Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 3780, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38710714

ABSTRACT

Recombinant adeno-associated viruses (rAAVs) have emerged as promising gene therapy vectors due to their proven efficacy and safety in clinical applications. In non-human primates (NHPs), rAAVs are administered via suprachoroidal injection at a higher dose. However, high doses of rAAVs tend to increase additional safety risks. Here, we present a novel AAV capsid (AAVv128), which exhibits significantly enhanced transduction efficiency for photoreceptors and retinal pigment epithelial (RPE) cells, along with a broader distribution across the layers of retinal tissues in different animal models (mice, rabbits, and NHPs) following intraocular injection. Notably, the suprachoroidal delivery of AAVv128-anti-VEGF vector completely suppresses the Grade IV lesions in a laser-induced choroidal neovascularization (CNV) NHP model for neovascular age-related macular degeneration (nAMD). Furthermore, cryo-EM analysis at 2.1 Å resolution reveals that the critical residues of AAVv128 exhibit a more robust advantage in AAV binding, the nuclear uptake and endosome escaping. Collectively, our findings highlight the potential of AAVv128 as a next generation ocular gene therapy vector, particularly using the suprachoroidal delivery route.


Subject(s)
Choroidal Neovascularization , Dependovirus , Genetic Therapy , Genetic Vectors , Retinal Pigment Epithelium , Animals , Dependovirus/genetics , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Genetic Therapy/methods , Mice , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/virology , Choroidal Neovascularization/therapy , Choroidal Neovascularization/genetics , Rabbits , Humans , Gene Transfer Techniques , Macular Degeneration/therapy , Macular Degeneration/genetics , Macular Degeneration/pathology , Disease Models, Animal , Capsid Proteins/genetics , Capsid Proteins/metabolism , Transduction, Genetic , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Mice, Inbred C57BL , Retina/metabolism , Retina/virology , Male , HEK293 Cells
2.
Int J Mol Sci ; 25(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38731826

ABSTRACT

Although Herpes simplex virus type 1 (HSV-1) has been deeply studied, significant gaps remain in the fundamental understanding of HSV-host interactions: our work focused on studying the Infected Cell Protein 27 (ICP27) as an inhibitor of the Absent-in-melanoma-2 (AIM 2) inflammasome pathway, leading to reduced pro-inflammatory cytokines that influence the activation of a protective innate immune response to infection. To assess the inhibition of the inflammasome by the ICP27, hTert-immortalized Retinal Pigment Epithelial cells (hTert-RPE 1) infected with HSV-1 wild type were compared to HSV-1 lacking functional ICP27 (HSV-1∆ICP27) infected cells. The activation of the inflammasome by HSV-1∆ICP27 was demonstrated by quantifying the gene and protein expression of the inflammasome constituents using real-time PCR and Western blot. The detection of the cleavage of the pro-caspase-1 into the active form was performed by using a bioluminescent assay, while the quantification of interleukins 1ß (IL-1ß) and 18 (IL-18)released in the supernatant was quantified using an ELISA assay. The data showed that the presence of the ICP27 expressed by HSV-1 induces, in contrast to HSV-1∆ICP27 vector, a significant downregulation of AIM 2 inflammasome constituent proteins and, consequently, the release of pro-inflammatory interleukins into the extracellular environment reducing an effective response in counteracting infection.


Subject(s)
Cytokines , Herpesvirus 1, Human , Immediate-Early Proteins , Inflammasomes , Retinal Pigment Epithelium , Humans , Inflammasomes/metabolism , Herpesvirus 1, Human/physiology , Cytokines/metabolism , Immediate-Early Proteins/metabolism , Immediate-Early Proteins/genetics , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/virology , Epithelial Cells/metabolism , Epithelial Cells/virology , Cell Line , Herpes Simplex/immunology , Herpes Simplex/metabolism , Herpes Simplex/virology , DNA-Binding Proteins
3.
Genes (Basel) ; 15(4)2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38674337

ABSTRACT

Ebola virus (EBOV) is a highly pathogenic virus that causes a severe illness called Ebola virus disease (EVD). EVD has a high mortality rate and remains a significant threat to public health. Research on EVD pathogenesis has traditionally focused on host transcriptional responses. Limited recent studies, however, have revealed some information on the significance of cellular microRNAs (miRNAs) in EBOV infection and pathogenic mechanisms, but further studies are needed. Thus, this study aimed to identify and validate additional known and novel human miRNAs in EBOV-infected adult retinal pigment epithelial (ARPE) cells and predict their potential roles in EBOV infection and pathogenic mechanisms. We analyzed previously available small RNA-Seq data obtained from ARPE cells and identified 23 upregulated and seven downregulated miRNAs in the EBOV-infected cells; these included two novel miRNAs and 17 additional known miRNAs not previously identified in ARPE cells. In addition to pathways previously identified by others, these miRNAs are associated with pathways and biological processes that include WNT, FoxO, and phosphatidylinositol signaling; these pathways were not identified in the original study. This study thus confirms and expands on the previous study using the same datasets and demonstrates further the importance of human miRNAs in the host response and EVD pathogenesis during infection.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , MicroRNAs , Retinal Pigment Epithelium , Humans , MicroRNAs/genetics , Hemorrhagic Fever, Ebola/genetics , Hemorrhagic Fever, Ebola/virology , Ebolavirus/genetics , Ebolavirus/pathogenicity , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/virology , Retinal Pigment Epithelium/pathology , Cell Line
4.
Mol Biol Rep ; 48(7): 5379-5392, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34282543

ABSTRACT

Resveratrol (RES) is a polyphenol with increasing interest for its inhibitory effects on a wide variety of viruses. Zika virus (ZIKV) is an arbovirus which causes a broad spectrum of ophthalmological manifestations in humans. Currently there is no certified therapy or vaccine to treat it, thus it has become a major global health threat. Retinal pigment epithelium (RPE) is highly permissive and susceptible to ZIKV. This work explored the protective effects of RES on ZIKV-infected human RPE cells. RES treatment resulted in a significant reduction of infectious viral particles in infected male ARPE-19 and female hTERT-RPE1 cells. This protection was positively influenced by the action of RES on mitochondrial dynamics. Also, docking studies predicted that RES has a high affinity for two enzymes of the rate-limiting steps of pyrimidine and purine biosynthesis and viral polymerase. This evidence suggests that RES might be a potential antiviral agent to treat ZIKV-induced ocular abnormalities.


Subject(s)
Antiviral Agents/pharmacology , Resveratrol/pharmacology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/virology , Zika Virus/drug effects , Antiviral Agents/chemistry , Binding Sites , Cell Line , Cell Survival/drug effects , Cells, Cultured , Drug Development , Epithelial Cells/drug effects , Epithelial Cells/virology , Humans , Ligands , Mitochondrial Dynamics/drug effects , Models, Biological , Models, Molecular , Protein Binding , Resveratrol/chemistry , Structure-Activity Relationship , Virus Replication/drug effects , Zika Virus Infection/drug therapy , Zika Virus Infection/pathology , Zika Virus Infection/virology
5.
Biomolecules ; 11(5)2021 05 18.
Article in English | MEDLINE | ID: mdl-34069869

ABSTRACT

Several RNA viruses, including SARS-CoV-2, can infect or use the eye as an entry portal to cause ocular or systemic diseases. Povidone-Iodine (PVP-I) is routinely used during ocular surgeries and eye banking as a cost-effective disinfectant due to its broad-spectrum antimicrobial activity, including against viruses. However, whether PVP-I can exert antiviral activities in virus-infected cells remains elusive. In this study, using Zika (ZIKV) and Chikungunya (CHIKV) virus infection of human corneal and retinal pigment epithelial cells, we report antiviral mechanisms of PVP-I. Our data showed that PVP-I, even at the lowest concentration (0.01%), drastically reduced viral replication in corneal and retinal cells without causing cellular toxicity. Antiviral effects of PVP-I against ZIKV and CHIKV were mediated by direct viral inactivation, thus attenuating the ability of the virus to infect host cells. Moreover, one-minute PVP-I exposure of infected ocular cells drastically reduced viral replication and the production of infectious progeny virions. Furthermore, viral-induced (CHIKV) expression of inflammatory genes (TNF-α, IL-6, IL-8, and IL1ß) were markedly reduced in PVP-I treated corneal epithelial cells. Together, our results demonstrate potent antiviral effects of PVP-I against ZIKV and CHIKV infection of ocular cells. Thus, a low dose of PVP-I can be used during tissue harvesting for corneal transplants to prevent potential transmission of RNA viruses via infected cells.


Subject(s)
Antiviral Agents/pharmacology , Povidone-Iodine/pharmacology , RNA Viruses/physiology , Virus Replication/drug effects , Animals , Cell Line , Chikungunya virus/physiology , Chlorocebus aethiops , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/virology , SARS-CoV-2/physiology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vero Cells , Zika Virus/physiology
6.
Curr Eye Res ; 45(9): 1136-1143, 2020 09.
Article in English | MEDLINE | ID: mdl-31951764

ABSTRACT

PURPOSE: Epstein-Barr virus is a γ-herpes virus that infects primary B cells and can transform infected cells into immortalized lymphoblastoid cell lines (LCL). The role of EBV in malignancies such as Burkitt's lymphoma and nasopharyngeal carcinoma is well understood, however, its role in EBV-infected retinal cells remains poorly understood. Therefore, we investigated the effect of EBV on the growth of retinal cells. METHODS: Previously, we established and reported a cell line model to address the relationship between EBV infection and retinal cell proliferation that used adult retinal pigment epithelium (ARPE-19) and EBV infection. To determine the effect of EBV on ARPE-19 cells, cell death was measured by propidium iodine/annexin V staining and reactive oxygen species (ROS) were measured by FACS, and protein expression was evaluated using western blot analysis. Also, downregulation of LMP1 and NADPH oxidase 4 (NOX4) expression was accomplished using siRNA technology. RESULTS: We found that ROS were dramatically increased in EBV-infected ARPE19 cells (APRE19/EBV) relative to the parental cell line. Additionally, the expression level of NOX4, a main source of ROS, was upregulated by EBV infection. Interestingly, downregulation of LMP1, one of the EBV viral onco-proteins, completely decreased EBV-induced ROS accumulation and the upregulation of NOX4. Treatment with APX-115A, a pan-NOX inhibitor, induced apoptotic cell death of only the EBV-infected ARPE19 cells but not the parental cell line. Pretreatment with z-VAD, a pan-caspase inhibitor, inhibited NOX inhibitor-induced cell death in ARPE19/EBV cells. Furthermore, APX-115A-induced cell death mediated the activation of JNK and ERK. Finally, we confirmed the expression level of NOX4, and APX-115A induced cell death of EBV-infected human primary retina epithelial cells and the activation of JNK and ERK. CONCLUSION: Taken together, these our results suggest that APX-115A could be a therapeutic agent for treating EBV-infected retinal cells or diseases by inhibiting LMP1-NOX4-ROS signaling.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Herpesvirus 4, Human/physiology , NADPH Oxidase 4/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology , Reactive Oxygen Species/metabolism , Retinal Pigment Epithelium/pathology , Blotting, Western , Cell Line , Cell Proliferation , Down-Regulation , Enzyme Inhibitors/pharmacology , Flow Cytometry , Humans , NADPH Oxidases/antagonists & inhibitors , Retinal Pigment Epithelium/enzymology , Retinal Pigment Epithelium/virology , Signal Transduction/drug effects
7.
Gene ; 724: 144157, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31629820

ABSTRACT

Cellular microRNAs are known to modulate the life-cycle of different viruses. Surprisingly, very little data exists on AAV-induced changes to the cellular microRNAome in general and in hepatic and retinal cells, in particular. We reasoned that inducible microRNA in response to recombinant AAV infection may regulate immediate and long-lived cellular responses necessary for the cell's own survival as well as its ability to control several aspects of viral life-cycle. To study this, we performed a global small RNA sequencing analysis in Adeno-associated virus (AAV) serotypes 2 and 3 infected hepatic and retinal cell models. This screen identified multiple differentially expressed microRNAs, in AAV infected Huh-7 and ARPE-19 cells. Among these, one microRNA (miR-4488) was found to be significantly down regulated (-2.24 fold for AAV2 and -3.32 fold for ARPE-19) in AAV infected cells. An enrichment and pathway analysis of miR-4488 predicted its possible effects on gene targets involved in multiple biological processes including cell-cycle regulation, endoplasmic reticulum stress response and lipid-signalling pathways. Moreover, validation studies in miR-4488 mimic or sponge transfected cells revealed modulation of these target pathways in a cell-specific manner. Further studies demonstrated that overexpression of miR-4488, modestly increased gene expression (126-128%) from AAV2 and AAV3 vectors in Huh-7 cells whereas miR-4488 inhibition in ARPE-19 cells had a similar increase (142-158%) on AAV2 or AAV3 transduction. Our results highlight that recombinant AAV mediated microRNA expression is cell-type and serotype-specific and can target specific host cellular biological pathways.


Subject(s)
Dependovirus/genetics , MicroRNAs/genetics , Parvoviridae Infections/genetics , Retinal Pigment Epithelium/virology , Transduction, Genetic/methods , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Cycle/genetics , Cell Line , Cell Line, Tumor , Endoplasmic Reticulum Stress/genetics , Gene Expression Profiling , Humans , Lipid Metabolism/genetics , Liver Neoplasms/pathology , Liver Neoplasms/virology , Parvovirinae/genetics , Reproducibility of Results , Retinal Pigment Epithelium/cytology , Transgenes
8.
PLoS Pathog ; 15(10): e1007956, 2019 10.
Article in English | MEDLINE | ID: mdl-31589653

ABSTRACT

We report the analysis of a complex enveloped human virus, herpes simplex virus (HSV), assembled after in vivo incorporation of bio-orthogonal methionine analogues homopropargylglycine (HPG) or azidohomoalanine (AHA). We optimised protocols for the production of virions incorporating AHA (termed HSVAHA), identifying conditions which resulted in normal yields of HSV and normal particle/pfu ratios. Moreover we show that essentially every single HSVAHA capsid-containing particle was detectable at the individual particle level by chemical ligation of azide-linked fluorochromes to AHA-containing structural proteins. This was a completely specific chemical ligation, with no capsids assembled under normal methionine-containing conditions detected in parallel. We demonstrate by quantitative mass spectrometric analysis that HSVAHA virions exhibit no qualitative or quantitative differences in the repertoires of structural proteins compared to virions assembled under normal conditions. Individual proteins and AHA incorporation sites were identified in capsid, tegument and envelope compartments, including major essential structural proteins. Finally we reveal novel aspects of entry pathways using HSVAHA and chemical fluorochrome ligation that were not apparent from conventional immunofluorescence. Since ligation targets total AHA-containing protein and peptides, our results demonstrate the presence of abundant AHA-labelled products in cytoplasmic macrodomains and tubules which no longer contain intact particles detectable by immunofluorescence. Although these do not co-localise with lysosomal markers, we propose they may represent sites of proteolytic virion processing. Analysis of HSVAHA also enabled the discrimination from primary entering from secondary assembling virions, demonstrating assembly and second round infection within 6 hrs of initial infection and dual infections of primary and secondary virus in spatially restricted cytoplasmic areas of the same cell. Together with other demonstrated applications e.g., in genome biology, lipid and protein trafficking, this work further exemplifies the utility and potential of bio-orthogonal chemistry for studies in many aspects of virus-host interactions.


Subject(s)
Amino Acids/metabolism , Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Retinal Pigment Epithelium/virology , Viral Structural Proteins/metabolism , Virus Assembly , Virus Internalization , Cell Proliferation , Cells, Cultured , Herpes Simplex/metabolism , Humans , Retinal Pigment Epithelium/metabolism
9.
Int J Mol Sci ; 20(4)2019 Feb 16.
Article in English | MEDLINE | ID: mdl-30781494

ABSTRACT

Human retinal pigment epithelial (hRPE) cells are important for the establishment and maintenance of the immune privilege of the eye. They function as target cells for human cytomegalovirus (hCMV), but are able to restrict viral replication. hCMV causes opportunistic posterior uveitis such as retinitis and chorioretinitis. Both mainly occur in severely immunocompromised patients and rarely manifest in immunocompetent individuals. In this study, hRPE cells were infected with hCMV in vitro and activated with proinflammatory cytokines. The enzymatic activities of indoleamine 2,3-dioxygenase-1 (IDO1) and inducible nitric oxide synthase (iNOS) were determined. The antimicrobial capacity of both molecules was analyzed in co-infection experiments using Staphylococcus aureus (S. aureus) and Toxoplasma gondii (T. gondii), causing uveitis in patients. We show that an hCMV infection of hRPE cells blocks IDO1 and iNOS mediated antimicrobial defense mechanisms necessary for the control of S. aureus and T. gondii. hCMV also inhibits immune suppressive effector mechanisms in hRPE. The interferon gamma-induced IDO1 dependent immune regulation was severely blocked, as detected by the loss of T cell inhibition. We conclude that an active hCMV infection in the eye might favor the replication of pathogens causing co-infections in immunosuppressed individuals. An hCMV caused blockade of IDO1 might weaken the eye's immune privilege and favor the development of post-infectious autoimmune uveitis.


Subject(s)
Eye/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Retinal Pigment Epithelium/immunology , Uveitis/immunology , Cell Proliferation/genetics , Coinfection/immunology , Coinfection/microbiology , Coinfection/virology , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Eye/microbiology , Eye/virology , Flow Cytometry , Humans , Immune Privilege/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interferon-gamma/immunology , Nitric Oxide Synthase Type II/genetics , Retinal Pigment Epithelium/microbiology , Retinal Pigment Epithelium/virology , Staphylococcus aureus/growth & development , Staphylococcus aureus/pathogenicity , T-Lymphocytes/immunology , T-Lymphocytes/microbiology , T-Lymphocytes/virology , Toxoplasma/growth & development , Toxoplasma/pathogenicity , Uveitis/microbiology , Uveitis/virology
10.
EBioMedicine ; 39: 315-331, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30579862

ABSTRACT

BACKGROUND: Zika virus (ZIKV) has recently re-emerged as a pathogenic agent with epidemic capacities as was well illustrated in South America. Because of the extent of this health crisis, a number of more serious symptoms have become associated with ZIKV infection than what was initially described. In particular, neuronal and ocular disorders have been characterized, both in infants and in adults. Notably, the macula and the retina can be strongly affected by ZIKV, possibly by a direct effect of the virus. This is supported by the detection of replicative and infectious virus in lachrimal fluid in human patients and mouse models. METHODS: Here, we used an innovative, state-of-the-art iPSC-derived human retinal pigment epithelium (RPE) model to study ZIKV retinal impairment. FINDINGS: We showed that the human RPE is highly susceptible to ZIKV infection and that a ZIKV African strain was more virulent and led to a more potent epithelium disruption and stronger anti-viral response than an Asian strain, suggesting lineage differences. Moreover, ZIKV infection led to impaired membrane dynamics involved in endocytosis, organelle biogenesis and potentially secretion, key mechanisms of RPE homeostasis and function. INTERPRETATION: Taken together, our results suggest that ZIKV has a highly efficient ocular tropism, which creates a strong inflammatory environment that could have acute or chronic adverse effects. FUND: This work was funded by Retina France, REACTing and La Région Languedoc-Roussillon.


Subject(s)
Interferons/metabolism , Retinal Pigment Epithelium/virology , Zika Virus Infection/immunology , Zika Virus/pathogenicity , Cells, Cultured , Homeostasis , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/virology , Interferons/genetics , Models, Biological , Phagocytosis , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/immunology , Viral Tropism , Virus Replication , Zika Virus/classification , Zika Virus/physiology , Zika Virus Infection/genetics , Zika Virus Infection/virology
11.
Proc Natl Acad Sci U S A ; 115(37): E8803-E8810, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30150412

ABSTRACT

A fundamental signal-processing problem is how biological systems maintain phenotypic states (i.e., canalization) long after degradation of initial catalyst signals. For example, to efficiently replicate, herpesviruses (e.g., human cytomegalovirus, HCMV) rapidly counteract cell-mediated silencing using transactivators packaged in the tegument of the infecting virion particle. However, the activity of these tegument transactivators is inherently transient-they undergo immediate proteolysis but delayed synthesis-and how transient activation sustains lytic viral gene expression despite cell-mediated silencing is unclear. By constructing a two-color, conditional-feedback HCMV mutant, we find that positive feedback in HCMV's immediate-early 1 (IE1) protein is of sufficient strength to sustain HCMV lytic expression. Single-cell time-lapse imaging and mathematical modeling show that IE1 positive feedback converts transient transactivation signals from tegument pp71 proteins into sustained lytic expression, which is obligate for efficient viral replication, whereas attenuating feedback decreases fitness by promoting a reversible silenced state. Together, these results identify a regulatory mechanism enabling herpesviruses to sustain expression despite transient activation signals-akin to early electronic transistors-and expose a potential target for therapeutic intervention.


Subject(s)
Cytomegalovirus/genetics , Feedback, Physiological , Gene Expression Regulation, Viral , Virus Replication/genetics , Cell Line , Cells, Cultured , Cytomegalovirus/physiology , Host-Pathogen Interactions , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Microscopy, Fluorescence , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/virology , Time-Lapse Imaging/methods
12.
Sci Rep ; 8(1): 11209, 2018 07 25.
Article in English | MEDLINE | ID: mdl-30046058

ABSTRACT

Previously, we reported that Zika virus (ZIKV) causes ocular complications such as chorioretinal atrophy, by infecting cells lining the blood-retinal barrier, including the retinal pigment epithelium (RPE). To understand the molecular basis of ZIKV-induced retinal pathology, we performed a meta-analysis of transcriptome profiles of ZIKV-infected human primary RPE and other cell types infected with either ZIKV or other related flaviviruses (Japanese encephalitis, West Nile, and Dengue). This led to identification of a unique ZIKV infection signature comprising 43 genes (35 upregulated and 8 downregulated). The major biological processes perturbed include SH3/SH2 adaptor activity, lipid and ceramide metabolism, and embryonic organ development. Further, a comparative analysis of some differentially regulated genes (ABCG1, SH2B3, SIX4, and TNFSF13B) revealed that ZIKV induced their expression relatively more than dengue virus did in RPE. Importantly, the pharmacological inhibition of ABCG1, a membrane transporter of cholesterol, resulted in reduced ZIKV infectivity. Interestingly, the ZIKV infection signature revealed the downregulation of ALDH5A1 and CHML, genes implicated in neurological (cognitive impairment, expressive language deficit, and mild ataxia) and ophthalmic (choroideremia) disorders, respectively. Collectively, our study revealed that ZIKV induces differential gene expression in RPE cells, and the identified genes/pathways (e.g., ABCG1) could potentially contribute to ZIKV-associated ocular pathologies.


Subject(s)
Retinal Pigment Epithelium/metabolism , Transcriptome/genetics , Zika Virus Infection/genetics , Zika Virus/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , Adaptor Proteins, Signal Transducing , B-Cell Activating Factor/genetics , Dengue/genetics , Dengue/pathology , Dengue/virology , Dengue Virus/pathogenicity , Encephalitis Viruses, Japanese/pathogenicity , Flavivirus Infections/genetics , Flavivirus Infections/pathology , Flavivirus Infections/virology , Gene Expression Regulation/genetics , Homeodomain Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins , Primary Cell Culture , Proteins/genetics , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/virology , Trans-Activators/genetics , Virus Replication/genetics , West Nile Fever/genetics , West Nile Fever/pathology , West Nile Fever/virology , West Nile virus/pathogenicity , Zika Virus/pathogenicity , Zika Virus Infection/pathology , Zika Virus Infection/virology
13.
Mol Vis ; 24: 379-394, 2018.
Article in English | MEDLINE | ID: mdl-29853772

ABSTRACT

Purpose: The purpose of this study was to determine whether the blood-retina barrier is compromised by choroidal murine cytomegalovirus (MCMV) infection, using electron microscopy. Methods: BALB/c mice were immunosuppressed with methylprednisolone and monoclonal antibodies to CD4 and CD8. At several time points post-MCMV intraperitoneal inoculation, the eyes were removed and analyzed with western blotting and immunoelectron microscopy for the presence of MCMV early antigen (EA) and the host protein RIP3. Posterior eyecups from RIP3-/- and RIP3+/+ mice were cultured and inoculated with MCMV. At days 4, 7, and 11 post-infection, cultures were collected and analyzed with plaque assay, immunohistochemical staining, and real-time PCR (RT-PCR). Results: MCMV EA was observed in the nuclei of vascular endothelial cells and pericytes in the choriocapillaris. Disruption of Bruch's membrane was observed, especially at sites adjacent to activated platelets, and a few RPE cells containing some enlarged vesicles were found directly beneath disrupted Bruch's membrane. Some virus particles were also observed in the enlarged vesicles of RPE cells. Levels of the RIP3 protein, which was observed mainly in the RPE cells and the basement membrane of the choriocapillaris, were greatly increased following MCMV infection, while depletion of RIP3 resulted in greatly decreased inflammasome formation, as well as expression of downstream inflammation factors. Conclusions: The results suggest that systemic MCMV spreads to the choroid and replicates in vascular endothelia and pericytes of the choriocapillaris during immunosuppression. Choroidal MCMV infection is associated with in situ inflammation and subsequent disruption of Bruch's membrane and the outer blood-retina barrier.


Subject(s)
Choroid/immunology , Cytomegalovirus Infections/immunology , Eye Infections, Viral/immunology , Immunocompromised Host , Retina/immunology , Retinitis/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antigens, Viral/genetics , Blood Platelets/immunology , Blood Platelets/pathology , Blood Platelets/virology , Blood-Retinal Barrier/immunology , Blood-Retinal Barrier/pathology , Blood-Retinal Barrier/virology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Choroid/blood supply , Choroid/pathology , Choroid/virology , Cytomegalovirus Infections/pathology , Cytomegalovirus Infections/virology , Endothelial Cells , Eye Infections, Viral/pathology , Eye Infections, Viral/virology , Female , Immediate-Early Proteins/genetics , Inflammasomes/immunology , Methylprednisolone/administration & dosage , Mice , Mice, Inbred BALB C , Muromegalovirus/growth & development , Muromegalovirus/pathogenicity , Pericytes/immunology , Pericytes/pathology , Pericytes/virology , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Retina/pathology , Retina/virology , Retinal Pigment Epithelium/immunology , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/virology , Retinitis/pathology , Retinitis/virology
14.
Invest Ophthalmol Vis Sci ; 59(6): 2445-2458, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29847649

ABSTRACT

Purpose: The purpose of this study was to determine if the receptor-interacting protein kinase 3 (RIP3) plays a significant role in innate immune responses and death of bystander retinal neurons during murine cytomegalovirus (MCMV) retinal infection, by comparing the innate immune response and cell death in RIP3-depleted mice (Rip3-/-) and Rip3+/+ control mice. Methods: Rip3-/- and Rip3+/+ mice were immunosuppressed (IS) and inoculated with MCMV via the supraciliary route. Virus-injected and mock-injected control eyes were removed at days 4, 7, and 10 post infection (p.i.) and markers of innate immunity and cell death were analyzed. Results: Compared to Rip3+/+ mice, significantly more MCMV was recovered and more MCMV-infected RPE cells were observed in injected eyes of Rip3-/- mice at days 4 and 7 p.i. In contrast, fewer TUNEL-stained photoreceptors were observed in Rip3-/- eyes than in Rip3+/+ eyes at these times. Electron microscopy showed that significantly more apoptotic photoreceptor cells were present in Rip3+/+ mice than in Rip3-/- mice. Immunohistochemistry showed that the majority of TUNEL-stained photoreceptors died via mitochondrial flavoprotein apoptosis-inducing factor (AIF)-mediated, caspase 3-independent apoptosis. The majority of RIP3-expressing cells in infected eyes were RPE cells, microglia/macrophages, and glia, whereas retinal neurons contained much lower amounts of RIP3. Western blots showed significantly higher levels of activated nuclear factor-κB and caspase 1 were present in Rip3+/+ eyes compared to Rip3-/- eyes. Conclusions: Our results suggest that RIP3 enhances innate immune responses against ocular MCMV infection via activation of the inflammasome and nuclear factor-κB, which also leads to inflammation and death of bystander cells by multiple pathways including apoptosis and necroptosis.


Subject(s)
Apoptosis , Eye Infections, Viral/pathology , Herpesviridae Infections/pathology , Muromegalovirus/isolation & purification , Photoreceptor Cells, Vertebrate/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Retinal Diseases/pathology , Animals , Biomarkers/metabolism , Blotting, Western , Cell Survival/physiology , Eye Infections, Viral/metabolism , Eye Infections, Viral/virology , Female , Fluorescent Antibody Technique, Indirect , Herpesviridae Infections/metabolism , Herpesviridae Infections/virology , Immunity, Innate/physiology , In Situ Nick-End Labeling , Inflammasomes/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Electron , NF-kappa B/metabolism , Retinal Diseases/metabolism , Retinal Diseases/virology , Retinal Pigment Epithelium/virology
15.
Mol Med Rep ; 17(1): 763-770, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29115621

ABSTRACT

The present study aimed to explore the effects of a stabilizing ligand, Shield­1, on the replication of recombinant varicella­zoster virus (VZV) containing FK506 binding protein (FKPB) tags in essential open reading frames (ORF) 4 and 48. A specific galactokinase (galK) selection method was conducted, following the addition of galK labels to VZV ORF4 and 48, using a SW102 VZV bacterial artificial chromosome (BAC) system. Subsequently, recombinant VZV containing FKPB tags in ORF4 and 48 was constructed by counterselection and homologous recombination. Recombinant viral plasmids containing FKPB­tagged VZV ORF4 and 48 were extracted and transfected into human acute retinal pigment epithelial ARPE­19 cells. The results demonstrated that the FKPB­tagged viral protein was rapidly degraded by proteases in recombinant virus­infected ARPE­19 cells. In addition, the recombinant VZVORF4­FKBP­ORF48­FKBP virus could not grow if a synthetic ligand of FKBP, Shield1, was not added to the ARPE­19 cell culture medium; however, the degradation of FKPB­tagged viral protein was prevented if Shield1 was added to the ARPE­19 cell culture medium, thereby allowing viral replication in ARPE­19 cells. These results indicated that Shield1 may regulate replication of recombinant VZVORF4­FKBP­ORF48­FKBP following transfection into human epithelial cells.


Subject(s)
Herpesvirus 3, Human/genetics , Immediate-Early Proteins/genetics , Tacrolimus Binding Proteins/genetics , Virus Replication/genetics , Cell Line , Chromosomes, Artificial, Bacterial/genetics , Culture Media/pharmacology , Herpesvirus 3, Human/growth & development , Herpesvirus 3, Human/pathogenicity , Humans , Ligands , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/virology , Transfection , Virus Replication/drug effects
16.
Invest Ophthalmol Vis Sci ; 58(10): 4355­4365, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28810265

ABSTRACT

Purpose: Emerging evidence has shown that both congenital and adult Zika virus (ZIKV) infection can cause eye diseases. The goals of the current study were to explore mechanisms and pathophysiology of ZIKV-induced eye defects. Methods: Wild-type or A129 interferon type I receptor-deficient mice were infected by either FSS13025 or Mex1-7 strain of ZIKV. Retinal histopathology was measured at different time points after infection. The presence of viral RNA and protein in the retina was determined by in situ hybridization and immunofluorescence staining, respectively. Growth curves of ZIKV in permissive retinal cells were assessed in cultured retinal pigment epithelial (RPE) and Müller glial cells. Results: ZIKV-infected mice developed a spectrum of ocular pathologies that affected multiple layers of the retina. A primary target of ZIKV in the eye was Müller glial cells, which displayed decreased neurotrophic function and increased expression of proinflammatory cytokines after infection. ZIKV also infected RPE; and both the RPE and Müller cells expressed viral entry receptors TYRO3 and AXL. Retinitis, focal retinal degeneration, and ganglion cell loss were observed after the clearance of viral particles. Conclusions: Our data suggest that ZIKV can infect infant eyes with immature blood-retinal barrier and cause structural damages to the retina. The ocular findings in microcephalic infants may not be solely caused by ZIKV-induced impairment of neurodevelopment.


Subject(s)
Disease Models, Animal , Ependymoglial Cells/virology , Eye Infections, Viral/virology , Retinal Diseases/virology , Retinal Pigment Epithelium/virology , Zika Virus Infection/virology , Animals , Blood-Retinal Barrier , Cells, Cultured , Ependymoglial Cells/pathology , Eye Infections, Viral/pathology , Flow Cytometry , In Situ Hybridization, Fluorescence , Mice , Mice, Inbred C57BL , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Retinal Diseases/pathology , Retinal Pigment Epithelium/pathology , Uveitis, Posterior/pathology , Uveitis, Posterior/virology , Viral Nonstructural Proteins/metabolism , Virus Replication/physiology , Zika Virus/physiology , Zika Virus Infection/pathology
17.
JCI Insight ; 2(4): e92340, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28239662

ABSTRACT

Zika virus (ZIKV) is an important pathogen that causes not only neurologic, but also ocular, abnormalities. Thus, it is imperative that models to study ZIKV pathogenesis in the eye are developed to identify potential targets for interventions. Here, we studied ZIKV interactions with human retinal cells and evaluated ZIKV's pathobiology in mouse eyes. We showed that cells lining the blood-retinal barrier (BRB), the retinal endothelium, and retinal pigment epithelium (RPE) were highly permissive and susceptible to ZIKV-induced cell death. Direct inoculation of ZIKV in eyes of adult C57BL/6 and IFN-stimulated gene 15 (ISG15) KO mice caused chorioretinal atrophy with RPE mottling, a common ocular manifestation of congenital ZIKV infection in humans. This response was associated with induced expression of multiple inflammatory and antiviral (IFNs) response genes in the infected mouse retina. Interestingly, ISG15 KO eyes exhibited severe chorioretinitis, which coincided with increased retinal cell death and higher ZIKV replication. Collectively, our study provides the first evidence to our knowledge that ZIKV causes retinal lesions and infects the cells lining the BRB and that ISG15 plays a role in retinal innate defense against ZIKV infection. Our mouse model can be used to study mechanisms underlying ZIKV-induced chorioretinitis and to gauge ocular antiviral therapies.


Subject(s)
Blood-Retinal Barrier/virology , Chorioretinitis/virology , Choroid/virology , Endothelium/virology , Retinal Pigment Epithelium/virology , Zika Virus Infection/pathology , Zika Virus , Animals , Atrophy , Blood-Retinal Barrier/cytology , Cell Death , Cell Line , Chorioretinitis/pathology , Choroid/pathology , Cytokines/genetics , Disease Models, Animal , Endothelium/cytology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Retina/pathology , Retina/virology , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/pathology , Ubiquitins/genetics , Virus Replication
18.
Antiviral Res ; 139: 117-128, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28049006

ABSTRACT

An epidemic of Zika virus (ZIKV) infection associated with congenital abnormalities such as microcephaly, is ongoing in the Americas and the Pacific. Currently there are no approved therapies to treat this emerging viral disease. Here, we tested three cell-directed broad-spectrum antiviral compounds against ZIKV replication using human retinal pigment epithelial (RPE) cells and a low-passage ZIKV strain isolated from fetal brain. We found that obatoclax, SaliPhe, and gemcitabine inhibited ZIKV infections at noncytotoxic concentrations. Moreover, all three compounds prevented production of viral RNA and proteins as well as activation of cellular caspase 8, 3 and 7. However, these compounds differentially affected ZIKV-mediated transcription, translation and posttranslational modifications of cellular factors as well as metabolic pathways indicating that these agents possess different mechanisms of action. Interestingly, combination of obatoclax and SaliPhe at nanomolar concentrations had a synergistic effect against ZIKV infection. Thus, our results provided the foundation for development of broad-spectrum cell-directed antivirals or their combinations for treatment of ZIKV and other emerging viral diseases.


Subject(s)
Amides/pharmacology , Antiviral Agents/pharmacology , Deoxycytidine/analogs & derivatives , Pyrroles/pharmacology , Salicylates/pharmacology , Signal Transduction/drug effects , Zika Virus/drug effects , Brain/cytology , Caspases/metabolism , Deoxycytidine/pharmacology , Drug Combinations , Enzyme Inhibitors/pharmacology , Fetus/anatomy & histology , Humans , Indoles , Metabolic Networks and Pathways/drug effects , RNA, Viral/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/virology , Virus Replication/drug effects , Zika Virus/isolation & purification , Zika Virus/metabolism , Gemcitabine
20.
Invest Ophthalmol Vis Sci ; 56(9): 5162-73, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26244291

ABSTRACT

PURPOSE: The a-disintegrin-and-metalloprotease (ADAM) family proteins are widely expressed in the different layers of the retina throughout development. The effect of ADAM proteins on the epithelial-to-mesenchymal transition (EMT) in proliferative vitreoretinopathy (PVR) or AMD is yet to be elucidated. In this study we used Epstein-Barr virus (EBV)-transformed adult retinal pigment epithelial (ARPE) cells to investigate how sorafenib, a multikinase inhibitor, modulates ADAM proteins to control EMT. METHODS: Epithelial to mesenchymal transition and related mechanisms in EBV-infected ARPE cells were determined by RT-PCR, Western blot, invasion assay, ELISA assay, and gene silencing with siRNA. RESULTS: Mesenchymal-like ARPE/EBV cells exhibited considerably increased cellular migration and invasion compared with ARPE cells and produced EMT-related cytokines. Sorafenib significantly inhibited production of TGF-ß1, VEGF, IL-6, IL-8, MCP-1, and TNF-α and blocked the activation of migration-related signaling molecules, such as HIF-1α, p-STAT3, MMP2, and Ang-1. The expression of mature ADAM10, ADAM17, and cleaved Notch 1 proteins in ARPE/EBV cells was downregulated after treatment with sorafenib through the regulatory activity of nardilysin (NRD-1). Gene silencing of NRD-1 in ARPE/EBV cells attenuated secretion of EMT-related cytokines and expression of ADAM10 and 17 and upregulated epithelial markers. CONCLUSIONS: Sorafenib controls the mesenchymal characteristics of EBV-infected ARPE cells. Nardilysin and ADAM family proteins might be new targets for the prevention or control of EMT in retinal diseases.


Subject(s)
ADAM Proteins/genetics , Amyloid Precursor Protein Secretases/genetics , DNA/genetics , Gene Expression Regulation/drug effects , Herpesvirus 4, Human , Membrane Proteins/genetics , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Retinal Pigment Epithelium/metabolism , ADAM Proteins/biosynthesis , ADAM Proteins/drug effects , ADAM10 Protein , ADAM17 Protein , Amyloid Precursor Protein Secretases/biosynthesis , Amyloid Precursor Protein Secretases/drug effects , Cell Movement , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Humans , Immunoblotting , Membrane Proteins/biosynthesis , Membrane Proteins/drug effects , Niacinamide/pharmacology , Protein Kinase Inhibitors/pharmacology , Receptors, Vascular Endothelial Growth Factor , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/virology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Sorafenib , Tumor Necrosis Factor-alpha , Vitreoretinopathy, Proliferative/genetics , Vitreoretinopathy, Proliferative/pathology , Vitreoretinopathy, Proliferative/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...