Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Mol Oncol ; 6(1): 33-47, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22177802

ABSTRACT

The uPA/uPAR system is known to play a critical role in angiogenesis of glioblastoma. Previously, we have shown that shRNA against uPA and uPAR attenuates angiogenesis by blocking nuclear translocation of angiogenin, inhibition of angiopoietin/Tie2 signaling, and regulating several other pro-angiogenic, angiostatic and anti-angiogenic molecules. Further analysis revealed that GM-CSF, a pleiotropic cytokine, was significantly inhibited in U87MG and 4910 co-cultures with endothelial cells transfected with shRNA against uPA and uPAR. The role of the uPA/uPAR system in this process is not completely understood. Analysis of tumor conditioned medium of U87MG, 4910 and HMECs transfected with shRNA against uPA or uPAR alone or in combination (pU2) revealed inhibition of GM-CSF-enhanced secretion of SVEGFR1 as shown by Western blotting and ELISA. Moreover, phosphorylation of JAK2 and STAT5, the downstream effectors of GM-CSF signaling, was also inhibited in all three cell lines. Phosphorylation at Tyr 166 position of the GM-CSFRß subunit, the signal activating subunit of the GM-CSF receptor, was inhibited in HMEC, U87MG and 4910 cells. Further analysis revealed that shRNA against uPA and/or uPAR increased secretion of TIMP-1, which is known to enhance SVEGFR1 secretion in endothelial cells. Moreover, addition of purified uPA (with and without GM-CSF) activated JAK2/STAT5 signaling in HMEC. Exogenous addition of SVEGFR1 to pU2 tumor conditioned medium enhanced inhibition of VEGF-induced endothelial capillary tube formation as assessed by an in vitro angiogenesis assay. To determine the significance of these events in vivo, nude mice with pre-established tumors treated with shRNA against uPA and/or uPAR showed decreased levels of GM-CSF and increased levels of SVEGFR1 and TIMP-1 when compared with controls. Enhanced secretion of SVEGFR1 by puPA, puPAR and pU2 in endothelial and GBM cells was mediated indirectly by MMP-7 and augmented by ectodomain shedding of VEGFr1 by tyrosine phosphorylation at the 1213 position. Taken together, these results suggest that the uPA/uPAR system could prove beneficial as an indirect target for inhibition of angiogenesis in glioblastoma.


Subject(s)
Glioblastoma/enzymology , Neovascularization, Physiologic , RNA, Small Interfering/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Endothelial Cells/enzymology , Enzyme-Linked Immunosorbent Assay , Glioblastoma/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Janus Kinase 2/metabolism , Matrix Metalloproteinase 7/metabolism , Mice , Microvessels/cytology , Models, Biological , Phosphorylation , Phosphotyrosine/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , STAT5 Transcription Factor/metabolism , Solubility
2.
Mol Cancer ; 10: 130, 2011 Oct 17.
Article in English | MEDLINE | ID: mdl-22004682

ABSTRACT

BACKGROUND: uPA/uPAR is a multifunctional system that is over expressed in many cancers and plays a critical role in glioblastoma (GBM) invasion. Previous studies from our lab have also shown that uPA/uPAR down regulation inhibits cancer cell invasion in SNB 19 GBM cells. METHODS: As Notch 1 is known to be over expressed and promotes invasion in glioblastoma, we therefore tested our hypothesis of whether down regulation of uPA/uPAR, singly or in tandem, attenuates GBM invasion via Notch 1 receptor. Targeted down regulation of uPA/uPAR, either singly or simultaneously, inhibited the anchorage independent growth of U251MG and GBM xenograft cell lines 4910 and 5310 as assessed by soft agar colony formation assay. Expression of all four Notch receptors was confirmed in GBM tissue array analysis by immunohistochemistry. RESULTS: Down regulation of uPA/uPAR, either singly or simultaneously, in U251 MG and tumor xenografts inhibited the cleavage of the Notch receptor between the Gly 1743 and Val 1744 positions, thereby suggesting inhibition of activated cytosolic fragment-related Notch gene transcription. Morphological analysis confirmed inhibition of NICD when U251 MG cells were treated with puPA, puPAR or pU2. uPA/uPAR down regulation inhibited Notch 1 mRNA in all three examined cell lines. uPA/uPAR shRNA down regulated nuclear activation of NF-κB subunits and phosphorylation of AKT/mTOR pathway in U251 MG and GBM xenografts. puPA down regulated NICD and HES induced phosphorylation of AKT/ERK and NF-κB. Down regulation of Notch 1 using siRNA inhibited uPA activity as shown by fibrinogen zymography. It also decreased uPA expression levels as shown by western blotting. Exogenous addition of uPA activated Notch 1 in uPAR antisense U251 MG cells and also in uPAR antisense cells transfected with siRNA against Delta and Jagged. The Notch 1 receptor co-localized with LAMP-1, a marker for lysosomes in uPA, uPAR and U2, down regulated U251 MG cells which probably indicates inhibition of Notch 1 receptor trafficking in GBM cells. Notch 1 expression was significantly inhibited in puPA- and pU2-treated pre-established intracranial tumors in mice. CONCLUSIONS: Overall our results show that down regulation of uPA/uPAR, either singly or simultaneously, could be an effective approach to attenuate Notch 1 receptor cleavage, signaling and endosomal trafficking in U251MG cells and xenografts, and ultimately inhibiting GBM invasion.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Protein Transport , Receptor, Notch1/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , Brain Neoplasms/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Nucleus/metabolism , Gene Expression , Gene Knockdown Techniques , Glioblastoma/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Protein Structure, Tertiary , RNA Interference , Receptors, Urokinase Plasminogen Activator/genetics , Serrate-Jagged Proteins , Signal Transduction , Tissue Array Analysis , Transcriptional Activation , Urokinase-Type Plasminogen Activator/genetics
3.
Int J Oncol ; 37(6): 1483-93, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21042717

ABSTRACT

Oxidative stress has emerged as an important pathogenic factor in the development of breast cancer. Cholesterol-rich membrane rafts or lipid rafts (LRs) are reported to play an important role in oxidative stress-induced signal transduction. NADPH oxidase-dependent reactive oxygen species (ROS) production is implicated in oxidative stress in human mammary epithelial cells. In the present study, we determined the expression and regulation of membrane-bound subunits by LRs in human breast cancer cells. We report that basal levels of gp91phox and p22phox are expressed in breast cancer cells. We demonstrate for the first time that disruption of LRs resulted in the downregulation of NADPH oxidase subunits in breast cancer cells. Cholesterol depletion by 10 mM methyl-ß-cyclodextrin (MßCD) translocated both gp91phox and p22phox out of LRs. Moreover, lipid raft disruption decreased NADPH oxidase activity (21.1 ± 0.5% in MCF-7 and 28.9 ± 1.0 in BT-549 cells), which was reversed by cholesterol repletion (95%). Therefore, the results suggest that the integrity of LRs plays an important role in the regulation of NADPH oxidase activity in breast cancer cells.


Subject(s)
Breast Neoplasms/genetics , Carcinoma/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Microdomains/physiology , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Breast Neoplasms/metabolism , Carcinoma/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , NADPH Oxidase 2 , beta-Cyclodextrins/pharmacology
4.
BMC Cancer ; 10: 647, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21106094

ABSTRACT

BACKGROUND: uPAR and MMP-9, which play critical roles in tumor cell invasion, migration and angiogenesis, have been shown to be associated with lipid rafts. METHODS: To investigate whether cholesterol could regulate uPAR and MMP-9 in breast carcinoma, we used MßCD (methyl beta cyclodextrin, which extracts cholesterol from lipid rafts) to disrupt lipid rafts and studied its effect on breast cancer cell migration, invasion, angiogenesis and signaling. RESULTS: Morphological evidence showed the association of uPAR with lipid rafts in breast carcinoma cells. MßCD treatment significantly reduced the colocalization of uPAR and MMP-9 with lipid raft markers and also significantly reduced uPAR and MMP-9 at both the protein and mRNA levels. Spheroid migration and invasion assays showed inhibition of breast carcinoma cell migration and invasion after MßCD treatment. In vitro angiogenesis studies showed a significant decrease in the angiogenic potential of cells pretreated with MßCD. MßCD treatment significantly reduced the levels of MMP-9 and uPAR in raft fractions of MDA-MB-231 and ZR 751 cells. Phosphorylated forms of Src, FAK, Cav, Akt and ERK were significantly inhibited upon MßCD treatment. Increased levels of soluble uPAR were observed upon MßCD treatment. Cholesterol supplementation restored uPAR expression to basal levels in breast carcinoma cell lines. Increased colocalization of uPAR with the lysosomal marker LAMP1 was observed in MßCD-treated cells when compared with untreated cells. CONCLUSION: Taken together, our results suggest that cholesterol levels in lipid rafts are critical for the migration, invasion, and angiogenesis of breast carcinoma cells and could be a critical regulatory factor in these cancer cell processes mediated by uPAR and MMP-9.


Subject(s)
Breast Neoplasms/enzymology , Cell Movement , Endothelial Cells/enzymology , Matrix Metalloproteinase 9/metabolism , Membrane Microdomains/enzymology , Neovascularization, Pathologic/enzymology , Receptors, Urokinase Plasminogen Activator/metabolism , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Caveolins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cholesterol/deficiency , Culture Media, Conditioned/metabolism , Dose-Response Relationship, Drug , Endothelial Cells/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation , Humans , Matrix Metalloproteinase 9/genetics , Membrane Microdomains/drug effects , Neoplasm Invasiveness , Neovascularization, Pathologic/pathology , Nystatin/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Receptors, Urokinase Plasminogen Activator/genetics , Time Factors , beta-Cyclodextrins/pharmacology , src-Family Kinases/metabolism
5.
PLoS One ; 5(8): e12458, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20805979

ABSTRACT

BACKGROUND: In our earlier reports, we showed that downregulation of uPA and uPAR inhibited glioma tumor angiogenesis in SNB19 cells, and intraperitoneal injection of a hairpin shRNA expressing plasmid targeting uPA and uPAR inhibited angiogenesis in nude mice. The exact mechanism by which inhibition of angiogenesis takes place is not clearly understood. METHODOLOGY/PRINCIPAL FINDINGS: In the present study, we have attempted to investigate the mechanism by which uPA/uPAR downregulation by shRNA inhibits angiogenesis in endothelial and glioblastoma cell lines. uPA/uPAR downregulation by shRNA in U87 MG and U87 SPARC co-cultures with endothelial cells inhibited angiogenesis as assessed by in vitro angiogenesis assay and in vivo dorsal skin-fold chamber model in nude mice. Protein antibody array analysis of co-cultures of U87 and U87 SPARC cells with endothelial cells treated with pU2 (shRNA against uPA and uPAR) showed decreased angiogenin secretion and angiopoietin-1 as well as several other pro-angiogenic molecules. Therefore, we investigated the role of angiogenin and found that nuclear translocation, ribonucleolytic and 45S rRNA synthesis, which are all critical for angiogenic function of angiogenin, were significantly inhibited in endothelial cells transfected with uPA, uPAR and uPA/uPAR when compared with controls. Moreover, uPA and uPAR downregulation significantly inhibited the phosphorylation of Tie-2 receptor and also down regulated FKHR activation in the nucleus of endothelial cells via the GRB2/AKT/BAD pathway. Treatment of endothelial cells with ruPA increased angiogenin secretion and angiogenin expression as determined by ELISA and western blotting in a dose-dependent manner. The amino terminal fragment of uPA down regulated ruPA-induced angiogenin in endothelial cells, thereby suggesting that uPA plays a critical role in positively regulating angiogenin in glioblastoma cells. CONCLUSIONS/SIGNIFICANCE: Taken together, our results suggest that uPA/uPAR downregulation suppresses angiogenesis in endothelial cells induced by glioblastoma cell lines partially by downregulation of angiogenin and by inhibition of the angiopoietin-1/AKT/FKHR pathway.


Subject(s)
Endothelial Cells/metabolism , Glioblastoma/pathology , Receptors, Urokinase Plasminogen Activator/deficiency , Receptors, Urokinase Plasminogen Activator/genetics , Ribonuclease, Pancreatic/metabolism , Urokinase-Type Plasminogen Activator/deficiency , Urokinase-Type Plasminogen Activator/genetics , Activating Transcription Factors/metabolism , Active Transport, Cell Nucleus/genetics , Angiopoietin-1/metabolism , Angiopoietin-2/metabolism , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Coculture Techniques , Dose-Response Relationship, Drug , Down-Regulation/genetics , Endothelial Cells/drug effects , Gene Knockout Techniques , Glioblastoma/blood supply , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Phosphorylation/genetics , RNA, Ribosomal/biosynthesis , RNA, Small Interfering/genetics , Receptor, TIE-2/metabolism , Ribonuclease, Pancreatic/pharmacology , Signal Transduction/genetics , Transcription, Genetic/genetics , Urokinase-Type Plasminogen Activator/metabolism , Urokinase-Type Plasminogen Activator/pharmacology
6.
J Virol ; 83(10): 4895-911, 2009 May.
Article in English | MEDLINE | ID: mdl-19279100

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) utilizes clathrin-mediated endocytosis for its infectious entry into human foreskin fibroblast (HFF) cells (S. M. Akula, P. P. Naranatt, N.-S. Walia, F.-Z. Wang, B. Fegley, and B. Chandran, J. Virol. 77:7978-7990, 2003). Here, we characterized KSHV entry into primary human microvascular dermal endothelial (HMVEC-d) and human umbilical vein endothelial (HUVEC) cells. Similar to the results for HMVEC-d cells, KSHV infection of HUVEC cells also resulted in an initial high level and subsequent decline in the expression of the lytic switch gene, ORF50, while latent gene expression persisted. Internalized virus particles enclosed in irregular vesicles were observed by electron microscopy of infected HMVEC-d cells. At an early time of infection, colocalization of KSHV capsid with envelope was observed by immunofluorescence analysis, thus demonstrating endocytosis of intact enveloped virus particles. Chlorpromazine, an inhibitor of clathrin-mediated endocytosis, and filipin (C(35)H(58)O(11)), a caveolar endocytosis inhibitor, did not have any effect on KSHV binding, entry (DNA internalization), or gene expression in HMVEC-d and HUVEC cells. In contrast to the results for HFF cells, virus entry and gene expression in both types of endothelial cells were significantly blocked by macropinocytosis inhibitors (EIPA [5-N-ethyl-N-isoproamiloride] and rottlerin [C(30)H(28)O(8)]) and by cytochalasin D, which affects actin polymerization. Inhibition of lipid raft blocked viral gene expression in HMVEC-d cells but not in HUVEC or HFF cells. In HMVEC-d and HUVEC cells, KSHV induced the actin polymerization and formation of lamellipodial extensions that are essential for macropinocytosis. Inhibition of macropinocytosis resulted in the distribution of viral capsids at the HMVEC-d cell periphery, and capsids did not associate with microtubules involved in the nuclear delivery of viral DNA. Internalized KSHV in HMVEC-d and HUVEC cells colocalized with the macropinocytosis marker dextran and not with the clathrin pathway marker transferrin or with caveolin. Dynasore, an inhibitor of dynamin, did not block viral entry into endothelial cells but did inhibit entry into HFF cells. KSHV was not associated with the early endosome marker EEA-1 in HMVEC-d cells, but rather with the late endosome marker LAMP1, as well as with Rab34 GTPase that is known to regulate macropinocytosis. Silencing Rab34 with small interfering RNA dramatically inhibited KSHV gene expression. Bafilomycin-mediated disruption of endosomal acidification inhibited viral gene expression. Taken together, these findings suggest that KSHV utilizes the actin polymerization-dependent, dynamin-independent macropinocytic pathway that involves a Rab34 GTPase-dependent late endosome and low-pH environment for its infectious entry into HMVEC-d and HUVEC cells. These studies also demonstrate that KSHV utilizes different modes of endocytic entry in fibroblast and endothelial cells.


Subject(s)
Actins/metabolism , Herpesvirus 8, Human/physiology , Pinocytosis/drug effects , Virus Internalization/drug effects , Acetophenones/pharmacology , Amiloride/analogs & derivatives , Amiloride/pharmacology , Benzopyrans/pharmacology , Cells, Cultured , Chlorpromazine/pharmacology , Clathrin/drug effects , Clathrin/metabolism , Cytochalasin D/pharmacology , DNA, Viral/metabolism , Endothelial Cells/ultrastructure , Endothelial Cells/virology , Fibroblasts/ultrastructure , Fibroblasts/virology , Filipin/pharmacology , Gene Expression Regulation, Viral , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/metabolism , Humans , Hydrogen-Ion Concentration , Immediate-Early Proteins/metabolism , Trans-Activators/metabolism , Umbilical Veins/cytology , Umbilical Veins/virology , rab GTP-Binding Proteins/metabolism
7.
J Virol ; 82(24): 12126-44, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18829766

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) interacts with cell surface heparan sulfate (HS) and alpha3beta1 integrin during the early stages of infection of human dermal microvascular endothelial cells (HMVEC-d) and human foreskin fibroblasts (HFF), and these interactions are followed by virus entry overlapping with the induction of preexisting host cell signal pathways. KSHV also utilizes the amino acid transporter protein xCT for infection of adherent cells, and the xCT molecule is part of the cell surface heterodimeric membrane glycoprotein CD98 (4F2 antigen) complex known to interact with alpha3beta1 and alphaVbeta3 integrins. KSHV gB mediates adhesion of HMVEC-d, CV-1, and HT-1080 cells and HFF via its RGD sequence. Anti-alphaV and -beta1 integrin antibodies inhibited the cell adhesion mediated by KSHV-gB. Variable levels of neutralization of HMVEC-d and HFF infection were observed with antibodies against alphaVbeta3 and alphaVbeta5 integrins. Similarly, variable levels of inhibition of virus entry into adherent HMVEC-d, 293 and Vero cells, and HFF was observed by preincubating virus with soluble alpha3beta1, alphaVbeta3, and alphaVbeta5 integrins, and cumulative inhibition was observed with a combination of integrins. We were unable to infect HT1080 cells. Virus binding and DNA internalization studies suggest that alphaVbeta3 and alphaVbeta5 integrins also play roles in KSHV entry. We observed time-dependent temporal KSHV interactions with HMVEC-d integrins and CD98/xCT with three different patterns of association and dissociation. Integrin alphaVbeta5 interaction with CD98/xCT predominantly occurred by 1 min postinfection (p.i.) and dissociated at 10 min p.i., whereas alpha3beta1-CD98/xCT interaction was maximal at 10 min p.i. and dissociated at 30 min p.i., and alphaVbeta3-CD98/xCT interaction was maximal at 10 min p.i. and remained at the observed 30 min p.i. Fluorescence microscopy also showed a similar time-dependent interaction of alphaVbeta5-CD98. Confocal-microscopy studies confirmed the association of CD98/xCT with alpha3beta1 and KSHV. Preincubation of KSHV with soluble heparin and alpha3beta1 significantly inhibited this association, suggesting that the first contact with HS and integrin is an essential element in subsequent CD98-xCT interactions. Anti-CD98 and xCT antibodies did not block virus binding and entry and nuclear delivery of viral DNA; however, viral-gene expression was significantly inhibited, suggesting that CD98-xCT play roles in the post-entry stage of infection, possibly in mediating signal cascades essential for viral-gene expression. Together, these studies suggest that KSHV interacts with functionally related integrins (alphaVbeta3, alpha3beta1, and alphaVbeta5) and CD98/xCT molecules in a temporal fashion to form a multimolecular complex during the early stages of endothelial cell infection, probably mediating multiple roles in entry, signal transduction, and viral-gene expression.


Subject(s)
Endothelial Cells/metabolism , Fusion Regulatory Protein-1/metabolism , Herpesvirus 8, Human/metabolism , Integrins/metabolism , Microvessels/metabolism , Skin/metabolism , Biological Transport , Cell Adhesion , Cell Line , DNA, Viral/metabolism , Endothelial Cells/cytology , Fusion Regulatory Protein-1/immunology , Gene Expression Regulation, Viral , Herpesvirus 8, Human/genetics , Humans , Integrin alpha3beta1/immunology , Integrin alpha3beta1/metabolism , Integrin alphaVbeta3/immunology , Integrin alphaVbeta3/metabolism , Integrins/immunology , Ligands , Microvessels/cytology , Protein Binding , Receptors, Vitronectin/immunology , Receptors, Vitronectin/metabolism , Skin/cytology , Solubility , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Internalization
8.
J Virol ; 82(4): 1759-76, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18057235

ABSTRACT

Kaposi's sarcoma (KS), a vascular tumor associated with human immunodeficiency virus type 1 infection, is characterized by spindle-shaped endothelial cells, inflammatory cells, cytokines, growth and angiogenic factors, and angiogenesis. KS spindle cells are believed to be of the lymphatic endothelial cell (LEC) type. Kaposi's sarcoma-associated herpesvirus (KSHV, or human herpesvirus 8) is etiologically linked to KS, and in vitro KSHV infection of primary human dermal microvascular endothelial cells (HMVEC-d) is characterized by the induction of preexisting host signal cascades, sustained expression of latency-associated genes, transient expression of a limited number of lytic genes, sustained induction of NF-kappaB and several cytokines, and growth and angiogenic factors. KSHV induced robust vascular endothelial growth factor A (VEGF-A) and VEGF-C gene expression as early as 30 min postinfection (p.i.) in serum-starved HMVEC-d, which was sustained throughout the observation period of 72 h p.i. Significant amounts of VEGF-A and -C were also detected in the culture supernatant of infected cells. VEGF-A and -C were also induced by UV-inactivated KSHV and envelope glycoprotein gpK8.1A, thus suggesting a role for virus entry stages in the early induction of VEGF and requirement of KSHV viral gene expression for sustained induction. Exogenous addition of VEGF-A and -C increased KSHV DNA entry into target cells and moderately increased latent ORF73 and lytic ORF50 promoter activation and gene expression. KSHV infection also induced the expression of lymphatic markers Prox-1 and podoplanin as early as 8 h p.i., and a paracrine effect was seen in the neighboring uninfected cells. Similar observations were also made in the pure blood endothelial cell (BEC)-TIME cells, thus suggesting that commitment to the LEC phenotype is induced early during KSHV infection of blood endothelial cells. Treatment with VEGF-C alone also induced Prox-1 expression in the BEC-TIME cells. Collectively, these studies show that the in vitro microenvironments of KSHV-infected endothelial cells are enriched, with VEGF-A and -C molecules playing key roles in KSHV biology, such as increased infection and gene expression, as well as in angiogenesis and lymphangiogenesis, thus recapitulating the microenvironment of early KS lesions.


Subject(s)
Endothelium, Vascular/virology , Herpesvirus 8, Human/growth & development , Skin/virology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor C/metabolism , Capillaries , Cell Line , Endothelium, Vascular/metabolism , Gene Expression/drug effects , Gene Expression Regulation, Viral , Herpesvirus 8, Human/drug effects , Herpesvirus 8, Human/genetics , Homeodomain Proteins/metabolism , Humans , Membrane Glycoproteins/metabolism , Promoter Regions, Genetic/drug effects , Skin/blood supply , Skin/metabolism , Tumor Suppressor Proteins/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/pharmacology
9.
J Virol ; 81(15): 7941-59, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17507466

ABSTRACT

Early during de novo infection of human microvascular dermal endothelial (HMVEC-d) cells, Kaposi's sarcoma-associated herpesvirus (KSHV) (human herpesvirus 8 [HHV-8]) induces the host cell's preexisting FAK, Src, phosphatidylinositol 3-kinase (PI3-K), Rho-GTPases, Diaphanous-2 (Dia-2), Ezrin, protein kinase C-zeta, extracellular signal-regulated kinase 1/2 (ERK1/2), and NF-kappaB signal pathways that are critical for virus entry, nuclear delivery of viral DNA, and initiation of viral gene expression. Since several of these signal molecules are known to be associated with lipid raft (LR) domains, we investigated the role of LR during KSHV infection of HMVEC-d cells. Pretreatment of cells with LR-disrupting agents methyl beta-cyclo dextrin (MbetaCD) or nystatin significantly inhibited the expression of viral latent (ORF73) and lytic (ORF50) genes. LR disruption did not affect KSHV binding but increased viral DNA internalization. In contrast, association of internalized viral capsids with microtubules (MTs) and the quantity of infected nucleus-associated viral DNA were significantly reduced. Disorganized and disrupted MTs and thick rounded plasma membranes were observed in MbetaCD-treated cells. LR disruption did not affect KSHV-induced FAK and ERK1/2 phosphorylation; in contrast, it increased the phosphorylation of Src, significantly reduced the KSHV-induced PI3-K and RhoA-GTPase and NF-kappaB activation, and reduced the colocalizations of PI3-K and RhoA-GTPase with LRs. Biochemical characterization demonstrated the association of activated PI3-K with LR fractions which was inhibited by MbetaCD treatment. RhoA-GTPase activation was inhibited by PI3-K inhibitors, demonstrating that PI3-K is upstream to RhoA-GTPase. In addition, colocalization of Dia-2, a RhoA-GTPase activated molecule involved in MT activation, with LR was reduced. KSHV-RhoA-GTPase mediated acetylation and aggregation of MTs were also reduced. Taken together, these studies suggest that LRs of endothelial cells play critical roles in KSHV infection and gene expression, probably due to their roles in modulating KSHV-induced PI3-K, RhoA-GTPase, and Dia-2 molecules essential for postbinding and entry stages of infection such as modulation of microtubular dynamics, movement of virus in the cytoplasm, and nuclear delivery of viral DNA.


Subject(s)
Active Transport, Cell Nucleus/physiology , DNA, Viral/metabolism , Herpesvirus 8, Human/metabolism , Membrane Microdomains/metabolism , Microtubules/metabolism , Phosphatidylinositol 3-Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Butadienes/metabolism , Capsid/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Enzyme Activation , Enzyme Induction , Enzyme Inhibitors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Kinase 2/metabolism , Formins , Gene Expression Regulation, Viral , Herpesviridae Infections , Herpesvirus 8, Human/genetics , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Nitriles/metabolism , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction/physiology , rhoA GTP-Binding Protein/genetics , src-Family Kinases/metabolism
10.
J Virol ; 81(8): 3949-68, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17287275

ABSTRACT

In vitro Kaposi's sarcoma-associated herpesvirus (KSHV) infection of primary human dermal microvascular endothelial (HMVEC-d) cells and human foreskin fibroblast (HFF) cells is characterized by the induction of preexisting host signal cascades, sustained expression of latency-associated genes, transient expression of a limited number of lytic genes, and induction of several cytokines, growth factors, and angiogenic factors. Since NF-kappaB is a key molecule involved in the regulation of several of these factors, here, we examined NF-kappaB induction during de novo infection of HMVEC-d and HFF cells. Activation of NF-kappaB was observed as early as 5 to 15 min postinfection by KSHV, and translocation of p65-NF-kappaB into nuclei was detected by immunofluorescence assay, electrophoretic mobility shift assay, and p65 enzyme-linked immunosorbent assay. IkappaB phosphorylation inhibitor (Bay11-7082) reduced this activation significantly. A sustained moderate level of NF-kappaB induction was seen during the observed 72 h of in vitro KSHV latency. In contrast, high levels of ERK1/2 activation at earlier time points and a moderate level of activation at later times were observed. p38 mitogen-activated protein kinase was activated only at later time points, and AKT was activated in a cyclic manner. Studies with UV-inactivated KSHV suggested a role for virus entry stages in NF-kappaB induction and a requirement for KSHV viral gene expression in sustained induction. Inhibition of NF-kappaB did not affect target cell entry by KSHV but significantly reduced the expression of viral latent open reading frame 73 and lytic genes. KSHV infection induced the activation of several host transcription factors, including AP-1 family members, as well as several cytokines, growth factors, and angiogenic factors, which were significantly affected by NF-kappaB inhibition. These results suggest that during de novo infection, KSHV induces sustained levels of NF-kappaB to regulate viral and host cell genes and thus possibly regulates the establishment of latent infection.


Subject(s)
Endothelial Cells/virology , Gene Expression Regulation , Herpesvirus 8, Human/physiology , NF-kappa B/metabolism , Active Transport, Cell Nucleus , Angiogenesis Inducing Agents/metabolism , Cell Line , Cell Nucleus/metabolism , Cells, Cultured , Cytokines/biosynthesis , Endothelial Cells/metabolism , Fibroblasts/virology , Gene Expression , Genes, Viral , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 3/metabolism , Models, Biological , NF-kappa B/antagonists & inhibitors , Nitriles/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Sulfones/pharmacology , Transcription Factors/biosynthesis , Virus Internalization , p38 Mitogen-Activated Protein Kinases/metabolism
11.
J Virol ; 80(23): 11432-46, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17005646

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) (human herpesvirus 8) binds to adherent target cell surface heparan sulfate molecules via its envelope glycoproteins gB and gpK8.1A, to integrins via gB, to the transporter CD98/xCT complex, and possibly to another molecule(s). This is followed by virus entry overlapping with the induction of preexisting host cell signal pathways, such as focal adhesion kinase, Src, phosphatidylinositol 3-kinase (PI3-K), Rho-GTPases, protein kinase C-zeta, and extracellular signal-regulated kinase 1/2. Here, using hemagglutinin-tagged plasmids expressing wild-type, dominant-positive, and dominant-negative forms of RhoA in HEK (human embryonic kidney) 293 cells, we investigated the role of RhoA-GTPase in virus entry. The dominant-negative form of RhoA GTPase and treatment of target cells with Clostridium difficile toxin B (CdTxB), a specific inactivator of Rho-GTPases, significantly blocked KSHV entry. KSHV infection induced closely similar levels of FAK and PI3-K in all three cell types. In contrast, very strong Src activation was observed in KSHV-infected dominant-positive RhoA cells compared to wild-type cells, and only moderate Src activation was seen in dominant-negative cells. Inhibition of Src activation by CdTxB and reduction of RhoA activation by Src inhibitors suggest that KSHV-induced Src is involved in RhoA activation, which in turn is involved in a feedback-sustained activation of Src. Since the decreased entry in RhoA dominant-negative cells may be due to inefficient signaling downstream of RhoA, we examined the induction of RhoA-activated Dia-2, which is also known to induce Src. Dia-2 coimmunoprecipitated with activated Src, which was inhibited by Src inhibitors, in the infected cells. Together with the reduced virus entry in RhoA dominant-negative cells, these results suggest that activated RhoA-dependent Dia-2 probably functions as a link between RhoA and Src in KSHV-infected cells, mediating the sustained Src activation, and that KSHV-induced Src and RhoA play roles in facilitating entry into adherent target cells.


Subject(s)
Cell Adhesion/physiology , Herpesvirus 8, Human/physiology , rho GTP-Binding Proteins/physiology , src-Family Kinases/metabolism , Cell Line , Gene Expression Regulation, Viral , Herpesvirus 8, Human/genetics , Humans , Signal Transduction
12.
J Virol ; 80(13): 6534-52, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16775340

ABSTRACT

Infection of human dermal microvascular endothelial (HMVEC-d) cells and human foreskin fibroblast (HFF) cells in vitro by Kaposi's sarcoma-associated herpesvirus (KSHV) provides an excellent in vitro model system to study viral latency. KSHV infection is characterized by the induction of preexisting host signal cascades; sustained expression of the latency-associated open reading frame 73 (ORF73) (LANA-1), ORF72, and K13 genes; transient expression of a limited number of lytic genes, including the lytic cycle switch ORF50 (replication and transcription activator) gene; and reprogramming of host transcriptional machinery regulating a variety of cellular processes, including several proinflammatory responses. The cyclooxygenase 2 (COX-2) gene was one of the host cell genes that was highly up-regulated at 2 and 4 h postinfection (p.i.) of HMVEC-d and HFF cells (P. P. Naranatt, H. H. Krishnan, S. R. Svojanovsky, C. Bloomer, S. Mathur, and B. Chandran, Cancer Res. 64:72-84, 2004). Since COX-2 is an important mediator of inflammatory and angiogenic responses, here, using real-time PCR, Western blot, and immunofluorescence assays, we characterized the COX-2 stimulation and its role in KSHV infection. KSHV induced a robust COX-2 expression, which reached a maximum at 2 h p.i. in HMVEC-d cells and at 8 h p.i. in HFF cells, and significantly higher levels were continuously detected for up to 72 h p.i. Constitutive COX-1 protein levels were not modulated by KSHV infection. Moderate levels of COX-2 were also induced by UV-irradiated KSHV and by envelope glycoproteins gB and gpK8.1A; however, viral gene expression appears to be essential for the increased COX-2 induction. High levels of prostaglandin E(2) (PGE(2)), a COX-2 product, were released in the culture supernatant medium of infected cells. PGE(2) synthase, catalyzing the biosynthesis of PGE(2), also increased upon infection and inhibition of COX-2 by NS-398, and indomethacin drastically reduced the levels of PGE(2) and PGE(2) synthase. COX-2 inhibition did not affect KSHV binding, internalization of virus, or the trafficking to the infected cell nuclei. However, latent ORF73 gene expression and ORF73 promoter activity were significantly reduced by COX-2 inhibitors, and this inhibition was relieved by exogenous supplementation with PGE(2). In contrast, lytic ORF50 gene expression and ORF50 promoter activity were unaffected. These studies demonstrate that COX-2 and PGE(2) play roles in facilitating latent viral gene expression and the establishment and maintenance of latency and suggest that KSHV has evolved to utilize the inflammatory responses induced during infection of endothelial cells for the maintenance of viral latent gene expression.


Subject(s)
Cyclooxygenase 2/biosynthesis , Gene Expression Regulation, Viral/physiology , Herpesviridae Infections/enzymology , Herpesvirus 8, Human/physiology , Membrane Proteins/biosynthesis , Viral Envelope Proteins/metabolism , Virus Latency/physiology , Cell Line , Cyclooxygenase 1/biosynthesis , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/biosynthesis , Endothelial Cells/enzymology , Endothelial Cells/virology , Fibroblasts/enzymology , Fibroblasts/virology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Enzymologic/radiation effects , Gene Expression Regulation, Viral/drug effects , Gene Expression Regulation, Viral/radiation effects , Humans , Indomethacin/pharmacology , Inflammation/enzymology , Inflammation/virology , Nitrobenzenes/pharmacology , Open Reading Frames/physiology , Promoter Regions, Genetic/physiology , Sulfonamides/pharmacology , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , Transcription, Genetic/radiation effects , Ultraviolet Rays , Up-Regulation/drug effects , Up-Regulation/physiology , Up-Regulation/radiation effects , Virus Latency/drug effects , Virus Latency/radiation effects , Virus Replication/drug effects , Virus Replication/physiology , Virus Replication/radiation effects
13.
Am J Ophthalmol ; 138(2): 313-4, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15289153

ABSTRACT

PURPOSE: To report association of herpes simplex virus-type 1 (HSV-1) in four cases of congenital cataract. DESIGN: Prospective interventional case series. METHODS: Four infants younger than 12 months, presenting with unilateral or bilateral congenital cataract, were included. The cases were clinically evaluated by the pediatric ophthalmologist. The lens aspirates collected at the time of cataract surgery were processed for HSV-1 culture in rabbit corneal epithelial (SIRC) cell line and for HSV-1 DNA by polymerase chain (PCR). The sera of the children and the mother were tested for HSV-1 immunoglobulin (Ig) M and IgG by enzyme linked immunosorbent assay (ELISA). RESULTS: HSV-1 was isolated in tube cultures in three of four lens aspirates, and all four lens aspirates were positive for HSV-1 DNA by PCR. Serum HSV-1 IgM was positive in all babies and in three cases HSV-1 IgM was positive in the mother's serum. CONCLUSION: Based on a computerized literature search, we believe this may be the first report of HSV-1 associated congenital cataract.


Subject(s)
Cataract/congenital , Cataract/virology , Eye Infections, Viral/virology , Herpes Simplex/virology , Herpesvirus 1, Human/isolation & purification , Antibodies, Viral/blood , DNA, Viral/analysis , Enzyme-Linked Immunosorbent Assay , Female , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/immunology , Humans , Immunoglobulin G/analysis , Immunoglobulin M/analysis , Infant , Male , Polymerase Chain Reaction , Prospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL