Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 182
Filter
Add more filters

Publication year range
1.
Cell ; 149(4): 847-59, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22541070

ABSTRACT

Alu RNA accumulation due to DICER1 deficiency in the retinal pigmented epithelium (RPE) is implicated in geographic atrophy (GA), an advanced form of age-related macular degeneration that causes blindness in millions of individuals. The mechanism of Alu RNA-induced cytotoxicity is unknown. Here we show that DICER1 deficit or Alu RNA exposure activates the NLRP3 inflammasome and triggers TLR-independent MyD88 signaling via IL18 in the RPE. Genetic or pharmacological inhibition of inflammasome components (NLRP3, Pycard, Caspase-1), MyD88, or IL18 prevents RPE degeneration induced by DICER1 loss or Alu RNA exposure. These findings, coupled with our observation that human GA RPE contains elevated amounts of NLRP3, PYCARD, and IL18 and evidence of increased Caspase-1 and MyD88 activation, provide a rationale for targeting this pathway in GA. Our findings also reveal a function of the inflammasome outside the immune system and an immunomodulatory action of mobile elements.


Subject(s)
Alu Elements , DEAD-box RNA Helicases/metabolism , Geographic Atrophy/immunology , Geographic Atrophy/pathology , Inflammasomes/immunology , Myeloid Differentiation Factor 88/metabolism , Retinal Pigment Epithelium/metabolism , Ribonuclease III/metabolism , Animals , Carrier Proteins/metabolism , Geographic Atrophy/metabolism , Humans , Inflammasomes/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Retinal Pigment Epithelium/pathology , Toll-Like Receptors/metabolism
2.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Article in English | MEDLINE | ID: mdl-33526699

ABSTRACT

Alu retroelements propagate via retrotransposition by hijacking long interspersed nuclear element-1 (L1) reverse transcriptase (RT) and endonuclease activities. Reverse transcription of Alu RNA into complementary DNA (cDNA) is presumed to occur exclusively in the nucleus at the genomic integration site. Whether Alu cDNA is synthesized independently of genomic integration is unknown. Alu RNA promotes retinal pigmented epithelium (RPE) death in geographic atrophy, an untreatable type of age-related macular degeneration. We report that Alu RNA-induced RPE degeneration is mediated via cytoplasmic L1-reverse-transcribed Alu cDNA independently of retrotransposition. Alu RNA did not induce cDNA production or RPE degeneration in L1-inhibited animals or human cells. Alu reverse transcription can be initiated in the cytoplasm via self-priming of Alu RNA. In four health insurance databases, use of nucleoside RT inhibitors was associated with reduced risk of developing atrophic macular degeneration (pooled adjusted hazard ratio, 0.616; 95% confidence interval, 0.493-0.770), thus identifying inhibitors of this Alu replication cycle shunt as potential therapies for a major cause of blindness.


Subject(s)
Alu Elements/genetics , Long Interspersed Nucleotide Elements/genetics , Macular Degeneration/genetics , Retinal Pigments/metabolism , Animals , Cytoplasm/genetics , DNA, Complementary/genetics , Epithelium/metabolism , Epithelium/pathology , Humans , Macular Degeneration/pathology , Retinal Pigments/biosynthesis , Retroelements/genetics , Reverse Transcription/genetics
3.
Acta Neuropathol ; 145(4): 409-438, 2023 04.
Article in English | MEDLINE | ID: mdl-36773106

ABSTRACT

Alzheimer's disease (AD) pathologies were discovered in the accessible neurosensory retina. However, their exact nature and topographical distribution, particularly in the early stages of functional impairment, and how they relate to disease progression in the brain remain largely unknown. To better understand the pathological features of AD in the retina, we conducted an extensive histopathological and biochemical investigation of postmortem retina and brain tissues from 86 human donors. Quantitative examination of superior and inferior temporal retinas from mild cognitive impairment (MCI) and AD patients compared to those with normal cognition (NC) revealed significant increases in amyloid ß-protein (Aß42) forms and novel intraneuronal Aß oligomers (AßOi), which were closely associated with exacerbated retinal macrogliosis, microgliosis, and tissue atrophy. These pathologies were unevenly distributed across retinal layers and geometrical areas, with the inner layers and peripheral subregions exhibiting most pronounced accumulations in the MCI and AD versus NC retinas. While microgliosis was increased in the retina of these patients, the proportion of microglial cells engaging in Aß uptake was reduced. Female AD patients exhibited higher levels of retinal microgliosis than males. Notably, retinal Aß42, S100 calcium-binding protein B+ macrogliosis, and atrophy correlated with severity of brain Aß pathology, tauopathy, and atrophy, and most retinal pathologies reflected Braak staging. All retinal biomarkers correlated with the cognitive scores, with retinal Aß42, far-peripheral AßOi and microgliosis displaying the strongest correlations. Proteomic analysis of AD retinas revealed activation of specific inflammatory and neurodegenerative processes and inhibition of oxidative phosphorylation/mitochondrial, and photoreceptor-related pathways. This study identifies and maps retinopathy in MCI and AD patients, demonstrating the quantitative relationship with brain pathology and cognition, and may lead to reliable retinal biomarkers for noninvasive retinal screening and monitoring of AD.


Subject(s)
Alzheimer Disease , Male , Humans , Female , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Proteome/metabolism , Proteomics , Retina/pathology , Atrophy/pathology , Biomarkers/metabolism
4.
Exp Eye Res ; 215: 108918, 2022 02.
Article in English | MEDLINE | ID: mdl-34986369

ABSTRACT

Oxidative stress in the retinal pigment epithelium (RPE) can cause mitochondrial dysfunction and is likely a causative factor in the pathogenesis of age-related macular degeneration (AMD). Under oxidative stress conditions, some of the RPE cells become senescent and a contributory role for RPE senescence in AMD pathology has been proposed. The purpose of this study is to 1) characterize senescence in human RPE; 2) investigate the effect of an αB Crystallin chaperone peptide (mini Cry) in controlling senescence, in particular by regulating mitochondrial function and senescence-associated secretory phenotype (SASP) production and 3) develop mouse models for studying the role of RPE senescence in dry and nAMD. Senescence was induced in human RPE cells in two ways. First, subconfluent cells were treated with 0.2 µg/ml doxorubicin (DOX); second, subconfluent cells were treated with 500 µM H2O2. Senescence biomarkers (senescence-associated beta-galactosidase (SA-ßgal), p21, p16) and mitochondrial proteins (Fis1, DRP1, MFN2, PGC1-α, mtTFA) were analyzed in control and experimental groups. The effect of mini Cry on mitochondrial bioenergetics, glycolysis and SASP was determined. In vivo, retinal degeneration was induced by intravenous injection of NaIO3 (20 mg/kg) and subretinal fibrosis by laser-induced choroidal neovascularization. Increased SA-ßgal staining and p16 and p21 expression was observed after DOX- or H2O2-induced senescence and mini Cry significantly decreased senescence-positive cells. The expression of mitochondrial biogenesis proteins PGC-1 and mTFA increased with senescence, and mini Cry reduced expression significantly. Senescent RPE cells were metabolically active, as evidenced by significantly enhanced oxidative phosphorylation and anaerobic glycolysis, mini Cry markedly reduced rates of respiration and glycolysis. Senescent RPE cells maintain a proinflammatory phenotype characterized by significantly increased production of cytokines (IFN-Ë , TNF-α, IL1-α IL1-ß, IL-6, IL-8, IL-10), and VEGF-A; mini Cry significantly inhibited their secretion. We identified and localized senescent RPE cells for the first time in NaIO3-induced retinal degeneration and laser-induced subretinal fibrosis mouse models. We conclude that mini Cry significantly impairs stress-induced senescence by modulating mitochondrial biogenesis and fission proteins in RPE cells. Characterization of senescence could provide further understanding of the metabolic changes that accompany the senescent phenotype in ocular disease. Future studies in vivo may better define the role of senescence in AMD and the therapeutic potential of mini Cry as a senotherapeutic.


Subject(s)
Macular Degeneration , Retinal Degeneration , Animals , Cellular Senescence , Disease Models, Animal , Fibrosis , Hydrogen Peroxide/pharmacology , Macular Degeneration/metabolism , Mice , Oxidative Stress , Peptides/pharmacology , Retinal Degeneration/metabolism , Retinal Pigment Epithelium/metabolism , alpha-Crystallin B Chain/genetics
5.
Acta Neuropathol ; 139(5): 813-836, 2020 05.
Article in English | MEDLINE | ID: mdl-32043162

ABSTRACT

Pericyte loss and deficient vascular platelet-derived growth factor receptor-ß (PDGFRß) signaling are prominent features of the blood-brain barrier breakdown described in Alzheimer's disease (AD) that can predict cognitive decline yet have never been studied in the retina. Recent reports using noninvasive retinal amyloid imaging, optical coherence tomography angiography, and histological examinations support the existence of vascular-structural abnormalities and vascular amyloid ß-protein (Aß) deposits in retinas of AD patients. However, the cellular and molecular mechanisms of such retinal vascular pathology were not previously explored. Here, by modifying a method of enzymatically clearing non-vascular retinal tissue and fluorescent immunolabeling of the isolated blood vessel network, we identified substantial pericyte loss together with significant Aß deposition in retinal microvasculature and pericytes in AD. Evaluation of postmortem retinas from a cohort of 56 human donors revealed an early and progressive decrease in vascular PDGFRß in mild cognitive impairment (MCI) and AD compared to cognitively normal controls. Retinal PDGFRß loss significantly associated with increased retinal vascular Aß40 and Aß42 burden. Decreased vascular LRP-1 and early apoptosis of pericytes in AD retina were also detected. Mapping of PDGFRß and Aß40 levels in pre-defined retinal subregions indicated that certain geometrical and cellular layers are more susceptible to AD pathology. Further, correlations were identified between retinal vascular abnormalities and cerebral Aß burden, cerebral amyloid angiopathy (CAA), and clinical status. Overall, the identification of pericyte and PDGFRß loss accompanying increased vascular amyloidosis in Alzheimer's retina implies compromised blood-retinal barrier integrity and provides new targets for AD diagnosis and therapy.


Subject(s)
Alzheimer Disease/pathology , Amyloidosis/pathology , Brain/pathology , Pericytes/pathology , Retina/pathology , Aged , Aged, 80 and over , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloidosis/complications , Blood-Brain Barrier/pathology , Cerebral Amyloid Angiopathy/pathology , Cognition/physiology , Female , Humans , Male
6.
Nanomedicine ; 24: 102111, 2020 02.
Article in English | MEDLINE | ID: mdl-31655204

ABSTRACT

Humanin (HN) is a hydrophobic 24-amino acid peptide derived from mitochondrial DNA that modulates cellular responses to oxidative stress and protects human retinal pigment epithelium (RPE) cells from apoptosis. To solubilize HN, this report describes two genetically-encoded fusions between HN and elastin-like polypeptides (ELP). ELPs provide steric stabilization and/or thermo-responsive phase separation. Fusions were designed to either remain soluble or phase separate at the physiological temperature of the retina. Interestingly, the soluble fusion assembles stable colloids with a hydrodynamic radius of 39.1 nm at 37°C. As intended, the thermo-responsive fusion forms large coacervates (>1,000 nm) at 37°C. Both fusions bind human RPE cells and protect against oxidative stress-induction of apoptosis (TUNEL, caspase-3 activation). Their activity is mediated through STAT3; furthermore, STAT3 inhibition eliminates their protection. These findings suggest that HN polypeptides may facilitate cellular delivery of biodegradable nanoparticles with potential protection against age-related diseases, including macular degeneration.


Subject(s)
Elastin , Epithelial Cells/metabolism , Intracellular Signaling Peptides and Proteins , Nanoparticles/chemistry , Oxidative Stress/drug effects , Peptides , Retinal Pigment Epithelium/metabolism , Apoptosis/drug effects , Cells, Cultured , Elastin/chemistry , Elastin/pharmacology , Epithelial Cells/pathology , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/pharmacology , Macular Degeneration/metabolism , Macular Degeneration/pathology , Peptides/chemistry , Peptides/pharmacology , Retinal Pigment Epithelium/pathology
7.
Retina ; 39(2): 265-273, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29190236

ABSTRACT

PURPOSE: We sought to characterize the angiofibrotic and apoptotic effects of vascular endothelial growth factor (VEGF)-inhibition on fibrovascular epiretinal membranes in eyes with traction retinal detachment because of proliferative diabetic retinopathy. METHODS: Membranes were excised from 20 eyes of 19 patients (10 randomized to intravitreal bevacizumab, 10 controls) at vitrectomy. Membranes were stained with antibodies targeting connective tissue growth factor (CTGF) or VEGF and colabeled with antibodies directed against endothelial cells (CD31), myofibroblasts, or retinal pigment epithelium markers. Quantitative and colocalization analyses of antibody labeling were obtained through immunofluorescence confocal microscopy. Masson trichrome staining, cell counting of hematoxylin and eosin sections, and terminal dUTP nick-end labeling staining were performed. RESULTS: High levels of fibrosis were observed in both groups. Cell apoptosis was higher (P = 0.05) in bevacizumab-treated membranes compared with controls. The bevacizumab group had a nonsignificant reduction in colocalization in CD31-CTGF and cytokeratin-VEGF studies compared with controls. Vascular endothelial growth factor in extracted membranes was positively correlated with vitreous levels of VEGF; CTGF in extracted membranes was negatively correlated with vitreous levels of CTGF. CONCLUSION: Bevacizumab suppresses vitreous VEGF levels, but does not significantly alter VEGF or CTGF in diabetic membranes that may be explained by high baseline levels of fibrosis. Bevacizumab may cause apoptosis within fibrovascular membranes.


Subject(s)
Apoptosis , Bevacizumab/administration & dosage , Diabetic Retinopathy/pathology , Epiretinal Membrane/surgery , Retina/pathology , Vitrectomy/methods , Actins/biosynthesis , Angiogenesis Inhibitors/administration & dosage , Cell Proliferation , Connective Tissue Growth Factor/biosynthesis , Diabetic Retinopathy/complications , Diabetic Retinopathy/drug therapy , Epiretinal Membrane/complications , Epiretinal Membrane/pathology , Fibrosis/pathology , Humans , Intravitreal Injections , Keratins/biosynthesis , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Prospective Studies , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Retina/metabolism , Vascular Endothelial Growth Factor A/biosynthesis
8.
Int J Mol Sci ; 20(19)2019 Sep 27.
Article in English | MEDLINE | ID: mdl-31569695

ABSTRACT

Age-related macular degeneration (AMD) is a leading cause of blindness in the developed world. The retinal pigment epithelium (RPE) is a critical site of pathology in AMD. Oxidative stress plays a key role in the development of AMD. We generated a chimeric high-density lipoprotein (HDL), mimetic peptide named HM-10/10, with anti-oxidant properties and investigated its potential for the treatment of retinal disease using cell culture and animal models of RPE and photoreceptor (PR) degeneration. Treatment with HM-10/10 peptide prevented human fetal RPE cell death caused by tert-Butyl hydroperoxide (tBH)-induced oxidative stress and sodium iodate (NaIO3), which causes RPE atrophy and is a model of geographic atrophy in mice. We also show that HM-10/10 peptide ameliorated photoreceptor cell death and significantly improved retinal function in a mouse model of N-methyl-N-nitrosourea (MNU)-induced PR degeneration. Our results demonstrate that HM-10/10 protects RPE and retina from oxidant injury and can serve as a potential therapeutic agent for the treatment of retinal degeneration.


Subject(s)
Lipoproteins, HDL/metabolism , Peptides/pharmacology , Photoreceptor Cells/drug effects , Photoreceptor Cells/metabolism , Retinal Degeneration/metabolism , Retinal Pigment Epithelium/metabolism , Animals , Apoptosis , Caspase 3/metabolism , Caspase 7/metabolism , Disease Models, Animal , Iodates , Mice , Oxidative Stress/drug effects , Retinal Degeneration/diagnosis , Retinal Degeneration/etiology , Retinal Pigment Epithelium/pathology , Tomography, Optical Coherence
9.
Int J Cancer ; 143(11): 2932-2942, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29978915

ABSTRACT

We previously described an in vitro model in which serous ovarian cystadenomas were transfected with SV40 large T antigen, resulting in loss of RB and P53 functions and thus mimicking genetic defects present in early high-grade serous extra-uterine Müllerian (traditionally called high-grade serous ovarian) carcinomas including those associated with the BRCA1 mutation carrier state. We showed that replicative aging in this cell culture model leads to a mitotic arrest at the spindle assembly checkpoint. Here we show that this arrest is due to a reduction in microtubule anchoring that coincides with decreased expression of the BUB1 kinase and of the phosphorylated form of its substrate, BUB3. The ensuing prolonged mitotic arrest leads to cohesion fatigue resulting in cell death or, in cells that recover from this arrest, in cytokinesis failure and polyploidy. Down-regulation of BRCA1 to levels similar to those present in BRCA1 mutation carriers leads to increased and uncontrolled microtubule anchoring to the kinetochore resulting in overcoming the spindle assembly checkpoint. Progression to anaphase under those conditions is associated with formation of chromatin bridges between chromosomal plates due to abnormal attachments to the kinetochore, significantly increasing the risk of cytokinesis failure. The dependence of this scenario on accelerated replicative aging can, at least in part, account for the site specificity of the cancers associated with the BRCA1 mutation carrier state, as epithelia of the mammary gland and of the reproductive tract are targets of cell-nonautonomous consequences of this carrier state on cellular proliferation associated with menstrual cycle progressions.


Subject(s)
BRCA1 Protein/genetics , Cystadenoma/genetics , Cytokinesis/genetics , Ovarian Neoplasms/genetics , Signal Transduction/genetics , Tumor Suppressor Protein p53/genetics , Cell Cycle Proteins/genetics , Chromosomes/genetics , Female , Humans , Microtubules/genetics , Mitosis/genetics , Polyploidy , Spindle Apparatus/genetics
10.
Graefes Arch Clin Exp Ophthalmol ; 256(11): 2113-2125, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30215097

ABSTRACT

PURPOSE: To create new immunodeficient Royal College of Surgeons (RCS) rats by introducing the defective MerTK gene into athymic nude rats. METHODS: Female homozygous RCS (RCS-p+/RCS-p+) and male nude rats (Hsd:RH-Foxn1mu, mutation in the foxn1 gene; no T cells) were crossed to produce heterozygous F1 progeny. Double homozygous F2 progeny obtained by crossing the F1 heterozygotes was identified phenotypically (hair loss) and genotypically (RCS-p+ gene determined by PCR). Retinal degenerative status was confirmed by optical coherence tomography (OCT) imaging, electroretinography (ERG), optokinetic (OKN) testing, superior colliculus (SC) electrophysiology, and by histology. The effect of xenografts was assessed by transplantation of human embryonic stem cell-derived retinal pigment epithelium (hESC-RPE) and human-induced pluripotent stem cell-derived RPE (iPS-RPE) into the eye. Morphological analysis was conducted based on hematoxylin and eosin (H&E) and immunostaining. Age-matched pigmented athymic nude rats were used as control. RESULTS: Approximately 6% of the F2 pups (11/172) were homozygous for RCS-p+ gene and Foxn1mu gene. Homozygous males crossed with heterozygous females resulted in 50% homozygous progeny for experimentation. OCT imaging demonstrated significant loss of retinal thickness in homozygous rats. H&E staining showed photoreceptor thickness reduced to 1-3 layers at 12 weeks of age. Progressive loss of visual function was evidenced by OKN testing, ERG, and SC electrophysiology. Transplantation experiments demonstrated survival of human-derived cells and absence of apparent immune rejection. CONCLUSIONS: This new rat animal model developed by crossing RCS rats and athymic nude rats is suitable for conducting retinal transplantation experiments involving xenografts.


Subject(s)
Disease Models, Animal , Human Embryonic Stem Cells/transplantation , Immunologic Deficiency Syndromes/therapy , Induced Pluripotent Stem Cells/transplantation , Retinal Dystrophies/therapy , Retinal Pigment Epithelium/transplantation , Animals , Cell Survival , Electroretinography , Female , Genotyping Techniques , Graft Survival/physiology , Human Embryonic Stem Cells/physiology , Humans , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/physiopathology , Induced Pluripotent Stem Cells/physiology , Male , Phenotype , Rats , Rats, Nude , Retina/physiopathology , Retinal Dystrophies/diagnosis , Retinal Dystrophies/physiopathology , Retinal Pigment Epithelium/physiology , Tomography, Optical Coherence , c-Mer Tyrosine Kinase/genetics
11.
Biochim Biophys Acta ; 1860(1 Pt B): 258-68, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26026469

ABSTRACT

BACKGROUND: αA- and αB crystallins are principal members of the small heat shock protein family and elicit both a cell protective function and a chaperone function. α-Crystallins have been found to be prominent proteins in normal and pathological retina emphasizing the importance for in-depth understanding of their function and significance. SCOPE OF REVIEW: Retinal pigment epithelial cells (RPE) play a vital role in the pathogenesis of age-related macular degeneration (AMD). This review addresses a number of cellular functions mediated by α-crystallins in the retina. Prominent expression of αB crystallin in mitochondria may serve to protect cells from oxidative injury. αB crystallin as secretory protein via exosomes can offer neuroprotection to adjacent RPE cells and photoreceptors. The availability of chaperone-containing minipeptides of αB crystallin could prove to be a valuable new tool for therapeutic treatment of retinal disorders. MAJOR CONCLUSIONS: α-Crystallins are expressed in cytosol and mitochondria of RPE cells and are regulated during oxygen-induced retinopathy and during development. α-Crystallins protect RPE from oxidative-and ER stress-induced injury and autophagy. αB-Crystallin is a modulator of angiogenesis and vascular endothelial growth factor. αB Crystallin is secreted via exosomal pathway. Minichaperone peptides derived from αB Crystallin prevent oxidant induced cell death and have therapeutic potential. GENERAL SIGNIFICANCE: Overall, this review summarizes several novel properties of α-crystallins and their relevance to maintaining normal retinal function. In particular, the use of α-crystallin derived peptides is a promising therapeutic strategy to combat retinal diseases such as AMD. This article is part of a Special Issue entitled Crystallin biochemistry in health and disease.


Subject(s)
Macular Degeneration/drug therapy , Oxidative Stress/drug effects , Peptides/therapeutic use , Retinal Pigment Epithelium/metabolism , alpha-Crystallins/metabolism , alpha-Crystallins/therapeutic use , Animals , Humans , Molecular Chaperones/chemistry , Molecular Chaperones/therapeutic use , Peptides/chemistry , Retinal Pigment Epithelium/drug effects , alpha-Crystallins/chemistry
12.
Am J Pathol ; 186(4): 859-73, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26878210

ABSTRACT

Subretinal fibrosis is an end stage of neovascular age-related macular degeneration, characterized by fibrous membrane formation after choroidal neovascularization. An initial step of the pathogenesis is an epithelial-mesenchymal transition (EMT) of retinal pigment epithelium cells. αB-crystallin plays multiple roles in age-related macular degeneration, including cytoprotection and angiogenesis. However, the role of αB-crystallin in subretinal EMT and fibrosis is unknown. Herein, we showed attenuation of subretinal fibrosis after regression of laser-induced choroidal neovascularization and a decrease in mesenchymal retinal pigment epithelium cells in αB-crystallin knockout mice compared with wild-type mice. αB-crystallin was prominently expressed in subretinal fibrotic lesions in mice. In vitro, overexpression of αB-crystallin induced EMT, whereas suppression of αB-crystallin induced a mesenchymal-epithelial transition. Transforming growth factor-ß2-induced EMT was further enhanced by overexpression of αB-crystallin but was inhibited by suppression of αB-crystallin. Silencing of αB-crystallin inhibited multiple fibrotic processes, including cell proliferation, migration, and fibronectin production. Bone morphogenetic protein 4 up-regulated αB-crystallin, and its EMT induction was inhibited by knockdown of αB-crystallin. Furthermore, inhibition of αB-crystallin enhanced monotetraubiquitination of SMAD4, which can impair its nuclear localization. Overexpression of αB-crystallin enhanced nuclear translocation and accumulation of SMAD4 and SMAD5. Thus, αB-crystallin is an important regulator of EMT, acting as a molecular chaperone for SMAD4 and as its potential therapeutic target for preventing subretinal fibrosis development in neovascular age-related macular degeneration.


Subject(s)
Choroidal Neovascularization/metabolism , Epithelial-Mesenchymal Transition/genetics , Fibrosis/metabolism , Macular Degeneration/pathology , Retinal Pigment Epithelium/metabolism , alpha-Crystallin B Chain/metabolism , Animals , Choroidal Neovascularization/genetics , Fibronectins/metabolism , Humans , Macular Degeneration/genetics , Male , Mice, Knockout , Retinal Pigment Epithelium/pathology , alpha-Crystallin B Chain/genetics
13.
Nature ; 471(7338): 325-30, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21297615

ABSTRACT

Geographic atrophy (GA), an untreatable advanced form of age-related macular degeneration, results from retinal pigmented epithelium (RPE) cell degeneration. Here we show that the microRNA (miRNA)-processing enzyme DICER1 is reduced in the RPE of humans with GA, and that conditional ablation of Dicer1, but not seven other miRNA-processing enzymes, induces RPE degeneration in mice. DICER1 knockdown induces accumulation of Alu RNA in human RPE cells and Alu-like B1 and B2 RNAs in mouse RPE. Alu RNA is increased in the RPE of humans with GA, and this pathogenic RNA induces human RPE cytotoxicity and RPE degeneration in mice. Antisense oligonucleotides targeting Alu/B1/B2 RNAs prevent DICER1 depletion-induced RPE degeneration despite global miRNA downregulation. DICER1 degrades Alu RNA, and this digested Alu RNA cannot induce RPE degeneration in mice. These findings reveal a miRNA-independent cell survival function for DICER1 involving retrotransposon transcript degradation, show that Alu RNA can directly cause human pathology, and identify new targets for a major cause of blindness.


Subject(s)
Alu Elements/genetics , DEAD-box RNA Helicases/deficiency , Macular Degeneration/genetics , Macular Degeneration/pathology , RNA/genetics , RNA/metabolism , Ribonuclease III/deficiency , Animals , Cell Death , Cell Survival , Cells, Cultured , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Gene Knockdown Techniques , Humans , Mice , MicroRNAs/metabolism , Molecular Sequence Data , Oligonucleotides, Antisense , Phenotype , Retinal Pigment Epithelium/enzymology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Ribonuclease III/genetics , Ribonuclease III/metabolism
14.
Proc Natl Acad Sci U S A ; 111(47): 16754-9, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25385631

ABSTRACT

The retinal pigmented epithelial (RPE) layer is one of the major ocular tissues affected by oxidative stress and is known to play an important role in the etiology of age-related macular degeneration (AMD), the major cause of blinding in the elderly. In the present study, sulindac, a nonsteroidal antiinflammatory drug (NSAID), was tested for protection against oxidative stress-induced damage in an established RPE cell line (ARPE-19). Besides its established antiinflammatory activity, sulindac has previously been shown to protect cardiac tissue against ischemia/reperfusion damage, although the exact mechanism was not elucidated. As shown here, sulindac can also protect RPE cells from chemical oxidative damage or UV light by initiating a protective mechanism similar to what is observed in ischemic preconditioning (IPC) response. The mechanism of protection appears to be triggered by reactive oxygen species (ROS) and involves known IPC signaling components such as PKG and PKC epsilon in addition to the mitochondrial ATP-sensitive K(+) channel. Sulindac induced iNOS and Hsp70, late-phase IPC markers in the RPE cells. A unique feature of the sulindac protective response is that it involves activation of the peroxisome proliferator-activated receptor alpha (PPAR-α). We have also used low-passage human fetal RPE and polarized primary fetal RPE cells to validate the basic observation that sulindac can protect retinal cells against oxidative stress. These findings indicate a mechanism for preventing oxidative stress in RPE cells and suggest that sulindac could be used therapeutically for slowing the progression of AMD.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , PPAR alpha/physiology , Retinal Pigment Epithelium/drug effects , Sulindac/pharmacology , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress
15.
Proc Natl Acad Sci U S A ; 111(45): 16082-7, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25349431

ABSTRACT

Geographic atrophy, an advanced form of age-related macular degeneration (AMD) characterized by death of the retinal pigmented epithelium (RPE), causes untreatable blindness in millions worldwide. The RPE of human eyes with geographic atrophy accumulates toxic Alu RNA in response to a deficit in the enzyme DICER1, which in turn leads to activation of the NLRP3 inflammasome and elaboration of IL-18. Despite these recent insights, it is still unclear how RPE cells die during the course of the disease. In this study, we implicate the involvement of Caspase-8 as a critical mediator of RPE degeneration. Here we show that DICER1 deficiency, Alu RNA accumulation, and IL-18 up-regulation lead to RPE cell death via activation of Caspase-8 through a Fas ligand-dependent mechanism. Coupled with our observation of increased Caspase-8 expression in the RPE of human eyes with geographic atrophy, our findings provide a rationale for targeting this apoptotic pathway in this disease.


Subject(s)
Alu Elements , Apoptosis , Caspase 8/metabolism , DEAD-box RNA Helicases/metabolism , Eye Proteins/metabolism , Macular Degeneration/metabolism , RNA/metabolism , Ribonuclease III/metabolism , Animals , Caspase 8/genetics , DEAD-box RNA Helicases/genetics , Eye Proteins/genetics , Humans , Interleukin-18/genetics , Interleukin-18/metabolism , Macular Degeneration/pathology , Mice , Mice, Knockout , RNA/genetics , Ribonuclease III/genetics , Up-Regulation/genetics
16.
J Neuroinflammation ; 13: 46, 2016 Feb 22.
Article in English | MEDLINE | ID: mdl-26906225

ABSTRACT

BACKGROUND: Tumor necrosis factor (TNF) has pleiotropic functions during both the demyelinating autoimmune disease multiple sclerosis (MS) and its murine model experimental autoimmune encephalomyelitis (EAE). How TNF regulates disability during progressive disease remains unresolved. Using a progressive EAE model characterized by sustained TNF and increasing morbidity, this study evaluates the role of unregulated TNF in exacerbating central nervous system (CNS) pathology and inflammation. METHODS: Progressive MS was mimicked by myelin oligodendrocyte glycoprotein (MOG) peptide immunization of mice expressing a dominant negative IFN-γ receptor alpha chain under the human glial fibrillary acidic protein promoter (GFAPγR1∆). Diseased GFAPγR1∆ mice were treated with anti-TNF or control monoclonal antibody during acute disease to monitor therapeutic effects on sustained disability, demyelination, CNS inflammation, and blood brain barrier (BBB) permeability. RESULTS: TNF was specifically sustained in infiltrating macrophages. Anti-TNF treatment decreased established clinical disability and mortality rate within 7 days. Control of disease progression was associated with a decline in myelin loss and leukocyte infiltration, as well as macrophage activation. In addition to mitigating CNS inflammation, TNF neutralization restored BBB integrity and enhanced CNS anti-inflammatory responses. CONCLUSIONS: Sustained TNF production by infiltrating macrophages associated with progressive EAE exacerbates disease severity by promoting inflammation and disruption of BBB integrity, thereby counteracting establishment of an anti-inflammatory environment required for disease remission.


Subject(s)
Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Macrophages/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Antibodies/pharmacology , Antigens, CD/metabolism , Blood-Brain Barrier/physiopathology , Calcium-Binding Proteins/metabolism , Capillary Permeability/genetics , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Mice , Mice, Transgenic , Microfilament Proteins/metabolism , Myelin-Oligodendrocyte Glycoprotein/toxicity , Neuroglia/pathology , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/genetics , Peptide Fragments/toxicity , Tumor Necrosis Factor-alpha/immunology
17.
J Virol ; 89(18): 9299-312, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26136579

ABSTRACT

UNLABELLED: Myd88 signaling is critical to the control of numerous central nervous system (CNS) infections by promoting both innate and adaptive immune responses. Nevertheless, the extent to which Myd88 regulates type I interferon (IFN) versus proinflammatory factors and T cell function, as well as the anatomical site of action, varies extensively with the pathogen. CNS infection by neurotropic coronavirus with replication confined to the brain and spinal cord induces protective IFN-α/ß via Myd88-independent activation of melanoma differentiation-associated gene 5 (MDA5). However, a contribution of Myd88-dependent signals to CNS pathogenesis has not been assessed. Infected Myd88(-/-) mice failed to control virus, exhibited enhanced clinical disease coincident with increased demyelination, and succumbed to infection within 3 weeks. The induction of IFN-α/ß, as well as of proinflammatory cytokines and chemokines, was impaired early during infection. However, defects in both IFN-α/ß and select proinflammatory factors were rapidly overcome prior to T cell recruitment. Myd88 deficiency also specifically blunted myeloid and CD4 T cell recruitment into the CNS without affecting CD8 T cells. Moreover, CD4 T cells but not CD8 T cells were impaired in IFN-γ production. Ineffective virus control indeed correlated most prominently with reduced antiviral IFN-γ in the CNS of Myd88(-/-) mice. The results demonstrate a crucial role for Myd88 both in early induction of innate immune responses during coronavirus-induced encephalomyelitis and in specifically promoting protective CD4 T cell activation. In the absence of these responses, functional CD8 T cells are insufficient to control viral spread within the CNS, resulting in severe demyelination. IMPORTANCE: During central nervous system (CNS) infections, signaling through the adaptor protein Myd88 promotes both innate and adaptive immune responses. The extent to which Myd88 regulates antiviral type I IFN, proinflammatory factors, adaptive immunity, and pathology is pathogen dependent. These results reveal that Myd88 protects from lethal neurotropic coronavirus-induced encephalomyelitis by accelerating but not enhancing the induction of IFN-α/ß, as well as by promoting peripheral activation and CNS accumulation of virus-specific CD4 T cells secreting IFN-γ. By controlling both early innate immune responses and CD4 T cell-mediated antiviral IFN-γ, Myd88 signaling limits the initial viral dissemination and is vital for T cell-mediated control of viral loads. Uncontrolled viral replication in the absence of Myd88 leads to severe demyelination and pathology despite overall reduced inflammatory responses. These data support a vital role of Myd88 signaling in protective antimicrobial functions in the CNS by promoting proinflammatory mediators and T cell-mediated IFN-γ production.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Coronavirus Infections/immunology , Encephalitis, Viral/immunology , Immunity, Cellular , Immunity, Innate , Maus Elberfeld virus/immunology , Myeloid Differentiation Factor 88/immunology , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Coronavirus Infections/genetics , Coronavirus Infections/pathology , Encephalitis, Viral/genetics , Encephalitis, Viral/pathology , Interferon-alpha/genetics , Interferon-alpha/immunology , Interferon-beta/genetics , Interferon-beta/immunology , Maus Elberfeld virus/genetics , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics
18.
Mol Vis ; 22: 213-23, 2016.
Article in English | MEDLINE | ID: mdl-27011730

ABSTRACT

PURPOSE: Rare mutations in the human RGR gene lead to autosomal recessive retinitis pigmentosa or dominantly inherited peripapillary choroidal atrophy. Here, we analyze a common exon-skipping isoform of the human retinal G protein-coupled receptor opsin (RGR-d) to determine differences in subcellular targeting between RGR-d and normal RGR and possible association with abnormal traits in the human eye. METHODS: The terminal complement complex (C5b-9), vitronectin, CD46, syntaxin-4, and RGR-d were analyzed in human eye tissue from young and old donors or in cultured fetal RPE cells by means of immunofluorescent labeling and high-resolution confocal microscopy or immunohistochemical staining. RESULTS: We observed that RGR-d is targeted to the basolateral plasma membrane of the RPE. RGR-d, but not normal RGR, is expressed in cultured human fetal RPE cells in which the protein also trafficks to the plasma membrane. In young donors, the amount of RGR-d protein in the basolateral plasma membrane was much higher than that in the RPE cells of older subjects. In older donor eyes, the level of immunoreactive RGR-d within RPE cells was often low or undetectable, and immunostaining of RGR-d was consistently strongest in extracellular deposits in Bruch's membrane. Double immunofluorescent labeling in the basal deposits revealed significant aggregate and small punctate co-localization of RGR-d with C5b-9 and vitronectin. CONCLUSIONS: RGR-d may escape endoplasmic reticulum-associated degradation and in contrast to full-length RGR, traffick to the basolateral plasma membrane, particularly in younger subjects. RGR-d in the plasma membrane indicates that the protein is properly folded, as misfolded membrane proteins cannot otherwise sort to the plasma membrane. The close association of extracellular RGR-d with both vitronectin and C5b-9 suggests a potential role of RGR-d-containing deposits in complement activation.


Subject(s)
Alternative Splicing/physiology , Cell Membrane/metabolism , Complement Membrane Attack Complex/metabolism , Eye Proteins/metabolism , Opsins/metabolism , Receptors, G-Protein-Coupled/metabolism , Retinal Pigment Epithelium/metabolism , Aged , Blotting, Western , Bruch Membrane/metabolism , Cells, Cultured , Exons/genetics , Eye Proteins/genetics , Female , Fluorescent Antibody Technique, Indirect , Genetic Vectors , Humans , Male , Membrane Cofactor Protein/metabolism , Microscopy, Confocal , Middle Aged , RNA, Messenger/genetics , Receptors, G-Protein-Coupled/genetics , Tissue Donors , Transfection , Vitronectin/metabolism
19.
Exp Eye Res ; 142: 19-25, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25773985

ABSTRACT

Subretinal fibrosis is a result of a wound healing response that follows choroidal neovascularization in neovascular age-related macular degeneration (nAMD). Although anti-vascular endothelial growth factor therapy has become a standard treatment that improves visual acuity in many nAMD patients, unsuccessful treatment outcomes have often been attributed to the progression of subretinal fibrosis. In this review, we summarize the cellular and extracellular components of subretinal fibrous membranes and also discuss the possible molecular mechanisms including the functional involvement of growth factors and the inflammatory response in the process. Moreover, we present an murine animal model of subretinal fibrosis that might facilitate greater understanding of the pathophysiology and the development of novel therapeutic strategies for the inhibition of subretinal fibrosis in nAMD.


Subject(s)
Fibrosis/complications , Macular Degeneration , Angiogenesis Inhibitors/therapeutic use , Animals , Cytokines/metabolism , Disease Models, Animal , Epithelial-Mesenchymal Transition/physiology , Extracellular Matrix/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Macular Degeneration/drug therapy , Macular Degeneration/immunology , Macular Degeneration/metabolism , Macular Degeneration/physiopathology , Mice , Molecular Targeted Therapy/methods , Retinal Pigment Epithelium/pathology
20.
J Immunol ; 193(1): 285-94, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24890725

ABSTRACT

IL-27 is a pleiotropic member of the IL-6 and IL-12 cytokine family composed of the IL-27p28 and the EBV-induced gene 3. IL-27 and its receptor mRNA are both upregulated in the CNS during acute encephalomyelitis induced by the JHM strain of mouse hepatitis virus (JHMV) and sustained during viral persistence. Contributions of IL-27 to viral pathogenesis were evaluated by infection of IL-27Rα-chain-deficient (IL-27Rα(-/-)) mice. The absence of IL-27 signaling accelerated virus control within the CNS associated with increased IFN-γ secreting virus-specific CD4+ and CD8+ T cells. Abrogation of IL-27 signaling did not affect virus-specific CD8+ T cell-mediated IL-10 production or cytolytic activity or Foxp3+ regulatory T cell populations. However, IL-10 production by virus-specific CD4+ T cells was reduced significantly. Despite increased T cell-mediated antiviral function in IL-27Rα(-/-) mice, the virus persisted in the CNS at similar levels as in wild-type mice. Nevertheless, IL-27Rα(-/-) mice exhibited decreased clinical disease during persistence, coincident with less severe demyelination, the hallmark tissue damage associated with JHMV infection. Overall, these data demonstrate that in contrast to viral infections at other sites, IL-27 does not play a proinflammatory role during JHMV-induced encephalomyelitis. Rather, it limits CNS inflammation and impairs control of CNS virus replication via induction of IL-10 in virus-specific CD4+ T cells. Furthermore, in contrast to its protective role in limiting CNS autoimmunity and preventing immunopathology, these data define a detrimental role of IL-27 in promoting demyelination by delaying viral control.


Subject(s)
Central Nervous System/immunology , Coronavirus Infections/immunology , Encephalomyelitis, Acute Disseminated/immunology , Interleukin-10/immunology , Interleukins/immunology , Murine hepatitis virus/immunology , Signal Transduction/immunology , Animals , Central Nervous System/pathology , Coronavirus Infections/genetics , Coronavirus Infections/pathology , Demyelinating Diseases , Encephalomyelitis, Acute Disseminated/genetics , Encephalomyelitis, Acute Disseminated/pathology , Interleukin-10/genetics , Interleukins/genetics , Mice , Mice, Knockout , Signal Transduction/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
SELECTION OF CITATIONS
SEARCH DETAIL