Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Virol ; 88(21): 12599-611, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25142587

ABSTRACT

UNLABELLED: The serine-arginine-specific protein kinase SRPK1 is a common binding partner of the E1^E4 protein of diverse human papillomavirus types. We show here for the first time that the interaction between HPV1 E1^E4 and SRPK1 leads to potent inhibition of SRPK1 phosphorylation of host serine-arginine (SR) proteins that have critical roles in mRNA metabolism, including pre-mRNA processing, mRNA export, and translation. Furthermore, we show that SRPK1 phosphorylates serine residues of SR/RS dipeptides in the hinge region of the HPV1 E2 protein in in vitro kinase assays and that HPV1 E1^E4 inhibits this phosphorylation. After mutation of the putative phosphoacceptor serine residues, the localization of the E2 protein was altered in primary human keratinocytes; with a significant increase in the cell population showing intense E2 staining of the nucleolus. A similar effect was observed following coexpression of E2 and E1^E4 that is competent for inhibition of SRPK1 activity, suggesting that the nuclear localization of E2 is sensitive to E1^E4-mediated SRPK1 inhibition. Collectively, these data suggest that E1^E4-mediated inhibition of SRPK1 could affect the functions of host SR proteins and those of the virus transcription/replication regulator E2. We speculate that the novel E4 function identified here is involved in the regulation of E2 and SR protein function in posttranscriptional processing of viral transcripts. IMPORTANCE: The HPV life cycle is tightly linked to the epithelial terminal differentiation program, with the virion-producing phase restricted to differentiating cells. While the most abundant HPV protein expressed in this phase is the E4 protein, we do not fully understand the role of this protein. Few E4 interaction partners have been identified, but we had previously shown that E4 proteins from diverse papillomaviruses interact with the serine-arginine-specific protein kinase SRPK1, a kinase important in the replication cycles of a diverse range of DNA and RNA viruses. We show that HPV1 E4 is a potent inhibitor of this host cell kinase. We show that E4 inhibits SRPK1 phosphorylation, not only of cellular SR proteins involved in regulating alternative splicing of RNA but also the viral transcription/replication regulator E2. Our findings reveal a potential E4 function in regulation of viral late gene expression through the inhibition of a host cell kinase.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , Host-Pathogen Interactions , Oncogene Proteins, Viral/antagonists & inhibitors , Oncogene Proteins, Viral/metabolism , Papillomaviridae/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Transcription, Genetic , Viral Proteins/metabolism , Cell Line , Humans , Phosphorylation , Protein Interaction Mapping , Virus Replication
2.
Viruses ; 6(3): 1454-72, 2014 Mar 24.
Article in English | MEDLINE | ID: mdl-24662676

ABSTRACT

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.


Subject(s)
Carcinoma, Hepatocellular/pathology , Hepacivirus/physiology , Tetraspanin 28/metabolism , Animals , Cell Line , DNA-Binding Proteins/metabolism , Humans , Protein Interaction Mapping , Receptors, Virus/metabolism , Transcription Factors/metabolism
3.
J Virol ; 86(8): 4305-16, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22318146

ABSTRACT

Hepatitis C virus (HCV) leads to progressive liver disease and hepatocellular carcinoma. Current treatments are only partially effective, and new therapies targeting viral and host pathways are required. Virus entry into a host cell provides a conserved target for therapeutic intervention. Tetraspanin CD81, scavenger receptor class B member I, and the tight-junction proteins claudin-1 and occludin have been identified as essential entry receptors. Limited information is available on the role of receptor trafficking in HCV entry. We demonstrate here that anti-CD81 antibodies inhibit HCV infection at late times after virus internalization, suggesting a role for intracellular CD81 in HCV infection. Several tetraspanins have been reported to internalize via motifs in their C-terminal cytoplasmic domains; however, CD81 lacks such motifs, leading several laboratories to suggest a limited role for CD81 endocytosis in HCV entry. We demonstrate CD81 internalization via a clathrin- and dynamin-dependent process, independent of its cytoplasmic domain, suggesting a role for associated partner proteins in regulating CD81 trafficking. Live cell imaging demonstrates CD81 and claudin-1 coendocytosis and fusion with Rab5 expressing endosomes, supporting a role for this receptor complex in HCV internalization. Receptor-specific antibodies and HCV particles increase CD81 and claudin-1 endocytosis, supporting a model wherein HCV stimulates receptor trafficking to promote particle internalization.


Subject(s)
Endocytosis , Hepacivirus/metabolism , Membrane Proteins/metabolism , Tetraspanin 28/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/metabolism , Antibody Affinity/immunology , Cell Line , Claudin-1 , Humans , Protein Structure, Tertiary , Protein Transport , Receptors, Virus/metabolism , Tetraspanin 28/chemistry , Tetraspanin 28/immunology , Viral Envelope Proteins/metabolism , Virus Internalization
4.
J Hepatol ; 56(4): 803-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22178269

ABSTRACT

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.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Cell Movement/physiology , Hepacivirus/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Liver Neoplasms/physiopathology , Virus Replication/physiology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Polarity/physiology , Disease Progression , Glycoproteins/physiology , Hepatitis C/pathology , Hepatitis C/physiopathology , Humans , Liver Neoplasms/pathology , Tight Junctions/physiology , Transforming Growth Factor beta/physiology , Vascular Endothelial Growth Factor A/physiology
5.
J Virol ; 85(1): 596-605, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20962076

ABSTRACT

Hepatitis C virus (HCV) can initiate infection by cell-free particle and cell-cell contact-dependent transmission. In this study we use a novel infectious coculture system to examine these alternative modes of infection. Cell-to-cell transmission is relatively resistant to anti-HCV glycoprotein monoclonal antibodies and polyclonal immunoglobulin isolated from infected individuals, providing an effective strategy for escaping host humoral immune responses. Chimeric viruses expressing the structural proteins representing the seven major HCV genotypes demonstrate neutralizing antibody-resistant cell-to-cell transmission. HCV entry is a multistep process involving numerous receptors. In this study we demonstrate that, in contrast to earlier reports, CD81 and the tight-junction components claudin-1 and occludin are all essential for both cell-free and cell-to-cell viral transmission. However, scavenger receptor BI (SR-BI) has a more prominent role in cell-to-cell transmission of the virus, with SR-BI-specific antibodies and small-molecule inhibitors showing preferential inhibition of this infection route. These observations highlight the importance of targeting host cell receptors, in particular SR-BI, to control viral infection and spread in the liver.


Subject(s)
Antibodies, Neutralizing/immunology , Hepacivirus/physiology , Hepatitis C Antibodies/immunology , Scavenger Receptors, Class B/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Line, Tumor , Claudin-1 , Coculture Techniques , Hepacivirus/immunology , Hepacivirus/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Occludin , Receptors, Virus/genetics , Receptors, Virus/metabolism , Scavenger Receptors, Class B/genetics , Tetraspanin 28 , Tight Junctions/genetics , Tight Junctions/metabolism
6.
J Virol ; 82(17): 8797-811, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18579596

ABSTRACT

Viruses exploit signaling pathways to their advantage during multiple stages of their life cycle. We demonstrate a role for protein kinase A (PKA) in the hepatitis C virus (HCV) life cycle. The inhibition of PKA with H89, cyclic AMP (cAMP) antagonists, or the protein kinase inhibitor peptide reduced HCV entry into Huh-7.5 hepatoma cells. Bioluminescence resonance energy transfer methodology allowed us to investigate the PKA isoform specificity of the cAMP antagonists in Huh-7.5 cells, suggesting a role for PKA type II in HCV internalization. Since viral entry is dependent on the host cell expression of CD81, scavenger receptor BI, and claudin-1 (CLDN1), we studied the role of PKA in regulating viral receptor localization by confocal imaging and fluorescence resonance energy transfer (FRET) analysis. Inhibiting PKA activity in Huh-7.5 cells induced a reorganization of CLDN1 from the plasma membrane to an intracellular vesicular location(s) and disrupted FRET between CLDN1 and CD81, demonstrating the importance of CLDN1 expression at the plasma membrane for viral receptor activity. Inhibiting PKA activity in Huh-7.5 cells reduced the infectivity of extracellular virus without modulating the level of cell-free HCV RNA, suggesting that particle secretion was not affected but that specific infectivity was reduced. Viral particles released from H89-treated cells displayed the same range of buoyant densities as did those from control cells, suggesting that viral protein association with lipoproteins is not regulated by PKA. HCV infection of Huh-7.5 cells increased cAMP levels and phosphorylated PKA substrates, supporting a model where infection activates PKA in a cAMP-dependent manner to promote virus release and transmission.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Hepacivirus/physiology , Hepacivirus/pathogenicity , Virus Internalization , Antigens, CD/analysis , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Claudin-1 , Fluorescent Antibody Technique, Indirect , Genes, Reporter , Hepacivirus/genetics , Humans , Isoenzymes/metabolism , Kidney/cytology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Luciferases/metabolism , Membrane Proteins/analysis , Plasmids , Receptors, Virus/analysis , Receptors, Virus/physiology , Scavenger Receptors, Class B/analysis , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...