Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Cell Mol Med ; 13(8B): 1920-1932, 2009 Aug.
Article in English | MEDLINE | ID: mdl-18752634

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) has both latent and lytic phases of replication. The molecular switch that triggers a reactivation is still unclear. Cells from the S phase of the cell cycle provide apt conditions for an active reactivation. In order to specifically delineate the Raman spectra of cells supporting KSHV reactivation, we followed a novel approach where cells were sorted based on the state of infection (latent versus lytic) by a flow cytometer and then analysed by the Raman tweezers. The Raman bands at 785, 813, 830, 1095 and 1128 cm(-1) are specifically altered in cells supporting KSHV reactivation. These five peaks make up the Raman fingerprint of cells supporting KSHV reactivation. The physiological relevance of the changes in these peaks with respect to KSHV reactivation is discussed in the following report.


Subject(s)
Herpesvirus 8, Human/physiology , Spectrum Analysis, Raman/methods , Virus Activation , Blotting, Western , Fluorescent Antibody Technique , Humans , Reverse Transcriptase Polymerase Chain Reaction
2.
Cell Microbiol ; 10(7): 1546-58, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18363908

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) has two modes of replications: latent and lytic replications. Reactivation from latency is dictated, in part, by the cell cycle. Herein, we have attempted to delineate the importance of cell cycle in KSHV pathogenesis by exploring the expression pattern of cell-surface receptors during different phases of the cell cycle. alphaV integrin expression is augmented during S phase in fibroblasts, epithelial and KSHV-infected cells. Using a Matrigel system, we pioneer the concept that KSHV-infected primary effusion lymphoma cells can attach to extracellular matrix proteins. This attachment is mediated primarily via alphaV integrins or virally encoded gB, and occurs preferentially in cells from S phase or cells from S phase actively supporting a lytic infection respectively. Such an ability of infected B cells to attach to endothelial cells may also aid in the dissemination of infection. The keystone of this work is that for the first time, we describe the ability of KSHV-infected B cells to preferentially use cellular (alphaV) or viral (gB) receptors to specifically bind cells, depending upon the stage of the cell cycle and infection.


Subject(s)
B-Lymphocytes/virology , Cell Adhesion/physiology , Extracellular Matrix/metabolism , Herpesvirus 8, Human/metabolism , Integrin alphaV/metabolism , Viral Proteins/metabolism , Animals , Cell Cycle/physiology , Cells, Cultured , Collagen/metabolism , Drug Combinations , Endothelial Cells/cytology , Endothelial Cells/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation, Viral , Herpesvirus 8, Human/pathogenicity , Humans , Integrin alphaV/genetics , Laminin/metabolism , Proteoglycans/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Viral Proteins/genetics , Virus Latency , Virus Replication
3.
Intervirology ; 50(4): 245-53, 2007.
Article in English | MEDLINE | ID: mdl-17460413

ABSTRACT

OBJECTIVE: Angiogenesis is defined as the formation of new blood vessels. In a recently concluded study, we identified Kaposi's sarcoma-associated herpesvirus (KSHV)-infected cells derived from primary effusion lymphoma (PEL) to overexpress vascular endothelial growth factor (VEGF) that had the propensity to mediate tubule formation on a Matrigel, an indicator of angiogenesis. The objective of this study was to determine the receptor molecules that mediate the tubule formation induced by the supernatant derived from KSHV-infected PEL cells. METHODS: The identity of receptor(s) that play a role in mediating tubule formation driven by PEL supernatant was determined by the classical in vitro angiogenesis assay conducted on a Matrigel. RESULTS: RGD peptides, antibodies, and siRNA specific to beta1 integrins significantly lowered the ability of the PEL supernatants to induce tubule formation by endothelial cells. beta1 Integrins mediated tubule formation to comparable levels in endothelial cells that were incubated with supernatants derived from uninduced or TPA-induced PEL cells. Interestingly, the beta1 integrins did not seem to have a major role in cellular attachment. CONCLUSION: We report for the first time a critical role for beta1 integrins in angiogenesis supported by the supernatant from KSHV-infected PEL cells.


Subject(s)
Culture Media, Conditioned/pharmacology , Endothelial Cells/metabolism , Herpesvirus 8, Human/pathogenicity , Integrin beta1/metabolism , Neovascularization, Pathologic , B-Lymphocytes/virology , Cell Line, Transformed , Cell Transformation, Viral , Cells, Cultured , Collagen , Drug Combinations , Endothelium, Vascular/cytology , Herpesviridae Infections/virology , Humans , Laminin , Proteoglycans , Vascular Endothelial Growth Factor A/metabolism
4.
J Electron Microsc (Tokyo) ; 56(1): 27-36, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17392397

ABSTRACT

Following an infection, Kaposi's sarcoma-associated herpes virus (KSHV) exists predominantly in its latent state, with only 1-2% of infected cells undergoing lytic reactivation. We have previously demonstrated along with others a relationship between lytic reactivation and cell cycle progression (Bryan et al., 2006. J. Gen. Virol. 87: 519; McAllister et al., 2005. J. Virol. 79: 2626). Infected cells in the S phase are much more likely to undergo lytic reactivation when compared to those in G(0)/G(1) phase. Through the use of scanning electron microscopy (SEM), we analyzed changes occurring on the surface of cells undergoing KSHV reactivation. KSHV reactivation was observed predominantly in cells with smoother surface topology; a hallmark of cells derived from S phase. Interestingly, during the late stages of the reactivation process, we observed KSHV particles to egress cells through budding. Taken together, based on scanning electron microscopy and transmission electron microscopy evidences, we demonstrate for the first time the existence of a direct link between cell surface topology, cell cycle progression and KSHV reactivation.


Subject(s)
Cell Membrane/ultrastructure , Cell Membrane/virology , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Herpesvirus 8, Human/physiology , Herpesvirus 8, Human/ultrastructure , Base Sequence , Cell Cycle , Cell Line , DNA Primers/genetics , Herpesvirus 8, Human/drug effects , Herpesvirus 8, Human/genetics , Humans , Microscopy, Electron , Microscopy, Electron, Scanning , Polymerase Chain Reaction , Tetradecanoylphorbol Acetate/pharmacology , Virus Activation/drug effects , Virus Activation/physiology , Virus Assembly
5.
J Gen Virol ; 87(Pt 5): 1139-1144, 2006 May.
Article in English | MEDLINE | ID: mdl-16603514

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) causes Kaposi's sarcoma, primary effusion lymphoma and multicentric Castleman's disease. KSHV infection of cells produces both latent and lytic cycles of infection. In vivo, the virus is found predominantly in the latent state. In vitro, a lytic infection can be induced in KSHV-infected cells by treating with phorbol ester (TPA). However, the exact signalling events that lead to the reactivation of KSHV lytic infection are still elusive. Here, a role is demonstrated for B-Raf/MEK/ERK signalling in TPA-induced reactivation of KSHV latent infection. Inhibiting MEK/ERK signalling by using MEK-specific inhibitors decreased expression of the TPA-induced KSHV lytic-cycle gene ORF8. Transfection of BCBL-1 cells with B-Raf small interfering RNA inhibited TPA-induced KSHV lytic infection significantly. Additionally, overexpression of MEK1 induced a lytic cycle of KSHV infection in BCBL-1 cells. The significance of these findings in understanding the biology of KSHV-associated pathogenesis is discussed.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Herpesvirus 8, Human/physiology , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Cell Line, Tumor , Humans , Phorbol Esters/pharmacology , Signal Transduction , Virus Activation
6.
Virus Res ; 114(1-2): 172-6, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16105704

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is etiologically associated with Kaposi's sarcoma (KS), primary effusion lymphoma(PEL), multicentric Castleman disease, and other tumors. Progression of KS is dictated by an aberrant production of inflammatory cytokines and increase in KSHV infection of cells. In this study, we analyzed the effect of cigarette smoke concentrate (CSC) on KSHV infection of human foreskin fibroblasts (HFF) using real time quantitative RT-PCR. Our results demonstrated that the CSC-treated cells supported 50% lower infection of KSHV when compared to the untreated cells. Radiolabeled-binding assays indicated that CSC inhibited KSHV infection of cells at a post attachment stage of entry. Taken together, we report for the first time the ability of CSC to specifically inhibit KSHV infection of cells.


Subject(s)
Antiviral Agents/pharmacology , Herpesvirus 8, Human/pathogenicity , Nicotiana , Smoke , Cells, Cultured , Dimethyl Sulfoxide/pharmacology , Fibroblasts/virology , Herpesvirus 8, Human/drug effects , Humans , Male , Nicotiana/chemistry
7.
J Virol Methods ; 129(2): 145-51, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15992938

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV), also referred to as human herpesvirus-8 (HHV-8), is a tumor causing virus. KSHV is the cause of several disease conditions known as Kaposi's sarcoma, multicentric Castleman disease, and primary effusion lymphoma. Cell culture supernatants from KSHV infected hematopoietic cells induced angiogenic tubule formation to a significantly greater extent than uninfected hematopoietic cells. Raman spectrum profiles were generated to differentiate the uninfected from KSHV infected cells. In general, profiles from all the hematopoietic cells shared similar peaks; however, the relative abundance of specific components varied significantly between the cells. Subsequent use of the multivariate analysis of the Raman spectra revealed significant differences between the uninfected and the KSHV infected cells. Taken together, this study reports the use of Raman tweezers to distinguish and analyze the biological relevance of KSHV infected cell signaling.


Subject(s)
Herpesviridae Infections/pathology , Herpesvirus 8, Human/physiology , Spectrum Analysis, Raman , Animals , Cell Line/pathology , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/virology , Herpesviridae Infections/virology , Neovascularization, Pathologic
8.
Blood ; 105(11): 4516-22, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15705790

ABSTRACT

Kaposi sarcoma-associated herpesvirus/human herpesvirus 8 (KSHV/HHV-8) is etiologically linked to Kaposi sarcoma (KS), primary effusion lymphoma (PEL), and multicentric Castleman disease. Vascular endothelial growth factor-A (VEGF-A) is one of the essential factors required in KSHV pathogenesis, mainly due to its ability to mediate angiogenesis. In this report we analyzed the relationship between Raf and VEGF-A expression in KSHV-infected hematopoietic cells. All of the KSHV-infected cell lines (derived from PEL) expressed higher levels of B-Raf and VEGF-A when compared with uninfected cells. Inhibition of Raf to mitogen-induced extracellular kinase (MEK) to extracellular signal-related kinase (ERK) signaling, either by the use of MEK inhibitor (PD98059) or by siRNA specific to B-Raf, significantly lowered VEGF-A expression. In addition, B-Raf-induced VEGF-A expression was demonstrated to be sufficient to enhance tubule formation in endothelial cells. Interestingly, we did not observe mutation in the B-Raf gene of the KSHV-infected PEL cell lines. Taken together, we report for the first time the ability of Raf-associated signaling to play a role in the expression of VEGF-A in KSHV-infected hematopoietic cells.


Subject(s)
B-Lymphocytes/virology , Gene Expression Regulation , Herpesviridae Infections/metabolism , Herpesvirus 8, Human , Proto-Oncogene Proteins B-raf/physiology , Vascular Endothelial Growth Factor A/genetics , Cell Line , Endothelium, Vascular/pathology , Extracellular Signal-Regulated MAP Kinases , Herpesviridae Infections/pathology , Humans , Neovascularization, Pathologic , Signal Transduction
9.
J Virol ; 78(23): 13381-90, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15542692

ABSTRACT

Recombinant green fluorescent protein encoding Kaposi's sarcoma-associated herpesvirus (rKSHV.152) infection of beta-estradiol stimulated human foreskin fibroblasts (HFF) or HFF/DeltaB-Raf([FF]):ER (expressing a weaker form of B-Raf) could be enhanced to levels comparable to that of HFF/DeltaB-Raf([DD]):ER cells by pretreating cells with soluble vascular endothelial growth factor (VEGF). Conversely, VEGF expression and infection efficiency typically observed in beta-estradiol stimulated HFF/DeltaB-Raf([DD]):ER cells could be lowered significantly by treating with VEGF small interfering RNA. In addition, we observed enhancement of the KSHV infection in HFF cells transfected with human VEGF(121). These results confirm the ability of Raf-induced VEGF to augment KSHV infection of cells.


Subject(s)
Herpesvirus 8, Human/physiology , Vascular Endothelial Growth Factor A/physiology , raf Kinases/physiology , Cells, Cultured , Humans , RNA, Small Interfering/pharmacology , Vascular Endothelial Growth Factor Receptor-1/physiology , Vascular Endothelial Growth Factor Receptor-2/physiology
10.
Cancer Biol Ther ; 3(9): 876-81, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15254407

ABSTRACT

Human herpesvirus-8 (HHV-8/KSHV) is etiologically associated with Kaposi's sarcoma (KS) and other tumors. The Raf oncoprotein enhances HHV-8 infection of target cells. In addition, we have previously demonstrated that Raf induces vascular endothelial growth factor (VEGF) expression. VEGF is a growth factor that has autocrine growth activity and has been implicated in the formation of the spindle shape cell morphology characteristic of Kaposi's sarcoma (KS). The aim of this study was to test the hypothesis that VEGF enhances infection of HHV-8. Herein, we demonstrate that the soluble VEGF enhanced green florescence protein encoding (GFP)-HHV-8 (rKSHV.152) infection of human foreskin fibroblasts (HFF) and not of 293 cells. We found this to be in part, due to the fact that HFF inherently produces significantly lower concentrations of VEGF when compared to 293 cells. Treating 293 cells (but not HFF) with a VEGF receptor (VEGFR) inhibitor significantly lowered infection. Furthermore, transfecting 293 cells with VEGF specific si-RNA did not alter the binding of HHV-8 to cells; but significantly lowered VEGF expression and thus GFP-HHV-8 infection. Interestingly, lowering VEGF expression in 293 cells wtih VEGF specific si-RNA did not completely inhibit GFP-HHV-8 infection. We conclude that VEGF is not a requirement for HHV-8 infection; but VEGF plays a major role in augmenting infection at a post binding stage of entry. These findings suggest that targeting VEGF/VEGFR may prove efficacious in controlling HHV-8 associated pathogenesis.


Subject(s)
Herpesviridae Infections/physiopathology , Herpesvirus 8, Human/pathogenicity , Sarcoma, Kaposi/physiopathology , Sarcoma, Kaposi/virology , Vascular Endothelial Growth Factor A/pharmacology , Cell Culture Techniques , Fibroblasts , Humans , Kidney/cytology , Solubility
11.
Int J Oncol ; 25(2): 469-80, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15254746

ABSTRACT

The biology of acquired immune deficiency (AIDS) is yet to be completely understood partly because it is complicated by the manifestation of various viral infections and associated pathogenesis. Virus entry into target cells is a key step in the virus replication cycle which is characterized by intricate and complex interactions between virus and host cells. Analyses of virus entry are always hampered to some extent due to the inability to mimic in vivo conditions. Emphasis has been placed on understanding what the virus does during the entry process; for example the signaling it mediates during entry, or identifying the cellular receptors with which the virus interact. Often, the role of the cellular environment that is critical for the complex process of virus uptake has taken a back stage. Interestingly, most of the viruses associated with AIDS cause tumors. In a recently concluded study, we identified a role for intracellular oncogenic (Raf) signaling in human herpesvirus-8 (HHV-8/KSHV) infection of target cells. In this review we present an update on entry of various viruses commonly associated with AIDS and yet another novel way of analyzing virus entry.


Subject(s)
AIDS-Related Opportunistic Infections/enzymology , AIDS-Related Opportunistic Infections/virology , Virus Diseases/enzymology , Virus Diseases/virology , raf Kinases/physiology , Herpesviridae Infections/enzymology , Herpesviridae Infections/virology , Herpesvirus 8, Human/metabolism , Humans , Signal Transduction , raf Kinases/metabolism
12.
Oncogene ; 23(30): 5227-41, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15122343

ABSTRACT

Human herpesvirus-8 (HHV-8/KSHV) is etiologically associated with Kaposi's sarcoma (KS) and other tumors. Constitutive activation of the mitogen-activated protein kinase (MAPK) signaling pathway has been associated with a variety of tumors, including AIDS-related KS. The oncoprotein Raf is situated at a pivotal position in regulating the MAPK pathway. Hence, we analysed the effect of oncoprotein Raf on HHV-8 infectious entry into target cells. Here we report Raf expression to significantly enhance HHV-8 infection of target cells. These findings implicate a role for Raf not only in the infectious entry of HHV-8 but also in modulating KS pathogenesis.


Subject(s)
Herpesviridae Infections , Herpesvirus 8, Human/metabolism , Herpesvirus 8, Human/pathogenicity , Proto-Oncogene Proteins c-raf/metabolism , AIDS-Related Opportunistic Infections/virology , Blotting, Western , Butadienes/pharmacology , Cell Line , Enzyme Activation , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , Estradiol/pharmacology , Green Fluorescent Proteins , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins , Luminescent Proteins/metabolism , Mitogen-Activated Protein Kinase Kinases/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Nitriles/pharmacology , Receptors, Estrogen/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism , Sarcoma, Kaposi/etiology , Sarcoma, Kaposi/virology , Signal Transduction , Vascular Endothelial Growth Factor A/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...