Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
FASEB J ; 27(7): 2777-87, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23572232

RESUMO

Coxsackievirus B3 (CVB3) is a causative agent of viral myocarditis, hepatitis, pancreatitis, and meningitis in humans. The adenosine-uridine (AU)-rich element RNA binding factor 1 (AUF1) is an integral component in the regulation of gene expression. AUF1 destabilizes mRNAs and targets them for degradation by binding to AU-rich elements in the 3' untranslated region (UTR) of mRNAs. The 3'-UTR of the CVB3 genome contains canonical AU-rich sequences, raising the possibility that CVB3 RNA may also be subjected to AUF1-mediated degradation. Here, we reported that CVB3 infection led to cytoplasmic redistribution and cleavage of AUF1. These events are independent of CVB3-induced caspase activation but require viral protein production. Overexpression of viral protease 2A reproduced CVB3-induced cytoplasmic redistribution of AUF1, while in vitro cleavage assay revealed that viral protease 3C contributed to AUF1 cleavage. Furthermore, we showed that knockdown of AUF1 facilitated viral RNA, protein, and progeny production, suggesting an antiviral property for AUF1 against CVB3 infection. Finally, an immunoprecipitation study demonstrated the physical interaction between AUF1 and the 3'-UTR of CVB3, potentially targeting CVB3 genome toward degradation. Together, our results suggest that cleavage of AUF1 may be a strategy employed by CVB3 to enhance the stability of its viral genome.


Assuntos
Citoplasma/metabolismo , Enterovirus Humano B/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo D/metabolismo , RNA Viral/metabolismo , Regiões 3' não Traduzidas/genética , Western Blotting , Cisteína Endopeptidases/genética , Cisteína Endopeptidases/metabolismo , Citocinas/genética , Citocinas/metabolismo , Citoplasma/virologia , Enterovirus Humano B/genética , Enterovirus Humano B/fisiologia , Expressão Gênica , Genoma Viral/genética , Células HeLa , Ribonucleoproteína Nuclear Heterogênea D0 , Ribonucleoproteínas Nucleares Heterogêneas Grupo D/genética , Interações Hospedeiro-Patógeno/genética , Humanos , Microscopia Confocal , Modelos Genéticos , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Ligação Proteica , Transporte Proteico , Proteólise , Interferência de RNA , Estabilidade de RNA , RNA Viral/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral/genética
2.
Virol J ; 11: 192, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25410318

RESUMO

BACKGROUND: Stress granules (SGs) are granular aggregates in the cytoplasm that are formed under a variety of stress situations including viral infection. Previous studies indicate that poliovirus, a member of Picornaviridae, can induce SG formation. However, the exact mechanism by which the picornaviruses induce SG formation is unknown. METHOD: The localization of SG markers in cells infected with coxsackievirus B3 (CVB3) or enterovirus 71 (EV71) and in cells expressing each viral protein was determined via immunofluorescence assays or plasmid transfection. Eight plasmids expressing mutants of the 2A protease (2A(pro)) of CVB3 were generated using a site-directed mutagenesis strategy. The cleavage efficiencies of eIF4G by CVB3 2A(pro) and its mutants were determined via western blotting assays. RESULTS: In this study, we found that CVB3 infection induced SG formation, as evidenced by the co-localization of some accepted SG markers in viral infection-induced granules. Furthermore, we identified that 2A(pro) of CVB3 was the key viral component that triggered SG formation. A 2A(pro) mutant with the G122E mutation, which exhibited very low cleavage efficiency toward eIF4G, significantly attenuated its capacity for SG induction, indicating that the protease activity was required for 2A(pro) to initiate SG formation. Finally, we observed that SGs also formed in EV71-infected cells. Expression of EV71 2A(pro) alone was also sufficient to cause SG formation. CONCLUSION: Both CVB3 and EV71 infections can induce SG formation, and 2A(pro) plays a crucial role in the induction of SG formation during these infections. This finding may help us to better understand how picornaviruses initiate the SG response.


Assuntos
Cisteína Endopeptidases/metabolismo , Grânulos Citoplasmáticos/metabolismo , Enterovirus Humano A/fisiologia , Enterovirus Humano B/fisiologia , Interações Hospedeiro-Patógeno , Proteínas Virais/metabolismo , Fator de Iniciação Eucariótico 4G/metabolismo , Imunofluorescência , Humanos , Proteólise
3.
Proc Natl Acad Sci U S A ; 107(39): 17053-8, 2010 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-20833815

RESUMO

Signal transduction networks can be perturbed biochemically, genetically, and pharmacologically to unravel their functions. But at the systems level, it is not clear how such perturbations are best implemented to extract molecular mechanisms that underlie network function. Here, we combined pairwise perturbations with multiparameter phosphorylation measurements to reveal causal mechanisms within the signaling network response of cardiomyocytes to coxsackievirus B3 (CVB3) infection. Using all possible pairs of six kinase inhibitors, we assembled a dynamic nine-protein phosphorylation signature of perturbed CVB3 infectivity. Cluster analysis of the resulting dataset showed repeatedly that paired inhibitor data were required for accurate data-driven predictions of kinase substrate links in the host network. With pairwise data, we also derived a high-confidence network based on partial correlations, which identified phospho-IκBα as a central "hub" in the measured phosphorylation signature. The reconstructed network helped to connect phospho-IκBα with an autocrine feedback circuit in host cells involving the proinflammatory cytokines, TNF and IL-1. Autocrine blockade substantially inhibited CVB3 progeny release and improved host cell viability, implicating TNF and IL-1 as cell autonomous components of CVB3-induced myocardial damage. We conclude that pairwise perturbations, when combined with network-level intracellular measurements, enrich for mechanisms that would be overlooked by single perturbants.


Assuntos
Enterovirus Humano B , Infecções por Enterovirus/metabolismo , Interações Hospedeiro-Patógeno , Redes e Vias Metabólicas , Miócitos Cardíacos/virologia , Linhagem Celular , Humanos , Interleucina-1/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
4.
J Virol ; 84(21): 11359-73, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20702616

RESUMO

Respiratory viruses exert a heavy toll of morbidity and mortality worldwide. Despite this burden there are few specific treatments available for respiratory virus infections. Since many viruses utilize host cell enzymatic machinery such as protein kinases for replication, we determined whether pharmacological inhibition of kinases could, in principle, be used as a broad antiviral strategy for common human respiratory virus infections. A panel of green fluorescent protein (GFP)-expressing recombinant respiratory viruses, including an isolate of H1N1 influenza virus (H1N1/Weiss/43), was used to represent a broad range of virus families responsible for common respiratory infections (Adenoviridae, Paramyxoviridae, Picornaviridae, and Orthomyxoviridae). Kinase inhibitors were screened in a high-throughput assay that detected virus infection in human airway epithelial cells (1HAEo-) using a fluorescent plate reader. Inhibition of p38 mitogen-activated protein kinase (MAPK) signaling was able to significantly inhibit replication by all viruses tested. Therefore, the pathways involved in virus-mediated p38 and extracellular signal-regulated kinase (ERK) MAPK activation were investigated using bronchial epithelial cells and primary fibroblasts derived from MyD88 knockout mouse lungs. Influenza virus, which activated p38 MAPK to approximately 10-fold-greater levels than did respiratory syncytial virus (RSV) in 1HAEo- cells, was internalized about 8-fold faster and more completely than RSV. We show for the first time that p38 MAPK is a determinant of virus infection that is dependent upon MyD88 expression and Toll-like receptor 4 (TLR4) ligation. Imaging of virus-TLR4 interactions showed significant clustering of TLR4 at the site of virus-cell interaction, triggering phosphorylation of downstream targets of p38 MAPK, suggesting the need for a signaling receptor to activate virus internalization.


Assuntos
Infecções Respiratórias/virologia , Receptor 4 Toll-Like/fisiologia , Tropismo Viral , Internalização do Vírus , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Humanos , Fator 88 de Diferenciação Mieloide/biossíntese , Fosforilação
5.
Circ Res ; 104(6): 813-21, 2009 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-19213955

RESUMO

Reduced cardiac output is one of the consequences of myocarditis. Bosentan, an endothelin-1 receptor (ET1R) antagonist, could be useful to reduce cardiac afterload, preserving cardiac output. In this study, we investigated the potential therapeutic use of bosentan in an animal model of viral myocarditis. Using a mouse model of coxsackievirus B3 (CVB3)-induced myocarditis, we demonstrated preserved ejection fraction (EF) and fractional shortening (FS) by treatment with bosentan (68+/-5.8% EF and 40+/-3.7% FS for treated versus 48+/-2.2% EF and 25+/-2.6% FS for controls; P=0.028). However, bosentan enhanced cardiac viral load (10.4+/-6.7% in the bosentan group versus 5.0+/-5.5% in control group; P=0.02), likely through enhancement of p38 mitogen-activated protein kinase (MAPK) phosphorylation (0.77+/-0.40% ATF2 activation in the bosentan group versus 0.03+/-0.02% in controls; P=0.0002), mediated by endothelin receptor type-A. We further demonstrate that a water soluble inhibitor of p38 MAPK, SB203580 HCl, is a potent inhibitor of virus replication in the heart (0.28% antisense viral genome stained area for 3 mg/kg dose versus 2.9% stained area for controls; P=0.01), attenuates CVB3-induced myocardial damage (blinded cardiac histopathologic scores of 1.8+/-1.6 and 2.05+/-1.2 for the 3 mg/kg and 10 mg/kg doses, respectively, versus 3.25+/-1.2 for the controls), and preserves cardiac function (69+/-3.5% EF for 3 mg/kg dose and 71+/-6.7% EF for 10 mg/kg dose versus 60+/-1.5% EF control; P=0.038 and P=0.045, as compared to control, respectively). Bosentan, a prescribed vasodilator, improves cardiac function but enhances viral load and myocarditis severity through ETRA mediated p38 MAPK activation; p38 MAPK is a desirable antiviral target. Caution must be exercised during treatment of suspected infectious myocarditis with supportive vasoactive remedies.


Assuntos
Anti-Hipertensivos/farmacologia , Infecções por Coxsackievirus/enzimologia , Infecções por Coxsackievirus/fisiopatologia , Antagonistas do Receptor de Endotelina A , Enterovirus Humano B , Miocardite/enzimologia , Miocardite/fisiopatologia , Volume Sistólico/efeitos dos fármacos , Sulfonamidas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Bosentana , Baixo Débito Cardíaco/tratamento farmacológico , Baixo Débito Cardíaco/enzimologia , Baixo Débito Cardíaco/fisiopatologia , Baixo Débito Cardíaco/virologia , Infecções por Coxsackievirus/tratamento farmacológico , Modelos Animais de Doenças , Ativação Enzimática/efeitos dos fármacos , Humanos , Camundongos , Miocardite/tratamento farmacológico , Miocardite/virologia , Carga Viral/métodos
6.
J Virol ; 82(18): 9143-53, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18596087

RESUMO

Recent studies suggest a possible takeover of host antimicrobial autophagy machinery by positive-stranded RNA viruses to facilitate their own replication. In the present study, we investigated the role of autophagy in coxsackievirus replication. Coxsackievirus B3 (CVB3), a picornavirus associated with viral myocarditis, causes pronounced intracellular membrane reorganization after infection. We demonstrate that CVB3 infection induces an increased number of double-membrane vesicles, accompanied by an increase of the LC3-II/LC3-I ratio and an accumulation of punctate GFP-LC3-expressing cells, two hallmarks of cellular autophagosome formation. However, protein expression analysis of p62, a marker for autophagy-mediated protein degradation, showed no apparent changes after CVB3 infection. These results suggest that CVB3 infection triggers autophagosome formation without promoting protein degradation by the lysosome. We further examined the role of the autophagosome in CVB3 replication. We demonstrated that inhibition of autophagosome formation by 3-methyladenine or small interfering RNAs targeting the genes critical for autophagosome formation (ATG7, Beclin-1, and VPS34 genes) significantly reduced viral replication. Conversely, induction of autophagy by rapamycin or nutrient deprivation resulted in increased viral replication. Finally, we examined the role of autophagosome-lysosome fusion in viral replication. We showed that blockage of the fusion by gene silencing of the lysosomal protein LAMP2 significantly promoted viral replication. Taken together, our results suggest that the host's autophagy machinery is activated during CVB3 infection to enhance the efficiency of viral replication.


Assuntos
Autofagia/fisiologia , Enterovirus Humano B/fisiologia , Enterovirus Humano B/patogenicidade , Fagossomos/ultraestrutura , Replicação Viral/fisiologia , Adenina/análogos & derivados , Adenina/farmacologia , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteína 7 Relacionada à Autofagia , Proteína Beclina-1 , Linhagem Celular , Células HeLa , Humanos , Lisossomos/fisiologia , Lisossomos/ultraestrutura , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Fagossomos/fisiologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Enzimas Ativadoras de Ubiquitina/genética , Enzimas Ativadoras de Ubiquitina/metabolismo
7.
J Cell Biol ; 160(1): 137-46, 2003 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-12515828

RESUMO

We investigated the molecular and cellular actions of receptor protein tyrosine phosphatase (PTP) alpha in integrin signaling using immortalized fibroblasts derived from wild-type and PTP alpha-deficient mouse embryos. Defects in PTP alpha-/- migration in a wound healing assay were associated with altered cell shape and focal adhesion kinase (FAK) phosphorylation. The reduced haptotaxis to fibronectin (FN) of PTP alpha-/- cells was increased by expression of active (but not inactive) PTP alpha. Integrin-mediated formation of src-FAK and fyn-FAK complexes was reduced or abolished in PTP alpha-/- cells on FN, concomitant with markedly reduced phosphorylation of FAK at Tyr397. Reintroduction of active (but not inactive) PTP alpha restored FAK Tyr-397 phosphorylation. FN-induced cytoskeletal rearrangement was retarded in PTP alpha-/- cells, with delayed filamentous actin stress fiber assembly and focal adhesion formation. This mimicked the effects of treating wild-type fibroblasts with the src family protein tyrosine kinase (Src-PTK) inhibitor PP2. These results, together with the reduced src/fyn tyrosine kinase activity in PTP alpha-/- fibroblasts (Ponniah et al., 1999; Su et al., 1999), suggest that PTP alpha functions in integrin signaling and cell migration as an Src-PTK activator. Our paper establishes that PTP alpha is required for early integrin-proximal events, acting upstream of FAK to affect the timely and efficient phosphorylation of FAK Tyr-397.


Assuntos
Citoesqueleto/metabolismo , Integrinas/metabolismo , Fosforilação , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Fosfatases/fisiologia , Proteínas Tirosina Quinases/metabolismo , Receptores de Superfície Celular , Adenoviridae/metabolismo , Animais , Catálise , Adesão Celular , Movimento Celular , Meios de Cultura Livres de Soro/farmacologia , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Immunoblotting , Camundongos , Microscopia de Fluorescência , Testes de Precipitina , Proteínas Tirosina Fosfatases Classe 4 Semelhantes a Receptores , Tirosina/metabolismo , Vinculina/biossíntese
8.
Circ Res ; 99(4): 354-61, 2006 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-16840719

RESUMO

Viral myocarditis is a major cause of sudden cardiac death in children and young adults. Among viruses, coxsackievirus B3 (CVB3) is the most common agent for myocarditis. Recently, more consideration has been given to the role of signaling pathways in pathogenesis of enteroviral myocarditis, providing new platform for identifying a new potential therapeutic target for this, so far, incurable disease. Previously, we reported on the role of the protein kinase-B/Akt in CVB3 replication and virus-induced cell injury. Here, we report on regulation of virus-induced Akt activation by the integrin-linked kinase in infected mouse cardiomyocytes and HeLa cells. This study also presents the first observation that inhibition of ILK in CVB3-infected cells significantly improves the viability of infected cells, while blocking viral replication and virus release. Complementary experiments using a constitutively active form of Akt1 revealed that the observed protective effect of ILK inhibition is dependent on the associated downregulation of virus-induced Akt activation. To our knowledge, this is the first report of such beneficial effects of ILK inhibition in a viral infection model and conveys new insights in our efforts to characterize a novel therapeutic target for treatment of enteroviral myocarditis.


Assuntos
Enterovirus/fisiologia , Coração/virologia , Células Musculares/virologia , Miocardite/virologia , Proteínas Serina-Treonina Quinases/metabolismo , Replicação Viral , Animais , Linhagem Celular , Enterovirus/genética , Inibidores Enzimáticos/farmacologia , Células HeLa , Humanos , Hibridização In Situ , Camundongos , Células Musculares/enzimologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Viral/genética
9.
FEBS Lett ; 581(5): 879-84, 2007 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-17292893

RESUMO

Apoptosis repressor with caspase recruitment domain (ARC), an anti-apoptotic protein, is highly expressed in differentiated heart and skeletal muscle. Apoptosis and differentiation share numerous common pathways; therefore, we examined the impact of ARC on H9c2-myoblast differentiation. We demonstrate that ARC expression levels increase and stabilize upon differentiation. ARC-overexpression in pre-differentiated H9c2-cells suppresses differentiation; indicated by increased myotube formation, nuclear fusion and expression of the differentiation markers myogenin and troponin-T. ARC-overexpression inhibited myoblast differentiation associated caspase-3 activation, suggesting ARC inhibits myogenic differentiation through caspase inhibition. In summary, we show a novel role for ARC in the regulation of muscle differentiation.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Musculares/metabolismo , Mioblastos Cardíacos/citologia , Mioblastos Cardíacos/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Caspase 3/metabolismo , Diferenciação Celular/fisiologia , Linhagem Celular , Ativação Enzimática , Expressão Gênica , Desenvolvimento Muscular/fisiologia , Proteínas Musculares/genética , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfecção
10.
J Virol Methods ; 133(2): 211-8, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16375979

RESUMO

Liposome-mediated gene delivery provides a powerful strategy for the study of gene function and for gene therapy. Coxsackievirus B3 is an important human pathogen associated with various diseases. Here we reported that liposome-mediated transient transfection of plasmid cDNA inhibited coxsackieviral replication at the levels of RNA, protein and viral progeny release. These inhibitory effects were observed in various cell types and by using different liposome reagents. We further showed that the inhibition was likely due to the lack of virus attachment. Moreover, we showed that addition of cholesterol restored, at least in part, the viral infectivity. Interestingly, we found that membrane cholesterol levels were unchanged during transfection, indicating that disruption rather than depletion of membrane cholesterol contributes to the inhibitory effects of transfection. Our data suggest that liposome-mediated cDNA transient transfection inhibits coxsackievirus infectivity via inhibition of viral attachment, which is likely occurring through the changes of membrane cholesterol integrity.


Assuntos
Colesterol/metabolismo , Infecções por Coxsackievirus/prevenção & controle , Enterovirus Humano B/crescimento & desenvolvimento , Enterovirus Humano B/fisiologia , Lipossomos , Transfecção/métodos , Western Blotting , Colesterol/farmacologia , Enterovirus Humano B/efeitos dos fármacos , Técnica Indireta de Fluorescência para Anticorpo , Células HeLa , Humanos , Imuno-Histoquímica , Receptores Virais/metabolismo , Ensaio de Placa Viral
11.
Methods Mol Med ; 112: 291-303, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16010025

RESUMO

Cardiac hypertrophy is a compensatory response to a variety of physiological or pathological stimuli. However, prolonged hypertrophic responses may eventually lead to heart failure, arrhythmia, and sudden death. A number of intracellular signaling pathways have been implicated to play a critical role in the regulation of cardiac hypertrophy. In this chapter, the mitogen-activated protein kinase signaling pathway is used to illustrate conventional assays to detect the expression, phosphorylation, and activation of signaling proteins during cardiac hypertrophy, including Western blot, immunohistochemical staining, and immune complex kinase assays. Newly emerging techniques for analyzing cell signaling are also discussed in this chapter. Identifying and characterizing the expression and activation of these signaling proteins will provide important insights into the mechanisms that regulate hypertrophic cell growth and assist in development of new therapeutic approaches to limit cardiac hypertrophy.


Assuntos
Cardiomegalia/metabolismo , Cardiomegalia/patologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Transdução de Sinais , Adenoviridae/genética , Animais , Western Blotting , Células Cultivadas , Ativação Enzimática , Técnicas de Transferência de Genes , Vetores Genéticos , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/metabolismo , Fosforilação , Testes de Precipitina , Interferência de RNA
12.
Virus Res ; 195: 207-16, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25456405

RESUMO

Enterovirus 71 (EV71) is the main causative pathogen of hand, foot, and mouth disease (HFMD). The severe neurological complications caused by EV71 infection and the lack of effective therapeutic medicine underline the importance of searching for antiviral substances. Pyrrolidine dithiocarbamate (PDTC), an antioxidant, has been reported to inhibit the replication of coxsackievirus B (CVB) through dysregulating ubiquitin-proteasome system (UPS). In this study, we demonstrated that PDTC exerted potent antiviral effect on EV71. Viral RNA synthesis, viral protein expression, and the production of viral progeny were significantly reduced by the treatment of PDTC in Vero cells infected with EV71. Similar to the previous report about the inhibitory effect of PDTC on UPS, we found that PDTC treatment led to decreased levels of polyubiquitinated proteins in EV71-infected cells. The inhibitory effect of PDTC on UPS was further confirmed by the increased accumulation of cell cycle regulatory proteins p21 and p53, which are normally degraded through UPS, while the expression levels of both proteins remained unchanged. We also showed that PDTC had no impact on the activity of proteasome. Thus, we demonstrated that the down-regulation of PDTC on UPS was the result of its inhibition on ubiquitination. More importantly, this study provides evidence that the inhibition on UPS was required for the antiviral activity of PDTC, since MG132, a potent proteasome inhibitor, significantly inhibited the cytopathic effect and viral protein synthesis in EV71-infected cells. We also found that the antioxidant property of PDTC did not contribute to its antiviral effect, since N-acetyl-l-cysteine, a potent antioxidant, could not inhibit viral replication. In addition, CPE and viral protein synthesis were not inhibited in the cells pretreated with PDTC 2h before viral infection and then cultured in the media with no PDTC supplement, while the antioxidant effect of PDTC was retained. PDTC also showed significant inhibition on apoptosis induced by EV71 infection when it was applied at the early stage of viral infection. Our results collectively suggest that PDTC could be a potential anti-EV71 compound which possesses both antiviral and anti-apoptotic capacity.


Assuntos
Antivirais/farmacologia , Enterovirus Humano A/efeitos dos fármacos , Enterovirus Humano A/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Pirrolidinas/farmacologia , Tiocarbamatos/farmacologia , Ubiquitina/metabolismo , Replicação Viral/efeitos dos fármacos , Animais , Chlorocebus aethiops , Efeito Citopatogênico Viral/efeitos dos fármacos , RNA Viral/análise , Ubiquitinação/efeitos dos fármacos , Células Vero , Proteínas Virais/análise
13.
Acta Pharm Sin B ; 4(4): 284-94, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26579397

RESUMO

Human enterovirus 71 (EV71) is the main causative pathogen of hand, foot, and mouth disease (HFMD) in children. The epidemic of HFMD has been a public health problem in Asia-Pacific region for decades, and no vaccine and effective antiviral medicine are available. Curcumin has been used as a traditional medicine for centuries to treat a diversity of disorders including viral infections. In this study, we demonstrated that curcumin showed potent antiviral effect again EV71. In Vero cells infected with EV71, the addition of curcumin significantly suppressed the synthesis of viral RNA, the expression of viral protein, and the overall production of viral progeny. Similar with the previous reports, curcumin reduced the production of ROS induced by viral infection. However, the antioxidant property of curcumin did not contribute to its antiviral activity, since N-acetyl-l-cysteine, the potent antioxidant failed to suppress viral replication. This study also showed that extracellular signal-regulated kinase (ERK) was activated by either viral infection or curcumin treatment, but the activated ERK did not interfere with the antiviral effect of curcumin, indicating ERK is not involved in the antiviral mechanism of curcumin. Unlike the previous reports that curcumin inhibited protein degradation through ubiquitin-proteasome system (UPS), we found that curcumin had no impact on UPS in control cells. However, curcumin did reduce the activity of proteasomes which was increased by viral infection. In addition, the accumulation of the short-lived proteins, p53 and p21, was increased by the treatment of curcumin in EV71-infected cells. We further probed the antiviral mechanism of curcumin by examining the expression of GBF1 and PI4KB, both of which are required for the formation of viral replication complex. We found that curcumin significantly reduced the level of both proteins. Moreover, the decreased expression of either GBF1 or PI4KB by the application of siRNAs was sufficient to suppress viral replication. We also demonstrated that curcumin showed anti-apoptotic activity at the early stage of viral infection. The results of this study provide solid evidence that curcumin has potent anti-EV71 activity. Whether or not the down-regulated GBF1 and PI4KB by curcumin contribute to its antiviral effect needs further studies.

14.
Virus Res ; 192: 52-61, 2014 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-25148713

RESUMO

Stress granules (SGs) are cytoplasmic granules that are formed in cells when stress occurs. In this study, we found that SGs formed in cells infected with coxsackievirus B3 (CVB3), evidenced with the co-localization of some accepted SG markers in the viral infection-induced granules. We further discovered that adenosine-uridine (AU)-rich element RNA binding factor 1 (AUF1), which can bind to mRNAs and regulate their translation, was recruited to the SGs in response to high dose of CVB3 by detecting the co-localization of AUF1 with SG markers. Similar results were also observed in the enterovirus 71 (EV71)-infected cells. Finally, we demonstrated that AUF1 was also recruited to arsenite-induced SGs, suggesting that the recruitment of AUF1 to SG is not a specific response to viral infection. In summary, our data indicate that both CVB3 and EV71 infections can induce SG formation, and AUF1 is a novel SG component upon the viral infections. Our findings may shed light on understanding the picornavirus-host interaction.


Assuntos
Grânulos Citoplasmáticos/química , Enterovirus Humano B/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo D/análise , Interações Hospedeiro-Patógeno , Arsenitos/toxicidade , Enterovirus Humano A/fisiologia , Células HeLa , Ribonucleoproteína Nuclear Heterogênea D0 , Humanos , Microscopia Confocal , Microscopia de Fluorescência , Estresse Fisiológico
15.
Antiviral Res ; 101: 82-92, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24263113

RESUMO

Enterovirus 71 (EV71), a member of Picornaviridae, is one of the major pathogens of human hand, foot and mouth disease. EV71 mainly infects children and causes severe neurological complications and even death. The pathogenesis of EV71 infection is largely unknown, and no clinically approved vaccine or effective treatment is available to date. Here we described a novel bioluminescence imaging approach for EV71 detection. In this approach, a plasmid-based reporter was constructed to express the fusion protein AmN(Q/G)BC, a split firefly luciferase mutant, which can be specifically cleaved by EV71 protease 3C(pro). Upon cleavage, the splitting fusion protein restores luciferase activity. Our test confirmed that AmN(Q/G)BC was specifically cleaved by 3C(pro) and EV71 and restored the luciferase activity to a degree that corresponds to the 3C(pro) and virus doses in cells and mice. The anti-EV71 effect of GW5074 and U0126, two mitogen-activated protein kinase (MAPK) inhibitors, was evaluated using this approach to validate its application of screening anti-EV71 agents. We found that the AmN(Q/G)BC reporter efficiently monitored the inhibitory effect of GW5074 and U0126 on EV71 infection under in vitro and in vivo conditions. The data from AmN(Q/G)BC reporter were consistent with Western blotting and histopathology examination. Taken together, this real-time imaging approach can quantitatively monitor the efficacy of anti-EV71 agents and is valuable for anti-EV71 drug screening and evaluation, especially, under in vivo conditions.


Assuntos
Antivirais/farmacologia , Cisteína Endopeptidases/metabolismo , Enterovirus Humano A/efeitos dos fármacos , Medições Luminescentes/métodos , Imagem Óptica/métodos , Proteínas Virais/metabolismo , Proteases Virais 3C , Animais , Antivirais/isolamento & purificação , Linhagem Celular , Cisteína Endopeptidases/genética , Avaliação Pré-Clínica de Medicamentos/métodos , Luciferases de Vaga-Lume/genética , Luciferases de Vaga-Lume/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Proteínas Virais/genética
16.
Virology ; 433(2): 513-21, 2012 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-23010168

RESUMO

The capsid proteins of some RNA viruses can translocate to the nucleus and interfere with cellular phenotypes. In this study we found that the VP1 capsid protein of coxsackievirus B3 (CVB3) was dominantly localized in the nucleus of the cells transfected with VP1-expressing plasmid. The VP1 nuclear localization also occurred in the cells infected with CVB3. Truncation analysis indicated that the VP1 nuclear localization sequence located near the C-terminal. The substitution of His220 with threonine completely abolished its translocation. The VP1 proteins of other CVB types might have the nuclear localization potential because this region was highly conserved. Moreover, the VP1 nuclear localization induced cell cycle deregulation, including a prolonged S phase and shortened G2-M phase. Besides these findings, we also found a domain between Ala72 and Phe106 that caused the VP1 truncates dotted distributed in the cytoplasm. Our results suggest a new pathogenic mechanism of CVB.


Assuntos
Enterovirus Humano B/genética , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/metabolismo , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Ciclo Celular , Núcleo Celular/metabolismo , Núcleo Celular/virologia , Enterovirus Humano B/metabolismo , Enterovirus Humano B/patogenicidade , Células HeLa , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Transfecção
17.
J Gen Virol ; 90(Pt 4): 854-862, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19264606

RESUMO

Clathrin- and caveolae-mediated endocytosis have been implicated in the productive entry of many viruses into host cells. ADP-ribosylation factor 6 (Arf6)-dependent endocytosis is another endocytosis pathway that traffics from the cell surface and it is the only Arf that traffics at the plasma membrane. However, little is known about Arf6-dependent trafficking during virus entry. This study showed that coxsackievirus type B3 (CVB3) associated with decay-accelerating factor in non-polarized HeLa cells can be redirected into non-productive compartments by Arf6-dependent internalization, thus restricting infection. Overexpression of wild-type (WT) and constitutively active (CA) Arf6 in HeLa cells resulted in a 2.3- and 3.6-fold decrease in infection, respectively. A dominant-negative inhibitor of Arf6 recovered restriction of infection by WT-Arf6 and CA-Arf6. RNA interference of endogenous Arf6 resulted in a 3.3-fold increase in CVB3 titre in HeLa cells. It was shown that coxsackie-adenovirus receptor (CAR) ligation by virus or CAR-specific antibody could activate extracellular signal-regulated kinase (ERK) of the mitogen-activated protein kinase family and lead to Arf6-mediated viral restriction. In the absence of ERK activation, CVB3 internalization into early endosomes was inhibited and subsequent infection was reduced, but Arf6-mediated restriction was also abolished. In conclusion, receptor-mediated signalling enhances CVB3 entry whilst also activating non-productive pathways of virus entry; thus, virus infection is an equilibrium of productive and non-productive pathways of entry.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Enterovirus Humano B/patogenicidade , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/genética , Linhagem Celular , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Endocitose , Endossomos/virologia , Enterovirus Humano B/genética , Enterovirus Humano B/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/genética , Células HeLa , Humanos , Receptores Virais/genética , Receptores Virais/metabolismo , Transdução de Sinais
18.
Int J Mol Med ; 24(5): 623-32, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19787196

RESUMO

In the present study we investigated the in vitro apoptosis inducing effects of peroxisome proliferator-activated receptor-gamma (PPAR-gamma) ligand ciglitazone (CGZ) on acute promyelocytic leukemia (APL) NB4 cells and its mechanisms of action. The results revealed that CGZ (10-50 micromol/l) inhibited the growth of leukemia NB4 cells and caused apoptosis in a time- and dose-dependent manner. Apoptosis was observed clearly by flow cytometry (FCM) and DNA fragmentation analysis. After treatment by CGZ for 48 h, the percentage of disruption of mitochondrial membrane potential (Deltapsim) was increased in a dose-dependent manner. Western blotting demonstrated the cleavage of caspase-3 zymogen protein and a time-dependent cleavage of poly (ADP-ribose) polymerase (PARP). The results also demonstrated that PPAR-gamma expression was increased concomitantly when apoptosis occurred, and that CGZ-induced apoptosis was inhibited by the PPAR-gamma antagonist GW9662, suggesting a PPAR-gamma dependent signaling pathway in CZG-induced cell death. Moreover, CGZ treatment remarkably downregulated the expression of the X-linked inhibitor of apoptosis protein (XIAP), which was inhibited by GW9662. Of note, a small-molecule XIAP antagonist (1396-12) mimicked the effect of CGZ-induced apoptosis via activation of caspase-3, 7 and 9. The apoptosis-inducing effects by CGZ on fresh APL cells were also found to be remarkable by using FCM and Wright's staining observation. Taken together, our results suggest that downregulation of XIAP and activation of capase-3 play an important role in mediating the PPAR-gamma-dependent cell death induced by CGZ in APL cells. These data provide a novel insight into potential therapeutic strategies for treatment of leukemia.


Assuntos
Apoptose , Leucemia Promielocítica Aguda/patologia , PPAR gama/metabolismo , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo , Anilidas/farmacologia , Compostos de Anilina/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Caspase 3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Citometria de Fluxo , Humanos , Proteínas Inibidoras de Apoptose , Leucemia Promielocítica Aguda/enzimologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/metabolismo , PPAR gama/antagonistas & inibidores , Compostos de Fenilureia/farmacologia , Inibidores de Proteases/farmacologia , Coloração e Rotulagem , Survivina , Tiazolidinedionas/farmacologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/antagonistas & inibidores
19.
Am J Physiol Heart Circ Physiol ; 295(1): H401-8, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18515649

RESUMO

Coxsackievirus B3 (CVB3) is one of the most prevalent pathogens of viral myocarditis, which may persist chronically and progress to dilated cardiomyopathy. We previously demonstrated a critical role of the ubiquitin-proteasome system (UPS) in the regulation of coxsackievirus replication in mouse cardiomyocytes. In the present study, we extend our interest to an in vivo animal model to examine the regulation and role of the UPS in CVB3-induced murine myocarditis. Male myocarditis-susceptible A/J mice at age 4-5 wk were randomized to four groups: sham infection + vehicle (n = 10), sham infection + proteasome inhibitor (n = 10), virus + vehicle (n = 20), and virus + proteasome inhibitor (n = 20). Proteasome inhibitor was administered subcutaneously once a day for 3 days. Mice were killed on day 9 after infection, and infected hearts were harvested for Western blot analysis, plaque assay, immunostaining, and histological examination. We showed that CVB3 infection led to an accumulation of ubiquitin conjugates at 9 days after infection. Protein levels of ubiquitin-activating enzyme E1A/E1B, ubiquitin-conjugating enzyme UBCH7, as well as deubiquitinating enzyme UCHL1 were markedly increased in CVB3-infected mice compared with sham infection. However, there was no significant alteration in proteasome activities at 9 days after infection. Immunohistochemical staining revealed that increased expression of E1A/E1B was mainly localized to virus-damaged cells. Finally, we showed that application of a proteasome inhibitor significantly reduced CVB3-induced myocardial damage. This observation reveals a novel mechanism of coxsackieviral pathogenesis, and suggests that the UPS may be an attractive therapeutic target against coxsackievirus-induced myocarditis.


Assuntos
Antivirais/farmacologia , Infecções por Coxsackievirus/tratamento farmacológico , Enterovirus Humano B/patogenicidade , Miocardite/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , Inibidores de Proteases/farmacologia , Inibidores de Proteassoma , Animais , Antivirais/administração & dosagem , Western Blotting , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Infecções por Coxsackievirus/complicações , Infecções por Coxsackievirus/enzimologia , Infecções por Coxsackievirus/virologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Enterovirus Humano B/crescimento & desenvolvimento , Enterovirus Humano B/metabolismo , Imuno-Histoquímica , Injeções Subcutâneas , Masculino , Camundongos , Camundongos Endogâmicos A , Miocardite/enzimologia , Miocardite/patologia , Miocardite/virologia , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Miócitos Cardíacos/virologia , Inibidores de Proteases/administração & dosagem , Complexo de Endopeptidases do Proteassoma/metabolismo , Fatores de Tempo , Ubiquitina/metabolismo , Enzimas Ativadoras de Ubiquitina/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Ensaio de Placa Viral
20.
PLoS One ; 3(7): e2585, 2008 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-18612413

RESUMO

BACKGROUND: Protein ubiquitination and/or degradation by the ubiquitin/proteasome system (UPS) have been recognized as critical mechanisms in the regulation of numerous essential cellular functions. The importance of the UPS in viral pathogenesis has become increasingly apparent. Using murine cardiomyocytes, we have previously demonstrated that the UPS plays a key role in the replication of coxsackievirus B3 (CVB3), an important human pathogen associated with various diseases. To further elucidate the underlying mechanisms, we examined the interplay between the UPS and CVB3, focusing on the role of ubiquitination in viral lifecycle. METHODOLOGY/PRINCIPAL FINDINGS: As assessed by in situ hybridization, Western blot, and plaque assay, we showed that proteasome inhibition decreased CVB3 RNA replication, protein synthesis, and viral titers in HeLa cells. There were no apparent changes in 20S proteasome activities following CVB3 infection. However, we found viral infection led to an accumulation of protein-ubiquitin conjugates, accompanied by a decreased protein expression of free ubiquitin, implicating an important role of ubiquitination in the UPS-mediated viral replication. Using small-interfering RNA, we demonstrated that gene-silencing of ubiquitin significantly reduced viral titers, possibly through downregulation of protein ubiquitination and subsequent alteration of protein function and/or degradation. Inhibition of deubiquitinating enzymes apparently enhances the inhibitory effects of proteasome inhibitors on CVB3 replication. Finally, by immunoprecipitation, we showed that coxsackieviral polymerase 3D was post-translationally modified by ubiquitination and such modification might be a prerequisite for its function in transcriptional regulation of viral genome. CONCLUSION: Coxsackievirus infection promotes protein ubiquitination, contributing to effective viral replication, probably through ubiquitin modification of viral polymerase.


Assuntos
Enterovirus Humano B/fisiologia , Ubiquitinação/fisiologia , Replicação Viral/fisiologia , Infecções por Coxsackievirus/metabolismo , Enterovirus Humano B/patogenicidade , Células HeLa , Humanos , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma , RNA Interferente Pequeno , RNA Polimerase Dependente de RNA/metabolismo , Ubiquitina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA