Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters

Publication year range
1.
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 , Cell Line , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Epithelial Cells/metabolism , Epithelial Cells/virology , Herpes Simplex/immunology , Herpes Simplex/metabolism , Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Immediate-Early Proteins/metabolism , Immediate-Early Proteins/genetics , Inflammasomes/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/virology
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Gene Ther ; 21(10): 855-65, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24989814

ABSTRACT

Retinal gene therapy with adeno-associated viral (AAV) vectors is safe and effective in humans. However, the limited cargo capacity of AAV prevents their use for therapy of those inherited retinopathies (IRs) due to mutations in large (>5 kb) genes. Viral vectors derived from adenovirus (Ad), lentivirus (LV) and herpes virus (HV) can package large DNA sequences, but do not target efficiently retinal photoreceptors (PRs) where the majority of genes responsible for IRs are expressed. Here, we have evaluated the mouse retinal transduction profiles of vectors derived from 16 different Ad serotypes, 7 LV pseudotypes and from a bovine HV. Most of the vectors tested transduced efficiently the retinal pigment epithelium. We found that LV-GP64 tends to transduce more PRs than the canonical LV-VSVG, albeit this was restricted to a narrow region. We observed more extensive PR transduction with HdAd1, 2 and 5/F35++ than with LV, although none of them outperformed the canonical HdAd5 or matched the extension of PR transduction achieved with AAV2/8.


Subject(s)
Dependovirus/genetics , Herpesvirus 4, Bovine/genetics , Lentivirus/genetics , Retinal Pigment Epithelium/virology , Animals , Dependovirus/classification , Electroretinography , Epithelial Cells/virology , Genetic Vectors/administration & dosage , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Herpesvirus 4, Bovine/classification , Lentivirus/classification , Male , Mice , Mice, Inbred BALB C , Photoreceptor Cells, Vertebrate/metabolism , Retinal Pigment Epithelium/cytology , Transduction, Genetic
8.
Lab Invest ; 94(7): 777-87, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24840331

ABSTRACT

The breakdown of human retinal pigment epithelial (HRPE) barrier is considered as the etiology of retinopathy, which affects the quality of life of HIV/AIDS patients. Here we demonstrate that HIV-1 could directly impair HRPE barrier function, which leads to the translocation of HIV-1 and bacteria. HRPE cells (D407) were grown to form polarized, confluent monolayers and treated with different HIV-1 infectious clones. A significant increase of monolayer permeability, as measured by trans-epithelial electrical resistance (TEER) and apical-basolateral movements of sodium fluorescein, was observed. Disrupted tightness of HRPE barrier was associated with the downregulation of several tight junction proteins in D407 cells, including ZO-1, Occludin, Claudin-1, Claudin-2, Claudin-3, Claudin-4, and Claudin-5, after exposure to HIV-1, without affecting the viability of cells. HIV-1 gp120 was shown to participate in the alteration of barrier properties, as evidenced by decreased TEER and weakened expression of tight junction proteins in D407 monolayers after exposure to pseudotyped HIV-1, UV-inactivated HIV-1, and free gp120, but not to an envelope (Env)-defective mutant of HIV. Furthermore, exposure to HIV-1 particles could induce the release of pro-inflammatory cytokines in D407, including IL-6 and MCP-1, both of which downregulated the expression of ZO-1 in the HRPE barrier. Disrupted HRPE monolayer allowed translocation of HIV-1 and bacteria across the epithelium. Overall, these findings suggest that HIV-1 may exploit its Env glycoprotein to induce an inflammatory state in HRPE cells, which could result in impairment of HRPE monolayer integrity, allowing virus and bacteria existing in ocular fluids to cross the epithelium and penetrate the HRPE barrier. Our study highlights the role of HIV-1 in the pathogenesis of HIV/AIDS-related retinopathy and suggests potential therapeutic targets for this ocular complication.


Subject(s)
HIV Infections/physiopathology , HIV-1/metabolism , Retinal Diseases/physiopathology , Retinal Pigment Epithelium/physiopathology , Bacterial Translocation , Blotting, Western , Cell Line , Chemokine CCL2/metabolism , Epithelial Cells/metabolism , Escherichia coli/metabolism , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/metabolism , HIV Infections/metabolism , HIV Infections/virology , HIV-1/genetics , HIV-1/physiology , Host-Pathogen Interactions , Humans , Interleukin-6/metabolism , Microscopy, Fluorescence , Mutation , Permeability , Retinal Diseases/metabolism , Retinal Diseases/virology , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/virology , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism , Tight Junctions/metabolism , Tight Junctions/physiology
9.
J Virol ; 87(19): 10489-500, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23885075

ABSTRACT

The human cytomegalovirus (HCMV) virion envelope contains a complex consisting of glycoproteins gH and gL plus proteins encoded by the UL128 locus (UL128L): pUL128, pUL130, and pUL131A. UL128L is necessary for efficient infection of myeloid, epithelial, and endothelial cells but limits replication in fibroblasts. Consequently, disrupting mutations in UL128L are rapidly selected when clinical isolates are cultured in fibroblasts. In contrast, bacterial artificial chromosome (BAC)-cloned strains TB40-BAC4, FIX, and TR do not contain overt disruptions in UL128L, yet no virus reconstituted from them has been reported to acquire mutations in UL128L in vitro. We performed BAC mutagenesis and reconstitution experiments to test the hypothesis that these strains contain subtle mutations in UL128L that were acquired during passage prior to BAC cloning. Compared to strain Merlin containing wild-type UL128L, all three strains produced higher yields of cell-free virus. Moreover, TB40-BAC4 and FIX spread cell to cell more rapidly than wild-type Merlin in fibroblasts but more slowly in epithelial cells. The differential growth properties of TB40-BAC4 and FIX (but not TR) were mapped to single-nucleotide substitutions in UL128L. The substitution in TB40-BAC4 reduced the splicing efficiency of UL128, and that in FIX resulted in an amino acid substitution in UL130. Introduction of these substitutions into Merlin dramatically increased yields of cell-free virus and increased cell-to-cell spread in fibroblasts but reduced the abundance of pUL128 in the virion and the efficiency of epithelial cell infection. These substitutions appear to represent mutations in UL128L that permit virus to be propagated in fibroblasts while retaining epithelial cell tropism.


Subject(s)
Cytomegalovirus Infections/virology , Cytomegalovirus/physiology , Fibroblasts/virology , Membrane Glycoproteins/metabolism , Retinal Pigment Epithelium/virology , Viral Envelope Proteins/metabolism , Blotting, Western , Cells, Cultured , Chromosomes, Artificial, Bacterial/genetics , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , Endothelial Cells/metabolism , Endothelial Cells/virology , Fetus , Fibroblasts/metabolism , Foreskin/metabolism , Foreskin/virology , Genetic Variation , Humans , Male , Membrane Glycoproteins/genetics , Mutagenesis , Mutation/genetics , Plasmids/genetics , Retinal Pigment Epithelium/metabolism , Tropism , Viral Envelope Proteins/genetics , Virion/physiology , Virus Internalization
10.
Mol Vis ; 20: 1161-73, 2014.
Article in English | MEDLINE | ID: mdl-25324684

ABSTRACT

PURPOSE: Previous studies have demonstrated that autophagy is involved in the pathogenesis of human cytomegalovirus (HCMV) infection. However, whether autophagy is regulated by murine cytomegalovirus (MCMV) infection has not yet been investigated. The purpose of these studies was to determine how autophagy is affected by MCMV infection of the retinal pigment epithelial (RPE) cells and whether there is a functional relationship between autophagy and apoptosis; and if so, how regulation of autophagy impacts apoptosis. METHODS: RPE cells were isolated from C57BL/6 mice and infected with MCMV K181. The cells were cultured in medium containing rapamycin, chloroquine, or ammonium chloride. Green fluorescent protein-light chain 3 (GFP-LC3) plasmid was transfected to RPE cells, and the GFP-LC3 positive puncta were counted. Electron microscopic (EM) images were taken to visualize the structure of the autophagic vacuoles. Western blot was performed to detect the expression of related proteins. Trypan blue exclusion assay was used to measure the percentage of viable cells. RESULTS: Although the LC3B-II levels consistently increased during MCMV infection of RPE cells, administration of chloroquine or ammonium chloride increased LC3B-II expression only at the early stage of infection (6 h post-inoculation [p.i.] and 12 h p.i.), not at or after 24 h p.i. The punctate autophagic vacuoles in the GFP-LC3 transfected RPE cells were counted using light microscopy or by EM examination, the number of autophagic vacuoles was significantly increased in the MCMV-infected RPE cells compared to the uninfected controls. Compared to untreated MCMV-infected control cells, rapamycin treatment resulted in a significant decrease in the cleaved caspase 3 levels as well as a significant decrease in the ratio of phosphorylated mammalian target of rapamycin (mTOR) to total mTOR and in the ratio of phosphorylated P70S6K to total P70S6K. In contrast, chloroquine treatment resulted in a significant increase in the cleaved caspase 3 levels in the MCMV-infected RPE cells. CONCLUSIONS: Autophagic vacuole accumulation was detected during MCMV infection of RPE cells. In contrast, autophagic flux was greatly decreased at or after 24 h p.i. The results suggest that MCMV might have a strategy for inhibiting or blocking autophagy activity by targeting a later autophagy process, such as the formation of autolysosomes or degradation of their content. Our data also suggest that there is a functional relationship between autophagy and apoptosis, which plays an important role during MCMV infection of the RPE.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Muromegalovirus/pathogenicity , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/virology , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Chloroquine/pharmacology , Cytomegalovirus Infections/pathology , Cytomegalovirus Infections/virology , Disease Models, Animal , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Herpesviridae Infections/metabolism , Humans , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinal Pigment Epithelium/metabolism , Sirolimus/pharmacology , Transfection , Vacuoles/ultrastructure
11.
Genes (Basel) ; 15(4)2024 03 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
12.
Int Immunopharmacol ; 137: 112460, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38908082

ABSTRACT

There is growing interest in evaluating the safety and therapeutic potential of existing treatments such as tocilizumab (TCZ), an IL-6 receptor antagonist used to treat inflammatory diseases. However, there have been reports of increased inflammation in patients with HTLV-1 uveitis after TCZ treatment, and its ocular safety in the HTLV-1 infected state remains unknown. This study focused on assessing the impact of TCZ on HTLV-1-infected ocular cells using an in vitro model in which retinal pigment epithelial cells were cocultured with irradiated HTLV-1-infected T-cell lines. TCZ did not significantly affect cellular viability, inflammatory markers, or HTLV-1 proviral loads at various concentrations (25/50/100 µg/ml), indicating no increased risk of HTLV-1 viral infection and no exacerbation of the inflammatory aspects of HTLV-1 infection in the ocular cells. These promising results support the potential of TCZ as a safe treatment option for HTLV-1-infected patients, particularly those with eye infections.


Subject(s)
Antibodies, Monoclonal, Humanized , HTLV-I Infections , Human T-lymphotropic virus 1 , Humans , HTLV-I Infections/drug therapy , HTLV-I Infections/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/adverse effects , Cell Line , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/virology , Retinal Pigment Epithelium/immunology , Coculture Techniques , Cell Survival/drug effects , Viral Load/drug effects
13.
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
15.
J Virol ; 85(23): 12750-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21937636

ABSTRACT

Human cytomegalovirus (HCMV) remains a major cause of viral disease in immunosuppressed transplant patients. The ability of HCMV to establish lifelong infection in humans and reactivate with devastating clinical consequences underscores the importance of understanding the triggers of HCMV reactivation in mature myeloid cells. Dendritic cell (DC) differentiation is concomitant with the activation of cellular signaling pathways and inflammatory gene expression and also HCMV reactivation. Here, we show a major role for interleukin-6 (IL-6) through extracellular signal-regulated kinase-mitogen-activated protein kinase (ERK-MAPK) signaling upon DC differentiation to promote HCMV reactivation. IL-6 drives reactivation by transcriptional upregulation of the major immediate-early (IE) genes, resulting in efficient progression of the virus life cycle and, ultimately, higher titers of infectious virus. Furthermore, the interception of IL-6 signaling with biological inhibitors significantly abrogated HCMV reactivation from experimental latency. Crucially, using cells derived from healthy seropositive donors, we observed a key role for IL-6 during reactivation from natural latency ex vivo in interstitial DCs. Clinically, HCMV reactivation occurs in highly inflammatory environments (i.e., transplantation); thus, the implications of this study could potentially provide novel approaches for therapeutic intervention.


Subject(s)
Cytomegalovirus Infections/virology , Dendritic Cells/virology , Extracellular Signal-Regulated MAP Kinases/metabolism , Inflammation Mediators/antagonists & inhibitors , Interleukin-6/metabolism , Mitogen-Activated Protein Kinases/metabolism , Virus Activation/physiology , Blotting, Western , Cell Differentiation , Cells, Cultured , Cytomegalovirus , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , Dendritic Cells/metabolism , Gene Expression Regulation, Viral , Genes, Immediate-Early , Humans , Interleukin-6/antagonists & inhibitors , Monocytes/cytology , Monocytes/metabolism , Monocytes/virology , Myeloid Cells/cytology , Myeloid Cells/metabolism , Myeloid Cells/virology , RNA, Messenger/genetics , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/virology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transcriptional Activation , Virus Latency
16.
Mol Vis ; 18: 730-43, 2012.
Article in English | MEDLINE | ID: mdl-22509103

ABSTRACT

PURPOSE: To identify key genes differentially expressed in the human retinal pigment epithelium (hRPE) following low-level West Nile virus (WNV) infection. METHODS: Primary hRPE and retinal pigment epithelium cell line (ARPE-19) cells were infected with WNV (multiplicity of infection 1). RNA extracted from mock-infected and WNV-infected cells was assessed for differential expression of genes using Affymetrix microarray. Quantitative real-time PCR analysis of 23 genes was used to validate the microarray results. RESULTS: Functional annotation clustering of the microarray data showed that gene clusters involved in immune and antiviral responses ranked highly, involving genes such as chemokine (C-C motif) ligand 2 (CCL2), chemokine (C-C motif) ligand 5 (CCL5), chemokine (C-X-C motif) ligand 10 (CXCL10), and toll like receptor 3 (TLR3). In conjunction with the quantitative real-time PCR analysis, other novel genes regulated by WNV infection included indoleamine 2,3-dioxygenase (IDO1), genes involved in the transforming growth factor-ß pathway (bone morphogenetic protein and activin membrane-bound inhibitor homolog [BAMBI] and activating transcription factor 3 [ATF3]), and genes involved in apoptosis (tumor necrosis factor receptor superfamily, member 10d [TNFRSF10D]). WNV-infected RPE did not produce any interferon-γ, suggesting that IDO1 is induced by other soluble factors, by the virus alone, or both. CONCLUSIONS: Low-level WNV infection of hRPE cells induced expression of genes that are typically associated with the host cell response to virus infection. We also identified other genes, including IDO1 and BAMBI, that may influence the RPE and therefore outer blood-retinal barrier integrity during ocular infection and inflammation, or are associated with degeneration, as seen for example in aging.


Subject(s)
Epithelial Cells/immunology , Gene Expression/immunology , Retinal Pigment Epithelium/immunology , West Nile virus/physiology , Apoptosis/genetics , Apoptosis/immunology , Chemokines/genetics , Chemokines/immunology , Epithelial Cells/cytology , Epithelial Cells/virology , Gene Expression Profiling , Humans , Membrane Proteins/genetics , Membrane Proteins/immunology , Multigene Family , Oligonucleotide Array Sequence Analysis , Primary Cell Culture , Proteomics , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/virology , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Viral Load
17.
Proc Natl Acad Sci U S A ; 106(27): 11143-8, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19549835

ABSTRACT

Adenoviruses invading the organism via normal digestive or respiratory routes require the Coxsackie-adenovirus receptor (CAR) to infect the epithelial barrier cells. Because CAR is a component of tight junctions and the basolateral membrane and is normally excluded from the apical membrane, most epithelia are resistant to adenoviruses. However, we discovered that a specialized epithelium, the retinal pigment epithelium (RPE), anomalously expressed CAR at the apical surface and was highly susceptible to adenovirus infection. These properties of RPE cells correlated with the absence of the epithelial-specific clathrin adaptor AP1B. Furthermore, knockdown of this basolateral sorting adaptor in adenovirus-resistant MDCK cells promoted apical localization of CAR and increased dramatically Adenovirus infectivity. Targeting assays showed that AP1B is required for accurate basolateral recycling of CAR after internalization. AP1B knock down MDCK cells missorted CAR from recycling endosomes to the apical surface. In summary, we have characterized the cellular machinery responsible for normal sorting of an adenovirus receptor and illustrated how tissue-specific variations in such machinery result in drastic changes in tissue-susceptibility to adenoviruses.


Subject(s)
Adaptor Protein Complex beta Subunits/metabolism , Adenoviridae/pathogenicity , Clathrin/metabolism , Epithelial Cells/virology , Adaptor Protein Complex beta Subunits/deficiency , Animals , Cell Line , Cell Polarity , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Dogs , Endocytosis , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Knockdown Techniques , Humans , Protein Transport , Receptors, Virus/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/virology , Tight Junctions
18.
Clin Exp Ophthalmol ; 40(2): 195-204, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21883773

ABSTRACT

BACKGROUND: This study sought to inhibit herpes simplex virus type 1 replication using small interfering RNA which targeting infected-cell polypeptide 4 genes to mediate transcription of early and late viral genes in herpes simplex virus type 1 lytic (productive) infection in retina epithelial cells. METHODS: After pre- or post-infecting with herpes simplex virus type 1, small interfering RNAs were transfected into retina epithelial cells. The antiviral effects of small interfering RNA were evaluated by Western blot, plaque assays, indirect immunofluorescence and reverse transcription polymerase chain reaction. The viral titre was detected by the 50% tissue culture infective dose method. RESULTS: Small interfering RNA decreased infected-cell polypeptide 4 expression in retina epithelial cells that were infected with herpes simplex virus type 1 before or after small interfering RNA transfection. Compared with herpes simplex virus type 1 infection alone or transfection with negative control small interfering RNA, the viral titre and the retina epithelial cell cytopathic effect were significantly decreased in retina epithelial cells transfected with infected-cell polypeptide 4-targeting small interfering RNA (50 and 100nM) (P<0.05). The small interfering RNA effectively silenced herpes simplex virus type 1 infected-cell polypeptide 4 expression on both mRNA and the protein levels (P<0.05). The inhibition of infected-cell polypeptide 4-targeting small interfering RNA on infected-cell polypeptide 4 protein expression was also verified by Western blot in herpes simplex virus type 1 infected human cornea epithelial cell, human trabecular meshwork cells and Vero cells. CONCLUSIONS: Infected-cell polypeptide 4-targeting small interfering RNA can inhibit herpes simplex virus type 1 replication in retina epithelial cells, providing a foundation for development of RNA interference as an antiviral therapy.


Subject(s)
Gene Silencing/physiology , Herpesvirus 1, Human/physiology , Immediate-Early Proteins/genetics , RNA, Small Interfering/genetics , Retinal Pigment Epithelium/virology , Virus Replication/genetics , Animals , Blotting, Western , Chlorocebus aethiops , Cytopathogenic Effect, Viral , Fluorescent Antibody Technique, Indirect , Gene Targeting , Humans , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Vero Cells , Viral Plaque Assay
19.
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
20.
Biochem J ; 419(3): 533-43, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19191760

ABSTRACT

Haemochromatosis is a genetic disorder of iron overload resulting from loss-of-function mutations in genes coding for the iron-regulatory proteins HFE [HLA-like protein involved in iron (Fe) homoeostasis], transferrin receptor 2, ferroportin, hepcidin and HJV (haemojuvelin). Expression of the first four genes coding for these proteins in retina has been established. Here we report on the expression of HJV. Since infection of retina with CMV (cytomegalovirus) causes blindness, we also investigated the expression of HJV and other iron-regulatory proteins in retina during CMV infection. HJV (HJV gene) mRNA was expressed in RPE (retinal pigment epithelium)/eyecup and neural retina in mouse. In situ hybridization and immunohistochemistry confirmed the presence of HJV mRNA and protein in RPE, outer and inner nuclear layers, and ganglion cell layer. Immunocytochemistry with cell lines and primary cell cultures showed HJV expression in RPE and Müller cells. In RPE, the expression was restricted to apical membrane. Infection of primary cultures of mouse RPE with CMV increased HJV mRNA and protein levels. Under similar conditions, HFE (HFE gene) mRNA levels were not altered, but HFE protein was decreased. Hepcidin expression was, however, not altered. These findings were demonstrable in vivo with CMV-infected mouse retina. The CMV-induced up-regulation of HJV in RPE was independent of changes in HFE because the phenomenon was also seen in HFE-null RPE cells. CMV-infected primary RPE cells showed evidence of iron accumulation and oxidative stress, as indicated by increased levels of ferritin and hydroxynonenal. The observed changes in HJV expression and iron status during CMV infection in retina may have significance in the pathophysiology of CMV retinitis.


Subject(s)
Cytomegalovirus Infections/metabolism , Iron-Regulatory Proteins/metabolism , Membrane Proteins/metabolism , Retina/metabolism , Retina/virology , Animals , Antimicrobial Cationic Peptides/metabolism , Cells, Cultured , GPI-Linked Proteins , Gene Expression Regulation , Hemochromatosis Protein , Hepcidins , Histocompatibility Antigens Class I/metabolism , Iron/metabolism , Membrane Proteins/genetics , Mice , Muromegalovirus/physiology , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/pathology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/virology
SELECTION OF CITATIONS
SEARCH DETAIL