Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
Front Immunol ; 12: 644153, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33968035

RESUMO

During recent Zika epidemics, adults infected with Zika virus (ZIKV) have developed organ-specific inflammatory complications. The most serious Zika-associated inflammatory eye disease is uveitis, which is commonly anterior in type, affecting both eyes and responding to corticosteroid eye drops. Mechanisms of Zika-associated anterior uveitis are unknown, but ZIKV has been identified in the aqueous humor of affected individuals. The iris pigment epithelium is a target cell population in viral anterior uveitis, and it acts to maintain immune privilege within the anterior eye. Interactions between ZIKV and human iris pigment epithelial cells were investigated with infectivity assays and RNA-sequencing. Primary cell isolates were prepared from eyes of 20 cadaveric donors, and infected for 24 hours with PRVABC59 strain ZIKV or incubated uninfected as control. Cytoimmunofluorescence, RT-qPCR on total cellular RNA, and focus-forming assays of culture supernatant showed cell isolates were permissive to infection, and supported replication and release of infectious ZIKV. To explore molecular responses of cell isolates to ZIKV infection at the whole transcriptome level, RNA was sequenced on the Illumina NextSeq 500 platform, and results were aligned to the human GRCh38 genome. Multidimensional scaling showed clear separation between transcriptomes of infected and uninfected cell isolates. Differential expression analysis indicated a vigorous molecular response of the cell to ZIKV: 7,935 genes were differentially expressed between ZIKV-infected and uninfected cells (FDR < 0.05), and 99% of 613 genes that changed at least two-fold were up-regulated. Reactome and KEGG pathway and Gene Ontology enrichment analyses indicated strong activation of viral recognition and defense, in addition to biosynthesis processes. A CHAT network included 6275 molecular nodes and 24 contextual hubs in the cell response to ZIKV infection. Receptor-interacting serine/threonine kinase 1 (RIPK1) was the most significantly connected contextual hub. Correlation of gene expression with read counts assigned to the ZIKV genome identified a negative correlation between interferon signaling and viral load across isolates. This work represents the first investigation of mechanisms of Zika-associated anterior uveitis using an in vitro human cell model. The results suggest the iris pigment epithelium mounts a molecular response that limits intraocular pathology in most individuals.


Assuntos
Células Epiteliais , Regulação Viral da Expressão Gênica/imunologia , Epitélio Pigmentado Ocular , RNA Viral/imunologia , Infecção por Zika virus , Zika virus/imunologia , Células Cultivadas , Células Epiteliais/imunologia , Células Epiteliais/patologia , Células Epiteliais/virologia , Genoma Viral/imunologia , Humanos , Iris/imunologia , Iris/patologia , Iris/virologia , Epitélio Pigmentado Ocular/imunologia , Epitélio Pigmentado Ocular/patologia , Epitélio Pigmentado Ocular/virologia , Infecção por Zika virus/imunologia , Infecção por Zika virus/patologia
2.
PLoS Negl Trop Dis ; 14(4): e0008223, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32324736

RESUMO

Usutu virus (USUV), an African mosquito-borne flavivirus closely related to West Nile virus, was first isolated in South Africa in 1959. USUV emerged in Europe two decades ago, causing notably massive mortality in Eurasian blackbirds. USUV is attracting increasing attention due to its potential for emergence and its rapid spread in Europe in recent years. Although mainly asymptomatic or responsible for mild clinical signs, USUV was recently described as being associated with neurological disorders in humans such as encephalitis and meningoencephalitis, highlighting the potential health threat posed by the virus. Despite this, USUV pathogenesis remains largely unexplored. The aim of this study was to evaluate USUV neuropathogenicity using in vivo and in vitro approaches. Our results indicate that USUV efficiently replicates in the murine central nervous system. Replication in the spinal cord and brain is associated with recruitment of inflammatory cells and the release of inflammatory molecules as well as induction of antiviral-responses without major modulation of blood-brain barrier integrity. Endothelial cells integrity is also maintained in a human model of the blood-brain barrier despite USUV replication and release of pro-inflammatory cytokines. Furthermore, USUV-inoculated mice developed major ocular defects associated with inflammation. Moreover, USUV efficiently replicates in human retinal pigment epithelium. Our results will help to better characterize the physiopathology related to USUV infection in order to anticipate the potential threat of USUV emergence.


Assuntos
Flavivirus/patogenicidade , Modelos Biológicos , Sistema Nervoso/virologia , Animais , Encéfalo/virologia , Modelos Animais de Doenças , Células Endoteliais/virologia , Células Epiteliais/virologia , Flavivirus/crescimento & desenvolvimento , Humanos , Camundongos , Epitélio Pigmentado Ocular/virologia , Medula Espinal/virologia
3.
Hum Gene Ther ; 21(2): 199-209, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19778186

RESUMO

Lentiviral vectors are promising tools for the treatment of chronic retinal diseases, including age-related macular degeneration (AMD), as they enable stable transgene expression. On the other hand, Sendai virus (SeV) vectors provide the unique advantage of rapid gene transfer. Here we show that novel simian immunodeficiency viral vectors pseudotyped with SeV envelope proteins (SeV-F/HN-SIV) achieved rapid, efficient, and long-lasting gene transfer in the mouse retina. Subretinal exposure to SeV-F/HN-SIV vectors for only a few minutes resulted in high-level gene transfer to the retinal pigment epithelium, whereas several hours were required for gene transfer by standard vesicular stomatitis virus G-pseudotyped SIV vectors. Transgene expression continued over a 1-year period. SeV-F/HN-SIV vector-mediated retinal overexpression of soluble Fms-like tyrosine kinase-1 (sFlt-1) or pigment epithelium-derived factor (PEDF) significantly suppressed laser-induced choroidal neovascularization (CNV). Histologically, 6-month-long sustained overexpression of PEDF did not adversely affect the retina; however, that with sFlt-1 resulted in photoreceptor degeneration associated with choroidal circulation defects. These data demonstrate that brief subretinal administration of SeV-F/HN-SIV vectors may facilitate safe and efficient retinal gene transfer, and suggest the therapeutic potential of PEDF with a higher safety profile for treating CNV in AMD patients.


Assuntos
Neovascularização de Coroide/terapia , Técnicas de Transferência de Genes , Vetores Genéticos , Retina/virologia , Vírus Sendai/genética , Vírus Sendai/metabolismo , Vírus da Imunodeficiência Símia/genética , Proteínas do Envelope Viral , Animais , Linhagem Celular , Neovascularização de Coroide/virologia , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Lentivirus/genética , Luciferases/genética , Luciferases/metabolismo , Degeneração Macular/genética , Degeneração Macular/metabolismo , Degeneração Macular/terapia , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Epitélio Pigmentado Ocular/metabolismo , Epitélio Pigmentado Ocular/virologia , Retina/metabolismo , Serpinas/genética , Serpinas/metabolismo , Vírus da Imunodeficiência Símia/metabolismo , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
4.
J Clin Microbiol ; 46(6): 2122-4, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18400911

RESUMO

Here we describe for the first time the productive in vitro infection of human retinal pigment epithelial cells by varicella-zoster virus (VZV), resulting in a typical cytopathic effect (CPE) that is characterized by enlarged cells with increased granularity. Depending on the CPE dissemination, high titers of up to 1.6 x 10(6) PFU of cell-free and cryostable VZV/ml can be recovered.


Assuntos
Herpesvirus Humano 3/fisiologia , Epitélio Pigmentado Ocular/virologia , Retina/virologia , Replicação Viral , Adulto , Células Cultivadas , Criopreservação , Efeito Citopatogênico Viral , Fibroblastos/virologia , Herpesvirus Humano 3/patogenicidade , Humanos , Masculino , Pessoa de Meia-Idade , Retina/citologia
5.
J Immunol ; 180(6): 3789-96, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-18322185

RESUMO

The retinal pigment epithelial (RPE) cell is a potent regulatory cell that facilitates normal physiologic processes and plays a critical role in a variety of retinal diseases. We evaluated IFN-beta production in human RPE cells through TLR signaling and investigated the effects of IFN-beta on RPE cells. RPE cells treated with poly(I:C) or infected with an RNA virus produce IFN-beta. Kinetic studies revealed that IFN-beta levels continue to increase over a 48-h period and this was associated with the up-regulation of IRF-7 gene expression, a known positive feedback molecule for IFN-beta production. Microarray analysis revealed that in IFN-beta treated cells, 480 genes of 22,283 genes were up or down-regulated by >2-fold. We hypothesize that IFN-beta induction during TLR signaling in the retina is an immunosuppressive factor produced to limit immunopathologic damage. Cytokine activation of RPE cells results in the production of the chemokines, CXCL9 and CXCL10, and the adhesion molecule, ICAM-1. Pretreatment of RPE cells with IFN-beta resulted in inhibition of ICAM-1 production and elimination of CXCL9 production. This treatment did not alter CXCL10 production. Anti-IFN-beta Ab blocked the inhibitory action of IFN-beta. Real time PCR analysis revealed that IFN-beta treatment inhibited gene expression of sICAM-1 and CXCL9. The results indicate a critical role for RPE cell derived IFN-beta in the down-regulation of CXCL9 and ICAM-1 expression in the retina and suggest that the inhibition of CXCL9 is an immuno-suppressive mechanism that protects the retina from excessive inflammation.


Assuntos
Quimiocina CXCL9/antagonistas & inibidores , Molécula 1 de Adesão Intercelular/metabolismo , Interferon beta/fisiologia , Epitélio Pigmentado Ocular/imunologia , Células Cultivadas , Quimiocina CXCL10/biossíntese , Quimiocina CXCL9/biossíntese , Regulação Viral da Expressão Gênica/imunologia , Humanos , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/fisiologia , Molécula 1 de Adesão Intercelular/biossíntese , Interferon beta/biossíntese , Interferon beta/metabolismo , Cinética , Análise de Sequência com Séries de Oligonucleotídeos , Epitélio Pigmentado Ocular/metabolismo , Epitélio Pigmentado Ocular/patologia , Epitélio Pigmentado Ocular/virologia , Retinite/imunologia , Retinite/patologia , Retinite/prevenção & controle , Vírus da Estomatite Vesicular Indiana/imunologia
6.
Mol Vis ; 14: 471-80, 2008 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-18334957

RESUMO

PURPOSE: To develop an hypoxia-regulated retinal pigment epithelium (RPE)-specific adeno-associated virus (AAV) gene transfer platform that exploits hypoxia as a physiologic trigger for an early antiangiogenic treatment strategy directed at arresting neovascularization in the eye. METHODS: Tissue-specific and hypoxia-regulated expression vectors were constructed with tandem combinations of hypoxia responsive elements and aerobically silenced elements (HRSE) that together induce gene expression in hypoxia and suppress it in normoxia. For RPE-specific expression, the HRSE and a (6x) HRE (hypoxia responsive element) oligomer were ligated upstream of the Rpe65 promoter in a pGL3 firefly luciferase plasmid (pGL3-HRSE-6xHRE-Rpe65). The cell specificity of this novel hybrid promoter was tested in human RPE (ARPE-19), human glioblastoma, rat C6 glioma, mouse hippocampal neurons, and human embryonic kidney cell lines. Expression of all cell types in normoxia, and following 40 h hypoxia, was analyzed by dual luciferase assay. After confirmation of its tissue-specificity and hypoxia-inducibility, the hybrid promoter construct was integrated into a replication-deficient AAV delivery system and tested for cell-selectivity and hypoxia-inducible green fluorescent protein (GFP) reporter expression. RESULTS: The HRSE-6xHRE-Rpe65 promoter was highly selective for RPE cells, strongly induced in hypoxia, and similar in activation strength to the cytomegalovirus (CMV) promoter. The AAV.HRSE.6xHRE.Rpe65 vector induced robust GFP expression in hypoxic ARPE-19 cells, but elicited no GFP expression in hypoxia in other cell types or in normoxic ARPE-19 cells. CONCLUSIONS: The hypoxia-regulated, aerobically-silenced RPE-targeting vector forms a platform for focal autoregulated delivery of antiangiogenic agents in hypoxic regions of the RPE. Such autoinitiated therapy provides a means for early intervention in choroidal neovascularization, when it is most sensitive to inhibition.


Assuntos
Dependovirus/genética , Vetores Genéticos/genética , Epitélio Pigmentado Ocular/virologia , Transdução Genética/métodos , Animais , Hipóxia Celular , Linhagem Celular , Proteínas do Olho/genética , Inativação Gênica , Proteínas de Fluorescência Verde/metabolismo , Humanos , Luciferases/metabolismo , Camundongos , Especificidade de Órgãos , Elementos de Resposta/genética , Transfecção
7.
Proc Natl Acad Sci U S A ; 104(50): 20037-42, 2007 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-18077432

RESUMO

Human cytomegalovirus infects multiple cell types, including fibroblasts and epithelial cells. It penetrates fibroblasts by fusion at the cell surface but is endocytosed into epithelial cells. In this report, we demonstrate by electron microscopy that the virus uses two different routes to enter retinal pigmented epithelial cells, depending on the cell type in which the infecting virus was produced. Virus produced in epithelial cells preferentially fuses with the plasma membrane, whereas fibroblast-derived virus mostly enters by receptor-mediated endocytosis. Treatment of epithelial cells with agents that block endosome acidification inhibited infection by virus produced in fibroblasts but had only a modest effect on infection by virus from epithelial cells. Epithelial cell-generated virions had higher intrinsic "fusion-from-without" activity than fibroblast-generated particles, and the two virus preparations triggered different cellular signaling responses, as evidenced by markedly different transcriptional profiles. We propose that the cell type in which a human cytomegalovirus particle is produced likely influences its subsequent spread and its contribution to pathogenesis.


Assuntos
Citomegalovirus/fisiologia , Endocitose/fisiologia , Células Epiteliais/virologia , Epitélio Pigmentado Ocular/virologia , Retina/virologia , Internalização do Vírus , Linhagem Celular , Células Cultivadas , Fibroblastos/fisiologia , Fibroblastos/virologia , Humanos , Epitélio Pigmentado Ocular/citologia , Epitélio Pigmentado Ocular/fisiologia , Retina/citologia , Retina/fisiologia
8.
Microbes Infect ; 8(8): 2236-44, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16782382

RESUMO

Cytomegalovirus (CMV) retinitis is characterized by alterations in retinal cell function and host responses to virus replication. The goal of this study was to evaluate the induction of cyclooxygenase-2 (COX-2) and prostaglandin (PGE) in CMV infected human retinal pigment epithelial (RPE) cells and to determine their effect on virus replication. CMV immediate early (IE) protein and COX-2 proteins were identified in RPE cells in retinal tissue sections from patients with CMV retinitis. COX-2 mRNA and protein were induced after CMV infection of human RPE cell cultures. CMV infection of RPE cells induced translocation of NF-kappaB from the cytoplasm to the nucleus. PGE1 and PGE2 were significantly (p<0.001) increased in human RPE cell cultures infected with CMV. Inhibition of CMV IE gene by antisense oligonucleotides abrogated induction of mRNA for COX-2 and protein synthesis of COX-2 and PGE2. PGE enhanced CMV plaque formation and real time PCR analysis revealed that PGE treatment significantly increased CMV DNA copy numbers. These studies demonstrate that when CMV replicates within human RPE cells, COX-2 induction augments virus replication via the PGE pathway. The induction of COX-2 and PGE during retinal CMV infection may augment virus replication and alter a variety of retinal physiological responses.


Assuntos
Ciclo-Oxigenase 2/biossíntese , Citomegalovirus/fisiologia , Epitélio Pigmentado Ocular/virologia , Prostaglandinas/biossíntese , Replicação Viral , Núcleo Celular/química , Células Cultivadas , Ciclo-Oxigenase 2/genética , Citomegalovirus/crescimento & desenvolvimento , Retinite por Citomegalovirus/patologia , Retinite por Citomegalovirus/virologia , Citoplasma/química , DNA Viral/análise , Expressão Gênica , Humanos , Proteínas Imediatamente Precoces/análise , Proteínas Imediatamente Precoces/antagonistas & inibidores , Imuno-Histoquímica , Microscopia de Fluorescência , NF-kappa B/metabolismo , Oligonucleotídeos Antissenso/genética , RNA Mensageiro/análise , RNA Mensageiro/genética , Retina/química , Ensaio de Placa Viral , Proteínas Virais/análise
9.
J Virol ; 80(7): 3167-79, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16537585

RESUMO

Herpes simplex virus (HSV) glycoprotein heterodimer gE/gI is necessary for virus spread in epithelial and neuronal tissues. Deletion of the relatively large gE cytoplasmic (CT) domain abrogates the ability of gE/gI to mediate HSV spread. The gE CT domain is required for the sorting of gE/gI to the trans-Golgi network (TGN) in early stages of virus infection, and there are several recognizable TGN sorting motifs grouped near the center of this domain. Late in HSV infection, gE/gI, other viral glycoproteins, and enveloped virions redistribute from the TGN to epithelial cell junctions, and the gE CT domain is also required for this process. Without the gE CT domain, newly enveloped virions are directed to apical surfaces instead of to cell junctions. We hypothesized that the gE CT domain promotes virus envelopment into TGN subdomains from which nascent enveloped virions are sorted to cell junctions, a process that enhances cell-to-cell spread. To characterize elements of the gE CT domain involved in intracellular trafficking and cell-to-cell spread, we constructed a panel of truncation mutants. Specifically, these mutants were used to address whether sorting to the TGN and redistribution to cell junctions are necessary, and sufficient, for gE/gI to promote cell-to-cell spread. gE-519, lacking 32 C-terminal residues, localized normally to the TGN early in infection and then trafficked to cell junctions at late times and mediated virus spread. By contrast, mutants gE-495 (lacking 56 C-terminal residues) and gE-470 (lacking 81 residues) accumulated in the TGN but did not traffic to cell junctions and did not mediate cell-to-cell spread. A fourth mutant, gE-448 (lacking most of the CT domain), did not localize to cell junctions and did not mediate virus spread. Therefore, the capacity of gE/gI to promote cell-cell spread requires early localization to the TGN, but this is not sufficient for virus spread. Additionally, gE CT sequences between residues 495 and 519, which contain no obvious cell sorting motifs, are required to promote gE/gI traffic to cell junctions and cell-to-cell spread.


Assuntos
Herpesvirus Humano 1/fisiologia , Junções Intercelulares/metabolismo , Proteínas do Envelope Viral/metabolismo , Rede trans-Golgi/metabolismo , Animais , Transporte Biológico , Linhagem Celular , Chlorocebus aethiops , Herpesvirus Humano 1/patogenicidade , Humanos , Junções Intercelulares/virologia , Queratinócitos/citologia , Queratinócitos/metabolismo , Queratinócitos/virologia , Microscopia de Fluorescência , Modelos Biológicos , Mutagênese , Epitélio Pigmentado Ocular/citologia , Epitélio Pigmentado Ocular/metabolismo , Epitélio Pigmentado Ocular/virologia , Fatores de Tempo , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Rede trans-Golgi/virologia
10.
Invest Ophthalmol Vis Sci ; 47(4): 1510-5, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16565386

RESUMO

PURPOSE: To examine whether human T-cell leukemia virus type 1 (HTLV-1) could infect a human retinal pigment epithelial (RPE) cell line, ARPE-19, in vitro and to investigate its regulation. METHODS: A coculture system with ARPE-19 and irradiated cells of an HTLV-1-producing T-cell line, MT2 was used to determine the permissivity of RPE to HTLV-1 infection in vitro. The susceptibility to HTLV-1 was assessed by detection of viral DNA using the polymerase chain reaction (PCR), viral mRNA transcripts with reverse transcription PCR (RT-PCR) and viral antigen by immunofluorescence staining. An HTLV-1 Tax-activated HTLV-LTR-luciferase reporter assay was developed to measure viral infection quantitatively. The ICAM-1 expression on cocultured ARPE-19 cells was detected by flow cytometry and an ICAM-1-neutralizing antibody was used to test ICAM-1's role in the HTLV-1 infection of ARPE-19 cells. The regulation of HTLV-1 infection was investigated by culturing ARPE-19 cells with proinflammatory cytokines. RESULTS: HTLV-1 infected ARPE-19 cells in vitro. The infection correlated with elevated expression of intercellular adhesion molecule (ICAM)-1 on the surface of ARPE-19 cells. ICAM-1-neutralizing antibody dramatically inhibited viral infection. Furthermore, proinflammatory cytokines dramatically suppressed HTLV-1 viral infection. CONCLUSIONS: The tropism of HTLV-1 to retinal pigment epithelium could provide an explanation for the pathogenesis of HTLV-1-related ophthalmic diseases. A better understanding of specific roles of proinflammatory cytokines in the development of ophthalmic diseases may be beneficial for treatment.


Assuntos
Anticorpos Bloqueadores/farmacologia , Vírus Linfotrópico T Tipo 1 Humano/fisiologia , Molécula 1 de Adesão Intercelular/imunologia , Epitélio Pigmentado Ocular/virologia , Linhagem Celular , Técnicas de Cocultura , Citocinas/farmacologia , DNA Viral/análise , Citometria de Fluxo , Regulação Viral da Expressão Gênica/fisiologia , Antígenos HTLV-I/metabolismo , Humanos , Imuno-Histoquímica , Epitélio Pigmentado Ocular/efeitos dos fármacos , Epitélio Pigmentado Ocular/metabolismo , Provírus/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/virologia , Transfecção
11.
Curr Eye Res ; 31(2): 191-8, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16500770

RESUMO

PURPOSE: To test if nicotine counteracts the dampening effect of human cytomegalovirus (HCMV) infection of NF-kappaB in retinal pigment epithelial (RPE) cells, thereby increasing the permissiveness of RPE cells for HCMV replication. METHODS: Human ARPE-19 cells were transfected with NF-kappaB luciferase DNA, inoculated with HCMV at 24 hr post-transfection, and maintained in the absence or presence of a physiologic dose of nicotine at 1 hr prior to HCMV inoculation. RESULTS: Whereas HCMV-infected ARPE-19 cells without nicotine treatment showed a dramatic decrease in NF-kappaB levels, nicotine treatment reduced this decrease but did not abolish it completely. Nicotine treatment of uninfected ARPE-19 cells had no effect on baseline NF-kappaB levels. CONCLUSIONS: Treatment of HCMV-infected ARPE-19 cells with nicotine at a physiologic dose dampened the downregulation of NF-kappaB observed in HCMV-infected ARPE-19 cells without nicotine treatment. We conclude that nicotine can serve as a cofactor to stimulate productive, lytic replication of HCMV.


Assuntos
Citomegalovirus/fisiologia , Expressão Gênica/fisiologia , NF-kappa B/genética , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Epitélio Pigmentado Ocular/efeitos dos fármacos , Epitélio Pigmentado Ocular/virologia , Western Blotting , Linhagem Celular , Humanos , NF-kappa B/metabolismo , Epitélio Pigmentado Ocular/metabolismo , Transfecção , Replicação Viral
12.
Invest Ophthalmol Vis Sci ; 47(2): 645-51, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16431963

RESUMO

PURPOSE: In addition to neuroinvasive disease, West Nile virus (WNV) infection is frequently associated with self-limiting chorioretinitis and vitritis. However, the mechanisms of ophthalmic WNV infection are rarely investigated, in part because of the lack of reliable in vitro models. The authors therefore established the first model of ocular WNV infection and investigated interaction of WNV with IFN signal-transduction mechanisms. METHODS: Human retinal pigment epithelial (RPE) cells were infected with WNV strain NY385-99 at a multiplicity of infection of 5. Virus replication was evaluated by virus titers at different times after infection. The susceptibility of RPE cells to WNV infection was confirmed by transmission electron microscopy. IFN-beta expression was assessed by quantitative real-time PCR and by measurements of antiviral activity in cell culture supernatants. IFN signaling was evaluated by phosphorylation of transducer and activator of transcription 1 and 2 (STAT1/2) proteins, with immunoblot analysis. RESULTS: RPE cells appeared to be highly sensitive to WNV infection. Maximum viral titers were found 24 hours after infection, followed by a continuous decline during the course of infection. WNV infection of RPE cells was followed by increased IFN-beta expression associated with IFN signaling and subsequent inhibition of WNV replication. CONCLUSIONS: In this study, the first cell culture model of ophthalmic WNV infection was developed and characterized in RPE cells, and the molecular mechanisms of WNV infection were studied. The data suggest that WNV induces a general antiviral state in RPE cells. This general antiviral state correlates with WNV-induced IFN signaling in retinal cells.


Assuntos
Interferon beta/biossíntese , Epitélio Pigmentado Ocular/virologia , Transdução de Sinais/fisiologia , Vírus do Nilo Ocidental/fisiologia , Animais , Western Blotting , Técnicas de Cultura de Células , Chlorocebus aethiops , Humanos , Interferon beta/genética , Microscopia Eletrônica de Transmissão , Modelos Biológicos , Fosforilação , Epitélio Pigmentado Ocular/metabolismo , Epitélio Pigmentado Ocular/ultraestrutura , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT2/metabolismo , Células Vero , Replicação Viral/fisiologia , Vírus do Nilo Ocidental/ultraestrutura
13.
Invest Ophthalmol Vis Sci ; 47(2): 745-52, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16431976

RESUMO

PURPOSE: To determine whether adenoassociated virus (AAV) vectors transduced into iris pigment epithelial (IPE) cells and transplanted into the subretinal space of rats will transfer the AAV genome to the host cells and whether the vectors are disseminated systemically. METHODS: Recombinant (r)AAV was transduced into rat IPE cells and transplanted into the subretinal space of rats. For the control, rAAVs alone were injected subretinally. The transplanted IPE cells were detected by LacZ staining. Immunohistochemistry, electron microscopy, electroretinography, and fluorescein-dextran angiography were performed. DNA was extracted from various organs and blood and examined for the AAV genome by polymerase chain reaction. RESULTS: No toxicity from rAAV transduction was observed in vitro. LacZ was expressed in the transplanted cells 1 and 2 weeks after transplantation. At 4 and 12 weeks, fewer transplanted cells were detected than at 1 week, and LacZ expression was occasionally detected at the level of host retinal pigment epithelial (RPE) cells. Expression was also detected in ciliary body epithelial cells. The electroretinograms and fluorescein-dextran angiography were only mildly altered. Significantly lower levels of AAV genome were detected in the organs and blood of rats receiving rAAV-IPE cell transplants than with direct intravenous injection of AAV vectors. CONCLUSIONS: AAV-mediated LacZ was expressed in the transplanted cells after subretinal transplantation, and the transplanted IPE cells may transfer the rAAV to host tissues, such as RPE cells, long after the transplantation. This method of gene delivery did not lead to systemic dissemination of the vectors.


Assuntos
Dependovirus/genética , Vetores Genéticos , Iris/citologia , Epitélio Pigmentado Ocular/transplante , Epitélio Pigmentado Ocular/virologia , Retina/cirurgia , Transdução Genética , Animais , Sobrevivência Celular , Transplante de Células , Células Cultivadas , DNA Viral/análise , Dextranos , Eletrorretinografia , Espaço Extracelular , Fluoresceínas , Genoma Viral , Imuno-Histoquímica , Óperon Lac/fisiologia , Microscopia de Fluorescência , Reação em Cadeia da Polimerase , Ratos , Ratos Long-Evans , Retina/virologia , beta-Galactosidase/metabolismo
14.
J Clin Virol ; 35(4): 478-84, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16388985

RESUMO

BACKGROUND: Human cytomegalovirus (HCMV) replication in epithelial cells is crucial for its pathogenesis. To date, HCMV gene expression has been primarily studied in human foreskin fibroblasts (HFFs), although their importance for HCMV pathogenesis remains unclear. Primary retinal pigment epithelial (RPE) cells are permissive for HCMV. OBJECTIVES: Our objectives were to determine the production of alternatively processed HCMV major immediate-early (MIE) and UL37 RNAs and their essential products in infected, terminally differentiated immortalized RPE (hTERT-RPE) cells. STUDY DESIGN: hTERT-RPE cells were studied because of their notable similarities with primary RPE cells, and because they overcome key limitations of primary cells. hTERT-RPE cells were terminally differentiated in vitro and infected with HCMV. Total RNA or cell proteins were analyzed at various times post-infection. RESULTS: We show for the first time that HCMV-infected, differentiated hTERT-RPE cells produce IE1, IE2, UL37 exon 1 (UL37x1) and UL37 alternatively spliced RNAs, albeit with abundances and kinetics distinct from those observed in HCMV-infected HFFs. IE1-72 was produced in HCMV-infected, differentiated hTERT-RPEs within 24h post-infection (hpi); whereas, IE2-86 and pUL37x1 were produced within 72 hpi. IE2-86 was detected after IE1-72 even though its transcript appeared first. Early/late (pp65) and late (pp28) proteins were produced within 96-120 hpi. CONCLUSIONS: The temporal cascade of HCMV gene expression was observed in infected, differentiated hTERT-RPE cells. Moreover, HCMV IE RNAs are alternatively and accurately processed in differentiated hTERT-RPE cells. However, the delayed temporal expression suggests further regulation of HCMV gene expression at post-transcriptional/translational levels in differentiated hTERT-RPE cells.


Assuntos
Diferenciação Celular , Citomegalovirus/patogenicidade , Regulação Viral da Expressão Gênica , Epitélio Pigmentado Ocular/citologia , Epitélio Pigmentado Ocular/virologia , Processamento Alternativo , Linhagem Celular , Citomegalovirus/genética , Citomegalovirus/crescimento & desenvolvimento , Citomegalovirus/metabolismo , Células Epiteliais/virologia , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , RNA Viral/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
15.
Invest Ophthalmol Vis Sci ; 46(9): 3451-7, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16123451

RESUMO

PURPOSE: Human cytomegalovirus (HCMV) replication depends on different cellular pathways, including histone acetylation and extracellular-signal regulated kinases 1 and 2 (Erk 1/2). In the present study, the influence of therapeutic valproic acid (VPA) concentrations was investigated on HCMV replication in retinal pigment epithelial (RPE) cells. METHODS: HCMV antigen expression and replication were detected by immunostaining, real-time RT-PCR, and determination of virus titers. Histone acetylation and Erk 1/2 phosphorylation were detected by Western blot. RESULTS: Pretreatment with VPA < or =1 mM enhanced HCMV antigen expression and replication by up to ninefold. In addition to histone deacetylase (HDAC) inhibition, VPA stimulated Erk 1/2 phosphorylation in RPE cells. Investigation of six VPA derivatives revealed that S-2-pentyl-4-pentynoic acid was the only derivative that induced histone hyperacetylation, indicating HDAC inhibition, in the observed concentrations < or =1 mM and that increased HCMV antigen expression. Other derivatives did not enhance HCMV replication in the tested concentrations, although some were found to induce Erk 1/2 phosphorylation. The mitogen-activated protein kinase kinase (MEK) inhibitor PD98059 inhibited VPA-induced Erk 1/2 phosphorylation but did not affect VPA-induced increased HCMV replication. In addition, the structurally nonrelated HDACI trichostatin A enhanced HCMV replication but did not affect Erk 1/2 phosphorylation in RPE cells. CONCLUSIONS: The data demonstrate that VPA stimulates HCMV replication by HDAC inhibition independent of Erk 1/2 phosphorylation in therapeutic concentrations in RPE cells. Therefore, patients at risk of HCMV retinitis who are treated with VPA or other HDAC inhibitors should be carefully monitored.


Assuntos
Citomegalovirus/fisiologia , Inibidores Enzimáticos/farmacologia , Inibidores de Histona Desacetilases , Epitélio Pigmentado Ocular/virologia , Ácido Valproico/farmacologia , Replicação Viral/efeitos dos fármacos , Acetilação , Antígenos Virais/genética , Antígenos Virais/metabolismo , Western Blotting , Células Cultivadas , Histonas/metabolismo , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Epitélio Pigmentado Ocular/enzimologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Invest Ophthalmol Vis Sci ; 46(6): 2047-55, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15914622

RESUMO

PURPOSE: Previous results suggest that retinal neurons are infected early during murine cytomegalovirus (MCMV) infection of the inner retina. The purposes of this study were to identify which retinal neurons are infected and to determine the routes by which MCMV spreads in the retina. METHODS: Immunosuppressed (IS) BALB/c mice were inoculated with 5 x 10(3) PFU of MCMV (k181) through the supraciliary route. Injected eyes were collected at several times after inoculation, sectioned, and examined by electron microscopy and by staining for retinal cell antigens and for MCMV early (EA) or late (LA) antigen. RESULTS: MCMV-infected cells were observed in the choroid and RPE by day 3 after infection (PI) and in the inner retina beginning at day 5 PI. At this time, many horizontal and bipolar cells were MCMV-antigen-positive but only rare MCMV-infected amacrine cells (glycine positive or gamma-aminobutyric acid [GABA] positive) or MCMV-infected ganglion cells (NF positive) were observed in the inner retina. At day 10 PI, most virus-infected cells were glial fibrillary acidic protein (GFAP)- and GABA-positive glia. Virions were observed by electron microscopy in the choroid, RPE, and inner nuclear layer of the retina. Although virions were observed in the endothelium of the retinal vessels and the nearby retinal cells, the endothelial cell lining of the retinal vessels remained intact. Both apoptotic cells and necrotic cells were seen in the inner retina. CONCLUSIONS: In the inner retina, horizontal and bipolar cells were the early (< or = day 7 PI) targets of MCMV infection. Virus spread from the RPE and the photoreceptor layer to the inner retina through infected Muller cells and within the inner retina horizontally through infected horizontal cells.


Assuntos
Infecções Oculares Virais/virologia , Infecções por Herpesviridae/virologia , Muromegalovirus/patogenicidade , Neurônios/virologia , Retina/virologia , Retinite/virologia , Animais , Antígenos Virais/análise , Calbindinas , Corioide/microbiologia , Corioide/ultraestrutura , Infecções Oculares Virais/patologia , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Proteína Glial Fibrilar Ácida/metabolismo , Infecções por Herpesviridae/patologia , Camundongos , Camundongos Endogâmicos BALB C , Epitélio Pigmentado Ocular/ultraestrutura , Epitélio Pigmentado Ocular/virologia , Retinite/patologia , Proteína G de Ligação ao Cálcio S100/metabolismo , Vírion/ultraestrutura , Ácido gama-Aminobutírico/metabolismo
17.
Eur J Neurosci ; 21(1): 161-72, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15654853

RESUMO

The aim of this study was to establish synapses between a transplant and a degenerated retina. To tackle this difficult task, a little-known but well-established CNS method was chosen: trans-synaptic pseudorabies virus (PRV) tracing. Sheets of E19 rat retina with or without retinal pigment epithelium (RPE) were transplanted to the subretinal space in 33 Royal College of Surgeons (RCS) and transgenic s334ter-5 rats with retinal degeneration. Several months later, PRV-BaBlu (expressing E. colibeta-galactosidase) or PRV-Bartha was injected into an area of the exposed superior colliculus (SC), topographically corresponding to the transplant placement in the retina. Twenty normal rats served as controls. After survival times of 1-5 days, retinas were examined for virus by X-gal histochemistry, immunohistochemistry and electron microscopy. In normal controls, virus was first seen in retinal ganglion cells and Müller glia after 1-1.5 days, and had spread to all retinal layers after 2-3 days. Virus-labeled cells were found in 16 of 19 transplants where the virus injection had retrogradely labeled the topographically correct transplant area of the host retina. Electron microscopically, enveloped and nonenveloped virus could clearly be detected in infected cells. Enveloped virus was found only in neurons. Infected glial cells contained only nonenveloped virus. Neurons in retinal transplants are labeled after PRV injection into the host brain, indicating synaptic connectivity between transplants and degenerated host retinas. This study provides evidence that PRV spreads in the retina as in other parts of the CNS and is useful to outline transplant-host circuitry.


Assuntos
Encéfalo/virologia , Herpesvirus Suídeo 1/isolamento & purificação , Epitélio Pigmentado Ocular/transplante , Retina/virologia , Degeneração Retiniana/metabolismo , Sinapses/fisiologia , Animais , Animais Geneticamente Modificados , Encéfalo/metabolismo , Encéfalo/ultraestrutura , Embrião de Mamíferos , Transplante de Tecido Fetal/métodos , Galactosídeos/metabolismo , Herpesvirus Suídeo 1/fisiologia , Imuno-Histoquímica/métodos , Indóis/metabolismo , Camundongos , Microscopia Eletrônica de Transmissão/métodos , Epitélio Pigmentado Ocular/embriologia , Epitélio Pigmentado Ocular/ultraestrutura , Epitélio Pigmentado Ocular/virologia , Ratos , Ratos Endogâmicos , Ratos Long-Evans , Ratos Sprague-Dawley , Retina/metabolismo , Retina/ultraestrutura , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Degeneração Retiniana/cirurgia , Sinapses/ultraestrutura , Sinapses/virologia , Fatores de Tempo
18.
J Virol ; 78(20): 11327-33, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15452253

RESUMO

The expression of lentivirus-transduced enhanced green fluorescent protein (EGFP) was detectable in rabbit retinal pigment epithelium (RPE) within 3 to 5 days after subretinal injection of the vector. Within 2 to 3 weeks, EGFP-expressing cells were eliminated by rejection. In the current experiments, we monitor serum antibody titers for EGFP before and after transduction and determine whether systemic immunosuppression prevents recognition of EGFP by the immune system. While all control rabbits developed antibodies against EFGP and showed signs of rejection, no such evidence was observed with animals which received immunosuppression. One month of systemic immunosuppression permanently prevented rejection of RPE with EGFP expression. Fluorescence has been maintained for more than a year. If a control eye was injected with the same virus after terminating immunosuppression, both eyes showed signs of rejection. The lack of rejection is not due to tolerance but to a failure of the animals to detect the foreign protein. Detection must depend upon a brief window of time after surgery needed to introduce the vector, perhaps related to a concurrent but transient inflammation. This strategy may be useful in managing other types of rejection in the retina.


Assuntos
Rejeição de Enxerto , Terapia de Imunossupressão , Lentivirus/metabolismo , Proteínas Luminescentes/metabolismo , Retina/virologia , Transdução Genética , Animais , Vetores Genéticos , Proteínas de Fluorescência Verde , Lentivirus/genética , Proteínas Luminescentes/genética , Epitélio Pigmentado Ocular/virologia , Coelhos , Retina/metabolismo
19.
Ophthalmologe ; 101(7): 720-4, 2004 Jul.
Artigo em Alemão | MEDLINE | ID: mdl-15309488

RESUMO

BACKGROUND: Influenza A is one type of influenza virus that commonly causes acute respiratory illness. Outbreaks of influenza occur every year. Major antigenic variations preclude permanent immunity in the population. Often signs of conjunctivitis or photophobia are common during acute infection. Posterior uveitis is very rare. PATIENT: A young lady with a diagnosed anterior uveitis was sent for further evaluation to the eye department with a known history of flu. RESULTS: This patient had a severe ocular manifestation of influenza A infection. There was bilateral panuveitis with keratic precipitates, cells and flare, and an impressive retinopathy in both eyes. Serology was positive for influenza A. CONCLUSION: The course of an influenza A infection is usually uncomplicated. Severe affection of the choriocapillaris results in a complicated post-influenza retinal pigmentary degeneration. Treatment with amantadine and therapy with hyperimmunoglobulins seem to be useful.


Assuntos
Amantadina/uso terapêutico , Antivirais/uso terapêutico , Imunoglobulinas Intravenosas/uso terapêutico , Vírus da Influenza A/patogenicidade , Influenza Humana/virologia , Pan-Uveíte/virologia , Adulto , Atrofia , Quimioterapia Combinada , Feminino , Seguimentos , Humanos , Vírus da Influenza A/efeitos dos fármacos , Influenza Humana/diagnóstico , Influenza Humana/tratamento farmacológico , Pan-Uveíte/diagnóstico , Pan-Uveíte/tratamento farmacológico , Epitélio Pigmentado Ocular/patologia , Epitélio Pigmentado Ocular/virologia
20.
Invest Ophthalmol Vis Sci ; 45(9): 2906-14, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15326101

RESUMO

PURPOSE: The transactivator protein Tat encoded by the human immunodeficiency virus-1 (HIV-1) genome reduces glutathione levels in mammalian cells. Because the retina contains large amounts of glutathione, a study was undertaken to determine the influence of Tat on glutathione levels, gamma-glutamyl transpeptidase activity, and the expression and activity of the cystine-glutamate transporter xc- in the human retinal pigment epithelial cell line ARPE-19 and in retina from Tat-transgenic mice. METHODS: The transport function of xc- was measured as glutamate uptake in the absence of Na+. mRNA levels for xCT and 4F2hc, the two subunits of system xc-, were monitored by RT-PCR and Northern blot and protein levels by Western blot. The expression pattern of xCT and 4F2hc in the mouse retina was analyzed by immunofluorescence. RESULTS: Expression of Tat in ARPE-19 cells led to a decrease in glutathione levels and an increase in gamma-glutamyl transpeptidase activity. The transport function of xc- was upregulated, and this increase was accompanied by increases in the levels of mRNAs for xCT and 4F2hc and in corresponding protein levels. The influence of Tat on the expression of xc- was independent of the cellular status of glutathione. Most of these findings were confirmed in the retina of Tat-transgenic mice. CONCLUSIONS: Expression of HIV-1 Tat in the retina decreases glutathione levels and increases gamma-glutamyl transpeptidase activity. Tat also upregulates the expression of system xc-. Glutathione levels may be decreased and the expression of xc- enhanced in the retina of patients with HIV-1 infection, leading to oxidative stress and excitotoxicity.


Assuntos
Sistema y+ de Transporte de Aminoácidos/biossíntese , Produtos do Gene tat/fisiologia , HIV-1/metabolismo , Epitélio Pigmentado Ocular/virologia , Sistema y+ de Transporte de Aminoácidos/genética , Sistema y+ de Transporte de Aminoácidos/metabolismo , Animais , Linhagem Celular , Cadeia Pesada da Proteína-1 Reguladora de Fusão/genética , Cadeia Pesada da Proteína-1 Reguladora de Fusão/metabolismo , Produtos do Gene tat/genética , Glutamato-Cisteína Ligase/metabolismo , Glutationa/metabolismo , Homeostase , Humanos , Isoenzimas/metabolismo , Cinética , Camundongos , Camundongos Transgênicos/genética , Epitélio Pigmentado Ocular/citologia , Epitélio Pigmentado Ocular/metabolismo , RNA Mensageiro/metabolismo , Retina/metabolismo , Regulação para Cima , gama-Glutamiltransferase/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA