Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Am J Reprod Immunol ; 85(3): e13352, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32969101

RESUMO

PROBLEM: Gestational membrane (GM) infection provokes inflammation and can result in preterm prelabor rupture of membranes (PPROM). The choriodecidual layer of the GM includes decidual stromal cells (DSC), cytotrophoblasts (CTB), and macrophages (Mφ). Our laboratory has previously shown that DSCs suppress Mφ TNF-α production through secreted prostaglandin E2 . We hypothesized that CTBs would also inhibit Mφ cytokine expression through secreted mediators. METHOD OF STUDY: THP.1 Mφ-like cells with an NF-κB reporter construct or human blood monocyte-derived Mφ were co-cultured with the Jeg3 CTB cell line or primary human CTBs and challenged with group B streptococcus (GBS) or Toll-like receptor (TLR) agonists. Conditioned medium generated from CTB cultures was applied to Mφ cultures before infection or treatment. Alternatively, CTBs were co-incubated with, but physically separated from, Mφ and GBS or TLR-stimulated. NF-κB was assessed via alkaline phosphatase assay, and proinflammatory mediators were assessed by qRT-PCR and ELISA. RESULTS: CTBs suppressed GBS- or TLR-stimulated Mφ NF-κB activity, and TNF-α and MMP9 production. Direct physical contact between CTBs and Mφ was required for full immunosuppression. Immunosuppression could be overcome by increasing the ratio of Mφ to CTB. CONCLUSIONS: CTBs limit Mφ NF-κB activation and production of TNF-α and MMP9 through an as-yet unknown, cell-to-cell contact-mediated mechanism. This suppression is distinct from the PGE2 -mediated Mφ TNF-α suppression by DSC, suggesting that DSCs and CTBs regulate Mφ inflammation through distinct mechanisms. How Mφ integrates these signals in an intact GM will be paramount to determining causes and prevention of PPROM.


Assuntos
Âmnio/patologia , Decídua/patologia , Ruptura Prematura de Membranas Fetais/imunologia , Inflamação/imunologia , Macrófagos/imunologia , Infecções Estreptocócicas/imunologia , Streptococcus/fisiologia , Células Estromais/metabolismo , Adesão Celular , Feminino , Humanos , Tolerância Imunológica , Metaloproteinase 9 da Matriz/metabolismo , NF-kappa B/metabolismo , Gravidez , Transdução de Sinais , Células Estromais/patologia , Células THP-1 , Receptores Toll-Like/metabolismo , Trofoblastos , Fator de Necrose Tumoral alfa/metabolismo
2.
J Clin Med ; 8(9)2019 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-31533282

RESUMO

BK polyomavirus (BKPyV), or BKV infection, is ubiquitous and usually non-pathogenic, with subclinical infections in 80-90% of adults worldwide. BKV infection is often associated with pathology in immunocompromised individuals. BKV infection often is associated with renal impairment, including ureteral stenosis, hemorrhagic cystitis, and nephropathy. BKV infection is less commonly associated with pneumonitis, retinitis, liver disease, and meningoencephalitis. BKV is known to replicate, establish latency, undergo reactivation, and induce clinical pathology in renal tubular epithelial cells. However, recent in vitro studies support the notion that BKV has expanded tropism-targeting glomerular parenchymal cells of the human kidney, which could impact glomerular function, enhance inflammation, and serve as viral reservoirs for reactivation from latency during immunosuppression. The implications of BKV expanded tropism in the glomerulus, and how specific host and viral factors that would contribute to glomerular inflammation, cytolysis, and renal fibrosis are related to BKV associated nephropathy (BKVAN), have not been explored. The pathogenesis of BKV in human glomerular parenchymal cells is poorly understood. In this review, I examine target cell populations for BKV infectivity in the human glomerulus. Specifically, I explore the implications of BKV expanded tropism in the glomerulus with regard viral entry, replication, and dissemination via cell types exposed to BKV trafficking in glomerulus. I also describe cellular targets shown to be permissive in vitro and in vivo for BKV infection and lytic replication, the potential role that glomerular parenchymal cells play in BKV latency and/or reactivation after immunosuppression, and the rare occurrence of BKV pathology in glomerular parenchymal cells in patients with BKVAN.

3.
Viruses ; 11(7)2019 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-31252545

RESUMO

BACKGROUND: BK polyomavirus (BKV) reactivates from latency after immunosuppression in renal transplant patients, resulting in BKV-associated nephropathy (BKVAN). BKVAN has emerged as an important cause of graft dysfunction and graft loss among transplant patients. BKV infection in kidney transplant patients has increased over recent decades which correlates with the use of more potent immunosuppressive therapies. BKV infection of the Glomerular Vascular Unit (GVU) consisting of podocytes, mesangial cells, and glomerular endothelial cells could lead to glomerular inflammation and contribute to renal fibrosis. The effects of BKV on GVU infectivity have not been reported. METHODS: We infected GVU cells with the Dunlop strain of BKV. Viral infectivity was analyzed by microscopy, immunofluorescence, Western blot analysis, and quantitative RT-PCR (qRT-PCR). The expression of specific proinflammatory cytokines induced by BKV was analyzed by qRT-PCR. RESULTS: BKV infection of podocytes, mesangial cells, and glomerular endothelial cells was confirmed by qRT-PCR and positive staining with antibodies to the BKV VP1 major capsid protein, or the SV40 Large T-Antigen. The increased transcriptional expression of interferon gamma-induced protein 10 (CXCL10/IP-10) and interferon beta (IFNß) was detected in podocytes and mesangial cells at 96 h post-infection. CONCLUSIONS: All cellular components of the GVU are permissive for BKV replication. Cytopathic effects induced by BKV in podocytes and glomerular endothelial cells and the expression of CXCL10 and IFNß genes by podocytes and mesangial cells may together contribute to glomerular inflammation and cytopathology in BKVAN.


Assuntos
Vírus BK/fisiologia , Nefropatias/virologia , Glomérulos Renais/virologia , Infecções por Polyomavirus/virologia , Complicações Pós-Operatórias/virologia , Vírus BK/genética , Citocinas/genética , Citocinas/imunologia , Humanos , Nefropatias/genética , Nefropatias/imunologia , Glomérulos Renais/imunologia , Transplante de Rim/efeitos adversos , Infecções por Polyomavirus/genética , Infecções por Polyomavirus/imunologia , Complicações Pós-Operatórias/etiologia , Complicações Pós-Operatórias/genética , Complicações Pós-Operatórias/imunologia , Ativação Viral , Replicação Viral
4.
Int J Mol Sci ; 20(6)2019 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-30909422

RESUMO

Pericytes are multipotent cells of the vascular system with cytoplasmic extensions proximal to endothelial cells that occur along the abluminal surface of the endothelium. The interactions between endothelial cells and pericytes are essential for proper microvascular formation, development, stabilization, and maintenance. Pericytes are essential for the regulation of paracellular flow between cells, transendothelial fluid transport, angiogenesis, and vascular immunosurveillance. They also influence the chemical composition of the surrounding microenvironment to protect endothelial cells from potential harm. Dysregulation or loss of pericyte function can result in microvascular instability and pathological consequences. Human pericytes have been shown to be targets for human cytomegalovirus (HCMV) infection and lytic replication that likely contribute to vascular inflammation. This review focuses on human vascular pericytes and their permissiveness for HCMV infection. It also discusses their implication in pathogenesis in the blood⁻brain barrier (BBB), the inner blood⁻retinal barrier (IBRB), the placenta⁻blood barrier, and the renal glomerulus as well as their potential role in subclinical vascular disease.


Assuntos
Infecções por Citomegalovirus/virologia , Citomegalovirus/fisiologia , Interações Hospedeiro-Patógeno , Pericitos/metabolismo , Pericitos/virologia , Animais , Suscetibilidade a Doenças , Endotélio Vascular/metabolismo , Endotélio Vascular/virologia , Feminino , Humanos , Células Mesangiais/metabolismo , Células Mesangiais/virologia , Placenta/metabolismo , Placenta/virologia , Gravidez , Retina/metabolismo , Retina/virologia , Medição de Risco , Fatores de Risco
5.
Virology ; 519: 77-85, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29679791

RESUMO

BACKGROUND: Zika virus (ZIKV) infection has been associated with microcephaly in infants. Currently there is no treatment or vaccine. Here we explore the use of a morpholino oligonucleotide targeted to the 5' untranslated region (5'-UTR) of the ZIKV RNA to prevent ZIKV replication. METHODS: Morpholino DWK-1 inhibition of ZIKV replication in human glomerular podocytes was examined by qRT-PCR, reduction in ZIKV genome copy number, western blot analysis, immunofluorescence and proinflammatory cytokine gene expression. RESULTS: Podocytes pretreated with DWK-1 showed reduced levels of both viral mRNA and ZIKV E protein expression compared to controls. We observed suppression in proinflammatory gene expression for IFN-ß (interferon ß) RANTES (regulated on activation, normal T cell expressed and secreted), MIP-1α (macrophage inflammatory protein-1α), TNF-α (tumor necrosis factor-α) and IL1-α (interleukin 1-α) in ZIKV-infected podocytes pretreated with DWK-1. CONCLUSIONS: Morpholino DWK-1 targeting the ZIKV 5'-UTR effectively inhibits ZIKV replication and suppresses ZIKV-induced proinflammatory gene expression.


Assuntos
Regiões 5' não Traduzidas/efeitos dos fármacos , Morfolinos/farmacologia , Replicação Viral/efeitos dos fármacos , Zika virus/efeitos dos fármacos , Quimiocina CCL5/genética , Citocinas/genética , Imunofluorescência , Humanos , Interferon beta/genética , Podócitos/virologia , Reação em Cadeia da Polimerase em Tempo Real , Zika virus/genética , Zika virus/fisiologia
6.
J Oncobiomarkers ; 2(1): 1-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26191531

RESUMO

Kaposi's sarcoma (KS), a common cancer in individuals with HIV/AIDS, lacks a curative therapy. Few studies have examined changes in extracellular matrix (ECM) protein profiles in the development of KS. Here we used an in vitro (human dermal microvascular endothelial cells, DMVEC) and an in vivo mECK mouse model of Kaposi's to study the impact of infection on tropoelastin. Using DMVEC, Kaposi's sarcoma-associated herpesvirus (KSHV) reduced tropoelastin transcription when examined at 2, 5, 7, and 10 days post addition, a finding that was inversely correlated with a rise in viral latency associated nuclear antigen (LANA) transcription. Immunohistochemical/immunofluorescence data confirmed that DMVEC cells were KSHV-infected (evidenced by LANA production) and that there was a loss of tropoelastin protein compared to controls. Using the mECK36 mouse model of KS we observed a reduced expression of tropoelastin mRNA in 3 of 3 tumor biopsies compared to controls. Immunofluorescence staining showed high levels of viral LANA expression in the tumor core, while immunohistochemical staining showed high levels of LANA expression and spindle cells in tumors. Dual label immunohistochemistry on formalin-fixed paraffin-embedded tumor tissue revealed reduced expression of tropoelastin in LANA positive spindle cell regions quantified by Ariol SL-50 scanning analysis. Together, this suggests that alterations in tropoelastin may play an important role in the development of Kaposi's and could serve as an early marker of this disease. This information will also allow us to explore the potential role of tropoelastin anti angiogenic properties in an in vivo model for KS disease.

7.
J Neuroinflammation ; 9: 95, 2012 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-22607552

RESUMO

BACKGROUND: Congenital human cytomegalovirus (HCMV) infections can result in CNS abnormalities in newborn babies including vision loss, mental retardation, motor deficits, seizures, and hearing loss. Brain pericytes play an essential role in the development and function of the blood-brain barrier yet their unique role in HCMV dissemination and neuropathlogy has not been reported. METHODS: Primary human brain vascular pericytes were exposed to a primary clinical isolate of HCMV designated 'SBCMV'. Infectivity was analyzed by microscopy, immunofluorescence, Western blot, and qRT-PCR. Microarrays were performed to identify proinflammatory cytokines upregulated after SBCMV exposure, and the results validated by real-time quantitative polymerase chain reaction (qPCR) methodology. In situ cytokine expression of pericytes after exposure to HCMV was examined by ELISA and in vivo evidence of HCMV infection of brain pericytes was shown by dual-labeled immunohistochemistry. RESULTS: HCMV-infected human brain vascular pericytes as evidenced by several markers. Using a clinical isolate of HCMV (SBCMV), microscopy of infected pericytes showed virion production and typical cytomegalic cytopathology. This finding was confirmed by the expression of major immediate early and late virion proteins and by the presence of HCMV mRNA. Brain pericytes were fully permissive for CMV lytic replication after 72 to 96 hours in culture compared to human astrocytes or human brain microvascular endothelial cells (BMVEC). However, temporal transcriptional expression of pp65 virion protein after SBCMV infection was lower than that seen with the HCMV Towne laboratory strain. Using RT-PCR and dual-labeled immunofluorescence, proinflammatory cytokines CXCL8/IL-8, CXCL11/ITAC, and CCL5/Rantes were upregulated in SBCMV-infected cells, as were tumor necrosis factor-alpha (TNF-alpha), interleukin-1 beta (IL-1beta), and interleukin-6 (IL-6). Pericytes exposed to SBCMV elicited higher levels of IL-6 compared to both mock-infected as well as heat-killed virus controls. A 6.6-fold induction of IL-6 and no induction TNF-alpha was observed in SBCMV-infected cell supernatants at 24 hours postinfection. Using archival brain tissue from a patient coinfected with HCMV and HIV, we also found evidence of HCMV infection of pericytes using dual-label immunohistochemistry, as monitored by NG2 proteoglycan staining. CONCLUSION: HCMV lytic infection of primary human brain pericytes suggests that pericytes contribute to both virus dissemination in the CNS as well as neuroinflammation.


Assuntos
Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Citocinas/biossíntese , Infecções por Citomegalovirus/patologia , Citomegalovirus/metabolismo , Mediadores da Inflamação/metabolismo , Pericitos/metabolismo , Regulação para Cima/fisiologia , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/virologia , Encéfalo/patologia , Encéfalo/virologia , Células Cultivadas , Citocinas/sangue , Citomegalovirus/imunologia , Infecções por Citomegalovirus/sangue , Infecções por Citomegalovirus/virologia , Humanos , Mediadores da Inflamação/sangue , Pericitos/patologia , Pericitos/virologia
8.
Microbes Infect ; 14(6): 500-8, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22227318

RESUMO

Bacterial vaginosis (BV), a common condition seen in premenopausal women, is associated with preterm labor, pelvic inflammatory disease, and delivery of low birth weight infants. Gardnerella vaginalis is the predominant bacterial species associated with BV, although its exact role in the pathology of BV is unknown. Using immunofluorescence, confocal and transmission electron microscopy, we found that VK2 vaginal epithelial cells take up G. vaginalis after exposure to the bacteria. Confocal microscopy also indicated the presence of internalized G. vaginalis within vaginal epithelial cells obtained from a subject with BV. Using VK2 cells and (35)S labeled bacteria in an invasion assay, we found that a 1 h uptake of G. vaginalis was 21.8-fold higher than heat-killed G. vaginalis, 84-fold compared to Lactobacillus acidophilus and 6.6-fold compared to Lactobacillus crispatus. Internalization was inhibited by pre-exposure of cells to cytochalasin-D. In addition, the cytoskeletal protein vimentin was upregulated in VK2 cells exposed to G. vaginalis, but there was no change in actin cytoskeletal polymerization/rearrangements or vimentin subcellular relocalization post exposure. Cytoskeletal protein modifications could represent a potential mechanism for G. vaginalis mediated internalization by vaginal epithelial cells. Finally, understanding vaginal bacteria/host interactions will allow us to better understand the underlying mechanisms of BV pathogenesis.


Assuntos
Células Epiteliais/microbiologia , Gardnerella vaginalis/patogenicidade , Interações Hospedeiro-Patógeno , Vagina/microbiologia , Vaginose Bacteriana/microbiologia , Hidrocarboneto de Aril Hidroxilases/farmacologia , Células Epiteliais/ultraestrutura , Feminino , Gardnerella vaginalis/efeitos dos fármacos , Humanos , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Esteroide Hidroxilases/farmacologia , Vaginose Bacteriana/fisiopatologia , Virulência
9.
Am J Pathol ; 179(3): 1443-54, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21741351

RESUMO

Kaposi's sarcoma is an angioproliferative tumor caused by Kaposi's sarcoma-associated herpesvirus (KSHV) infection of vascular endothelial cells. Fibulins, proteins that associate with extracellular matrix (ECM) proteins, may have both tumor-suppressive and oncogenic activities. We found that the expression of fibulin-2 protein and mRNA were decreased 50-fold and 26-fold, respectively, in 10-day KSHV-infected dermal microvascular endothelial cells (DMVEC). Using quantitative RT-PCR, we found a fivefold and 25-fold decrease of fibulin-2 extracellular matrix binding partners, fibronectin and tropoelastin, respectively. Time-course transcriptional analyses over 10 days showed that in addition to that of fibulin-2, expression of fibulins 3 and 5 was decreased in KSHV-infected DMVEC, fibulins 1C/1D were increased, and fibulins 4, 6, and 7 were unchanged. KSHV latency-associated nuclear antigen (LANA) transcription levels rose consistently over the same period. Addition of recombinant fibulin-3 or -5 for 48 hours to 10-day KSHV-infected cells caused a suppression of KSHV-induced vascular endothelial growth factor (VEGF) protein and mRNA levels. Recombinant fibulin-3 also significantly reduced VEGF receptor 3 expression. In pleural effusion lymphoma cell lines that express variable levels of KSHV lytic replication, we observed no detectable fibulin-2 or -5 expression. Finally, fibulin-2 expression was decreased in tissue microarrays from KSHV-infected, LANA-positive patient cells as compared to that in patient nontumor controls. Understanding the interactions between KSHV and the fibulins may lead to the development of novel therapies for treatment of Kaposi's sarcoma.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Transformação Celular Neoplásica/patologia , Proteínas da Matriz Extracelular/metabolismo , Herpesvirus Humano 8/fisiologia , Sarcoma de Kaposi/virologia , Neoplasias Cutâneas/virologia , Linhagem Celular Tumoral , Regulação para Baixo , Endotélio Vascular/virologia , Proteínas da Matriz Extracelular/farmacologia , Fibronectinas/metabolismo , Humanos , Proteínas Recombinantes , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/patologia , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Tropoelastina/metabolismo , Regulação para Cima , Carga Viral
10.
Glycobiology ; 20(5): 521-32, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20040515

RESUMO

Galectins are a family of proteins that share an affinity for beta-galactoside containing glycoconjugates. In prostate, ovarian and breast cancer, downregulation of galectin-3 is associated with malignancy and tumor progression. Kaposi's sarcoma (KS) is characterized as an angioproliferative tumor of vascular endothelial cells and produces rare B cell lymphoproliferative diseases in the form of primary effusion lymphomas and some forms of multicentric Castleman's disease. Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiological agent of KS. We found reduced levels of galectin-3 expression in a significant fraction of latency-associated nuclear antigen (LANA)-positive spindle cell regions in human archival KS tissue and as measured in KS tissue microarrays. Here we demonstrate that galectin-3 protein expression is downregulated 10-fold in 10-day KSHV-infected dermal microvascular endothelial cells (DMVEC) accompanied by downregulation of message. There is loss of galectin-3 staining in KSHV-infected DMVEC by dual labeled immunohistochemistry in LANA-positive spindle cells. We observed a consistent downregulation of galectin-3 by time-course transcriptional analysis. Of the galectins assayed, only galectin-1 was also downregulated in KSHV-infected DMVEC. We examined 86 KS tumors; 19 were LANA positive (22%) and 67 LANA negative (78%). All 86 tumors were found to be galectin-3 positive; 11 of 19 showed reduced expression of galectin-3 in LANA-positive spindle cell regions. Our data suggest that KSHV vFLIP and LANA are the viral genes targeting galectin-3 downregulation. The contribution of host factors to the pathogenesis of KS is essential for early detection and development of innovative therapies for treatment.


Assuntos
Regulação para Baixo , Galectina 3/metabolismo , Herpesvirus Humano 8/fisiologia , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virologia , Antígenos Virais/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/virologia , Galectina 3/biossíntese , Galectina 3/genética , Células HeLa , Humanos , Imuno-Histoquímica , Microcirculação , Proteínas Nucleares/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise Serial de Tecidos , Células Tumorais Cultivadas
11.
J Clin Virol ; 40(1): 1-8, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17698410

RESUMO

Ever since the original identification of fragments of KSHV DNA in Kaposi's sarcoma (KS) tissue by Chang et al. in 1994, PCR has been used successfully and extensively to detect the virus in clinical samples from the accepted etiological diseases of KS, PEL and MCD. However, a number of other clinical and epidemiological studies claiming evidence for KSHV in multiple myeloma or sarcoid and more recently in primary pulmonary hypertension, as well as claims about the biological significance of DNA sequence polymorphisms based just on small ORF26 PCR DNA fragments have not been convincing. Here, we evaluate the validity and interpretations of previous results in the context of both the observed rates and global patterns of sequence variability within an extended ORF26 locus, as well as from the perspective of the overall levels of KSHV variability found after sampling multiple loci across the complete KSHV genome. The results cast doubts on most claims for biological significance for these polymorphisms, which instead correlate with viral subtype clustering arising from geographic and ethnic divergence of the ancestral human hosts. In addition, we describe several observations that help to explain likely sources of the often either unexpectedly high or unexpectedly low levels of sporadic variability seen in the PCR DNA sequence data reported in some of those studies.


Assuntos
Genoma Viral , Herpesvirus Humano 8/genética , Sarcoma de Kaposi/epidemiologia , Sarcoma de Kaposi/virologia , Infecções Oportunistas Relacionadas com a AIDS/epidemiologia , Infecções Oportunistas Relacionadas com a AIDS/virologia , Variação Genética , Genótipo , Infecções por Herpesviridae/epidemiologia , Infecções por Herpesviridae/virologia , Humanos , Fases de Leitura Aberta , Reação em Cadeia da Polimerase , Polimorfismo Genético
12.
J Virol ; 76(7): 3421-39, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11884567

RESUMO

The ORF74 or vGCR gene encoded by Kaposi's sarcoma-associated herpesvirus (KSHV; also called human herpesvirus 8) has properties of a ligand-independent membrane receptor signaling protein with angiogenic properties that is predicted to play a key role in the biology of the virus. We have examined the expression of vGCR mRNA and protein in primary effusion lymphoma (PEL) cell lines, PEL and multicentric Castleman's disease (MCD) tumors, Kaposi's sarcoma lesions and infected endothelial cell cultures. The vGCR gene proved to be expressed in PEL cell lines as a large spliced bicistronic mRNA of 3.2 kb that also encompasses the upstream vOX2 (K14) gene. This mRNA species was induced strongly by phorbol ester (TPA) and sodium butyrate treatment in the BCBL-1 cell line, but only weakly in the HBL6 cell line, and was classified as a relatively late and low-abundance delayed early class lytic cycle gene product. A complex bipartite upstream lytic cycle promoter for this mRNA was nestled within the intron of the 5'-overlapping but oppositely oriented latent-state transcription unit for LANA1/vCYC-D/vFLIP and responded strongly to both TPA induction and cotransfection with the KSHV RNA transactivator protein (RTA or ORF50) in transient reporter gene assays. A vGCR protein product of 45 kDa that readily dimerized was detected by Western blotting and in vitro translation and was localized in a cytoplasmic and membrane pattern in DNA-transfected Vero and 293T cells or adenovirus vGCR-transduced dermal microvascular endothelial cells (DMVEC) as detected by indirect immunofluorescence assay (IFA) and immunohistochemistry with a specific rabbit anti-vGCR antibody. Similarly, a subfraction of KSHV-positive cultured PEL cells and of KSHV (JSC-1) persistently infected DMVEC cells displayed cytoplasmic vGCR protein expression, but only after TPA or spontaneous lytic cycle induction, respectively. The vGCR protein was also detectable by immunohistochemical staining in a small fraction (0.5 to 3%) of the cells in PEL and MCD tumor and nodular Kaposi's sarcoma lesion specimens that were apparently undergoing lytic cycle expression. These properties are difficult to reconcile with the vGCR protein's playing a direct role in spindle cell proliferation, transformation, or latency, but could be compatible with proposed contributions to angiogenesis via downstream paracrine effects. The ability of vGCR to transactivate expression of both several KSHV promoter-driven luciferase (LUC) reporter genes and an NFkappaB motif containing the chloramphenicol acetyltransferase (CAT) reporter gene may also suggest an unexpected regulatory role in viral gene expression.


Assuntos
Regulação Viral da Expressão Gênica , Herpesvirus Humano 8/metabolismo , Receptores de Quimiocinas/genética , Transativadores/metabolismo , Proteínas Virais/genética , Sequência de Bases , Linhagem Celular , Membrana Celular/metabolismo , Citoplasma/metabolismo , Humanos , Proteínas Imediatamente Precoces/metabolismo , Dados de Sequência Molecular , Regiões Promotoras Genéticas , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , RNA Viral/análise , RNA Viral/biossíntese , Receptores de Quimiocinas/biossíntese , Receptores de Quimiocinas/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Transcrição Gênica , Proteínas Virais/biossíntese , Proteínas Virais/metabolismo , Latência Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA