Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Adv Clin Exp Med ; 28(11): 1441-1450, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31778596

RESUMO

BACKGROUND: Human cytomegalovirus (HCMV) infection is one of the risk factors of cardiovascular disease; the most important pathological change is the change of vascular endothelial cell (VEC) function, but its mechanism is still unclear. Transforming growth factor ß1 (TGF-ß1) is an important cytokine associated with fibrosis; it can induce the occurrence of endothelial mesenchymal transition (EndMT) in VECs, which means endothelial cells acquire the characteristics and phenotypes of mesenchymal cells and secrete molecules associated with the deposition and remodeling of the extracellular matrix. Many in vivo and in vitro studies have shown that HCMV infection promotes the secretion and activation of TGF-ß1. OBJECTIVES: This study aims to observe the changes of endothelial cells after HCMV infection and EndMT occurrence induced by TGF-ß1 and to explore the possible mechanism of HCMV infection in the pathogenesis of cardiovascular disease. MATERIAL AND METHODS: Immunofluorescence staining, reverse transcription polymerase chain reaction (RT-PCR), enzyme-linked immunosorbent assay (ELISA), and immunoprecipitation methods were used in this study to analyze the changes in morphology and gene expression. RESULTS: We found that EndMT-related morphological and gene expression changes occurred in human umbilical vein endothelial cells (HUVECs) infected and uninfected with HCMV after treatment with TGF-ß1. Human umbilical vein endothelial cells infected with HCMV, which are treated with TGF-ß1, can activate the extracellular potential TGF-ß1 by activating matrix metalloproteinase 2 (MMP-2). CONCLUSIONS: Our findings provide a molecular basis for the association between HCMV infection, TGF-ß1 and cardiovascular disease.


Assuntos
Infecções por Citomegalovirus/tratamento farmacológico , Citomegalovirus , Transição Epitelial-Mesenquimal , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Metaloproteinase 2 da Matriz , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Veias Umbilicais/metabolismo , Veias Umbilicais/virologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Infecções por Citomegalovirus/diagnóstico , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Transdução de Sinais
2.
Cell Rep ; 26(6): 1598-1613.e8, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30726741

RESUMO

Flaviviruses cause systemic or neurotropic-encephalitic pathology in humans. The flavivirus nonstructural protein 1 (NS1) is a secreted glycoprotein involved in viral replication, immune evasion, and vascular leakage during dengue virus infection. However, the contribution of secreted NS1 from related flaviviruses to viral pathogenesis remains unknown. Here, we demonstrate that NS1 from dengue, Zika, West Nile, Japanese encephalitis, and yellow fever viruses selectively binds to and alters permeability of human endothelial cells from lung, dermis, umbilical vein, brain, and liver in vitro and causes tissue-specific vascular leakage in mice, reflecting the pathophysiology of each flavivirus. Mechanistically, each flavivirus NS1 leads to differential disruption of endothelial glycocalyx components, resulting in endothelial hyperpermeability. Our findings reveal the capacity of a secreted viral protein to modulate endothelial barrier function in a tissue-specific manner both in vitro and in vivo, potentially influencing virus dissemination and pathogenesis and providing targets for antiviral therapies and vaccine development.


Assuntos
Vírus da Dengue/genética , Células Endoteliais/virologia , Glicocálix/virologia , Proteínas não Estruturais Virais/genética , Animais , Encéfalo/patologia , Encéfalo/virologia , Linhagem Celular , Permeabilidade da Membrana Celular , Dengue/genética , Dengue/metabolismo , Dengue/patologia , Vírus da Dengue/metabolismo , Vírus da Dengue/patogenicidade , Derme/patologia , Derme/virologia , Vírus da Encefalite Japonesa (Espécie)/genética , Vírus da Encefalite Japonesa (Espécie)/metabolismo , Vírus da Encefalite Japonesa (Espécie)/patogenicidade , Células Endoteliais/patologia , Expressão Gênica , Glicocálix/química , Humanos , Fígado/patologia , Fígado/virologia , Pulmão/patologia , Pulmão/virologia , Masculino , Camundongos , Especificidade de Órgãos , Cultura Primária de Células , Veias Umbilicais/patologia , Veias Umbilicais/virologia , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/metabolismo , Vírus do Nilo Ocidental/patogenicidade , Vírus da Febre Amarela/genética , Vírus da Febre Amarela/metabolismo , Vírus da Febre Amarela/patogenicidade , Zika virus/genética , Zika virus/metabolismo , Zika virus/patogenicidade
3.
Antiviral Res ; 155: 20-27, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29709564

RESUMO

Dengue virus (DENV) infection has become a severe public health problem worldwide. However, there is no specific antiviral drug available yet. In this study, we found that DENV serotype 2 (DENV2) infection enhanced the expression of ß3 integrin on human umbilical vein endothelial cells (HUVECs) and that DENV2 antigens co-localized with ß3 integrin. DENV2 envelope protein (E) directly interacted with ß3 integrin, and their interacting sites were located at domain III of E protein (EDIII). Several synthetic peptides were designed based on the amino acid sequence of EDIII, and peptides P4 and P7 could inhibit DENV2 entry into HUVECs in a dose-dependent manner. The inhibitory concentration (IC50) of the two peptides was 19.08 ±â€¯2.52 µM for P4 and 12.86 ±â€¯5.96 µM for P7. Moreover, P7 containing an FG-loop, but not P4, could also inhibit DENV1 entry into HUVECs. Our results suggest a novel mechanism in which interaction between ß3 integrin and EDIII is involved in DENV entry. The findings on the inhibitory effect of the peptides on viral entry have significance for anti-DENV drug design.


Assuntos
Células Endoteliais/virologia , Interações entre Hospedeiro e Microrganismos , Integrina beta3/metabolismo , Peptídeos/farmacologia , Proteínas do Envelope Viral/metabolismo , Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Desenho de Fármacos , Humanos , Concentração Inibidora 50 , Peptídeos/síntese química , Domínios e Motivos de Interação entre Proteínas , Sorogrupo , Veias Umbilicais/citologia , Veias Umbilicais/virologia , Internalização do Vírus/efeitos dos fármacos
4.
PLoS One ; 12(10): e0186477, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29059211

RESUMO

Severe influenza infections are featured by acute lung injury, a syndrome of increased pulmonary microvascular permeability. A growing number of evidences have shown that influenza A virus induces cytoskeletal rearrangement and permeability increase in endothelial cells. Although miRNA's involvement in the regulation of influenza virus infection and endothelial cell (EC) function has been well documented, little is known about the miRNA profiles in influenza-infected endothelial cells. Using human umbilical vein endothelial cells (HUVECs) as cell models, the present study aims to explore the differential miRNAs in influenza virus-infected ECs and analyze their target genes involved in EC permeability regulation. As the results showed, permeability increased and F-actin cytoskeleton reorganized after HUVECs infected with influenza A virus (CA07 or PR8) at 30 MOI. MicroRNA microarray revealed a multitude of miRNAs differentially expressed in HUVECs after influenza virus infection. Through target gene prediction, we found that a series of miRNAs were involved in PKC, Rho/ROCK, HRas/Raf/MEK/ERK, and Ca2+/CaM pathways associated with permeability regulation, and most of these miRNAs were down-regulated after flu infection. It has been reported that PKC, Rho/ROCK, HRas/Raf/MEK/ERK, and Ca2+/CaM pathways are activated by flu infection and play important roles in permeability regulation. Therefore, the cumulative effects of these down-regulated miRNAs which synergistically enhanced activation of PKC, Rho/ROCK, Ras/Raf/MEK/ERK, and Ca2+/CaM pathways, can eventually lead to actin rearrangement and hyperpermeability in flu-infected HUVECs.


Assuntos
Permeabilidade Capilar/genética , Vírus da Influenza A/fisiologia , MicroRNAs/fisiologia , Veias Umbilicais/virologia , Perfilação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos
5.
PLoS One ; 9(10): e110911, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25340775

RESUMO

Classical Swine Fever (CSF) is a highly infectious fatal pig disease, resulting in huge economic loss to the swine industry. Integrins are membrane-bound signal mediators, expressed on a variety of cell surfaces and are known as receptors or co-receptors for many viruses. However, the role of integrin ß3 in CSFV infection is unknown. Here, through quantitive PCR, immunofluorescence (IFC) and immunocytohistochemistry (ICC), we revealed that ST (swine testicles epithelial) cells have a prominent advantage in CSFV proliferation as compared to EC (swine umbilical vein endothelial cell), IEC (swine intestinal epithelial cell) and PK (porcine kidney epithelial) cells. Meanwhile, ST cells had remarkably more integrin ß3 expression as compared to EC, IEC and PK cells, which was positively correlated with CSFV infection and proliferation. Integrin ß3 was up-regulated post CSFV infection in all the four cell lines, while the CSFV proliferation rate was decreased in integrin ß3 function-blocked cells. ShRNA1755 dramatically decreased integrin ß3, with a deficiency of 96% at the mRNA level and 80% at the protein level. CSFV proliferation was dramatically reduced in integrin ß3 constantly-defected cells (ICDC), with the deficiencies of 92.6%, 99% and 81.7% at 24 h, 48 h and 72 h post CSFV infection, respectively. These results demonstrate that integrin ß3 is required in CSFV infection and proliferation, which provide a new insight into the mechanism of CSFV infection.


Assuntos
Vírus da Febre Suína Clássica/fisiologia , Peste Suína Clássica/virologia , Células Endoteliais/virologia , Integrina beta3/metabolismo , Animais , Adesão Celular , Técnicas de Cultura de Células , Primers do DNA/química , Células Epiteliais/virologia , Intestinos/virologia , Masculino , Microscopia de Fluorescência , Reação em Cadeia da Polimerase , RNA Interferente Pequeno/metabolismo , Suínos , Testículo/virologia , Fatores de Tempo , Tripsina/química , Veias Umbilicais/virologia
6.
Vet Res ; 45: 48, 2014 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-24758593

RESUMO

Infection of domestic swine with the highly virulent Shimen strain of classical swine fever virus causes hemorrhagic lymphadenitis and diffuse hemorrhaging in infected swine. We analyzed patterns of gene expression for CSFV Shimen in swine umbilical vein endothelial cells (SUVECs). Transcription of the vascular endothelial growth factor (VEGF) C gene (VEGF-C) and translation of the corresponding protein were significantly up-regulated in SUVECs. Our findings suggest that VEGF-C is involved in mechanisms of acute infection caused by virulent strains of CSFV.


Assuntos
Vírus da Febre Suína Clássica/fisiologia , Vírus da Febre Suína Clássica/patogenicidade , Peste Suína Clássica/genética , Doenças dos Suínos/genética , Fator C de Crescimento do Endotélio Vascular/genética , Animais , Peste Suína Clássica/virologia , Vírus da Febre Suína Clássica/genética , Células Endoteliais/metabolismo , Perfilação da Expressão Gênica/veterinária , Regulação Viral da Expressão Gênica , Suínos , Doenças dos Suínos/virologia , Veias Umbilicais/metabolismo , Veias Umbilicais/virologia , Regulação para Cima , Fator C de Crescimento do Endotélio Vascular/metabolismo , Virulência
7.
Virology ; 443(2): 338-48, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23830076

RESUMO

Hantaviruses are negative strand RNA species that replicate predominantly in the cytoplasm. They also activate numerous cellular responses, but their involvement in nuclear processes is yet to be established. Using human umbilical vein endothelial cells (HUVECs), this study investigates the molecular finger-print of nuclear transcription factors during hantavirus infection. The viral-replication-dependent activation of pro-myelocytic leukemia protein (PML) was followed by subsequent localization in nuclear bodies (NBs). PML was also found in close proximity to activated Sp100 nuclear antigen and interferon-stimulated gene 20 kDa protein (ISG-20), but co-localization with death-domain associated protein-6 (DAXX) was not observed. These data demonstrate that hantavirus triggers PML activation and localization in NBs in the absence of DAXX-PLM-NB co-localization. The results suggest that viral infection interferes with DAXX-mediated apoptosis, and expression of interferon-activated Sp100 and ISG-20 proteins may indicate intracellular intrinsic antiviral attempts.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Antígenos Nucleares/metabolismo , Apoptose , Autoantígenos/metabolismo , Exonucleases/metabolismo , Regulação da Expressão Gênica , Infecções por Hantavirus/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Antígenos Nucleares/genética , Autoantígenos/genética , Proteínas Correpressoras , Células Endoteliais/imunologia , Células Endoteliais/virologia , Exonucleases/genética , Exorribonucleases , Infecções por Hantavirus/patologia , Humanos , Interferons/farmacologia , Corpos de Inclusão Intranuclear/metabolismo , Chaperonas Moleculares , Proteínas Nucleares/genética , Proteína da Leucemia Promielocítica , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Veias Umbilicais/citologia , Veias Umbilicais/virologia
8.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 41(3): 424-7, 2010 May.
Artigo em Chinês | MEDLINE | ID: mdl-20629313

RESUMO

OBJECTIVE: To detect whether the primary culture of human umbilical vein endothelial cells (HUVEC) is susceptible to human cytomegalovirus (HCMV) infection so as to explain how virus from mothers traverses the placenta to fetus. METHODS: Thirty umbilical cords of healthy parturient were used in this study. The HUVEC were harvested with 0.1% collagenase for 15 minutes. The cells were identified by morphology and immunofluorescence. Then the HUVEC were infected with HCMV in vitro. The DNA of cytomegalovirus and protein of infected cells were detected 72 hours later. RESULTS: The HUVEC with adequate quality were harvested, which expressed VIII related antigen. Positive expressions of CMV gB gene and virus proteins were identified in all of the HUVEC infected with HCMV. CONCLUSION: HUVEC is susceptible to HCMV infection, which suggests a possible route of HCMV infection through the fetal-maternal interface.


Assuntos
Citomegalovirus/patogenicidade , Células Endoteliais/virologia , Veias Umbilicais/virologia , Células Cultivadas , Infecções por Citomegalovirus/transmissão , Células Endoteliais/citologia , Humanos , Transmissão Vertical de Doenças Infecciosas , Veias Umbilicais/citologia
9.
Mol Cell Biochem ; 342(1-2): 215-21, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20526654

RESUMO

Dengue virus (DV) causes a non-specific febrile illness known as Dengue fever (DF), and a severe life-threatening illness, Dengue hemorrhagic fever/Dengue shock syndrome (DHF/DSS). Hemostatic changes induced by this virus involve three main factors: thrombocytopenia, endothelial cell damage, and significant abnormalities of the coagulation and fibrinolysis systems. The pathogenesis of bleeding in DV infections remains unknown. In this article, we focused on the DV activating endothelial cells and altering the parameters of hemostasis system. The expression of hemostasis-related factors, Thrombomodulin, TF, TFPI, t-PA, and PAI-1, in DV-infected cells were determined by RT-PCR. Flow cytometry analysis and immunofluorescence staining confirmed that the expression levels of TM in the DV-infected HMEC-1 and THP-1 cells were increased. In addition, the purified recombinant domain III of the envelope glycoprotein of DV (EIII) could induce the expression of TM in the HMEC-1 cells and THP-1 cells. The TM expression induced by DV or EIII in the endothelial cells and monocytic cells suggests that the EIII of DV plays an important role in the pathogenesis of DHF/DSS.


Assuntos
Anticoagulantes/metabolismo , Vírus da Dengue/fisiologia , Dengue/metabolismo , Endotélio Vascular/metabolismo , Leucemia Monocítica Aguda/metabolismo , Veias Umbilicais/metabolismo , Proteínas do Envelope Viral/metabolismo , Biomarcadores/metabolismo , Western Blotting , Dengue/patologia , Dengue/virologia , Endotélio Vascular/citologia , Endotélio Vascular/virologia , Citometria de Fluxo , Imunofluorescência , Hemostasia , Humanos , Leucemia Monocítica Aguda/patologia , Leucemia Monocítica Aguda/virologia , RNA Mensageiro/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Veias Umbilicais/citologia , Veias Umbilicais/virologia , Proteínas do Envelope Viral/genética
10.
J Thromb Thrombolysis ; 30(2): 164-71, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20155436

RESUMO

Human cytomegalovirus (HCMV) establishes a life-long persistent infection. HCMV infection could be associated with chronic inflammatory diseases, such as cardiovascular disease and atherosclerosis. Here we observed that in HCMV (AD-169) pre-exposed human umbilical vein endothelial cells (HUVEC), thrombin-induced expression of IL-1alpha and M-CSF is markedly enhanced compared to the un-exposed cells. Study of the expression of thrombin receptor genes in HUVEC showed that HCMV triggered a time- and concentration-dependent expression of the thrombin receptors PAR1, PAR3 and PAR4 at the mRNA level. Induction of PAR1 and PAR3 mRNA expression is due to transcriptional activation of their promoters as shown by gene reporter assay. Furthermore, the virus induced expression of PAR1 and PAR3 but not PAR4 proteins, as analyzed by Western immunoblotting. However, flow cytometric analysis revealed that only PAR3, expressed at very low level in control HUVEC, is induced at the surface during the exposure to the virus. Our data suggest that although exposure to HCMV induces a minor increase of cell-surface receptors expression, it does make endothelial cells more responsive to additional thrombin stimulation.


Assuntos
Citomegalovirus/patogenicidade , Células Endoteliais/virologia , Receptores de Trombina/metabolismo , Trombina/metabolismo , Veias Umbilicais/virologia , Western Blotting , Células Cultivadas , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Citometria de Fluxo , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-1beta/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Receptor PAR-1/metabolismo , Receptores de Trombina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Ativação Transcricional , Veias Umbilicais/citologia , Veias Umbilicais/imunologia , Veias Umbilicais/metabolismo , Regulação para Cima
11.
J Virol ; 83(10): 4895-911, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19279100

RESUMO

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.


Assuntos
Actinas/metabolismo , Herpesvirus Humano 8/fisiologia , Pinocitose/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Acetofenonas/farmacologia , Amilorida/análogos & derivados , Amilorida/farmacologia , Benzopiranos/farmacologia , Células Cultivadas , Clorpromazina/farmacologia , Clatrina/efeitos dos fármacos , Clatrina/metabolismo , Citocalasina D/farmacologia , DNA Viral/metabolismo , Células Endoteliais/ultraestrutura , Células Endoteliais/virologia , Fibroblastos/ultraestrutura , Fibroblastos/virologia , Filipina/farmacologia , Regulação Viral da Expressão Gênica , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Proteínas Imediatamente Precoces/metabolismo , Transativadores/metabolismo , Veias Umbilicais/citologia , Veias Umbilicais/virologia , Proteínas rab de Ligação ao GTP/metabolismo
12.
Cancer Res ; 68(12): 4640-8, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18559509

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) is associated with three different human malignancies, including Kaposi's sarcoma (KS), primary effusion lymphoma, and multicentric Castleman's disease. The KS lesion is of endothelial cell in origin and is highly dependent on autocrine and paracrine factors for survival and growth. In this study, we show that KSHV infection of endothelial cells induces the activation of the prosurvival phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin pathway. KSHV infection of endothelial cells augmented cell survival in the presence of apoptotic inducers, including etoposide and staurosporine, and under conditions of serum deprivation. We found that KSHV infection of endothelial cells also increased the ability of these cells to form an in vitro tubular network under conditions of stress and growth factor deprivation. Finally, we show that the nuclear factor-kappaB and PI3K pathways are also required for endothelial tubular network formation. Collectively, these results suggest that KSHV infection of endothelial cells modulates cell signaling pathways and induces cell survival and angiogenesis, thereby contributing to the pathogenesis induced by KSHV.


Assuntos
Sobrevivência Celular/fisiologia , Transformação Celular Neoplásica/patologia , Endotélio Vascular/citologia , Endotélio Vascular/virologia , Herpesvirus Humano 8/fisiologia , Sarcoma de Kaposi/virologia , Transdução de Sinais , Proteínas Quinases Ativadas por AMP , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Caspase 3/metabolismo , Transformação Celular Neoplásica/metabolismo , Meios de Cultura Livres de Soro , Endotélio Vascular/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Etoposídeo/farmacologia , Fator 2 de Crescimento de Fibroblastos , Humanos , Complexos Multienzimáticos , Fosfatidilinositol 3-Quinases/metabolismo , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/patologia , Estaurosporina/farmacologia , Taxa de Sobrevida , Serina-Treonina Quinases TOR , Veias Umbilicais/citologia , Veias Umbilicais/metabolismo , Veias Umbilicais/virologia , Fator A de Crescimento do Endotélio Vascular , Replicação Viral
13.
Am J Pathol ; 172(4): 1127-40, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18349127

RESUMO

Human cytomegalovirus (CMV) infection is a major cause of morbidity in immunosuppressed individuals, and congenital CMV infection is a leading cause of birth defects in newborns. Infection with pathogenic viral strains alters cell-cell and cell-matrix interactions, affecting extracellular matrix remodeling and endothelial cell migration. The multifunctional cytokine transforming growth factor (TGF)-beta1 regulates cell proliferation, differentiation, and extracellular matrix remodeling. Secreted as a latent protein complex, TGF-beta1 requires activation before binding to receptors that phosphorylate intracellular effectors. TGF-beta1 is activated by integrin alphavbeta6, which is strongly induced in the epithelium by injury and inflammation but has not previously been found in endothelial cells. Here, we report that CMV infection induces integrin alphavbeta6 expression in endothelial cells, leading to activation of TGF-beta1, signaling through its receptor ALK5, and phosphorylation of its intracellular effector Smad3. Infection of endothelial cells was also found to stimulate collagen synthesis through a mechanism dependent on both TGF-beta1 and integrin alphavbeta6. Immunohistochemical analysis showed integrin alphavbeta6 up-regulation in capillaries proximal to foci of CMV infection in lungs, salivary glands, uterine decidua, and injured chorionic villi of the placenta, demonstrating both its induction in endothelium and up-regulation in epithelium in vivo. Our results suggest that activation of TGF-beta1 by integrin alphavbeta6 contributes to pathological changes and may impair endothelial cell functions in tissues that are chronically infected with CMV.


Assuntos
Antígenos de Neoplasias/metabolismo , Colágeno/biossíntese , Infecções por Citomegalovirus/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/virologia , Células Epiteliais/metabolismo , Integrinas/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Receptores de Activinas Tipo II/metabolismo , Animais , Células Cultivadas , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/virologia , Células Endoteliais/patologia , Matriz Extracelular/metabolismo , Humanos , Cadeias beta de Integrinas/metabolismo , Vison , Especificidade de Órgãos , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Proteína Smad3/metabolismo , Veias Umbilicais/patologia , Veias Umbilicais/virologia , Regulação para Cima , Replicação Viral
14.
J Gen Virol ; 89(Pt 3): 642-652, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18272754

RESUMO

Plasma leakage is one of the characteristic features of dengue haemorrhagic fever. The interaction among peripheral blood mononuclear cells (PBMCs), dengue virus and endothelial cells was analysed in vitro. Human umbilical vein endothelial cells (HUVECs) were infected with dengue-2 virus (DV-2) at an m.o.i. of 0.5 p.f.u. per cell. PBMCs were added to DV-2-infected HUVECs, and transendothelial electrical resistance (TEER) and transalbumin permeability were assessed. Dengue virus infection at an m.o.i. of 0.5 p.f.u. per cell alone did not decrease the TEER, but addition of PBMCs decreased the TEER, increased the albumin permeability and induced morphological changes of HUVECs. The extent of the decrease was more profound with adherent PBMCs than with non-adherent PBMCs. The expression of vascular endothelial cadherin (VE-cadherin) was examined using real-time RT-PCR and immunofluorescence. Addition of PBMCs to DV-2-infected HUVECs decreased the levels of mRNA transcripts and cell-surface expression of VE-cadherin. The results indicate that PBMCs increased the permeability of DV-2-infected HUVECs and that the increased permeability was concomitant with morphological change and the decrease in VE-cadherin expression. The results suggest that functional impairment of the DV-2-infected HUVEC monolayer was caused by interaction with PBMCs.


Assuntos
Antígenos CD/metabolismo , Caderinas/metabolismo , Permeabilidade da Membrana Celular , Vírus da Dengue/patogenicidade , Regulação para Baixo , Células Endoteliais/virologia , Leucócitos Mononucleares/fisiologia , Adulto , Albuminas/fisiologia , Adesão Celular , Células Cultivadas , Impedância Elétrica , Células Endoteliais/metabolismo , Células Endoteliais/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/virologia , Humanos , Veias Umbilicais/citologia , Veias Umbilicais/virologia
15.
Cell Biol Int ; 31(10): 1207-13, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17544707

RESUMO

Tight junction (TJ) plays a pivotal role in preventing the invasion of pathogens from the blood to extracellular environment. However, the mechanisms by which Group B coxsackievirus 3 (CVB(3)) can get through TJ from the apical surface still remain obscure. In the present study, the human umbilical vein endothelial cell (HUVEC) was utilized to investigate the alterations in F-actin and ZO-1 status, permeability as well as p38 mitogen-activated protein kinase (MAPK) activity in response to CVB(3) by means of fluorescence labeling, flow cytometry, and macromolecule permeability assay. We found that CVB(3) was able to induce reorganization of F-actin and redistribution of ZO-1, increase the level of F-actin, and elevate the permeability of FITC-albumin. Moreover, CVB(3)-mediated the above effects involve in P38 MAPK activation. Our preliminary study indicates that CVB(3)-induced alteration in permeability may be attributed to disruption of F-actin and ZO-1 organizations and that SB203580, a specific P38 MAPK inhibitor, can reverse these effects. The precise mechanisms underlying the CVB(3)-mediated effects on HUVECs need to be studied further.


Assuntos
Actinas/metabolismo , Endotélio Vascular/virologia , Enterovirus Humano B/patogenicidade , Proteínas de Membrana/metabolismo , Fosfoproteínas/metabolismo , Junções Íntimas/virologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Permeabilidade da Membrana Celular , Células Cultivadas/metabolismo , Células Cultivadas/virologia , Infecções por Coxsackievirus/metabolismo , Infecções por Coxsackievirus/virologia , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Imidazóis/farmacologia , Piridinas/farmacologia , Junções Íntimas/metabolismo , Veias Umbilicais/citologia , Veias Umbilicais/metabolismo , Veias Umbilicais/virologia , Proteína da Zônula de Oclusão-1 , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
16.
J Virol ; 81(13): 7001-10, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17442715

RESUMO

Matrix metalloproteinases (MMPs) play important roles in cancer invasion, angiogenesis, and inflammatory infiltration. Kaposi's sarcoma is a highly disseminated angiogenic tumor of proliferative endothelial cells linked to infection by Kaposi's sarcoma-associated herpesvirus (KSHV). In this study, we showed that KSHV infection increased the invasiveness of primary human umbilical vein endothelial cells (HUVEC) in a Matrigel-based cell invasion assay. KSHV-induced cell invasion was abolished by an inhibitor of MMPs, BB-94, and occurred in both autocrine- and paracrine-dependent fashions. Analysis by zymography and Western blotting showed that KSHV-infected HUVEC cultures had increased secretion of MMP-1, -2, and -9. KSHV increased the secretion of MMP-2 within 1 h following infection without upregulating its mRNA expression level. In contrast, the secretion of MMP-1 and -9 was not increased until 6 h after KSHV infection and was correlated with the upregulation of their mRNA expression levels. Promoter analysis by reporter assays and electrophoretic mobility shift assays identified an AP-1 cis-element as the dominant KSHV-responsive site in the MMP-1 promoter. Together, these results suggest that KSHV infection modulates the production of multiple MMPs to increase cell invasiveness and thus contributes to the pathogenesis of KSHV-induced malignancies.


Assuntos
Colagenases/biossíntese , Células Endoteliais/enzimologia , Herpesvirus Humano 8/metabolismo , Neovascularização Patológica/enzimologia , Sarcoma de Kaposi/enzimologia , Veias Umbilicais/enzimologia , Comunicação Autócrina/efeitos dos fármacos , Transformação Celular Viral , Células Cultivadas , Células Endoteliais/patologia , Células Endoteliais/virologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Invasividade Neoplásica/patologia , Neovascularização Patológica/patologia , Neovascularização Patológica/virologia , Comunicação Parácrina/efeitos dos fármacos , Fenilalanina/análogos & derivados , Fenilalanina/farmacologia , Inibidores de Proteases/farmacologia , RNA Mensageiro/biossíntese , RNA Neoplásico/biossíntese , Elementos de Resposta , Sarcoma de Kaposi/patologia , Tiofenos/farmacologia , Fator de Transcrição AP-1/metabolismo , Veias Umbilicais/patologia , Veias Umbilicais/virologia , Regulação para Cima/efeitos dos fármacos
17.
Nan Fang Yi Ke Da Xue Xue Bao ; 26(3): 316-20, 2006 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-16546736

RESUMO

OBJECTIVE: To investigate the mechanisms for the cytopathic effect (CPE) of human cytomegalovirus (HCMV) in ECV304 endothelial-like cells. METHODS: PCR and indirect immunofluorescence were used to detect HCMV infection by examining immediate-early (IE) gene and protein expression of the virus in ECV304 cells. Phase-contrast and electron microscopies were performed to observe the morphological changes of the infected and uninfected cells, and DNA ladder analysis and flow cytometry were carried out to study HCMV-induced cell apoptosis. RESULTS: In HCMV-infected ECV304 cells, cytopathic effects were first observed at approximately 72 h post-infection. The cells with CPE changes exhibited detachment from the monolayer, cell rounding and shrinkage. The expression of the IE gene was detected. Chromatin condensation and nuclear fragmentation along with dramatic changes of the mitochondria were observed by electron microscopy at 96 h post-infection. Cellular DNA fragmentation was observed in the infected cells, which had cells apoptotic rates of 4.1% and 45.7% at 96 h and 144 h post-infection, respectively. CONCLUSION: HCMV can induce apoptosis of ECV304 endothelial-like cells.


Assuntos
Apoptose/fisiologia , Citomegalovirus/crescimento & desenvolvimento , Células Endoteliais/citologia , Antígenos Virais/genética , Antígenos Virais/metabolismo , Linhagem Celular , Citomegalovirus/genética , Citomegalovirus/metabolismo , DNA Viral/análise , Células Endoteliais/ultraestrutura , Células Endoteliais/virologia , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Microscopia Eletrônica , Microscopia de Contraste de Fase , Reação em Cadeia da Polimerase , Veias Umbilicais/citologia , Veias Umbilicais/virologia
18.
J Virol Methods ; 121(2): 171-80, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15381354

RESUMO

Electrical resistance across human umbilical vein endothelial cells (HUVECs) was measured using an electrical cell sensor system. The transendothelial electrical resistance (TEER) value was used to estimate the permeability through endothelial cells in vitro. Decrease in the TEER value was associated with increase in the passage of albumin through endothelial cells in the albumin permeability assay. The effects of cytokines and dengue virus infection on the permeability of HUVECs were examined by measuring the TEER value. Tumor necrosis factor alpha (TNF-alpha) at 1 and 0.1 microg/ml decreased the TEER value, but TNF-alpha at lower dose did not. Interferon-gamma (IFN-gamma) at 1 microg/ml also decreased the TEER value. In contrast, interleukin-2 (IL-2), interleukin-6 (IL-6), interleukin-8 (IL-8), interleukin-10 (IL-10) or interferon-beta (IFN-beta) did not decrease the TEER value. The decrease in the TEER value was associated with the morphological changes of HUVECs. Dengue virus infection at a multiplicities of infection (m.o.i.) of 5 pfu/cell decreased the TEER value. Infection at an m.o.i. of 0.5 pfu/cell did not decrease the TEER value; however, addition of 0.01 microg/ml of TNF-alpha to these infected endothelial cells decreased the TEER value. The results suggest that TNF-alpha and dengue virus infection decrease synergistically the TEER value of endothelial cells. The TEER method is easy, reliable and can be applicable to further analysis of the increase in the permeability of endothelial cells in vitro induced by inflammatory cytokines and dengue virus infection.


Assuntos
Permeabilidade da Membrana Celular , Citocinas/farmacologia , Vírus da Dengue/fisiologia , Células Endoteliais/metabolismo , Veias Umbilicais/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Impedância Elétrica , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/virologia , Humanos , Interferons/farmacologia , Fatores de Tempo , Fator de Necrose Tumoral alfa/farmacologia , Veias Umbilicais/efeitos dos fármacos , Veias Umbilicais/virologia
19.
Arterioscler Thromb Vasc Biol ; 23(9): 1535-40, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12842839

RESUMO

OBJECTIVE: Endothelial cells play a pivotal role in vascular homeostasis. In this study, we investigated the function of the nerve growth factor-induced protein-B (NGFI-B) subfamily of nuclear receptors comprising the TR3 orphan receptor (TR3), mitogen-induced nuclear orphan receptor (MINOR), and nuclear orphan receptor of T cells (NOT) in endothelial cells. METHODS AND RESULTS: The mRNA expression of TR3, MINOR, and NOT in atherosclerotic lesions was assessed in human vascular specimens. Each of these factors is expressed in smooth muscle cells, as described before, and in subsets of endothelial cells, implicating that they might affect endothelial cell function. Adenoviral overexpression of TR3 in cultured endothelial cells resulted in decreased [3H]thymidine incorporation, whereas a dominant-negative TR3 variant that inhibits the activity of endogenous TR3-like factors enhanced DNA synthesis. TR3 interfered with progression of the cell cycle by upregulating p27Kip1 and downregulating cyclin A, whereas expression levels of a number of other cell cycle-associated proteins remained unchanged. CONCLUSIONS: These findings demonstrate that TR3 is a modulator of endothelial cell proliferation and arrests endothelial cells in the G1 phase of the cell cycle by influencing cell cycle protein levels. We hypothesize involvement of TR3 in the maintenance of integrity of the vascular endothelium.


Assuntos
Ciclo Celular/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Receptores de Esteroides/biossíntese , Receptores de Esteroides/fisiologia , Receptores dos Hormônios Tireóideos/biossíntese , Receptores dos Hormônios Tireóideos/fisiologia , Adenoviridae/genética , Divisão Celular/genética , Divisão Celular/fisiologia , Células Cultivadas , DNA Complementar/genética , Endotélio Vascular/virologia , Regulação da Expressão Gênica/genética , Técnicas de Transferência de Genes , Variação Genética/genética , Humanos , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares , Peptídeos/genética , Peptídeos/fisiologia , Estrutura Terciária de Proteína/genética , Receptores de Esteroides/deficiência , Receptores de Esteroides/genética , Receptores dos Hormônios Tireóideos/deficiência , Receptores dos Hormônios Tireóideos/genética , Deleção de Sequência/genética , Células-Tronco/química , Células-Tronco/metabolismo , Células-Tronco/virologia , Ativação Transcricional/genética , Veias Umbilicais/citologia , Veias Umbilicais/virologia , Replicação Viral/genética
20.
Thromb Haemost ; 84(2): 319-24, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10959707

RESUMO

Influenza virus epidemics are associated with excess mortality due to cardiovascular diseases. There are several case reports of excessive coagulation during generalised influenza virus infection. In this study, we demonstrate the ability of respiratory viruses (influenza A, influenza B, parainfluenza-1, respiratory syncytial virus, adenovirus, cytomegalovirus) to infect lung fibroblasts and human umbilical vein endothelial cells in culture. All viral pathogens induced procoagulant activity in infected endothelial cells, as determined in a one-stage clotting assay, by causing an average 55% reduction in the clotting time. When factor VII deficient plasma was used clotting time was not reduced. The induction of procoagulant activity was associated with a 4- to 5-fold increase in the expression of tissue factor, as measured by the generation of factor Xa. Both experiments indicate that the procoagulant activity of endothelial cells in response to infection with respiratory viruses is caused by upregulation of the extrinsic pathway. Although both enveloped viruses and a non-enveloped virus (adenovirus) induced procoagulant activity in endothelial cells by stimulating tissue factor expression, the role of the viral envelope in the assembly of the prothrombinase complex remains uncertain. We conclude that both enveloped and non-enveloped respiratory viruses are capable of infecting cultured human endothelial cells and causing a shift from anticoagulant to procoagulant activity associated with the induction of tissue factor expression.


Assuntos
Endotélio Vascular/virologia , Infecções Respiratórias/sangue , Coagulação Sanguínea , Testes de Coagulação Sanguínea , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Fator Xa/biossíntese , Fibroblastos/metabolismo , Fibroblastos/virologia , Hemaglutininas Virais/metabolismo , Humanos , Pulmão/citologia , Pulmão/patologia , Pulmão/virologia , Infecções Respiratórias/patologia , Infecções Respiratórias/virologia , Tromboplastina/biossíntese , Fatores de Tempo , Células Tumorais Cultivadas/virologia , Veias Umbilicais/patologia , Veias Umbilicais/fisiopatologia , Veias Umbilicais/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...