Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Proc Natl Acad Sci U S A ; 114(37): 9948-9953, 2017 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-28851829

RESUMEN

Glucocorticoid (GC) and hypoxic transcriptional responses play a central role in tissue homeostasis and regulate the cellular response to stress and inflammation, highlighting the potential for cross-talk between these two signaling pathways. We present results from an unbiased in vivo chemical screen in zebrafish that identifies GCs as activators of hypoxia-inducible factors (HIFs) in the liver. GCs activated consensus hypoxia response element (HRE) reporters in a glucocorticoid receptor (GR)-dependent manner. Importantly, GCs activated HIF transcriptional responses in a zebrafish mutant line harboring a point mutation in the GR DNA-binding domain, suggesting a nontranscriptional route for GR to activate HIF signaling. We noted that GCs increase the transcription of several key regulators of glucose metabolism that contain HREs, suggesting a role for GC/HIF cross-talk in regulating glucose homeostasis. Importantly, we show that GCs stabilize HIF protein in intact human liver tissue and isolated hepatocytes. We find that GCs limit the expression of Von Hippel Lindau protein (pVHL), a negative regulator of HIF, and that treatment with the c-src inhibitor PP2 rescued this effect, suggesting a role for GCs in promoting c-src-mediated proteosomal degradation of pVHL. Our data support a model for GCs to stabilize HIF through activation of c-src and subsequent destabilization of pVHL.


Asunto(s)
Glucocorticoides/farmacología , Glucocorticoides/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Animales , Hipoxia de la Célula/fisiología , Humanos , Hipoxia , Ligasas/metabolismo , Hígado/metabolismo , Unión Proteica , Transducción de Señal/fisiología , Transactivadores/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Pez Cebra , Enfermedad de von Hippel-Lindau/metabolismo
2.
Hepatology ; 65(1): 294-309, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27770554

RESUMEN

The recruitment of lymphocytes via the hepatic sinusoidal channels and positioning within liver tissue is a critical event in the development and persistence of chronic inflammatory liver diseases. The hepatic sinusoid is a unique vascular bed lined by hepatic sinusoidal endothelial cells (HSECs), a functionally and phenotypically distinct subpopulation of endothelial cells. Using flow-based adhesion assays to study the migration of lymphocytes across primary human HSECs, we found that lymphocytes enter into HSECs, confirmed by electron microscopy demonstrating clear intracellular localization of lymphocytes in vitro and by studies in human liver tissues. Stimulation by interferon-γ increased intracellular localization of lymphocytes within HSECs. Furthermore, using confocal imaging and time-lapse recordings, we demonstrated "intracellular crawling" of lymphocytes entering into one endothelial cell from another. This required the expression of intracellular adhesion molecule-1 and stabilin-1 and was facilitated by the junctional complexes between HSECs. CONCLUSION: Lymphocyte migration is facilitated by the unique structure of HSECs. Intracellular crawling may contribute to optimal lymphocyte positioning in liver tissue during chronic hepatitis. (Hepatology 2017;65:294-309).


Asunto(s)
Capilares/citología , Movimiento Celular , Células Endoteliales/fisiología , Linfocitos/fisiología , Citoplasma , Endotelio Vascular/citología , Humanos , Hígado/irrigación sanguínea
3.
J Hepatol ; 66(5): 919-929, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28126468

RESUMEN

BACKGROUND & AIMS: Chronic hepatitis C is a global health problem with an estimated 170 million hepatitis C virus (HCV) infected individuals at risk of progressive liver disease and hepatocellular carcinoma (HCC). Autotaxin (ATX, gene name: ENPP2) is a phospholipase with diverse roles in the physiological and pathological processes including inflammation and oncogenesis. Clinical studies have reported increased ATX expression in chronic hepatitis C, however, the pathways regulating ATX and its role in the viral life cycle are not well understood. METHODS: In vitro hepatocyte and ex vivo liver culture systems along with chimeric humanized liver mice and HCC tissue enabled us to assess the interplay between ATX and the HCV life cycle. RESULTS: HCV infection increased hepatocellular ATX RNA and protein expression. HCV infection stabilizes hypoxia inducible factors (HIFs) and we investigated a role for these transcription factors to regulate ATX. In vitro studies show that low oxygen increases hepatocellular ATX expression and transcriptome analysis showed a positive correlation between ATX mRNA levels and hypoxia gene score in HCC tumour tissue associated with HCV and other aetiologies. Importantly, inhibiting ATX-lysophosphatidic acid (LPA) signalling reduced HCV replication, demonstrating a positive role for this phospholipase in the viral life cycle. LPA activates phosphoinositide-3-kinase that stabilizes HIF-1α and inhibiting the HIF signalling pathway abrogates the pro-viral activity of LPA. CONCLUSIONS: Our data support a model where HCV infection increases ATX expression which supports viral replication and HCC progression. LAY SUMMARY: Chronic hepatitis C is a global health problem with infected individuals at risk of developing liver disease that can progress to hepatocellular carcinoma. Autotaxin generates the biologically active lipid lysophosphatidic acid that has been reported to play a tumorigenic role in a wide number of cancers. In this study we show that hepatitis C virus infection increases autotaxin expression via hypoxia inducible transcription factor and provides an environment in the liver that promotes fibrosis and liver injury. Importantly, we show a new role for lysophosphatidic acid in positively regulating hepatitis C virus replication.


Asunto(s)
Hepacivirus/fisiología , Hidrolasas Diéster Fosfóricas/fisiología , Receptores del Ácido Lisofosfatídico/fisiología , Replicación Viral , Animales , Línea Celular , Hepatitis C Crónica/complicaciones , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Neoplasias Hepáticas/etiología , Ratones , Hidrolasas Diéster Fosfóricas/genética , Regiones Promotoras Genéticas , ARN Mensajero/análisis , Transducción de Señal
4.
J Gen Virol ; 96(Pt 6): 1369-1373, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25667327

RESUMEN

The major cell type supporting hepatitis C virus (HCV) infection is the hepatocyte; however, most reports studying viral entry and replication utilize transformed hepatoma cell lines. We demonstrate that HCV pseudoparticles (HCVpp) infect primary hepatocytes with comparable rates to hepatoma cells, demonstrating the limited variability in donor hepatocytes to support HCV receptor-glycoprotein-dependent entry. In contrast, we observed a 2-log range in viral replication between the same donor hepatocytes. We noted that cell proliferation augments pseudoparticle reporter activity and arresting hepatoma cells yields comparable levels of infection to hepatocytes. This study demonstrates comparable rates of HCVpp entry into primary hepatocytes and hepatoma cells, validating the use of transformed cells as a model system to study HCV entry.


Asunto(s)
Hepacivirus/fisiología , Hepatocitos/virología , Tropismo Viral , Células Cultivadas , Humanos , Internalización del Virus , Replicación Viral
5.
J Gen Virol ; 96(Pt 6): 1380-1388, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25701818

RESUMEN

Hepatitis C virus (HCV) infects the liver and hepatocytes are the major cell type supporting viral replication. Hepatocytes and cholangiocytes derive from a common hepatic progenitor cell that proliferates during inflammatory conditions, raising the possibility that cholangiocytes may support HCV replication and contribute to the hepatic reservoir. We screened cholangiocytes along with a panel of cholangiocarcinoma-derived cell lines for their ability to support HCV entry and replication. While primary cholangiocytes were refractory to infection and lacked expression of several entry factors, two cholangiocarcinoma lines, CC-LP-1 and Sk-ChA-1, supported efficient HCV entry; furthermore, Sk-ChA-1 cells supported full virus replication. In vivo cholangiocarcinomas expressed all of the essential HCV entry factors; however, cholangiocytes adjacent to the tumour and in normal tissue showed a similar pattern of receptor expression to ex vivo isolated cholangiocytes, lacking SR-BI expression, explaining their inability to support infection. This study provides the first report that HCV can infect cholangiocarcinoma cells and suggests that these heterogeneous tumours may provide a reservoir for HCV replication in vivo.


Asunto(s)
Células Epiteliales/virología , Hepacivirus/fisiología , Tropismo Viral , Línea Celular Tumoral , Hepacivirus/crecimiento & desarrollo , Humanos , Internalización del Virus , Replicación Viral
6.
Hepatology ; 59(2): 375-84, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23775568

RESUMEN

UNLABELLED: Hepatitis C virus (HCV) is a major cause of global morbidity, causing chronic liver injury that can progress to cirrhosis and hepatocellular carcinoma. The liver is a large and complex organ containing multiple cell types, including hepatocytes, sinusoidal endothelial cells (LSEC), Kupffer cells, and biliary epithelial cells. Hepatocytes are the major reservoir supporting HCV replication; however, the role of nonparenchymal cells in the viral lifecycle remains largely unexplored. LSEC secrete factors that promote HCV infection and transcript analysis identified bone morphogenetic protein 4 (BMP4) as a candidate endothelial-expressed proviral molecule. Recombinant BMP4 increased HCV replication and neutralization of BMP4 abrogated the proviral activity of LSEC-conditioned media. Importantly, BMP4 expression was negatively regulated by vascular endothelial growth factor A (VEGF-A) by way of a VEGF receptor-2 (VEGFR-2) primed activation of p38 MAPK. Consistent with our in vitro observations, we demonstrate that in normal liver VEGFR-2 is activated and BMP4 expression is suppressed. In contrast, in chronic liver disease including HCV infection where there is marked endothelial cell proliferation, we observed reduced endothelial cell VEGFR-2 activation and a concomitant increase in BMP4 expression. CONCLUSION: These studies identify a role for LSEC and BMP4 in HCV infection and highlight BMP4 as a new therapeutic target for treating individuals with liver disease.


Asunto(s)
Células Endoteliales/metabolismo , Hepacivirus/fisiología , Hígado/metabolismo , Comunicación Paracrina/fisiología , Replicación Viral/fisiología , Proteína Morfogenética Ósea 4/metabolismo , Proliferación Celular , Células Cultivadas , Células Endoteliales/patología , Hepatocitos/patología , Hepatocitos/virología , Humanos , Técnicas In Vitro , Hígado/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
J Hepatol ; 61(6): 1397-406, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25157983

RESUMEN

Hypoxia inducible transcription factors (HIFs) activate diverse pathways that regulate cellular metabolism, angiogenesis, proliferation, and migration, enabling a cell to respond to a low oxygen or hypoxic environment. HIFs are regulated by oxygen-dependent and independent signals including: mitochondrial dysfunction, reactive oxygen species, endoplasmic reticular stress, and viral infection. HIFs have been reported to play a role in the pathogenesis of liver disease of diverse aetiologies. This review explores the impact of HIFs on hepatocellular biology and inflammatory responses, highlighting the therapeutic potential of targeting HIFs for an array of liver pathologies.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Carcinoma Hepatocelular/fisiopatología , Factor 1 Inducible por Hipoxia/fisiología , Hepatopatías/fisiopatología , Neoplasias Hepáticas/fisiopatología , Animales , Carcinoma Hepatocelular/etiología , Modelos Animales de Enfermedad , Hepatovirus/fisiología , Humanos , Hígado/metabolismo , Hígado/virología , Hepatopatías/etiología , Neoplasias Hepáticas/etiología , Ratones , Oxígeno/metabolismo , Virosis/fisiopatología
8.
J Hepatol ; 58(6): 1074-80, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23353869

RESUMEN

BACKGROUND & AIMS: Hepatitis C virus (HCV) poses a global health problem, with over 170 million chronically infected individuals at risk of developing progressive liver disease. The ability of a virus to spread within a host is a key determinant of its persistence and virulence. HCV can transmit in vitro by cell-free particle diffusion or via contact(s) between infected and naïve hepatocytes. However, limited information is available on the relative efficiency of these routes, our aim is to develop physiologically relevant assays to quantify these processes. METHODS: We developed a single-cycle infection assay to measure HCV transmission rates. RESULTS: We compared HCV spread in proliferating and arrested cell systems and demonstrated a significant reduction in cell-to-cell infection of arrested target cells. Comparison of cell-free and cell-to-cell virus spread demonstrated relatively poor transmission rates, with 10-50 infected producer cells required to infect a single naïve target cell. We found HCV strain J6/JFH to be 10-fold more efficient at spreading via the cell-to-cell route than cell-free, whereas SA13/JFH and HK6/JFH strains showed comparable rates of infection via both routes. Importantly, the level of infectious virus released from cells did not predict the ability of a virus to spread in vitro, highlighting the importance of studying cell-associated viruses. CONCLUSIONS: These studies demonstrate the relatively poor infectivity of HCV and highlight differences between strains in their efficiency and preferred route of transmission that may inform future therapeutic strategies that target virus entry.


Asunto(s)
Hepacivirus/fisiología , Hepatocitos/virología , Adhesión Celular , Comunicación Celular , Línea Celular , Humanos , Receptores Depuradores de Clase B/fisiología
9.
Gastroenterology ; 142(3): 634-643.e6, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22138189

RESUMEN

BACKGROUND & AIMS: Hepatitis C virus (HCV) infection leads to progressive liver disease and is associated with a variety of extrahepatic syndromes, including central nervous system (CNS) abnormalities. However, it is unclear whether such cognitive abnormalities are a function of systemic disease, impaired hepatic function, or virus infection of the CNS. METHODS: We measured levels of HCV RNA and expression of the viral entry receptor in brain tissue samples from 10 infected individuals (and 3 uninfected individuals, as controls) and human brain microvascular endothelial cells by using quantitative polymerase chain reaction and immunochemical and confocal imaging analyses. HCV pseudoparticles and cell culture-derived HCV were used to study the ability of endothelial cells to support viral entry and replication. RESULTS: Using quantitative polymerase chain reaction, we detected HCV RNA in brain tissue of infected individuals at significantly lower levels than in liver samples. Brain microvascular endothelia and brain endothelial cells expressed all of the recognized HCV entry receptors. Two independently derived brain endothelial cell lines, hCMEC/D3 and HBMEC, supported HCV entry and replication. These processes were inhibited by antibodies against the entry factors CD81, scavenger receptor BI, and claudin-1; by interferon; and by reagents that inhibit NS3 protease and NS5B polymerase. HCV infection promotes endothelial permeability and cellular apoptosis. CONCLUSIONS: Human brain endothelial cells express functional receptors that support HCV entry and replication. Virus infection of the CNS might lead to HCV-associated neuropathologies.


Asunto(s)
Barrera Hematoencefálica/virología , Células Endoteliales/virología , Hepacivirus/patogenicidad , Hepatitis C/virología , Microvasos/virología , Adulto , Antivirales/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Permeabilidad Capilar , Estudios de Casos y Controles , Línea Celular Tumoral , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Células HEK293 , Hepacivirus/genética , Hepatitis C/complicaciones , Hepatitis C/mortalidad , Humanos , Inmunohistoquímica , Hígado/virología , Masculino , Microscopía Confocal , Microvasos/efectos de los fármacos , Microvasos/metabolismo , Microvasos/patología , Persona de Mediana Edad , ARN Viral/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Virales/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Virión/metabolismo , Internalización del Virus , Replicación Viral
10.
Rev Med Virol ; 22(3): 182-93, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22392805

RESUMEN

HCV is a blood-borne pathogen that affects approximately 3% of the global population and leads to progressive liver disease. Recent advances have identified an essential role for host cell molecules: tetraspanin CD81, scavenger receptor B1 and the tight junction proteins claudin-1 and occludin in HCV entry, suggesting a complex multi-step process. The conserved nature of this receptor-dependent step in the viral life cycle offers an attractive target for therapeutic intervention. Evidence is emerging that additional factors other than classical receptors, such as inflammatory mediators regulate the ability of hepatocytes to support HCV entry, and as such may provide potential avenues for drug design and development. In this review, we summarise the recent literature on HCV entry mechanisms with a view to realising the future potential of therapeutically targeting this process.


Asunto(s)
Hepacivirus/fisiología , Hepatitis C/metabolismo , Hepatitis C/virología , Receptores Virales/metabolismo , Internalización del Virus , Animales , Hepacivirus/genética , Hepatitis C/genética , Humanos , Receptores Virales/genética
11.
J Hepatol ; 56(4): 803-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22178269

RESUMEN

BACKGROUND & AIMS: Hepatitis C virus (HCV) causes progressive liver disease and is a major risk factor for the development of hepatocellular carcinoma (HCC). However, the role of infection in HCC pathogenesis is poorly understood. We investigated the effect(s) of HCV infection and viral glycoprotein expression on hepatoma biology to gain insights into the development of HCV associated HCC. METHODS: We assessed the effect(s) of HCV and viral glycoprotein expression on hepatoma polarity, migration and invasion. RESULTS: HCV glycoproteins perturb tight and adherens junction protein expression, and increase hepatoma migration and expression of epithelial to mesenchymal transition markers Snail and Twist via stabilizing hypoxia inducible factor-1α (HIF-1α). HIF-1α regulates many genes involved in tumor growth and metastasis, including vascular endothelial growth factor (VEGF) and transforming growth factor-beta (TGF-ß). Neutralization of both growth factors shows different roles for VEGF and TGFß in regulating hepatoma polarity and migration, respectively. Importantly, we confirmed these observations in virus infected hepatoma and primary human hepatocytes. Inhibition of HIF-1α reversed the effect(s) of infection and glycoprotein expression on hepatoma permeability and migration and significantly reduced HCV replication, demonstrating a dual role for HIF-1α in the cellular processes that are deregulated in many human cancers and in the viral life cycle. CONCLUSIONS: These data provide new insights into the cancer-promoting effects of HCV infection on HCC migration and offer new approaches for treatment.


Asunto(s)
Carcinoma Hepatocelular/fisiopatología , Movimiento Celular/fisiología , Hepacivirus/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Neoplasias Hepáticas/fisiopatología , Replicación Viral/fisiología , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Polaridad Celular/fisiología , Progresión de la Enfermedad , Glicoproteínas/fisiología , Hepatitis C/patología , Hepatitis C/fisiopatología , Humanos , Neoplasias Hepáticas/patología , Uniones Estrechas/fisiología , Factor de Crecimiento Transformador beta/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología
13.
Sci Rep ; 6: 29417, 2016 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-27388959

RESUMEN

Angiogenesis is an essential physiological process and an important factor in disease pathogenesis. However, its exploitation as a clinical target has achieved limited success and novel molecular targets are required. Although heme oxygenase-1 (HO-1) acts downstream of vascular endothelial growth factor (VEGF) to modulate angiogenesis, knowledge of the mechanisms involved remains limited. We set out identify novel HO-1 targets involved in angiogenesis. HO-1 depletion attenuated VEGF-induced human endothelial cell (EC) proliferation and tube formation. The latter response suggested a role for HO-1 in EC migration, and indeed HO-1 siRNA negatively affected directional migration of EC towards VEGF; a phenotype reversed by HO-1 over-expression. EC from Hmox1(-/-) mice behaved similarly. Microarray analysis of HO-1-depleted and control EC exposed to VEGF identified cyclins A1 and E1 as HO-1 targets. Migrating HO-1-deficient EC showed increased p27, reduced cyclin A1 and attenuated cyclin-dependent kinase 2 activity. In vivo, cyclin A1 siRNA inhibited VEGF-driven angiogenesis, a response reversed by Ad-HO-1. Proteomics identified structural protein vimentin as an additional VEGF-HO-1 target. HO-1 depletion inhibited VEGF-induced calpain activity and vimentin cleavage, while vimentin silencing attenuated HO-1-driven proliferation. Thus, vimentin and cyclins A1 and E1 represent VEGF-activated HO-1-dependent targets important for VEGF-driven angiogenesis.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Ciclina A1/genética , Ciclina E/genética , Células Madre Embrionarias/citología , Hemo-Oxigenasa 1/genética , Factor A de Crecimiento Endotelial Vascular/farmacología , Vimentina/genética , Animales , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quinasa 2 Dependiente de la Ciclina/metabolismo , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Hemo-Oxigenasa 1/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Proteómica
14.
Nat Biotechnol ; 33(5): 549-554, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25798937

RESUMEN

Hepatitis C virus (HCV) infection is a leading cause of liver cirrhosis and cancer. Cell entry of HCV and other pathogens is mediated by tight junction (TJ) proteins, but successful therapeutic targeting of TJ proteins has not been reported yet. Using a human liver-chimeric mouse model, we show that a monoclonal antibody specific for the TJ protein claudin-1 (ref. 7) eliminates chronic HCV infection without detectable toxicity. This antibody inhibits HCV entry, cell-cell transmission and virus-induced signaling events. Antibody treatment reduces the number of HCV-infected hepatocytes in vivo, highlighting the need for de novo infection by means of host entry factors to maintain chronic infection. In summary, we demonstrate that an antibody targeting a virus receptor can cure chronic viral infection and uncover TJ proteins as targets for antiviral therapy.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Claudina-1/inmunología , Hepatitis C/terapia , Cirrosis Hepática/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/inmunología , Claudina-1/uso terapéutico , Hepacivirus/inmunología , Hepacivirus/patogenicidad , Hepatitis C/inmunología , Hepatitis C/virología , Hepatocitos/inmunología , Humanos , Cirrosis Hepática/terapia , Cirrosis Hepática/virología , Ratones
15.
Viruses ; 6(3): 1454-72, 2014 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-24662676

RESUMEN

Tetraspanins are a family of small proteins that interact with themselves, host transmembrane and cytosolic proteins to form tetraspanin enriched microdomains (TEMs) that regulate important cellular functions. Several tetraspanin family members are linked to tumorigenesis. Hepatocellular carcinoma (HCC) is an increasing global health burden, in part due to the increasing prevalence of hepatitis C virus (HCV) associated HCC. The tetraspanin CD81 is an essential receptor for HCV, however, its role in hepatoma biology is uncertain. We demonstrate that antibody engagement of CD81 promotes hepatoma spread, which is limited by HCV infection, in an actin-dependent manner and identify an essential role for the C-terminal interaction with Ezrin-Radixin-Moesin (ERM) proteins in this process. We show enhanced hepatoma migration and invasion following expression of CD81 and a reduction in invasive potential upon CD81 silencing. In addition, we reveal poorly differentiated HCC express significantly higher levels of CD81 compared to adjacent non-tumor tissue. In summary, these data support a role for CD81 in regulating hepatoma mobility and propose CD81 as a tumour promoter.


Asunto(s)
Carcinoma Hepatocelular/patología , Hepacivirus/fisiología , Tetraspanina 28/metabolismo , Animales , Línea Celular , Proteínas de Unión al ADN/metabolismo , Humanos , Mapeo de Interacción de Proteínas , Receptores Virales/metabolismo , Factores de Transcripción/metabolismo
16.
Int J Hepatol ; 2012: 292591, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23056952

RESUMEN

The study of a virus is made possible by the availability of culture systems in which the viral lifecycle can be realized. Such systems support robust virus entry, replication, assembly, and secretion of nascent virions. Furthermore, culture models provide a platform in which therapeutic interventions can be devised or monitored. Hepatitis C virus (HCV) has a restricted tropism to human and chimpanzees; thus investigations of HCV biology have been hindered for many years due to a lack of small animal models. Nevertheless, significant efforts have been directed at developing cell culture models to elucidate the viral lifecycle in vitro. HCV primarily infects liver parenchymal cells commonly known as hepatocytes. The liver is a highly specialized and complex organ and the development of in vitro systems that reflects this complexity has proven difficult. Consequently, host cell receptor molecules that potentiate HCV infection were identified over a decade after the virus was discovered. A summary of the various HCV in vitro culture models, their advantages, and disadvantages are described.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA