Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Front Immunol ; 12: 644153, 2021.
Article in English | MEDLINE | ID: mdl-33968035

ABSTRACT

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.


Subject(s)
Epithelial Cells , Gene Expression Regulation, Viral/immunology , Pigment Epithelium of Eye , RNA, Viral/immunology , Zika Virus Infection , Zika Virus/immunology , Cells, Cultured , Epithelial Cells/immunology , Epithelial Cells/pathology , Epithelial Cells/virology , Genome, Viral/immunology , Humans , Iris/immunology , Iris/pathology , Iris/virology , Pigment Epithelium of Eye/immunology , Pigment Epithelium of Eye/pathology , Pigment Epithelium of Eye/virology , Zika Virus Infection/immunology , Zika Virus Infection/pathology
2.
PLoS Negl Trop Dis ; 14(4): e0008223, 2020 04.
Article in English | MEDLINE | ID: mdl-32324736

ABSTRACT

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.


Subject(s)
Flavivirus/pathogenicity , Models, Biological , Nervous System/virology , Animals , Brain/virology , Disease Models, Animal , Endothelial Cells/virology , Epithelial Cells/virology , Flavivirus/growth & development , Humans , Mice , Pigment Epithelium of Eye/virology , Spinal Cord/virology
3.
Hum Gene Ther ; 21(2): 199-209, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19778186

ABSTRACT

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.


Subject(s)
Choroidal Neovascularization/therapy , Gene Transfer Techniques , Genetic Vectors , Retina/virology , Sendai virus/genetics , Sendai virus/metabolism , Simian Immunodeficiency Virus/genetics , Viral Envelope Proteins , Animals , Cell Line , Choroidal Neovascularization/virology , Eye Proteins/genetics , Eye Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Lentivirus/genetics , Luciferases/genetics , Luciferases/metabolism , Macular Degeneration/genetics , Macular Degeneration/metabolism , Macular Degeneration/therapy , Mice , Mice, Inbred C57BL , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Pigment Epithelium of Eye/metabolism , Pigment Epithelium of Eye/virology , Retina/metabolism , Serpins/genetics , Serpins/metabolism , Simian Immunodeficiency Virus/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
4.
J Clin Microbiol ; 46(6): 2122-4, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18400911

ABSTRACT

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.


Subject(s)
Herpesvirus 3, Human/physiology , Pigment Epithelium of Eye/virology , Retina/virology , Virus Replication , Adult , Cells, Cultured , Cryopreservation , Cytopathogenic Effect, Viral , Fibroblasts/virology , Herpesvirus 3, Human/pathogenicity , Humans , Male , Middle Aged , Retina/cytology
5.
J Immunol ; 180(6): 3789-96, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18322185

ABSTRACT

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.


Subject(s)
Chemokine CXCL9/antagonists & inhibitors , Intercellular Adhesion Molecule-1/metabolism , Interferon-beta/physiology , Pigment Epithelium of Eye/immunology , Cells, Cultured , Chemokine CXCL10/biosynthesis , Chemokine CXCL9/biosynthesis , Gene Expression Regulation, Viral/immunology , Humans , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Intercellular Adhesion Molecule-1/biosynthesis , Interferon-beta/biosynthesis , Interferon-beta/metabolism , Kinetics , Oligonucleotide Array Sequence Analysis , Pigment Epithelium of Eye/metabolism , Pigment Epithelium of Eye/pathology , Pigment Epithelium of Eye/virology , Retinitis/immunology , Retinitis/pathology , Retinitis/prevention & control , Vesicular stomatitis Indiana virus/immunology
6.
Mol Vis ; 14: 471-80, 2008 Mar 07.
Article in English | MEDLINE | ID: mdl-18334957

ABSTRACT

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.


Subject(s)
Dependovirus/genetics , Genetic Vectors/genetics , Pigment Epithelium of Eye/virology , Transduction, Genetic/methods , Animals , Cell Hypoxia , Cell Line , Eye Proteins/genetics , Gene Silencing , Green Fluorescent Proteins/metabolism , Humans , Luciferases/metabolism , Mice , Organ Specificity , Response Elements/genetics , Transfection
7.
Proc Natl Acad Sci U S A ; 104(50): 20037-42, 2007 Dec 11.
Article in English | MEDLINE | ID: mdl-18077432

ABSTRACT

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.


Subject(s)
Cytomegalovirus/physiology , Endocytosis/physiology , Epithelial Cells/virology , Pigment Epithelium of Eye/virology , Retina/virology , Virus Internalization , Cell Line , Cells, Cultured , Fibroblasts/physiology , Fibroblasts/virology , Humans , Pigment Epithelium of Eye/cytology , Pigment Epithelium of Eye/physiology , Retina/cytology , Retina/physiology
8.
Microbes Infect ; 8(8): 2236-44, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16782382

ABSTRACT

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.


Subject(s)
Cyclooxygenase 2/biosynthesis , Cytomegalovirus/physiology , Pigment Epithelium of Eye/virology , Prostaglandins/biosynthesis , Virus Replication , Cell Nucleus/chemistry , Cells, Cultured , Cyclooxygenase 2/genetics , Cytomegalovirus/growth & development , Cytomegalovirus Retinitis/pathology , Cytomegalovirus Retinitis/virology , Cytoplasm/chemistry , DNA, Viral/analysis , Gene Expression , Humans , Immediate-Early Proteins/analysis , Immediate-Early Proteins/antagonists & inhibitors , Immunohistochemistry , Microscopy, Fluorescence , NF-kappa B/metabolism , Oligonucleotides, Antisense/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics , Retina/chemistry , Viral Plaque Assay , Viral Proteins/analysis
9.
Invest Ophthalmol Vis Sci ; 47(4): 1510-5, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16565386

ABSTRACT

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.


Subject(s)
Antibodies, Blocking/pharmacology , Human T-lymphotropic virus 1/physiology , Intercellular Adhesion Molecule-1/immunology , Pigment Epithelium of Eye/virology , Cell Line , Coculture Techniques , Cytokines/pharmacology , DNA, Viral/analysis , Flow Cytometry , Gene Expression Regulation, Viral/physiology , HTLV-I Antigens/metabolism , Humans , Immunohistochemistry , Pigment Epithelium of Eye/drug effects , Pigment Epithelium of Eye/metabolism , Proviruses/genetics , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/virology , Transfection
10.
J Virol ; 80(7): 3167-79, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16537585

ABSTRACT

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.


Subject(s)
Herpesvirus 1, Human/physiology , Intercellular Junctions/metabolism , Viral Envelope Proteins/metabolism , trans-Golgi Network/metabolism , Animals , Biological Transport , Cell Line , Chlorocebus aethiops , Herpesvirus 1, Human/pathogenicity , Humans , Intercellular Junctions/virology , Keratinocytes/cytology , Keratinocytes/metabolism , Keratinocytes/virology , Microscopy, Fluorescence , Models, Biological , Mutagenesis , Pigment Epithelium of Eye/cytology , Pigment Epithelium of Eye/metabolism , Pigment Epithelium of Eye/virology , Time Factors , Vero Cells , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , trans-Golgi Network/virology
11.
Curr Eye Res ; 31(2): 191-8, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16500770

ABSTRACT

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.


Subject(s)
Cytomegalovirus/physiology , Gene Expression/physiology , NF-kappa B/genetics , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Pigment Epithelium of Eye/drug effects , Pigment Epithelium of Eye/virology , Blotting, Western , Cell Line , Humans , NF-kappa B/metabolism , Pigment Epithelium of Eye/metabolism , Transfection , Virus Replication
12.
J Clin Virol ; 35(4): 478-84, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16388985

ABSTRACT

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.


Subject(s)
Cell Differentiation , Cytomegalovirus/pathogenicity , Gene Expression Regulation, Viral , Pigment Epithelium of Eye/cytology , Pigment Epithelium of Eye/virology , Alternative Splicing , Cell Line , Cytomegalovirus/genetics , Cytomegalovirus/growth & development , Cytomegalovirus/metabolism , Epithelial Cells/virology , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , RNA, Viral/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
13.
Invest Ophthalmol Vis Sci ; 47(2): 645-51, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16431963

ABSTRACT

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.


Subject(s)
Interferon-beta/biosynthesis , Pigment Epithelium of Eye/virology , Signal Transduction/physiology , West Nile virus/physiology , Animals , Blotting, Western , Cell Culture Techniques , Chlorocebus aethiops , Humans , Interferon-beta/genetics , Microscopy, Electron, Transmission , Models, Biological , Phosphorylation , Pigment Epithelium of Eye/metabolism , Pigment Epithelium of Eye/ultrastructure , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , Vero Cells , Virus Replication/physiology , West Nile virus/ultrastructure
14.
Invest Ophthalmol Vis Sci ; 47(2): 745-52, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16431976

ABSTRACT

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.


Subject(s)
Dependovirus/genetics , Genetic Vectors , Iris/cytology , Pigment Epithelium of Eye/transplantation , Pigment Epithelium of Eye/virology , Retina/surgery , Transduction, Genetic , Animals , Cell Survival , Cell Transplantation , Cells, Cultured , DNA, Viral/analysis , Dextrans , Electroretinography , Extracellular Space , Fluoresceins , Genome, Viral , Immunohistochemistry , Lac Operon/physiology , Microscopy, Fluorescence , Polymerase Chain Reaction , Rats , Rats, Long-Evans , Retina/virology , beta-Galactosidase/metabolism
15.
Invest Ophthalmol Vis Sci ; 46(9): 3451-7, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16123451

ABSTRACT

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.


Subject(s)
Cytomegalovirus/physiology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Pigment Epithelium of Eye/virology , Valproic Acid/pharmacology , Virus Replication/drug effects , Acetylation , Antigens, Viral/genetics , Antigens, Viral/metabolism , Blotting, Western , Cells, Cultured , Histones/metabolism , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Pigment Epithelium of Eye/enzymology , Reverse Transcriptase Polymerase Chain Reaction
16.
Invest Ophthalmol Vis Sci ; 46(6): 2047-55, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15914622

ABSTRACT

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.


Subject(s)
Eye Infections, Viral/virology , Herpesviridae Infections/virology , Muromegalovirus/pathogenicity , Neurons/virology , Retina/virology , Retinitis/virology , Animals , Antigens, Viral/analysis , Calbindins , Choroid/microbiology , Choroid/ultrastructure , Eye Infections, Viral/pathology , Female , Fluorescent Antibody Technique, Indirect , Glial Fibrillary Acidic Protein/metabolism , Herpesviridae Infections/pathology , Mice , Mice, Inbred BALB C , Pigment Epithelium of Eye/ultrastructure , Pigment Epithelium of Eye/virology , Retinitis/pathology , S100 Calcium Binding Protein G/metabolism , Virion/ultrastructure , gamma-Aminobutyric Acid/metabolism
17.
Eur J Neurosci ; 21(1): 161-72, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15654853

ABSTRACT

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.


Subject(s)
Brain/virology , Herpesvirus 1, Suid/isolation & purification , Pigment Epithelium of Eye/transplantation , Retina/virology , Retinal Degeneration/metabolism , Synapses/physiology , Animals , Animals, Genetically Modified , Brain/metabolism , Brain/ultrastructure , Embryo, Mammalian , Fetal Tissue Transplantation/methods , Galactosides/metabolism , Herpesvirus 1, Suid/physiology , Immunohistochemistry/methods , Indoles/metabolism , Mice , Microscopy, Electron, Transmission/methods , Pigment Epithelium of Eye/embryology , Pigment Epithelium of Eye/ultrastructure , Pigment Epithelium of Eye/virology , Rats , Rats, Inbred Strains , Rats, Long-Evans , Rats, Sprague-Dawley , Retina/metabolism , Retina/ultrastructure , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Degeneration/surgery , Synapses/ultrastructure , Synapses/virology , Time Factors
18.
J Virol ; 78(20): 11327-33, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15452253

ABSTRACT

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.


Subject(s)
Graft Rejection , Immunosuppression Therapy , Lentivirus/metabolism , Luminescent Proteins/metabolism , Retina/virology , Transduction, Genetic , Animals , Genetic Vectors , Green Fluorescent Proteins , Lentivirus/genetics , Luminescent Proteins/genetics , Pigment Epithelium of Eye/virology , Rabbits , Retina/metabolism
19.
Invest Ophthalmol Vis Sci ; 45(9): 2906-14, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15326101

ABSTRACT

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.


Subject(s)
Amino Acid Transport System y+/biosynthesis , Gene Products, tat/physiology , HIV-1/metabolism , Pigment Epithelium of Eye/virology , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Animals , Cell Line , Fusion Regulatory Protein 1, Heavy Chain/genetics , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Gene Products, tat/genetics , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Homeostasis , Humans , Isoenzymes/metabolism , Kinetics , Mice , Mice, Transgenic/genetics , Pigment Epithelium of Eye/cytology , Pigment Epithelium of Eye/metabolism , RNA, Messenger/metabolism , Retina/metabolism , Up-Regulation , gamma-Glutamyltransferase/metabolism , tat Gene Products, Human Immunodeficiency Virus
20.
Ophthalmologe ; 101(7): 720-4, 2004 Jul.
Article in German | MEDLINE | ID: mdl-15309488

ABSTRACT

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.


Subject(s)
Amantadine/therapeutic use , Antiviral Agents/therapeutic use , Immunoglobulins, Intravenous/therapeutic use , Influenza A virus/pathogenicity , Influenza, Human/virology , Panuveitis/virology , Adult , Atrophy , Drug Therapy, Combination , Female , Follow-Up Studies , Humans , Influenza A virus/drug effects , Influenza, Human/diagnosis , Influenza, Human/drug therapy , Panuveitis/diagnosis , Panuveitis/drug therapy , Pigment Epithelium of Eye/pathology , Pigment Epithelium of Eye/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...