Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Med Chem ; 63(10): 5031-5073, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-31930920

RESUMEN

Nonalcoholic steatohepatitis (NASH) is a severe form of nonalcoholic fatty liver disease (NAFLD) characterized by liver steatosis, inflammation, and hepatocellular damage. NASH is a serious condition that can progress to cirrhosis, liver failure, and hepatocellular carcinoma. The association of NASH with obesity, type 2 diabetes mellitus, and dyslipidemia has led to an emerging picture of NASH as the liver manifestation of metabolic syndrome. Although diet and exercise can dramatically improve NASH outcomes, significant lifestyle changes can be challenging to sustain. Pharmaceutical therapies could be an important addition to care, but currently none are approved for NASH. Here, we review the most promising targets for NASH treatment, along with the most advanced therapeutics in development. These include targets involved in metabolism (e.g., sugar, lipid, and cholesterol metabolism), inflammation, and fibrosis. Ultimately, combination therapies addressing multiple aspects of NASH pathogenesis are expected to provide benefit for patients.


Asunto(s)
Sistemas de Liberación de Medicamentos/tendencias , Desarrollo de Medicamentos/tendencias , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Animales , Anticolesterolemiantes/administración & dosificación , Anticolesterolemiantes/química , Anticolesterolemiantes/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Desarrollo de Medicamentos/métodos , Humanos , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/química , Hipoglucemiantes/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/fisiología , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/epidemiología , Síndrome Metabólico/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/epidemiología , Obesidad/metabolismo , PPAR gamma/agonistas , PPAR gamma/química , Estructura Terciaria de Proteína
2.
PLoS Pathog ; 11(11): e1005297, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26588073

RESUMEN

The hepatitis C virus (HCV) p7 protein is required for infectious virus production via its role in assembly and ion channel activity. Although NMR structures of p7 have been reported, the location of secondary structural elements and orientation of the p7 transmembrane domains differ among models. Furthermore, the p7 structure-function relationship remains unclear. Here, extensive mutagenesis, coupled with infectious virus production phenotyping and molecular modeling, demonstrates that the N-terminal helical region plays a previously underappreciated yet critical functional role, especially with respect to E2/p7 cleavage efficiency. Interrogation of specific N-terminal helix residues identified as having p7-specific defects and predicted to point toward the channel pore, in a context of independent E2/p7 cleavage, further supports p7 as a structurally plastic, minimalist ion channel. Together, our findings indicate that the p7 N-terminal helical region is critical for E2/p7 processing, protein-protein interactions, ion channel activity, and infectious HCV production.


Asunto(s)
Hepacivirus/metabolismo , Canales Iónicos/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Línea Celular , Humanos , Modelos Moleculares , Ensamble de Virus , Replicación Viral
3.
J Neurosci ; 33(47): 18553-65, 2013 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-24259577

RESUMEN

V3 interneurons (INs) are a major group of excitatory commissural interneurons in the spinal cord, and they are essential for producing a stable and robust locomotor rhythm. V3 INs are generated from the ventral-most progenitor domain, p3, but migrate dorsally and laterally during postmitotic development. At birth, they are located in distinctive clusters in the ventral horn and deep dorsal horn. To assess the heterogeneity of this genetically identified group of spinal INs, we combined patch-clamp recording and anatomical tracing with cluster analysis. We examined electrophysiological and morphological properties of mature V3 INs identified by their expression of tdTomato fluorescent proteins in Sim1(Cre/+); Rosa(floxstop26TdTom) mice. We identified two V3 subpopulations with distinct intrinsic properties and spatial distribution patterns. Ventral V3 INs, primarily located in lamina VIII, possess a few branching processes and were capable of generating rapid tonic firing spikes. By contrast, dorsal V3 INs exhibited a more complex morphology and relatively slow average spike frequency with strong adaptation, and they also displayed large sag voltages and post-inhibitory rebound potentials. Our data suggested that hyperpolarization-activated cation channel currents and T-type calcium channel currents may account for some of the membrane properties of V3 INs. Finally, we observed that ventral and dorsal V3 INs were active in different ways during running and swimming, indicating that ventral V3 INs may act as premotor neurons and dorsal V3 INs as relay neurons mediating sensory inputs. Together, we detected two physiologically and topographically distinct subgroups of V3 INs, each likely playing different roles in locomotor activities.


Asunto(s)
Potenciales de Acción/fisiología , Interneuronas/clasificación , Interneuronas/fisiología , Médula Espinal/citología , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Bencimidazoles/farmacología , Fármacos Cardiovasculares/farmacología , Análisis por Conglomerados , Ciclopropanos/farmacología , Estimulación Eléctrica , Técnicas In Vitro , Ratones , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Naftalenos/farmacología , Níquel/farmacología , Técnicas de Placa-Clamp , Proteínas Proto-Oncogénicas c-fos/farmacología , Pirimidinas/farmacología , ARN no Traducido/genética , Proteínas Represoras/genética , Estadísticas no Paramétricas
4.
PLoS One ; 8(3): e59776, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23527266

RESUMEN

Although a cure for HCV is on the near horizon, emerging drug cocktails will be expensive, associated with side-effects and resistance making a global vaccine an urgent priority given the estimated high incidence of infection around the world. Due to the highly heterogeneous nature of HCV, an effective HCV vaccine which could elicit broadly cross-neutralizing antibodies has represented a major challenge. In this study, we tested for the presence of cross-neutralizing antibodies in human volunteers who were immunized with recombinant glycoproteins gpE1/gpE2 derived from a single HCV strain (HCV1 of genotype 1a). Cross neutralization was tested in Huh-7.5 human hepatoma cell cultures using infectious recombinant HCV (HCVcc) expressing structural proteins of heterologous HCV strains from all known major genotypes, 1-7. Vaccination induced significant neutralizing antibodies against heterologous HCV genotype 1a virus which represents the most common genotype in North America. Of the 16 vaccinees tested, 3 were selected on the basis of strong 1a virus neutralization for testing of broad cross-neutralizing responses. At least 1 vaccinee was shown to elicit broad cross-neutralization against all HCV genotypes. Although observed in only a minority of vaccinees, our results prove the key concept that a vaccine derived from a single strain of HCV can elicit broad cross-neutralizing antibodies against all known major genotypes of HCV and provide considerable encouragement for the further development of a human vaccine against this common, global pathogen.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Hepacivirus/inmunología , Proteínas Recombinantes/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Vacunas contra Hepatitis Viral/inmunología , Línea Celular Tumoral , Reacciones Cruzadas , Electroforesis en Gel de Poliacrilamida , Hepacivirus/genética , Hepacivirus/metabolismo , Humanos , Sueros Inmunes/inmunología , Pruebas de Neutralización , Colorantes de Rosanilina
5.
Hepatology ; 55(2): 364-72, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21953761

RESUMEN

UNLABELLED: Endstage liver disease caused by chronic hepatitis C virus (HCV) infection is the leading indication for liver transplantation in the Western world. However, immediate reinfection of the grafted donor liver by circulating virus is inevitable and liver disease progresses much faster than the original disease. Standard antiviral therapy is not well tolerated and usually ineffective in liver transplant patients, whereas anti-HCV immunotherapy is hampered by the extreme genetic diversity of the virus and its ability to spread by way of cell-cell contacts. We generated a human monoclonal antibody against scavenger receptor class B type I (SR-BI), monoclonal antibody (mAb)16-71, which can efficiently prevent infection of Huh-7.5 hepatoma cells and primary hepatocytes by cell-culture-derived HCV (HCVcc). Using an Huh7.5 coculture system we demonstrated that mAb16-71 interferes with direct cell-to-cell transmission of HCV. Finally we evaluated the in vivo efficacy of mAb16-71 in "human liver urokinase-type plasminogen activator, severe combined immune deficiency (uPA-SCID) mice" (chimeric mice). A 2-week anti-SR-BI therapy that was initiated 1 day before viral inoculation completely protected all chimeric mice from infection with serum-derived HCV of different genotypes. Moreover, a 9-day postexposure therapy that was initiated 3 days after viral inoculation (when viremia was already observed in the animals) suppressed the rapid viral spread observed in untreated control animals. After cessation of anti-SR-BI-specific antibody therapy, a rise of the viral load was observed. CONCLUSION: Using in vitro cell culture and human liver-chimeric mouse models, we show that a human mAb targeting the HCV coreceptor SR-BI completely prevents infection and intrahepatic spread of multiple HCV genotypes. This strategy may be an efficacious way to prevent infection of allografts following liver transplantation in chronic HCV patients, and may even hold promise for the prevention of virus rebound during or following antiviral therapy.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos CD36/inmunología , Hepatitis C/prevención & control , Animales , Línea Celular Tumoral , Quimera , Genotipo , Humanos , Trasplante de Hígado , Ratones , Ratones SCID , Prevención Secundaria
6.
Hepatology ; 54(6): 1913-23, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21800339

RESUMEN

UNLABELLED: Hepatitis C virus (HCV) replication in primary liver cells is less robust than that in hepatoma cell lines, suggesting that innate antiviral mechanisms in primary cells may limit HCV replication or spread. Here we analyzed the expression of 47 genes associated with interferon (IFN) induction and signaling following HCV infection of primary human fetal liver cell (HFLC) cultures from 18 different donors. We report that cell culture-produced HCV (HCVcc) induced expression of Type III (λ) IFNs and of IFN-stimulated genes (ISGs). Little expression of Type I IFNs was detected. Levels of IFNλ and ISG induction varied among donors and, often, between adapted and nonadapted HCV chimeric constructs. Higher levels of viral replication were associated with greater induction of ISGs and of λ IFNs. Gene induction was dependent on HCV replication, as ultraviolet light-inactivated virus was not stimulatory and an antiviral drug, 2'-C-methyladenosine, reduced induction of λ IFNs and ISGs. The level of IFNλ protein induced was sufficient to inhibit HCVcc infection of naïve cultures. CONCLUSION: Together, these results indicate that despite its reported abilities to blunt the induction of an IFN response, HCV infection is capable of inducing antiviral cytokines and pathways in primary liver cell cultures. Induction of ISGs and λ IFNs may limit the growth and spread of HCV in primary cell cultures and in the infected liver. HCV infection of HFLC may provide a useful model for the study of gene induction by HCV in vivo.


Asunto(s)
Hepacivirus/fisiología , Interferones/biosíntesis , Interleucinas/genética , Adenosina/análogos & derivados , Adenosina/farmacología , Células Cultivadas , Perfilación de la Expresión Génica , Hepacivirus/efectos de los fármacos , Hepacivirus/efectos de la radiación , Humanos , Interferones/farmacología , Interleucinas/biosíntesis , Interleucinas/farmacología , Quinasas Janus/metabolismo , Hígado/citología , Cultivo Primario de Células , Proteínas Recombinantes/farmacología , Activación Transcripcional , Rayos Ultravioleta , Replicación Viral/efectos de los fármacos
7.
Nature ; 474(7350): 208-11, 2011 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-21654804

RESUMEN

Hepatitis C virus (HCV) remains a major medical problem. Antiviral treatment is only partially effective and a vaccine does not exist. Development of more effective therapies has been hampered by the lack of a suitable small animal model. Although xenotransplantation of immunodeficient mice with human hepatocytes has shown promise, these models are subject to important challenges. Building on the previous observation that CD81 and occludin comprise the minimal human factors required to render mouse cells permissive to HCV entry in vitro, we attempted murine humanization via a genetic approach. Here we show that expression of two human genes is sufficient to allow HCV infection of fully immunocompetent inbred mice. We establish a precedent for applying mouse genetics to dissect viral entry and validate the role of scavenger receptor type B class I for HCV uptake. We demonstrate that HCV can be blocked by passive immunization, as well as showing that a recombinant vaccinia virus vector induces humoral immunity and confers partial protection against heterologous challenge. This system recapitulates a portion of the HCV life cycle in an immunocompetent rodent for the first time, opening opportunities for studying viral pathogenesis and immunity and comprising an effective platform for testing HCV entry inhibitors in vivo.


Asunto(s)
Modelos Animales de Enfermedad , Hepacivirus/fisiología , Hepatitis C/genética , Hepatitis C/virología , Hepatocitos/metabolismo , Hepatocitos/virología , Adenoviridae/genética , Adenoviridae/fisiología , Animales , Anticuerpos Bloqueadores/inmunología , Antígenos CD/genética , Antígenos CD/metabolismo , Células Cultivadas , Claudina-1 , Genotipo , Hepacivirus/genética , Hepacivirus/metabolismo , Hepatocitos/citología , Humanos , Inmunización Pasiva , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Receptores Virales/genética , Receptores Virales/metabolismo , Receptores Depuradores de Clase B/genética , Receptores Depuradores de Clase B/metabolismo , Tetraspanina 28 , Transfección , Tropismo Viral
8.
Expert Rev Gastroenterol Hepatol ; 4(5): 541-50, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20932139

RESUMEN

Approximately 2% of the worldwide population is infected with hepatitis C virus (HCV), the major causative agent of non-A, non-B hepatitis. Although substantial progress has been made in developing tools to dissect the viral life cycle, most in vitro studies rely on hepatoma cell lines, which are functionally disparate from the natural in vivo target of the virus ­ hepatocytes. To gain insights into virus­host interactions, there is a need for HCV-model systems that more closely mimic the physiological environment of the liver. Here, we discuss recent advances in culture and detection systems that facilitate the study of HCV in primary cells. Use of these new models may help bridge the gap between in vitro studies and clinical research.


Asunto(s)
Antivirales/uso terapéutico , Hepacivirus/efectos de los fármacos , Hepatitis C/tratamiento farmacológico , Hepatocitos/efectos de los fármacos , Animales , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Modelos Animales de Enfermedad , Hepacivirus/crecimiento & desarrollo , Hepacivirus/patogenicidad , Hepatitis C/virología , Hepatocitos/virología , Humanos
9.
Proc Natl Acad Sci U S A ; 107(7): 3141-5, 2010 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-20133632

RESUMEN

Hepatitis C virus (HCV) remains a major public health problem, affecting approximately 130 million people worldwide. HCV infection can lead to cirrhosis, hepatocellular carcinoma, and end-stage liver disease, as well as extrahepatic complications such as cryoglobulinemia and lymphoma. Preventative and therapeutic options are severely limited; there is no HCV vaccine available, and nonspecific, IFN-based treatments are frequently ineffective. Development of targeted antivirals has been hampered by the lack of robust HCV cell culture systems that reliably predict human responses. Here, we show the entire HCV life cycle recapitulated in micropatterned cocultures (MPCCs) of primary human hepatocytes and supportive stroma in a multiwell format. MPCCs form polarized cell layers expressing all known HCV entry factors and sustain viral replication for several weeks. When coupled with highly sensitive fluorescence- and luminescence-based reporter systems, MPCCs have potential as a high-throughput platform for simultaneous assessment of in vitro efficacy and toxicity profiles of anti-HCV therapeutics.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Hepacivirus/fisiología , Hepatitis C/fisiopatología , Hepatocitos/virología , Ingeniería de Tejidos/métodos , Anticuerpos Monoclonales/farmacología , Células Cultivadas , Humanos , Internalización del Virus/efectos de los fármacos , Replicación Viral/fisiología
10.
J Virol ; 84(9): 4504-12, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20181706

RESUMEN

The zinc finger antiviral protein (ZAP) is a host factor with potent antiviral activity when overexpressed in cells. ZAP blocks replication of the prototype alphavirus Sindbis virus (SINV) at a step at or before translation of the incoming viral genome. The mechanism of ZAP anti-SINV activity and the determinants of its antiviral function, however, have not been defined. Here, we have identified a dominant negative inhibitor of human ZAP. Rat ZAP with a cysteine-to-arginine mutation at position 88 (rZAPC88R), previously reported as a nonfunctional form of ZAP, increases SINV growth in cells. These results led us to discover a previously undetectable pool of endogenous functional ZAP within human cells. Investigation of the mechanism of dominant negative inhibition, combined with a comprehensive mutational analysis of the antiviral factor, revealed that homotypic associations are required for ZAP function in limiting SINV propagation.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/farmacología , Inhibidores Enzimáticos/farmacología , Proteínas Mutantes/genética , Proteínas Mutantes/farmacología , Proteínas de Unión al ARN/antagonistas & inhibidores , Virus Sindbis/crecimiento & desarrollo , Sustitución de Aminoácidos/genética , Animales , Línea Celular , Células Cultivadas , Cricetinae , Humanos , Mutación Missense , Unión Proteica , Ratas , Virus Sindbis/inmunología , Carga Viral
11.
J Virol ; 84(4): 1666-73, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20007277

RESUMEN

Hepatitis C virus (HCV) is a liver-tropic pathogen with severe health consequences for infected individuals. Chronic HCV infection can progress to cirrhosis and hepatocellular carcinoma and is a leading indicator for liver transplantation. The HCV core protein is an essential component of the infectious virus particle, but many aspects of its role remain undefined. The C-terminal region of the core protein acts as a signal sequence for the E1 glycoprotein and undergoes dual processing events during infectious virus assembly. The exact C terminus of the mature, virion-associated core protein is not known. Here, we performed genetic analyses to map the essential determinants of the HCV core C-terminal region, as well as to define the minimal length of the protein that can function for infectious virus production in trans.


Asunto(s)
Hepacivirus/genética , Proteínas del Núcleo Viral/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Línea Celular , Genes Virales , Prueba de Complementación Genética , Hepacivirus/patogenicidad , Hepacivirus/fisiología , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Señales de Clasificación de Proteína/genética , ARN Viral/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Transfección , Proteínas del Núcleo Viral/química , Virulencia , Ensamble de Virus/genética , Ensamble de Virus/fisiología , Replicación Viral/genética , Replicación Viral/fisiología
12.
Hepatology ; 48(6): 1843-50, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19003912

RESUMEN

UNLABELLED: Hepatitis C virus (HCV) replicates primarily in the liver, but HCV RNA has been observed in association with other tissues and cells including B and T lymphocytes, monocytes, and dendritic cells. We have taken advantage of a recently described, robust system that fully recapitulates HCV entry, replication and virus production in vitro to re-examine the issue of HCV infection of blood cell subsets. The HCV replicase inhibitor 2'C-methyl adenosine was used to distinguish HCV RNA replication from RNA persistence. Whereas cell culture-grown HCV replicated in Huh-7.5 hepatoma cells, no HCV replication was detected in B or T lymphocytes, monocytes, macrophages, or dendritic cells from healthy donors. No blood cell subset tested expressed significant levels of Claudin-1, a tight junction protein needed for HCV infection of Huh-7.5 cells. A B cell line expressing high levels of Claudin-1, CD81, and scavenger receptor BI remained resistant to HCV pseudoparticle infection. We bypassed the block in HCV entry by transfecting HCV RNA into blood cell subsets. Transfected RNA was not detectably translated and induced high levels of interferon-alpha. Supernatants from HCV RNA-transfected macrophages inhibited HCV replication in Huh-7.5 cells. CONCLUSION: We conclude that multiple blocks prevent blood cells from supporting HCV infection.


Asunto(s)
Hepacivirus/fisiología , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/virología , Replicación Viral/fisiología , Antígenos CD/metabolismo , Linfocitos B/citología , Linfocitos B/metabolismo , Linfocitos B/virología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/virología , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Claudina-1 , Células Dendríticas/citología , Células Dendríticas/metabolismo , Células Dendríticas/virología , Hepacivirus/genética , Hepacivirus/patogenicidad , Humanos , Leucocitos Mononucleares/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Proteínas de la Membrana/metabolismo , ARN Viral/genética , Receptores Depuradores de Clase B/metabolismo , Tetraspanina 28 , Transfección
13.
J Virol ; 81(19): 10220-31, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17634240

RESUMEN

Hepatitis C virus (HCV) is an important human pathogen affecting an estimated 3% of the world's population. Recent advances have enabled in vitro propagation of the virus and allow assembly and egress to be investigated for the first time. As a component of the virion, the HCV core protein likely functions primarily in infectious virus production, although little is known about the determinants of this activity. To investigate the roles of core in the viral life cycle, we performed a comprehensive deletion and alanine scanning mutagenesis study of this protein in the context of a genotype 2a reporter virus. We have confirmed that core protein is essential for infectious virion production and have identified numerous residues required for this role. The infectivity of several assembly-defective core mutants could be rescued by compensatory mutations identified in p7 and NS2, suggesting genetic interactions with core and highlighting the importance of these nonstructural proteins in infectious virion morphogenesis.


Asunto(s)
Hepacivirus/fisiología , Proteínas del Núcleo Viral/fisiología , Virión/fisiología , Replicación Viral/genética , Alanina/química , Alanina/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Línea Celular , Genoma Viral/genética , Hepacivirus/genética , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Serina/química , Serina/genética , Proteínas del Núcleo Viral/genética , Virión/genética
14.
J Virol ; 80(22): 11331-42, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16943299

RESUMEN

Hepatitis C virus (HCV) entry is dependent on CD81. To investigate whether the CD81 sequence is a determinant of HCV host range, we expressed a panel of diverse CD81 proteins and tested their ability to interact with HCV. CD81 large extracellular loop (LEL) sequences were expressed as recombinant proteins; the human and, to a low level, the African green monkey sequences bound soluble HCV E2 (sE2) and inhibited infection by retrovirus pseudotype particles bearing HCV glycoproteins (HCVpp). In contrast, mouse or rat CD81 proteins failed to bind sE2 or to inhibit HCVpp infection. However, CD81 proteins from all species, when expressed in HepG2 cells, conferred susceptibility to infection by HCVpp and cell culture-grown HCV to various levels, with the rat sequence being the least efficient. Recombinant human CD81 LEL inhibited HCVpp infectivity only if present during the virus-cell incubation, consistent with a role for CD81 after virus attachment. Amino acid changes that abrogate sE2 binding (I182F, N184Y, and F186S, alone or in combination) were introduced into human CD81. All three amino acid changes in human CD81 resulted in a molecule that still supported HCVpp infection, albeit with reduced efficiency. In summary, there is a remarkable plasticity in the range of CD81 sequences that can support HCV entry, suggesting that CD81 polymorphism may contribute to, but alone does not define, the HCV susceptibility of a species. In addition, the capacity to support viral entry is only partially reflected by assays measuring sE2 interaction with recombinant or full-length CD81 proteins.


Asunto(s)
Antígenos CD/fisiología , Hepacivirus/fisiología , Receptores Virales/fisiología , Internalización del Virus , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Antígenos CD/genética , Línea Celular Tumoral , Cricetinae , Citometría de Flujo , Haplorrinos , Humanos , Inmunohistoquímica , Ratones , Datos de Secuencia Molecular , Mutación Missense , Unión Proteica , Ratas , Receptores Virales/genética , Especificidad de la Especie , Tetraspanina 28 , Proteínas del Envoltorio Viral/metabolismo
15.
J Virol ; 80(4): 1734-41, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16439530

RESUMEN

Hepatitis C virus (HCV) is an important human pathogen associated with chronic liver disease. Recently, based on a genotype 2a isolate, tissue culture systems supporting complete replication and infectious virus production have been developed. In this study, we used cell culture-produced infectious HCV to analyze the viral entry pathway into Huh-7.5 cells. Bafilomycin A1 and concanamycin A, inhibitors of vacuolar ATPases, prevented HCV entry when they were present prior to infection and had minimal effect on downstream replication events. HCV entry therefore appears to be pH dependent, requiring an acidified intracellular compartment. For many other enveloped viruses, acidic pH triggers an irreversible conformational change, which promotes virion-endosomal membrane fusion. Such viruses are often inactivated by low pH. In the case of HCV, exposure of virions to acidic pH followed by return to neutral pH did not affect their infectivity. This parallels the observation made for the related pestivirus bovine viral diarrhea virus. Low pH could activate the entry of cell surface-bound HCV but only after prolonged incubation at 37 degrees C. This suggests that there are rate-limiting, postbinding events that are needed to render HCV competent for low-pH-triggered entry. Such events may involve interaction with a cellular coreceptor or other factors but do not require cathepsins B and L, late endosomal proteases that activate Ebola virus and reovirus for entry.


Asunto(s)
Hepacivirus/fisiología , Replicación Viral , Catepsina B/fisiología , Catepsina L , Catepsinas/fisiología , Línea Celular , Cisteína Endopeptidasas/fisiología , Inhibidores Enzimáticos/farmacología , Humanos , Concentración de Iones de Hidrógeno , Macrólidos/farmacología , Temperatura , Factores de Tiempo , Inactivación de Virus
16.
Virology ; 331(2): 247-59, 2005 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-15629769

RESUMEN

Subgenomic replicons of yellow fever virus (YFV) were constructed to allow expression of heterologous reporter genes in a replication-dependent manner. Expression of the antibiotic resistance gene neomycin phosphotransferase II (Neo) from one of these YFV replicons allowed selection of a stable population of cells (BHK-REP cells) in which the YFV replicon persistently replicated. BHK-REP cells were successfully used to trans-complement replication-defective YFV replicons harboring large internal deletions within either the NS1 or NS3 proteins. Although replicons with large deletions in either NS1 or NS3 were trans-complemented in BHK-REP, replicons that contained deletions of NS3 were trans-complemented at lower levels. In addition, replicons that retained the N-terminal protease domain of NS3 in cis were trans-complemented with higher efficiency than replicons in which both the protease and helicase domains of NS3 were deleted. To study packaging of YFV replicons, Sindbis replicons were constructed that expressed the YFV structural proteins in trans. Using these Sindbis replicons, both replication-competent and trans-complemented, replication-defective YFV replicons could be packaged into pseudo-infectious particles (PIPs). Although these results eliminate a potential role of either NS1 or full-length NS3 in cis for packaging and assembly of the flavivirus virion, they do not preclude the possibility that these proteins may act in trans during these processes.


Asunto(s)
Flavivirus/genética , Vectores Genéticos , Replicón , Virus Sindbis/genética , Proteínas Estructurales Virales/metabolismo , Ensamble de Virus/fisiología , Virus de la Fiebre Amarilla/genética , Línea Celular , Expresión Génica , Genes , Genes Reporteros , Prueba de Complementación Genética , Ingeniería Genética , Genoma Viral , Proteínas Fluorescentes Verdes , Proteínas Estructurales Virales/genética , Replicación Viral , Virus de la Fiebre Amarilla/fisiología
17.
Cell ; 108(5): 717-25, 2002 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-11893341

RESUMEN

The first structure of a flavivirus has been determined by using a combination of cryoelectron microscopy and fitting of the known structure of glycoprotein E into the electron density map. The virus core, within a lipid bilayer, has a less-ordered structure than the external, icosahedral scaffold of 90 glycoprotein E dimers. The three E monomers per icosahedral asymmetric unit do not have quasiequivalent symmetric environments. Difference maps indicate the location of the small membrane protein M relative to the overlaying scaffold of E dimers. The structure suggests that flaviviruses, and by analogy also alphaviruses, employ a fusion mechanism in which the distal beta barrels of domain II of the glycoprotein E are inserted into the cellular membrane.


Asunto(s)
Virus del Dengue/química , Virus del Dengue/fisiología , Cápside/química , Microscopía por Crioelectrón , Virus del Dengue/genética , Virus del Dengue/ultraestructura , Humanos , Concentración de Iones de Hidrógeno , Procesamiento de Imagen Asistido por Computador , Membrana Dobles de Lípidos/química , Fusión de Membrana/fisiología , Modelos Moleculares , Proteínas del Envoltorio Viral/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA