Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Biomed Pharmacother ; 170: 116077, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38154274

RESUMEN

Hepatitis D virus (HDV), which co-infects or superinfects patients with hepatitis B virus, is estimated to affect 74 million people worldwide. Chronic hepatitis D is the most severe form of viral hepatitis and can result in liver cirrhosis, liver failure, and hepatocellular carcinoma (HCC). Currently, there are no efficient HDV-specific drugs. Therefore, there is an urgent need for novel HDV therapies that can achieve a functional cure or even eliminate the viral infection. In the HDV life cycle, agents targeting the entry step of HDV infection preemptively reduce the intrahepatic viral RNA. Human sodium taurocholate co-transporting polypeptide (hNTCP), a transporter of bile acids on the plasma membrane of hepatocytes, is an essential entry receptor of HDV and is a promising molecular target against HDV infection. Here, we investigated the effect of ergosterol peroxide (EP) on HDV infection in vitro and in vivo. EP inhibited HDV infection of hNTCP-expressing dHuS-E/2 hepatocytes by interrupting the early fusion/endocytosis step of HDV entry. Furthermore, molecular modeling suggested that EP hinders LHBsAg binding to hNTCP by blocking access to S267 and V263. In addition, we generated hNTCP-expressing transgenic (Tg) C57BL/6 mice using the Cre/loxP system for in vivo study. EP reduced the liver HDV RNA level of HDV-challenged hNTCP-Cre Tg mice. Intriguingly, EP downregulated the mRNA level of liver IFN-γ. We demonstrate that EP is a bona fide HDV entry inhibitor that acts on hNTCP and has the potential for use in HDV therapies.


Asunto(s)
Carcinoma Hepatocelular , Hepatitis D , Neoplasias Hepáticas , Simportadores , Ratones , Animales , Humanos , Virus de la Hepatitis Delta/genética , Virus de la Hepatitis Delta/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Ratones Endogámicos C57BL , Hepatitis D/tratamiento farmacológico , Hepatitis D/patología , Virus de la Hepatitis B/fisiología , Hepatocitos , Ratones Transgénicos , Simportadores/metabolismo
2.
Vaccine ; 41(21): 3337-3346, 2023 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-37085450

RESUMEN

Middle East respiratory syndrome coronavirus (MERS-CoV) outbreaks have constituted a public health issue with drastic mortality higher than 34%, necessitating the development of an effective vaccine. During MERS-CoV infection, the trimeric spike protein on the viral envelope is primarily responsible for attachment to host cellular receptor, dipeptidyl peptidase 4 (DPP4). With the goal of generating a protein-based prophylactic, we designed a subunit vaccine comprising the recombinant S1 protein with a trimerization motif (S1-Fd) and examined its immunogenicity and protective immune responses in combination with various adjuvants. We found that sera from immunized wild-type and human DPP4 transgenic mice contained S1-specific antibodies that can neutralize MERS-CoV infection in susceptible cells. Vaccination with S1-Fd protein in combination with a saponin-based QS-21 adjuvant provided long-term humoral as well as cellular immunity in mice. Our findings highlight the significance of the trimeric S1 protein in the development of MERS-CoV vaccines and offer a suitable adjuvant, QS-21, to induce robust and prolonged memory T cell response.


Asunto(s)
Infecciones por Coronavirus , Coronavirus del Síndrome Respiratorio de Oriente Medio , Vacunas Virales , Animales , Ratones , Humanos , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Dipeptidil Peptidasa 4 , Inmunidad Celular , Ratones Transgénicos , Adyuvantes Inmunológicos , Proteínas Recombinantes , Vacunas de Subunidad , Glicoproteína de la Espiga del Coronavirus
3.
J Virol ; 94(13)2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32295922

RESUMEN

Type I and type III interferons (IFNs) are the frontline of antiviral defense mechanisms that trigger hundreds of downstream antiviral genes. In this study, we observed that MERS-CoV nucleocapsid (N) protein suppresses type I and type III IFN gene expression. The N protein suppresses Sendai virus-induced IFN-ß and IFN-λ1 by reducing their promoter activity and mRNA levels, as well as downstream IFN-stimulated genes (ISGs). Retinoic acid-inducible gene I (RIG-I) is known to recognize viral RNA and induce IFN expression through tripartite motif-containing protein 25 (TRIM25)-mediated ubiquitination of RIG-I caspase activation and recruitment domains (CARDs). We discovered that MERS-CoV N protein suppresses RIG-I-CARD-induced, but not MDA5-CARD-induced, IFN-ß and IFN-λ1 promoter activity. By interacting with TRIM25, N protein impedes RIG-I ubiquitination and activation and inhibits the phosphorylation of transcription factors IFN-regulatory factor 3 (IRF3) and NF-κB that are known to be important for IFN gene activation. By employing a recombinant Sindbis virus-EGFP replication system, we showed that viral N protein downregulated the production of not only IFN mRNA but also bioactive IFN proteins. Taken together, MERS-CoV N protein functions as an IFN antagonist. It suppresses RIG-I-induced type I and type III IFN production by interfering with TRIM25-mediated RIG-I ubiquitination. Our study sheds light on the pathogenic mechanism of how MERS-CoV causes disease.IMPORTANCE MERS-CoV causes death of about 35% of patients. Published studies showed that some coronaviruses are capable of suppressing interferon (IFN) expression in the early phase of infection and MERS-CoV proteins can modulate host immune response. In this study, we demonstrated that MERS-CoV nucleocapsid (N) protein suppresses the production of both type I and type III IFNs via sequestering TRIM25, an E3 ubiquitin ligase that is essential for activating the RIG-I signaling pathway. Ectopic expression of TRIM25 rescues the suppressive effect of the N protein. In addition, the C-terminal domain of the viral N protein plays a pivotal role in the suppression of IFN-ß promoter activity. Our findings reveal how MERS-CoV evades innate immunity and provide insights into the interplay between host immune response and viral pathogenicity.


Asunto(s)
Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/virología , Proteína 58 DEAD Box/metabolismo , Interferón Tipo I/biosíntesis , Interferones/biosíntesis , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Proteínas de la Nucleocápside/metabolismo , Transducción de Señal , Proteínas Adaptadoras de Señalización CARD/metabolismo , Línea Celular , Infecciones por Coronavirus/genética , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Humanos , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/genética , Interferones/genética , Regiones Promotoras Genéticas , Unión Proteica , Receptores Inmunológicos , Factores de Transcripción , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas , Interferón lambda
4.
J Virol ; 93(22)2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31462559

RESUMEN

Hepatitis C virus (HCV) NS3 protein possesses protease and helicase activities and is considered an oncoprotein in virus-derived hepatocellular carcinoma. The NS3-associated oncogenesis has been studied but not fully understood. In this study, we have identified novel interactions of the NS3 protein with DNA repair factors, Werner syndrome protein (WRN) and Ku70, in both an HCV subgenomic replicon system and Huh7 cells expressing NS3. HCV NS3 protein inhibits WRN-mediated DNA repair and reduces the repair efficiency of nonhomologous end joining. It interferes with Ku70 recruitment to the double-strand break sites and alters the nuclear distribution of WRN-Ku repair complex. In addition, WRN is a substrate of the NS3/4A protease; the level of WRN protein is regulated by both the proteasome degradation pathway and HCV NS3/4A protease activity. The dual role of HCV NS3 and NS3/4A proteins in regulating the function and expression level of the WRN protein intensifies the effect of impairment on DNA repair. This may lead to an accumulation of DNA mutations and genome instability and, eventually, tumor development.IMPORTANCE HCV infection is a worldwide problem of public health and a major contributor to hepatocellular carcinoma. The single-stranded RNA virus with RNA-dependent RNA polymerase experiences a high error rate and develops strategies to escape the immune system and hepatocarcinogenesis. Studies have revealed the involvement of HCV proteins in the impairment of DNA repair. The present study aimed to further elucidate mechanisms by which the viral NS3 protein impairs the repair of DNA damage. Our results clearly indicate that HCV NS3/4A protease targets WRN for degradation, and, at the same time, diminishes the repair efficiency of nonhomologous end joining by interfering with the recruitment of Ku protein to the DNA double-strand break sites. The study describes a novel mechanism by which the NS3 protein influences DNA repair and provides new insight into the molecular mechanism of HCV pathogenesis.


Asunto(s)
Reparación del ADN por Unión de Extremidades , Hepacivirus/genética , Hepacivirus/metabolismo , Proteínas no Estructurales Virales/metabolismo , Helicasa del Síndrome de Werner/metabolismo , Línea Celular , ADN/metabolismo , Roturas del ADN de Doble Cadena , Daño del ADN , Reparación del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Exodesoxirribonucleasas/metabolismo , Células HEK293 , Hepatitis C Crónica/genética , Humanos , Autoantígeno Ku/genética , ARN Polimerasa Dependiente del ARN/genética , Proteínas no Estructurales Virales/genética , Helicasa del Síndrome de Werner/fisiología
5.
J Biomed Sci ; 25(1): 47, 2018 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-29793506

RESUMEN

BACKGROUND: Middle East respiratory syndrome coronavirus (MERS-CoV) consists of a positive-sense, single-stranded RNA genome and four structural proteins: the spike, envelope, membrane, and nucleocapsid protein. The assembly of the viral genome into virus particles involves viral structural proteins and is believed to be mediated through recognition of specific sequences and RNA structures of the viral genome. METHODS AND RESULTS: A culture system for the production of MERS coronavirus-like particles (MERS VLPs) was determined and established by electron microscopy and the detection of coexpressed viral structural proteins. Using the VLP system, a 258-nucleotide RNA fragment, which spans nucleotides 19,712 to 19,969 of the MERS-CoV genome (designated PS258(19712-19969)ME), was identified to function as a packaging signal. Assembly of the RNA packaging signal into MERS VLPs is dependent on the viral nucleocapsid protein. In addition, a 45-nucleotide stable stem-loop substructure of the PS258(19712-19969)ME interacted with both the N-terminal domain and the C-terminal domain of the viral nucleocapsid protein. Furthermore, a functional SARS-CoV RNA packaging signal failed to assemble into the MERS VLPs, which indicated virus-specific assembly of the RNA genome. CONCLUSIONS: A MERS-oV RNA packaging signal was identified by the detection of GFP expression following an incubation of MERS VLPs carrying the heterologous mRNA GFP-PS258(19712-19969)ME with virus permissive Huh7 cells. The MERS VLP system could help us in understanding virus infection and morphogenesis.


Asunto(s)
Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Proteínas de la Nucleocápside/metabolismo , ARN Viral/metabolismo , Ensamble de Virus/genética , Línea Celular Tumoral , Células HEK293 , Humanos , ARN Mensajero/metabolismo
6.
J Biol Chem ; 291(50): 26226-26238, 2016 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-27807029

RESUMEN

Hepatitis delta virus (HDV) is a satellite virus of hepatitis B virus (HBV). HDV genome encodes two forms of hepatitis delta antigen (HDAg), small HDAg (HDAg-S), which is required for viral replication, and large HDAg (HDAg-L), which is essential for viral assembly. HDAg-L is identical to HDAg-S except that it bears a 19-amino acid extension at the C terminus. Both HDAgs contain a nuclear localization signal (NLS), but only HDAg-L contains a CRM1-independent nuclear export signal at its C terminus. The nuclear export activity of HDAg-L is important for HDV particle formation. However, the mechanisms of HDAg-L-mediated nuclear export of HDV ribonucleoprotein are not clear. In this study, the host cellular RNA export complex TAP-Aly was found to form a complex with HDAg-L, but not with an export-defective HDAg-L mutant, in which Pro205 was replaced by Ala. HDAg-L was found to colocalize with TAP and Aly in the nucleus. The C-terminal domain of HDAg-L was shown to directly interact with the N terminus of TAP, whereas an HDAg-L mutant lacking the NLS failed to interact with full-length TAP. In addition, small hairpin RNA-mediated down-regulation of TAP or Aly reduced nuclear export of HDAg-L and assembly of HDV virions. Furthermore, a peptide, TAT-HDAg-L(198-210), containing the 10-amino acid TAT peptide and HDAg-L(198-210), inhibited the interaction between HDAg-L and TAP and blocked HDV virion assembly and secretion. These data demonstrate that formation and release of HDV particles are mediated by TAP and Aly.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Núcleo Celular/metabolismo , Virus de la Hepatitis Delta/fisiología , Antígenos de Hepatitis delta/metabolismo , Señales de Localización Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Virión/metabolismo , Ensamble de Virus/fisiología , Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/genética , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/genética , Núcleo Celular/genética , Núcleo Celular/virología , Células Hep G2 , Antígenos de Hepatitis delta/genética , Humanos , Señales de Localización Nuclear/genética , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Péptidos/farmacología , Dominios Proteicos , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Virión/genética , Ensamble de Virus/efectos de los fármacos
7.
Liver Int ; 34(9): 1358-68, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25360475

RESUMEN

BACKGROUND & AIMS: Hepatitis C virus (HCV) infection is highly associated with the type 2 diabetes mellitus, but the detailed mechanisms by which the viral proteins are involved in the clinical outcome remain unclear. METHODS: A cDNA microarray analysis was performed following introducing an NS5A-encoding plasmid or a control vector into a mouse system by hydrodynamics- based transfection. Differentially expressed genes that are associated with gluconeogenesis were selected and their expression levels in HCV patients, in NS5A-expressing systems, and in the viral subgenomic replicon system were further examined by real-time quantitative polymerase chain reaction and Western blot analysis. RESULTS: Differential gene expression including an upregulation of the gluconeogenic rate-limiting enzyme phosphoenolpyruvate carboxykinase (PEPCK) compared with controls was detected in mouse hepatocytes expressing HCV NS5A and in HCV patients with diabetes. In addition, an NS5A-dependent increase in glucose production was demonstrated in human primary hepatocytes. The upregulation of PEPCK and peroxisome proliferator-activated receptor-c coactivator-1a (PGC-1a) were also detected in NS5A-expressing cells and in the viral genotype 1b subgenomic replicon system. Further studies demonstrated that the NS5A-mediated upregulation of PEPCK and PGC-1a genes were resulted from the activation of PI3K-Akt and JNK signalling pathways. In addition, the expression levels of the forkhead transcription factor FoxO1 and the liver-enriched transcription factor HNF-4a were increased in HCV NS5A expressing cells. CONCLUSIONS: By upregulating the expression of PEPCK gene via its transactivators FoxO1 and HNF-4a, and the coactivator PGC-1a, the NS5A promotes the production of hepatic glucose which may contribute to the development of HCV-associated type 2 diabetes mellitus.


Asunto(s)
Diabetes Mellitus Tipo 2/virología , Regulación de la Expresión Génica/fisiología , Gluconeogénesis/fisiología , Hepacivirus/metabolismo , Hepatitis C/complicaciones , Transducción de Señal/fisiología , Proteínas no Estructurales Virales/metabolismo , Animales , Western Blotting , Diabetes Mellitus Tipo 2/etiología , Glucosa/metabolismo , Hepatocitos/metabolismo , Humanos , MAP Quinasa Quinasa 4/metabolismo , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína Oncogénica v-akt/metabolismo , Fosfoenolpiruvato Carboxiquinasa (ATP)/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección
8.
J Biol Chem ; 288(45): 32424-32432, 2013 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-24078630

RESUMEN

Energy homeostasis is crucial for maintaining normally functioning cells; disturbances in this balance often cause various diseases. The limb region 1 (LMBR1) domain containing 1 gene (lmbrd1) encodes the LMBD1 protein that possesses 9 putative transmembrane domains. LMBD1 has been suggested to be involved in the lysosome in aiding the export of cobalamin. In this study, we determined that LMBD1 plays a regulatory role in the plasma membrane. A micro-positron emission tomography analysis showed that a single-allele knock-out of lmbrd1 increased the (18)F-fluorodeoxyglucose uptake in murine hearts. In addition, the knockdown of lmbrd1 resulted in an up-regulated signaling of the insulin receptor (IR) and its downstream signaling molecule, Akt. Confocal and live total internal reflection fluorescence microscopy showed that LMBD1 co-localized and co-internalized with clathrin and the IR, but not with the transferrin receptor. The results of the mutation analysis and phenotypic rescue experiments indicate that LMBD1 interacts with adaptor protein-2 and is involved in the unique clathrin-mediated endocytosis of the IR. LMBD1 selectively interacts with the IR. The knockdown of lmbrd1 attenuated IR endocytosis, resulting in the perturbation of the IR recycling pathway and consequential enhancement of the IR signaling cascade. In summary, LMBD1 plays an imperative role in mediating and regulating the endocytosis of the IR.


Asunto(s)
Endocitosis/fisiología , Miocardio/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Receptor de Insulina/metabolismo , Transducción de Señal/fisiología , Complejo 2 de Proteína Adaptadora/genética , Complejo 2 de Proteína Adaptadora/metabolismo , Animales , Línea Celular , Clatrina/genética , Clatrina/metabolismo , Fluorodesoxiglucosa F18/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Mutantes , Proteínas de Transporte Nucleocitoplasmático/genética , Tomografía de Emisión de Positrones , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Radiofármacos/farmacología , Ratas , Receptor de Insulina/genética
9.
J Virol ; 84(15): 7703-12, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20484496

RESUMEN

Severe acute respiratory syndrome coronavirus (SARS-CoV) was identified to be the causative agent of SARS with atypical pneumonia. Angiotensin-converting enzyme 2 (ACE2) is the major receptor for SARS-CoV. It is not clear whether ACE2 conveys signals from the cell surface to the nucleus and regulates expression of cellular genes upon SARS-CoV infection. To understand the pathogenesis of SARS-CoV, human type II pneumocyte (A549) cells were incubated with the viral spike protein or with SARS-CoV virus-like particles containing the viral spike protein to examine cytokine modulation in lung cells. Results from oligonucleotide-based microarray, real-time PCR, and enzyme-linked immunosorbent assays indicated an upregulation of the fibrosis-associated chemokine (C-C motif) ligand 2 (CCL2) by the viral spike protein and the virus-like particles. The upregulation of CCL2 by SARS-CoV spike protein was mainly mediated by extracellular signal-regulated kinase 1 and 2 (ERK1/2) and AP-1 but not the IkappaBalpha-NF-kappaB signaling pathway. In addition, Ras and Raf upstream of the ERK1/2 signaling pathway were involved in the upregulation of CCL2. Furthermore, ACE2 receptor was activated by casein kinase II-mediated phosphorylation in cells pretreated with the virus-like particles containing spike protein. These results indicate that SARS-CoV spike protein triggers ACE2 signaling and activates fibrosis-associated CCL2 expression through the Ras-ERK-AP-1 pathway.


Asunto(s)
Quimiocina CCL2/biosíntesis , Glicoproteínas de Membrana/inmunología , Peptidil-Dipeptidasa A/metabolismo , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , Transducción de Señal , Proteínas del Envoltorio Viral/inmunología , Enzima Convertidora de Angiotensina 2 , Línea Celular , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/virología , Perfilación de la Expresión Génica , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Glicoproteína de la Espiga del Coronavirus , Regulación hacia Arriba
10.
FEBS Lett ; 584(3): 482-6, 2010 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-19958771

RESUMEN

Hepatitis C virus (HCV) internal non-structural protein 3 (NS3) cleavage can occur in trans in the presence of NS4A. In this study, we have further demonstrated a critical role of the helicase domain in the internal NS3 cleavage, different from HCV polyprotein processing which requires only the serine protease domain. The NTPase domain of NS3 helicase interacts with the RNA binding domain to facilitate internal NS3 cleavage. In addition, NS3 protease activity contributes to the transforming ability of the major internal cleavage product NS3(1-402). These findings imply important roles of the internal cleavage and protease activity of the NS3 protein in the pathogenesis of HCV.


Asunto(s)
Hepacivirus/enzimología , ARN Helicasas/metabolismo , Proteínas no Estructurales Virales/metabolismo , Animales , Sitios de Unión , Línea Celular , Línea Celular Tumoral , Hepacivirus/genética , Humanos , Ratones , Células 3T3 NIH , Poliproteínas/genética , Poliproteínas/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína/genética , Estructura Terciaria de Proteína/fisiología , ARN Helicasas/química , ARN Helicasas/genética , Serina Proteasas/química , Serina Proteasas/genética , Serina Proteasas/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética
11.
Cell Transplant ; 19(2): 231-43, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19906331

RESUMEN

Oval cells and hepatocytes rarely proliferate simultaneously. This study aimed to determine the impacts of hepatocyte transplantation on the response and fate of oval cells that are activated to proliferate in acute severe hepatic injury. Retrorsine + D-galactosamine (R+D-gal) treatment was used to induce acute hepatic injury and to elicit extensive activation of oval cells in male dipeptidyl peptidase IV-deficient F344 rats. These rats were then randomized to receive wild-type hepatocyte transplantation or vehicle intraportally. The kinetics of oval cell response and their differentiation fate were analyzed. Results showed that oval cells were activated early and differentiated into hepatocytes in R+D-gal-treated rats without hepatocyte transplantation. With hepatocyte transplantation, the oval cells were recruited later and continued to proliferate in parallel with the massive proliferation of transplanted hepatocytes. They formed ductules and differentiated into biliary cells. When hepatocytes were transplanted at the day when oval cells were at their peak response, the numerous activated oval cells ceased to differentiate into hepatocytes and remained in ductular form. The ductular oval cells were capable of differentiating into hepatocytes again when the donor hepatocytes were inhibited to proliferate. We conclude that hepatocyte transplantation changes the mechanism of liver reconstitution and affects the differentiation fate of host oval cells in acute severe hepatic injury.


Asunto(s)
Diferenciación Celular , Enfermedad Hepática Inducida por Sustancias y Drogas , Hepatocitos/trasplante , Hígado/lesiones , Animales , Antineoplásicos Fitogénicos/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Enfermedad Hepática Inducida por Sustancias y Drogas/cirugía , Dipeptidil Peptidasa 4/deficiencia , Galactosamina/toxicidad , Hepatocitos/citología , Humanos , Hígado/citología , Masculino , Alcaloides de Pirrolicidina/toxicidad , Distribución Aleatoria , Ratas , Ratas Endogámicas F344
12.
Hepatology ; 50(4): 1184-93, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19676133

RESUMEN

UNLABELLED: Liver cirrhosis is characterized by progressive accumulation of extracellular matrix following chronic liver injuries. In the extracellular space, the constant turnover of liver matrix is regulated by the matrix metalloproteinase (MMP) class of enzyme. To assess whether genetic variations in MMP would result in diversity of liver cirrhosis, a case-control study of 320 patients with hepatocellular carcinoma, with or without cirrhosis, was conducted. Ten single-nucleotide polymorphism markers from four potential fibrosis-associated genes were selected for genotyping. Among these genes, a nonsynonymous single-nucleotide polymorphism which generates the variation of Gly-137 and Asp-137 in the MMP-7 gene was found to be strongly associated with the development of liver cirrhosis. In contrast to MMP-7(Gly-137) that predominantly secretes out into the cell culture medium, the cirrhosis-associated MMP-7(Asp-137) variant is preferentially localized on the extracellular membranes where it exerts its proteolytic activity on pericellular substrates. Functional analysis demonstrated an increased ability of the MMP-7(Asp-137) variant to associate with the cell surface CD151 molecule. In wound-healing and Boyden chamber assays, cell motility was specifically enhanced with the expression of MMP-7(Asp-137) as compared to the cells expressing MMP-7(Gly-137). These results demonstrate that the MMP-7(Asp-137) variant confers a gain-of-function phenotype for MMP-7. CONCLUSION: We have identified a novel genetic association of MMP-7(Asp-137) variant with liver cirrhosis in patients with hepatocellular carcinoma. Whether the MMP-7 variant can be a new marker for liver cirrhosis will be further studied.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Predisposición Genética a la Enfermedad/genética , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Neoplasias Hepáticas/metabolismo , Metaloproteinasa 7 de la Matriz/genética , Metaloproteinasa 7 de la Matriz/metabolismo , Adulto , Anciano , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Estudios de Casos y Controles , Membrana Celular/metabolismo , Femenino , Genotipo , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo , Tetraspanina 24
13.
J Biol Chem ; 283(43): 29396-404, 2008 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-18728014

RESUMEN

Hepatitis C virus often causes persistent infection and hepatocellular carcinoma. Studies have demonstrated the roles of viral nonstructural protein 5A (NS5A) in the induction of chromosome aneuploidy, but the molecular mechanisms are not clear. In this study, hydrodynamics-based in vivo transfection was applied to a mouse system. Mouse hepatocytes that successfully expressed NS5A protein were isolated by laser capture microdissection. Gene expression profiles of the NS5A-expressing hepatocytes were examined by an Affymetrix oligonucleotide microarray system. Aspm (abnormal spindle-like, microcephaly associated), which encodes the mitotic spindle protein ASPM, was identified to be differentially expressed in the absence and the presence of NS5A. The down-regulation of Aspm mRNA and ASPM protein was confirmed by real time polymerase chain reaction and Western blot analysis, respectively, both in mouse model systems and in viral subgenomic replicon and in vitro transfection culturing systems. In addition, cultured cells that constitutively expressed NS5A protein showed G(2)/M cell cycle block and chromosome aneuploidy. Overexpression of ASPM relieved the G(2)/M cell cycle block. Furthermore, NS5A protein repressed the promoter activity of Aspm gene in a dose-dependent manner. The regulatory effect was abolished when amino acid substitutions P2209L, T2214A, and T2217G known to interrupt the NS5A-PKR interaction were introduced into the NS5A protein. This indicates that the down-regulation of Aspm expression is via the PKR-p38 signaling pathway. These results suggest that NS5A protein down-regulates the expression of the mitotic spindle protein ASPM and induces aberrant mitotic cell cycle associated with chromosome instability and hepatocellular carcinoma.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Regulación Viral de la Expresión Génica , Proteínas del Tejido Nervioso/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/fisiología , eIF-2 Quinasa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Células COS , Proteínas de Unión a Calmodulina , Chlorocebus aethiops , Regulación hacia Abajo , Perfilación de la Expresión Génica , Humanos , Ratones , Modelos Biológicos , Células 3T3 NIH
14.
J Virol ; 81(15): 7999-8008, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17522200

RESUMEN

The NS3 protein of hepatitis C virus (HCV) possesses protease activity responsible for the proteolytic cleavage of the viral polyprotein at the junctions of nonstructural proteins downstream of NS3. The NS3 protein was also found to be internally cleaved. In this study, we demonstrated that internal cleavages occurred on the NS3 protein of genotype 1b in the presence of NS4A, both in culture cells and with a mouse model system. No internal cleavage products were detected with the NS3 and NS4A proteins of genotype 2a. Three potential cleavage sites were detected in the NS3 protein (genotype 1b), with IPT(402)|S being the major one. The internal cleavage requires the polyprotein processing activity of NS3 protease, but when supplemented in trans, the internal cleavage efficiency is reduced. In addition, several mutations in NS4A disrupted the internal cleavage of NS3 but did not affect polyprotein processing, indicating that NS4A contributes differently to these two proteolytic activities. Furthermore, Ile-25, Val-26, and Ile-29 of the NS4A protein, important for the NS4A-dependent internal cleavages, were also shown to be critical for the transforming activity of NS3, but mutations at these critical residues resulted only in a slight increase of HCV replicating efficiency. The internal cleavage-associated enhancement of the transforming activity of NS3 was reduced when a T402A substitution at the major internal cleavage site was introduced. The multiple roles of NS4A in viral multiplication and pathogenesis make NS4A an ideal molecular target for HCV therapy.


Asunto(s)
Proteínas Portadoras/metabolismo , Hepacivirus/metabolismo , Poliproteínas/metabolismo , Proteínas no Estructurales Virales/metabolismo , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas Portadoras/genética , Línea Celular , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Datos de Secuencia Molecular , Alineación de Secuencia , Proteínas Virales/genética
15.
J Biomed Sci ; 13(6): 861-74, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16927014

RESUMEN

The genomic RNA of hepatitis C virus (HCV) encodes the viral polyprotein precursor that undergoes proteolytic cleavage into structural and nonstructural proteins by cellular and the viral NS3 and NS2-3 proteases. Nonstructural protein 4A (NS4A) is a cofactor of the NS3 serine protease and has been demonstrated to inhibit protein synthesis. In this study, GST pull-down assay was performed to examine potential cellular factors that interact with the NS4A protein and are involved in the pathogenesis of HCV. A trypsin digestion followed by LC-MS/MS analysis revealed that one of the GST-NS4A-interacting proteins to be eukaryotic elongation factor 1A (eEF1A). Both the N-terminal domain of NS4A from amino acid residues 1-20, and the central domain from residues 21-34 interacted with eEF1A, but the central domain was the key player involved in the NS4A-mediated translation inhibition. NS4A(21-34) diminished both cap-dependent and HCV IRES-mediated translation in a dose-dependent manner. The translation inhibitory effect of NS4A(21-34) was relieved by the addition of purified recombinant eEF1A in an in vitro translation system. Taken together, NS4A inhibits host and viral translation through interacting with eEF1A, implying a possible mechanism by which NS4A is involved in the pathogenesis and chronic infection of HCV.


Asunto(s)
Proteínas Portadoras/metabolismo , Factor 1 de Elongación Peptídica/metabolismo , Biosíntesis de Proteínas , Caperuzas de ARN , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Proteínas Portadoras/química , Cartilla de ADN , Péptidos y Proteínas de Señalización Intracelular , Datos de Secuencia Molecular , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas no Estructurales Virales , Proteínas Virales/química
16.
J Virol ; 79(22): 13848-55, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16254320

RESUMEN

The severe acute respiratory syndrome coronavirus (SARS-CoV) was recently identified as the etiology of SARS. The virus particle consists of four structural proteins: spike (S), small envelope (E), membrane (M), and nucleocapsid (N). Recognition of a specific sequence, termed the packaging signal (PS), by a virus N protein is often the first step in the assembly of viral RNA, but the molecular mechanisms involved in the assembly of SARS-CoV RNA are not clear. In this study, Vero E6 cells were cotransfected with plasmids encoding the four structural proteins of SARS-CoV. This generated virus-like particles (VLPs) of SARS-CoV that can be partially purified on a discontinuous sucrose gradient from the culture medium. The VLPs bearing all four of the structural proteins have a density of about 1.132 g/cm(3). Western blot analysis of the culture medium from transfection experiments revealed that both E and M expressed alone could be released in sedimentable particles and that E and M proteins are likely to form VLPs when they are coexpressed. To examine the assembly of the viral genomic RNA, a plasmid representing the GFP-PS580 cDNA fragment encompassing the viral genomic RNA from nucleotides 19715 to 20294 inserted into the 3' noncoding region of the green fluorescent protein (GFP) gene was constructed and applied to the cotransfection experiments with the four structural proteins. The SARS-CoV VLPs thus produced were designated VLP(GFP-PS580). Expression of GFP was detected in Vero E6 cells infected with the VLP(GFP-PS580), indicating that GFP-PS580 RNA can be assembled into the VLPs. Nevertheless, when Vero E6 cells were infected with VLPs produced in the absence of the viral N protein, no green fluorescence was visualized. These results indicate that N protein has an essential role in the packaging of SARS-CoV RNA. A filter binding assay and competition analysis further demonstrated that the N-terminal and C-terminal regions of the SARS-CoV N protein each contain a binding activity specific to the viral RNA. Deletions that presumably disrupt the structure of the N-terminal domain diminished its RNA-binding activity. The GFP-PS-containing SARS-CoV VLPs are powerful tools for investigating the tissue tropism and pathogenesis of SARS-CoV.


Asunto(s)
Nucleocápside/fisiología , ARN Viral/genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Animales , Secuencia de Bases , Chlorocebus aethiops , Cartilla de ADN , ADN Complementario/genética , ADN Viral/genética , Genes Reporteros , Conformación de Ácido Nucleico , Proteínas de la Nucleocápside/genética , Proteínas de la Nucleocápside/aislamiento & purificación , Plásmidos , ARN Viral/química , Proteínas Recombinantes/aislamiento & purificación , Células Vero , Proteínas Estructurales Virales/metabolismo
17.
Biochem Biophys Res Commun ; 333(1): 194-9, 2005 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-15950190

RESUMEN

SARS-CoV 3CL protease is essential for viral protein processing and is regarded as a good drug target to prevent SARS-CoV replication. In the present study, we established a high-throughput FRET technique for screening for anti-SARS-CoV 3CL protease drugs. Of a thousand existing drugs examined, hexachlorophene was identified as the most potent in inhibiting SARS-CoV 3CL protease. Further characterization showed that it was effective at micromolar concentrations (K(i) = 4 microM). The binding mode was competitive, and the inhibitory effect was dependent on preincubation time. Two other drugs, triclosan and nelfinavir, were about 10 times less potent. The structure-based search and biological evaluation of various hexachlorophene analogues were described. These analogues gave optimal inhibitory activity against SARS-CoV 3CL protease with IC(50) values ranging from 7.6 to 84.5 microM. Optimization of hexachlorophene analogues was shown to provide several active 3CL protease inhibitors that function as potential anti-SARS agents.


Asunto(s)
Diseño de Fármacos , Evaluación Preclínica de Medicamentos/métodos , Transferencia Resonante de Energía de Fluorescencia/métodos , Modelos Moleculares , Inhibidores de Proteasas/química , Mapeo de Interacción de Proteínas/métodos , Proteínas Virales/antagonistas & inhibidores , Sitios de Unión , Simulación por Computador , Proteasas 3C de Coronavirus , Cisteína Endopeptidasas , Endopeptidasas/análisis , Activación Enzimática , Cinética , Inhibidores de Proteasas/análisis , Unión Proteica , Proteínas Virales/análisis
18.
J Biol Chem ; 280(5): 3129-37, 2005 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-15563453

RESUMEN

p160 coregulators were initially identified as nuclear hormone receptor coactivators. In this study, functional data demonstrate that members of the three p160 families can have opposing roles in regulating gene expression by the same transcription factor. Both SRC1A and p/CIP function as coactivators for MyoD-mediated transcription whereas GRIP1 acts negatively as a (co)repressor. SRC1A and p/CIP predominantly interact with distinct sites on the NH2-terminal activation domain of MyoD. GRIP1 binds to both these regions but it alone, and neither SRC1A nor p/CIP, also interacts with specific sites on MyoD that are critical for the binding of the essential MyoD coactivator, p300. This suggests that competition by GRIP1 for SRC1A, p/CIP, and p300 binding sites on a transcription factor may regulate the activity of the factor.


Asunto(s)
Proteína MioD/genética , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Células Cultivadas , Fibroblastos/citología , Fibroblastos/fisiología , Eliminación de Gen , Regulación de la Expresión Génica/fisiología , Histona Acetiltransferasas , Ratones , Ratones Endogámicos C3H , Proteína MioD/química , Coactivador 1 de Receptor Nuclear , Coactivador 2 del Receptor Nuclear , Coactivador 3 de Receptor Nuclear , Regiones Promotoras Genéticas/fisiología , Estructura Terciaria de Proteína , Factores de Transcripción/química , Factores de Transcripción/genética
19.
FEBS Lett ; 565(1-3): 111-6, 2004 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-15135062

RESUMEN

The open reading frame 3 of the severe acute respiratory syndrome coronavirus (SARS-CoV) genome encodes a predicted protein 3a, consisting of 274 amino acids, that lacks any significant similarities to any known protein. We generated specific antibodies against SARS protein 3a by using a synthetic peptide (P2) corresponding to amino acids 261-274 of the putative protein. Anti-P2 antibodies and the sera from SARS patients could specifically detect the recombinant SARS protein 3a expressed in Escherichia coli and in Vero E6 cells. Expression of SARS protein 3a was detected at 8-12 h after infection and reached a higher level after approximately 24 h in SARS-CoV-infected Vero E6 cells. Protein 3a was also detected in the alveolar lining pneumocytes and some intra-alveolar cells of a SARS-CoV-infected patient's lung specimen. Recombinant protein 3a expressed in Vero E6 cells and protein 3a in the SARS-CoV-infected cells was distributed over the cytoplasm in a fine punctate pattern with partly concentrated staining in the Golgi apparatus. Our study demonstrates that SARS-CoV indeed expresses a novel protein 3a, which is present only in SARS-CoV and not in other known CoVs.


Asunto(s)
Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo , Proteínas Virales/química , Animales , Anticuerpos/química , Chlorocebus aethiops , Clonación Molecular , Grupo Citocromo b/química , Citoplasma/metabolismo , ADN Complementario/metabolismo , Electroforesis en Gel de Poliacrilamida , Escherichia coli/metabolismo , Aparato de Golgi/metabolismo , Humanos , Inmunohistoquímica , Pulmón/virología , Microscopía Fluorescente , Nucleocápside/metabolismo , Sistemas de Lectura Abierta , Péptidos/química , Estructura Terciaria de Proteína , Alveolos Pulmonares/virología , Conejos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Fracciones Subcelulares/metabolismo , Factores de Tiempo , Células Vero , Proteínas del Envoltorio Viral , Proteínas Viroporinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA