the IFN system in the context of virus-host interactions. Recently, we demonstrated that the IFN-induced 3' --> 5' exonuclease ISG20 possesses antiviral activities against RNA viruses. Here we show that ISG20 induction by synthetic dsRNA (pIpC) is stronger and faster than its induction by IFN. Two families of transcription factors are implicated in the transcriptional activation of ISG20 by dsRNA. Initially, the NF-kappaB factors p50 and p65 bind and activate the kappaB element of the Isg20 promoter. This is followed by IRF1 binding to the ISRE. As pIpC often induces protein movements in the cells, we questioned whether it could influence ISG20 localization. Interestingly and contrary to IFN, dsRNA induces a nuclear matrix enrichment of the ISG20 protein. dsRNA induction of ISG20 via NF-kappaB and its antiviral activity led us to suggest that ISG20 could participate in the cellular response to virus infection.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Ligação a DNA/metabolismo , Exonucleases/metabolismo , NF-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , RNA de Cadeia Dupla/farmacologia , Sítios de Ligação , Proteínas de Transporte/efeitos dos fármacos , Proteínas de Transporte/genética , Citoplasma/metabolismo , Proteínas de Ligação a DNA/genética , Exonucleases/genética , Exorribonucleases , Células HeLa , Humanos , Fator Regulador 1 de Interferon , Interferon Tipo I/farmacologia , Mutagênese Sítio-Dirigida , NF-kappa B/genética , Proteínas Nucleares/efeitos dos fármacos , Proteínas Nucleares/genética , Fosfoproteínas/genética , Regiões Promotoras Genéticas , RNA de Cadeia Dupla/síntese química , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
13.
J Interferon Cytokine Res ; 35(11): 859-74, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26237181

RESUMO

Activation of innate immune response, induced after the recognition of double-stranded RNA (dsRNA), formed during replication of most viruses, results in intracellular signaling cascades ultimately culminating in the expression of type I interferon (IFN). In this study, we provide the first evidence that FoxO4 triggers the activation of the innate immune signaling pathway in coupling stimulation of TLR3 and RIG-like receptors by the synthetic dsRNA analog, poly(I:C), to IFN-ß and IFN-induced gene induction, whereas knockdown of FoxO4 had opposite effects. Similar effects of FoxO4 were observed during paramyxovirus-mediated IFN-ß transcriptional induction. We further found that knockdown of FoxO4 did not affect IRF3 and NF-κB activation by poly(I:C), suggesting that FoxO4 would act downstream in the signaling pathway. In addition, we show that the IFN-induced TRIM22 ubiquitin ligase targets FoxO4 and antagonizes its activity through an unrelated ubiquitin/autophagosomic-lysosomal pathway. Unexpectedly, TRIM22 knockdown strongly sensitizes cells to dsRNA-induced caspase-dependent apoptosis, as early as 2 h after poly(I:C) stimulation, concomitantly to the inhibition of the expression of the antiapoptotic protein, Bcl-2, indicating that TRIM22 might be a key factor for controlling the cell survival after TLR3 stimulation. Taken together, our data demonstrate that the regulation of FoxO4 protein expression and cell survival by TRIM22 controls TLR3-mediated IFN type I gene induction, preventing excessive antiviral response through dsRNA-induced apoptosis.


Assuntos
Apoptose/imunologia , Regulação da Expressão Gênica/imunologia , Interferon beta/biossíntese , Proteínas Repressoras/metabolismo , Receptor 3 Toll-Like/metabolismo , Fatores de Transcrição/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/genética , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Sobrevivência Celular/imunologia , Fatores de Transcrição Forkhead , Células HEK293 , Células HeLa , Humanos , Imunidade Inata/imunologia , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/genética , Antígenos de Histocompatibilidade Menor , NF-kappa B/metabolismo , Poli I-C/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , RNA de Cadeia Dupla/genética , Proteínas Repressoras/genética , Transdução de Sinais/imunologia , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Ativação Transcricional/imunologia , Proteínas com Motivo Tripartido , Ubiquitinação
14.
FEBS Lett ; 529(2-3): 232-6, 2002 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-12372606

RESUMO

To investigate acquired resistance to oxaliplatin, we selected two resistant clones from the HCT116 cell line. We found that the resistant phenotype was associated with resistance to oxaliplatin-induced apoptosis as demonstrated by FACS analysis and by Western blotting of caspase 3 activation. In addition, the resistant phenotype showed a concomitant resistance to lonidamine and arsenic trioxide which are inducers of mitochondrial apoptosis. Furthermore, a complete loss of Bax expression due to a frameshift mutation was observed in the most resistant clone. Taken together, these findings suggest that altered mitochondrial-mediated apoptosis could play a role in oxaliplatin resistance.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Neoplasias do Colo/patologia , Modelos Biológicos , Compostos Organoplatínicos/farmacologia , Trióxido de Arsênio , Arsenicais/farmacologia , Sequência de Bases , Western Blotting , Caspase 3 , Caspases/genética , Primers do DNA , Resistencia a Medicamentos Antineoplásicos , Citometria de Fluxo , Humanos , Indazóis/farmacologia , Oxaliplatina , Óxidos/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/genética
15.
Bull Cancer ; 90(2): 131-41, 2003 Feb.
Artigo em Francês | MEDLINE | ID: mdl-12660132

RESUMO

15 % of human cancers have virus origin, meaning that viruses are the second cause of cancers after tabagism. The knowledge of antiviral mechanisms is essential for treatment and prevention of infection evolution towards cancers. Interferons (IFNs) are a large family of multifunctional cytokines. They are involved in regulation of cell growth and modulation of immune response. But, all these functions seem to converge toward the most important of them : the antiviral activity. IFN secretion is the first event induced by viral infection, and will act on specific receptors on neighbour cells and prevent their infection by inducing numbers of antiviral genes. Although few of them are well known like the PKR, the 2-5OAS/RNase L pathway and the Mx proteins, many others need extensive studies to understand the wide range of IFN effect. Viruses have evolved to circumvent the IFN antiviral activity, and are able not only to divert the cellular machinery but also to lure the antiviral mechanisms of the host cell. The purpose of this review is to describe the many antiviral pathways and proteins induced by IFNs and to summarize the strategies of viral escape.


Assuntos
Interferons/fisiologia , Viroses/virologia , Animais , Endorribonucleases/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Humanos , Interferon Tipo I/fisiologia , Camundongos , Proteínas de Resistência a Myxovirus , Neoplasias/virologia , Transdução de Sinais/fisiologia , Viroses/metabolismo , eIF-2 Quinase/metabolismo
16.
J Gen Virol ; 90(Pt 3): 536-545, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19218198

RESUMO

The interferon (IFN) system is a major effector of the innate immunity that allows time for the subsequent establishment of an adaptive immune response against a wide-range of pathogens. Their diverse biological actions are thought to be mediated by the products of specific but usually overlapping sets of cellular genes induced in the target cells. Ubiquitin ligase members of the tripartite motif (TRIM) protein family have emerged as IFN-induced proteins involved in both innate and adaptive immunity. In this report, we provide evidence that TRIM22 is a functional E3 ubiquitin ligase that is also ubiquitinated itself. We demonstrate that TRIM22 expression leads to a viral protection of HeLa cells against encephalomyocarditis virus infections. This effect is dependent upon its E3 ubiquitinating activity, since no antiviral effect was observed in cells expressing a TRIM22-deletion mutant defective in ubiquitinating activity. Consistent with this, TRIM22 interacts with the viral 3C protease (3C(PRO)) and mediates its ubiquitination. Altogether, our findings demonstrate that TRIM22 E3 ubiquitin ligase activity represents a new antiviral pathway induced by IFN against picornaviruses.


Assuntos
Antivirais/metabolismo , Cisteína Endopeptidases/metabolismo , Vírus da Encefalomiocardite/efeitos dos fármacos , Proteínas Repressoras/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Virais/metabolismo , Proteases Virais 3C , Antivirais/farmacologia , Linhagem Celular , Cisteína Endopeptidases/genética , Vírus da Encefalomiocardite/enzimologia , Vírus da Encefalomiocardite/genética , Células HeLa , Humanos , Antígenos de Histocompatibilidade Menor , Proteínas Repressoras/genética , Proteínas Repressoras/farmacologia , Proteínas com Motivo Tripartido , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/farmacologia , Proteínas Virais/genética
17.
Virology ; 393(2): 311-8, 2009 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-19733381

RESUMO

RNA helicase-like receptors MDA-5 but not RIG-I has been shown to be essential for triggering innate immune responses against picornaviruses. However, virus-host co-evolution has selected for viruses capable of replicating despite host cells antiviral defences. In this report, we demonstrate that RIG-I is degraded during encephalomyocarditis virus (EMCV) infection. This effect is mediated by both the viral-encoded 3C protease and caspase proteinase. In addition, we show that RIG-I overexpression confers IFN-beta promoter activation during EMCV infection, in MDA-5 knockout (MDA-5(-/-)) mouse embryo fibroblasts. This induction is followed by a strong inhibition reflecting the ability of EMCV to disrupt RIG-I signalling. Taken together, our data strongly suggest that during evolution RIG-I has been involved for triggering innate immune response to picornavirus infections.


Assuntos
Infecções por Cardiovirus/imunologia , RNA Helicases DEAD-box/metabolismo , Vírus da Encefalomiocardite/imunologia , Imunidade Inata , Interferon beta/imunologia , Proteases Virais 3C , Animais , Infecções por Cardiovirus/virologia , Caspases/imunologia , Caspases/metabolismo , Cisteína Endopeptidases/imunologia , Cisteína Endopeptidases/metabolismo , Proteína DEAD-box 58 , RNA Helicases DEAD-box/imunologia , Vírus da Encefalomiocardite/metabolismo , Regulação da Expressão Gênica , Células HeLa , Humanos , Helicase IFIH1 Induzida por Interferon , Camundongos , Regiões Promotoras Genéticas , Proteínas Virais/imunologia , Proteínas Virais/metabolismo
18.
Microbiol Immunol ; 52(1): 30-5, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18352910

RESUMO

ISG20 is an ribonuclease specific for single-stranded RNA and considered to play a role in innate immunity against virus infections. We herein show that both poly IC, an authentic double-stranded RNA, and IFN-gamma induced ISG20 expression in cultured HUVEC. Poly IC-induced ISG20 expression was inhibited by LY294002, an inhibitor of PI3K, or by RNA interference against IFN regulatory factor three. ISG20 expression was not induced by IFN-beta, loxoribine or CpG oligonucleotide. These results suggest that ISG20 induction by poly IC may not be dependent on the IRF-3-mediated type I IFN induction pathway in HUVEC. ISG20 may be involved in innate immunity against viral infection in vascular endothelial cells.


Assuntos
Células Endoteliais/imunologia , Exonucleases/biossíntese , Perfilação da Expressão Gênica , Interferons/imunologia , Células Cultivadas , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Exorribonucleases , Guanosina/análogos & derivados , Guanosina/farmacologia , Humanos , Fatores Imunológicos/farmacologia , Interferon beta/farmacologia , Morfolinas/farmacologia , Oligodesoxirribonucleotídeos/farmacologia , Poli I-C/imunologia
19.
J Interferon Cytokine Res ; 28(12): 733-40, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18937547

RESUMO

Interferons (IFNs) have proven antitumor activity against a variety of human malignancies, which may result, at least in part, from inhibition of angiogenesis. The objective of this study was to identify IFN-stimulated genes (ISGs) that played a role in mediation of angiogenic inhibition. IFN-beta was a more potent antiangiogenic agent compared to IFN-alpha2b (80% versus 20%, respectively) and suggests that IFNs inhibited angiogenesis by preventing endothelial cell differentiation, and not by direct antiproliferative effects. To identify ISGs that were key inhibitors of angiogenesis, we utilized an in vitro fibrin gel angiogenic assay which closely recapitulated the in vivo processes of angiogenesis. DNA microarray analysis of IFN-beta-treated endothelial cells in the fibrin gel assay identified 11 ISGs that were induced >10-fold during angiogenesis inhibition. Recombinant IP-10 inhibited angiogenesis in a dose-dependent fashion, but was a less effective inhibitor compared to IFN-beta, suggesting that additional ISGs are involved in inhibiting angiogenesis. ISG20 was upregulated by microarray analysis, but did not inhibit angiogenesis when overexpressed in human umbilical vein endothelial cells (HUVECs). However, a dominant negative mutant of ISG20 inhibited angiogenesis by 43%. Results suggest that IFN-induced angiogenic inhibition was likely mediated by multiple ISGs; our novel finding is that decreased exonuclease activity in HUVECs associated with expression of the ISG20 ExoII mutant inhibited angiogenesis.


Assuntos
Inibidores da Angiogênese/farmacologia , Endotélio Vascular/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Interferon beta/farmacologia , Neovascularização Patológica/imunologia , Neovascularização Fisiológica/efeitos dos fármacos , Animais , Quimiocina CXCL10/farmacologia , Quimiocina CXCL11/farmacologia , Exonucleases/genética , Exorribonucleases , Perfilação da Expressão Gênica , Humanos , Camundongos , Camundongos Nus , Neovascularização Patológica/genética , Neovascularização Fisiológica/genética , Análise de Sequência com Séries de Oligonucleotídeos , Veias Umbilicais/efeitos dos fármacos , Veias Umbilicais/metabolismo , Regulação para Cima/efeitos dos fármacos
20.
J Cell Biochem ; 98(5): 1320-33, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16514659

RESUMO

We have previously shown that ISG20, an interferon (IFN)-induced gene, encodes a 3' to 5' exoribonuclease member of the DEDD superfamily of exonucleases. ISG20 specifically degrades single-stranded RNA. In this report, using immunofluorescence analysis, we demonstrate that in addition to a diffuse cytoplasmic and nucleoplasmic localization, the endogenous ISG20 protein was present in the nucleus both in the nucleolus and in the Cajal bodies (CBs). In addition, we show that the ectopic expression of the CBs signature protein, coilin, fused to the red fluorescent protein (coilin-dsRed) increased the number of nuclear dots containing both ISG20 and coilin-dsRed. Using electron microcopy analysis, ISG20 appeared principally concentrated in the dense fibrillar component of the nucleolus, the major site for rRNA processing. We also present evidences that ISG20 was associated with survival of motor neuron (SMN)-containing macromolecular nuclear complexes required for the biogenesis of various small nuclear ribonucleoproteins. Finally, we demonstrate that ISG20 was associated with U1 and U2 snRNAs, and U3 snoRNA. The accumulation of ISG20 in the CBs after IFN treatment strongly suggests its involvement in a new route for IFN-mediated inhibition of protein synthesis by modulating snRNA and rRNA maturation.


Assuntos
Nucléolo Celular/enzimologia , Corpos Enovelados/enzimologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Exonucleases/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas de Ligação a RNA/metabolismo , Nucléolo Celular/efeitos dos fármacos , Nucléolo Celular/ultraestrutura , Corpos Enovelados/efeitos dos fármacos , Corpos Enovelados/ultraestrutura , Exorribonucleases , Células HeLa , Humanos , Interferons/farmacologia , Microscopia Imunoeletrônica , Ligação Proteica , RNA Nuclear Pequeno/metabolismo , Proteínas do Complexo SMN
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA