ABSTRACT
DNA damage accumulates in aged postmitotic retinal pigment epithelium (RPE) cells, a phenomenon associated with the development of age-related macular degeneration. In this study, we have experimentally induced DNA damage by ultraviolet B (UVB) irradiation in interleukin-1α (IL-1α)-primed ARPE-19 cells and examined inflammasome-mediated signaling. To reveal the mechanisms of inflammasome activation, cells were additionally exposed to high levels of extracellular potassium chloride, n-acetyl-cysteine, or mitochondria-targeted antioxidant MitoTEMPO, prior to UVB irradiation. Levels of interleukin-18 (IL-18) and IL-1ß mRNAs were detected with qRT-PCR and secreted amounts of IL-1ß, IL-18, and caspase-1 were measured with ELISA. The role of nucleotide-binding domain and leucine-rich repeat pyrin containing protein 3 (NLRP3) in UVB-induced inflammasome activation was verified by using the NLRP3-specific siRNA. Reactive oxygen species (ROS) levels were measured immediately after UVB exposure using the cell-permeant 2',7'-dichlorodihydrofluorescein diacetate (H2 DCFDA) indicator, the levels of cyclobutane pyrimidine dimers were assayed by cell-based ELISA, and the extracellular levels of adenosine triphosphate (ATP) determined using a commercial bioluminescence assay. We found that pro-IL-18 was constitutively expressed by ARPE-19 cells, whereas the expression of pro-IL-1ß was inducible by IL-1α priming. UVB induced the release of mature IL-18 and IL-1ß but NLRP3 contributed only to the secretion of IL-1ß. At the mechanistic level, the release of IL-1ß was regulated by K+ efflux, whereas the secretion of IL-18 was dependent on ROS production. As well as K+ efflux, the cells released ATP following UVB exposure. Collectively, our data suggest that UVB clearly stimulates the secretion of mature IL-18 as a result of ROS induction, and this response is associated with DNA damage. Moreover, in human RPE cells, K+ efflux mediates the UVB-activated NLRP3 inflammasome signaling, leading to the processing of IL-1ß.
Subject(s)
Inflammasomes/metabolism , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Retinal Pigment Epithelium/metabolism , Ultraviolet Rays , DNA Damage , DNA Repair , Humans , Inflammasomes/immunology , Inflammasomes/radiation effects , Reactive Oxygen Species/metabolism , Retinal Pigment Epithelium/immunology , Retinal Pigment Epithelium/radiation effects , Signal TransductionABSTRACT
Age-related macular degeneration (AMD), the leading cause of vision loss in the elderly, is a degenerative disease of the macula, where retinal pigment epithelium (RPE) cells are damaged in the early stages of the disease, and chronic inflammatory processes may be involved. Besides aging and lifestyle factors as drivers of AMD, a strong genetic association to AMD is found in genes of the complement system, with a single polymorphism in the complement factor H gene (CFH), accounting for the majority of AMD risk. However, the exact mechanism of CFH dysregulation confers such a great risk for AMD and its role in RPE cell homeostasis is unclear. To explore the role of endogenous CFH locally in RPE cells, we silenced CFH in human hTERT-RPE1 cells. We demonstrate that endogenously expressed CFH in RPE cells modulates inflammatory cytokine production and complement regulation, independent of external complement sources, or stressors. We show that loss of the factor H protein (FH) results in increased levels of inflammatory mediators (e.g., IL-6, IL-8, GM-CSF) and altered levels of complement proteins (e.g., C3, CFB upregulation, and C5 downregulation) that are known to play a role in AMD. Moreover, our results identify the NF-κB pathway as the major pathway involved in regulating these inflammatory and complement factors. Our findings suggest that in RPE cells, FH and the NF-κB pathway work in synergy to maintain inflammatory and complement balance, and in case either one of them is dysregulated, the RPE microenvironment changes towards a proinflammatory AMD-like phenotype.
Subject(s)
Cytokines/metabolism , Gene Silencing , Macular Degeneration/genetics , Retinal Pigment Epithelium/immunology , Cell Line , Complement Factor H/genetics , Complement System Proteins/metabolism , Cytokines/genetics , Gene Expression Regulation , Humans , Macular Degeneration/immunology , Models, Biological , NF-kappa B/genetics , NF-kappa B/metabolism , Retinal Pigment Epithelium/metabolism , Signal TransductionABSTRACT
The choroid plexus epithelium within the brain ventricles orchestrates blood-derived monocyte entry to the central nervous system under injurious conditions, including when the primary injury site is remote from the brain. Here, we hypothesized that the retinal pigment epithelium (RPE) serves a parallel role, as a gateway for monocyte trafficking to the retina following direct or remote injury. We found elevated expression of genes encoding leukocyte trafficking determinants in mouse RPE as a consequence of retinal glutamate intoxication or optic nerve crush (ONC). Blocking VCAM-1 after ONC interfered with monocyte infiltration into the retina and resulted in a local pro-inflammatory cytokine bias. Live imaging of the injured eye showed monocyte accumulation first in the RPE, and subsequently in the retina, and peripheral leukocytes formed close contact with the RPE Our findings further implied that the ocular milieu can confer monocytes a phenotype advantageous for neuroprotection. These results suggest that the eye utilizes a mechanism of crosstalk with the immune system similar to that of the brain, whereby epithelial barriers serve as gateways for leukocyte entry.
Subject(s)
Monocytes/immunology , Retinal Pigment Epithelium/immunology , Animals , Autoimmune Diseases/immunology , CX3C Chemokine Receptor 1 , Chemotaxis, Leukocyte , Gene Expression , Glutamic Acid/toxicity , Green Fluorescent Proteins/genetics , Male , Mice, Inbred C57BL , Mice, Transgenic , Optic Nerve Injuries/immunology , Receptors, Cytokine/genetics , Receptors, HIV/genetics , Retinitis/immunology , Vascular Cell Adhesion Molecule-1/immunologyABSTRACT
Tissue-specific immune regulation is an important component of the immune response relevant to many areas of immunology. The focus of this study is on tissue-specific mechanisms that contribute to autoimmune uveitis. Precise gene regulation is necessary for the proper expression of an inflammatory or regulatory response. This precision gene regulation can be accomplished by microRNA at the level of the mRNA transcript. miR-155, in particular, has a complicated role in the immune response with positive and negative inflammatory effects. In this work, we identify a decrease in miR-155 in suppressor macrophages and further examine how tissue-specific production of miR-155 impacts experimental autoimmune uveitis. Importantly, we show that eliminating miR-155 expression by the target tissue before initiation reduces disease severity, but elimination of miR-155 after the onset of inflammation does not alter the course of disease. Additionally, expression of miR-155 by the target tissue before initiation is necessary for the induction of regulatory immunity that protects from further autoimmune disease, but not after the onset of inflammation. In summary, we find a MC5r-dependent decrease in miR-155 in postexperimental autoimmune uveitis APC, miR-155 production by the target tissue is necessary for the initiation of autoimmune uveitis, and may have a role in establishing protective regulatory immunity.
Subject(s)
Autoimmune Diseases/genetics , Macrophages/immunology , MicroRNAs/genetics , Receptors, Melanocortin/genetics , Retinal Pigment Epithelium/immunology , Uveitis/genetics , Animals , Autoimmune Diseases/chemically induced , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Freund's Adjuvant/administration & dosage , Gene Expression Regulation/immunology , Humans , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/immunology , Organ Specificity , Receptors, Melanocortin/immunology , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Signal Transduction , Spleen/drug effects , Spleen/immunology , Spleen/pathology , Uveitis/chemically induced , Uveitis/immunology , Uveitis/pathologyABSTRACT
It has become increasingly important to understand how retinal inflammation is regulated because inflammation plays a role in retinal degenerative diseases. Lipocalin 2 (LCN2), an acute stress response protein with multiple innate immune functions, is increased in ATP-binding cassette subfamily A member 4 (Abca4) -/- retinol dehydrogenase 8 (Rdh8) -/- double-knockout mice, an animal model for Stargardt disease and age-related macular degeneration (AMD). To examine roles of LCN2 in retinal inflammation and degeneration, Lcn2-/-Abca4-/-Rdh8-/- triple-knockout mice were generated. Exacerbated inflammation following light exposure was observed in Lcn2-/-Abca4-/-Rdh8-/- mice as compared with Abca4-/-Rdh8-/- mice, with upregulation of proinflammatory genes and microglial activation. RNA array analyses revealed an increase in immune response molecules such as Ccl8, Ccl2, and Cxcl10 To further probe a possible regulatory role for LCN2 in retinal inflammation, we examined the in vitro effects of LCN2 on NF-κB signaling in human retinal pigmented epithelial (RPE) cells differentiated from induced pluripotent stem cells derived from healthy donors. We found that LCN2 induced expression of antioxidant enzymes heme oxygenase 1 and superoxide dismutase 2 in these RPE cells and could inhibit the cytotoxic effects of H2O2 and LPS. ELISA revealed increased LCN2 levels in plasma of patients with Stargardt disease, retinitis pigmentosa, and age-related macular degeneration as compared with healthy controls. Finally, overexpression of LCN2 in RPE cells displayed protection from cell death. Overall these results suggest that LCN2 is involved in prosurvival responses during cell stress and plays an important role in regulating inflammation during retinal degeneration.
Subject(s)
Inflammation/metabolism , Lipocalin-2/metabolism , Retinal Degeneration/metabolism , Animals , Humans , Inflammation/immunology , Lipocalin-2/immunology , Mice , Retinal Degeneration/immunology , Retinal Pigment Epithelium/immunology , Retinal Pigment Epithelium/metabolismABSTRACT
Human retinal pigment epithelial (RPE) cells derived from induced pluripotent stem (iPS) cells have immunosuppressive properties. However, RPE cells are also known as immunogenic cells, and they have major histocompatibility complex expression and produce inflammatory proteins, and thus experience immune rejection after transplantation. In this study, to confirm the immunological properties of IPS-RPE cells, we examined whether human RPE cells derived from iPS cells could suppress or stimulate inflammatory T cells from uveitis patients via costimulatory signals. We established T cells from patients with active uveitis as target cells and used iPS-RPE cells as effector cells. As a result, cultured iPS-RPE cells inhibited cell proliferation and the production of IFN-γ by activated uveitis CD4+ T cells, especially Th1-type T cells. In contrast, iPS-RPE cells stimulated T cells of uveitis patients. The iPS-RPE cells constitutively expressed B7-H1/CD274 and B7-DC/CD273, and suppressed the activation of T cells via the PD-1 receptor. iPS-RPE expressed these negative costimulatory molecules, especially when RPE cells were pretreated with recombinant IFN-γ. In addition, iPS-RPE cells also expressed B7-H3/CD276 costimulatory molecules and activated uveitis T cells through the B7-H3-TLT-2 receptor. Thus, cultured iPS-derived retinal cells can suppress or activate inflammatory T cells in vitro through costimulatory interactions.
Subject(s)
Costimulatory and Inhibitory T-Cell Receptors/immunology , Retinal Pigment Epithelium/metabolism , T-Lymphocytes/physiology , B7 Antigens/metabolism , B7-H1 Antigen/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation/drug effects , Cells, Cultured , Costimulatory and Inhibitory T-Cell Receptors/metabolism , Epithelial Cells/metabolism , Flow Cytometry , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/physiology , Interferon-gamma/metabolism , Interleukin-2/metabolism , Lymphocyte Activation/immunology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Programmed Cell Death 1 Receptor/metabolism , Retinal Pigment Epithelium/immunology , Retinal Pigment Epithelium/physiology , Retinal Pigments/metabolism , Uveitis/immunology , Uveitis/metabolismABSTRACT
Lactobacillus paracasei KW3110 (KW3110) has anti-inflammatory effects and mitigates retinal pigment epithelium (RPE) cell damage caused by blue-light exposure. We investigated whether KW3110 suppresses chronic inflammatory stress-induced RPE cell damage by modulating immune cell activity and whether it improves ocular disorders in healthy humans. First, we showed that KW3110 treatment of mouse macrophages (J774A.1) produced significantly higher levels of interleukin-10 as compared with other lactic acid bacterium strains (all p < 0.01). Transferring supernatant from KW3110- and E. coli 0111:B4 strain and adenosine 5'-triphosphate (LPS/ATP)-stimulated J774A.1 cells to human retinal pigment epithelium (ARPE-19) cells suppressed senescence-associated phenotypes, including proliferation arrest, abnormal appearance, cell cycle arrest, and upregulation of cytokines, and also suppressed expression of tight junction molecule claudin-1. A randomized, double-blind, placebo-controlled parallel-group study of healthy subjects (n = 88; 35 to below 50 years) ingesting placebo or KW3110-containing supplements for 8 weeks showed that changes in critical flicker frequency, an indicator of eye fatigue, from the week-0 value were significantly larger in the KW3110 group at weeks 4 (p = 0.040) and 8 (p = 0.036). These results suggest that KW3110 protects ARPE-19 cells against premature senescence and aberrant expression of tight junction molecules caused by chronic inflammatory stress, and may improve chronic eye disorders including eye fatigue.
Subject(s)
Cellular Senescence/drug effects , Eye Diseases/drug therapy , Inflammation/drug therapy , Lacticaseibacillus paracasei , Probiotics/therapeutic use , Retinal Pigment Epithelium/drug effects , Adenosine Triphosphate/toxicity , Adult , Animals , Cell Cycle Checkpoints/drug effects , Cell Death/drug effects , Cell Line , Cell Proliferation/drug effects , Cytokines/metabolism , Escherichia coli , Female , Humans , Inflammation/immunology , Interleukin-10/metabolism , Lipopolysaccharides/toxicity , Macrophages/drug effects , Male , Mice , Middle Aged , Retina/drug effects , Retina/immunology , Retina/pathology , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/immunology , Retinal Pigment Epithelium/pathology , Tight Junctions/metabolismABSTRACT
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/virologyABSTRACT
Age-associated dysfunction of retinal pigment epithelial cells (RPEs) is considered to be the initial trigger of retinal diseases such as age-related macular degeneration. Although autophagy is upregulated in RPEs during the course of aging, little is known about how autophagy is regulated and its functional role in RPEs. In this study, we found that expression of Sirtuin 6 (SIRT6) and autophagic markers are upregulated in RPEs of aged mice where subretinal deposition of amyloid-ß is accumulated and in amyloid-ß stimulated RPEs. In addition, gain and loss-of-function studies confirmed the positive role of SIRT6 in regulating autophagy. Interesting, inhibition of autophagy attenuates amyloid-ß stimulated inflammatory response in RPEs. Collectively, our findings uncover the autophagy modulated by SIRT6 may be a proinflammatory mechanism for amyloid-ß induced RPE dysfunction.
Subject(s)
Amyloid beta-Peptides/immunology , Autophagy/immunology , Epithelial Cells/immunology , Inflammation Mediators/immunology , Retinal Pigment Epithelium/immunology , Retinitis/immunology , Sirtuins/immunology , Animals , Autophagy/drug effects , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Mice , Mice, Inbred C57BL , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Retinitis/chemically induced , Retinitis/pathologyABSTRACT
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/virologyABSTRACT
The molecular basis of intraocular tuberculosis (TB) is not well understood. In this study, we investigated the role of two constituents of viable Mycobacterium tuberculosis - Early Secreted Antigenic Target-6 (ESAT-6), and mycobacterial RNA- in inflammasome activation in the retinal pigment epithelium (RPE), a key site of inflammation in intraocular TB. We found that ESAT-6 induced caspase-1 activation and inflammasome priming in mouse RPE cells, substantially more in wild-type than in Tlr2/3/4/7/9-/-, Myd88-/- or Nlrp3-/- RPE cells. Sub-retinal ESAT-6 injection resulted in greater RPE degeneration in wild-type than in Nlrp3-/- mice. In human ocular TB tissue sections, NLRP3 staining was noted in retina as well as RPE. Mycobacterial RNA, specifically its double stranded component, also induced caspase-1 activation, and the double stranded RNA was immunolocalized to human ocular TB sections. Our observations suggest that inflammasome activation in RPE by viable M. tuberculosis could potentially contribute to human intraocular TB.
Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Inflammasomes/immunology , Mycobacterium tuberculosis/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , RNA, Bacterial/immunology , RNA, Double-Stranded/immunology , Tuberculosis, Ocular/immunology , Animals , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Caspase 1/genetics , Caspase 1/immunology , Host-Parasite Interactions , Humans , Inflammasomes/genetics , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , RNA, Bacterial/genetics , RNA, Double-Stranded/genetics , Retinal Pigment Epithelium/immunology , Retinal Pigment Epithelium/microbiology , Tuberculosis, Ocular/genetics , Tuberculosis, Ocular/microbiologyABSTRACT
γδ T cells located near the epithelial barrier are integral components of local inflammatory and innate immune responses. We have previously reported the presence of choroidal γδ T cells in a model of chronic degeneration of the retinal pigment epithelium (RPE). The goals of the current study were to further define the functions of choroidal γδ T cells and to explore the underlying mechanisms of their action. Our data demonstrate that choroidal γδ T cells are activated by RPE injury in response to NaIO3 treatment, and that they express genes that encode immunosuppressive cytokines, such as IL-4 and IL-10. γδ-T-cell-deficient mice developed profound RPE and retinal damage at doses that caused minimal effects in wild-type mice, and adoptive transfer of γδ T cells prevented sensitization. Intravitreal injection of IL-4 and IL-10 ameliorated RPE toxicity that was induced by NaIO3Ex vivo coculture of γδ T cells with RPE explants activated the production of anti-inflammatory cytokines via an aryl hydrocarbon receptor (AhR)-dependent mechanism. AhR deficiency abolished the protective effects of γδ T cells after adoptive transfer. Collectively, these findings define important roles for choroid γδ T cells in maintaining tissue homeostasis in the outer retina.-Zhao, Z., Liang, Y., Liu, Y., Xu, P., Flamme-Wiese, M. J., Sun, D., Sun, J., Mullins, R. F., Chen, Y., Cai, J. Choroidal γδ T cells in protection against retinal pigment epithelium and retinal injury.
Subject(s)
Adoptive Transfer , Corneal Dystrophies, Hereditary/immunology , Corneal Dystrophies, Hereditary/therapy , Receptors, Antigen, T-Cell, gamma-delta/immunology , Retinal Pigment Epithelium/immunology , T-Lymphocytes/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Corneal Dystrophies, Hereditary/chemically induced , Corneal Dystrophies, Hereditary/genetics , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Iodates/toxicity , Mice , Mice, Knockout , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/immunology , Retinal Pigment Epithelium/injuries , T-Lymphocytes/pathology , T-Lymphocytes/transplantationABSTRACT
Age-related macular degeneration (AMD) is the leading cause of central vision loss worldwide. Loss of retinal pigment epithelium (RPE) is a major pathological hallmark in AMD with or without pathological neovascularization. Although activation of the immune system is implicated in disease progression, pathological pathways remain diverse and unclear. Here, we report an unexpected protective role of a pro-inflammatory cytokine, interleukin-33 (IL-33), in ocular angiogenesis. IL-33 and its receptor (ST2) are expressed constitutively in human and murine retina and choroid. When RPE was activated, IL-33 expression was markedly elevated in vitro. We found that IL-33 regulated tissue remodelling by attenuating wound-healing responses, including reduction in the migration of choroidal fibroblasts and retinal microvascular endothelial cells, and inhibition of collagen gel contraction. In vivo, local administration of recombinant IL-33 inhibited murine choroidal neovascularization (CNV) formation, a surrogate of human neovascular AMD, and this effect was ST2-dependent. Collectively, these data demonstrate IL-33 as a potential immunotherapy and distinguishes pathways for subverting AMD pathology. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Subject(s)
Interleukin-33/immunology , Macular Degeneration/immunology , Adolescent , Adult , Aged , Animals , Cells, Cultured , Choroid/immunology , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/immunology , Fibroblasts/immunology , Humans , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/therapeutic use , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Recombinant Proteins/therapeutic use , Retinal Pigment Epithelium/immunology , Young AdultABSTRACT
Age-related macular degeneration (AMD) is a complex and progressive degenerative eye disease resulting in severe loss of central vision. Recent evidence indicates that immune system dysregulation could contribute to the development of AMD. We hypothesize that defective lysosome-mediated clearance causes accumulation of waste products in the retinal pigmented epithelium (RPE), activating the immune system and leading to retinal tissue injury and AMD. We have generated unique genetically engineered mice in which lysosome-mediated clearance (both by phagocytosis and autophagy) in RPE cells is compromised, causing the development of features of early AMD. Our recent data indicate a link between lipocalin-2 (LCN-2) and the inflammatory responses induced in this mouse model. We show that nuclear factor-κB (NF-κB) and STAT-1 may function as a complex in our animal model system, together controlling the upregulation of LCN-2 expression in the retina and stimulating an inflammatory response. This study revealed increased infiltration of LCN-2-positive neutrophils in the choroid and retina of early AMD patients as compared with age-matched controls. Our results demonstrate that, both in our animal model and in human AMD, the AKT2-NF-κB-LCN-2 signalling axis is involved in activating the inflammatory response, making this pathway a potential target for AMD treatment. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Subject(s)
Lipocalin-2/genetics , Lysosomes/immunology , Macular Degeneration/genetics , NF-kappa B/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , Age Factors , Animals , Autophagy , Choroid/immunology , Choroid/metabolism , Disease Models, Animal , Humans , Inflammation , Lipocalin-2/metabolism , Lysosomes/metabolism , Macular Degeneration/immunology , Macular Degeneration/pathology , Mice , NF-kappa B/metabolism , Neutrophils/immunology , Phagocytosis , Proto-Oncogene Proteins c-akt/metabolism , Retina/immunology , Retina/injuries , Retina/metabolism , Retinal Pigment Epithelium/immunology , Retinal Pigment Epithelium/metabolism , Up-RegulationABSTRACT
Age-related macular degeneration (AMD) is the leading cause of blindness in the USA. Polymorphisms in various complement components are associated with an increased risk for AMD, and it has been hypothesized that an overactive complement system is partially responsible for the pathology of AMD. AMD is classified as early, intermediate, or late AMD, depending on the degree of the associated pathologies. Late AMD can be characterized as either lesions associated with neovascular AMD or geographic atrophy. Both sets of lesions are associated with pathology at the RPE/choroid interface, which include a thickening of Bruch's membrane, presence of drusen, and pigmentary alterations, and deterioration of the blood-retina barrier has been reported. These changes can lead to the slow degeneration and atrophy of the photoreceptors in the macula in dry AMD, or progress to choroidal neovascularization (CNV) and leakage of these new vessels in wet AMD. It has been shown previously that complement anaphylatoxins C3a and C5a, signaling via their respective G-protein-coupled receptors, can alter RPE cell function and promote choroidal neovascularization. However, it is important to note these components also play a role in tissue repair. Here we discuss anaphylatoxin signaling in AMD-related target cells and the potential implications for the design of anti-complement therapeutics.
Subject(s)
Complement Activation , Complement C3/immunology , Complement C5a/immunology , Macular Degeneration/immunology , Receptor, Anaphylatoxin C5a/immunology , Aging/immunology , Animals , Bruch Membrane/pathology , Choroid/pathology , Choroidal Neovascularization/immunology , Cytokines/biosynthesis , Endothelial Cells/pathology , Fibrosis , Forecasting , Humans , Inflammasomes/metabolism , Inflammation , Macular Degeneration/drug therapy , Macular Degeneration/pathology , Mice , Retinal Pigment Epithelium/immunology , Retinal Pigment Epithelium/pathology , Retinal Rod Photoreceptor Cells/immunology , Retinal Rod Photoreceptor Cells/pathologyABSTRACT
Given the complex etiology of age-related macular degeneration (AMD), treatments are developed to target intermediate/late stages of the disease. Unfortunately, the design of therapies for early stages of the disease is limited by our understanding of the mechanisms involved in the formation of basal deposits and drusen, the first clinical signs of AMD. During the last decade, the identification of common and rare alleles in complement genes as risk AMD variants in addition to the presence of active complement components in basal deposits and drusen has provided compelling evidence that the complement system plays a key role in the pathobiology of AMD. However, the mechanisms for complement activation in AMD are unknown. Here we propose that the activation of the complement system is a consequence of alterations in the aged extracellular matrix (ECM) of the retinal pigment epithelium (RPE)/Bruch's membrane (BrM), which favors the anchoring of complement C3b generated by convertase-independent cleavage of C3 via tick-over and produces a chronic activation of the alternative complement pathway.
Subject(s)
Bruch Membrane/immunology , Complement Pathway, Alternative , Extracellular Matrix/immunology , Macular Degeneration/immunology , Retinal Pigment Epithelium/immunology , Animals , Bruch Membrane/metabolism , Bruch Membrane/pathology , Clinical Trials as Topic , Complement C3/metabolism , Complement C3b/immunology , Complement C3b/metabolism , Complement Pathway, Alternative/drug effects , Complement Pathway, Alternative/genetics , Drug Design , Extracellular Matrix/metabolism , Eye Diseases, Hereditary/immunology , Eye Diseases, Hereditary/metabolism , Humans , Immunoglobulin Fab Fragments/therapeutic use , Macular Degeneration/drug therapy , Macular Degeneration/genetics , Macular Degeneration/metabolism , Mice , Models, Immunological , Molecular Targeted Therapy , Peptides, Cyclic/therapeutic use , Retinal Drusen/immunology , Retinal Drusen/metabolism , Retinal Pigment Epithelium/metabolismABSTRACT
Inherited and age-related macular degenerations (AMDs) are important causes of vision loss. An early hallmark of these disorders is the formation of sub-retinal pigment epithelium (RPE) basal deposits. A role for the complement system in MDs was suggested by genetic association studies, but direct functional connections between alterations in the complement system and the pathogenesis of MD remain to be defined. We used primary RPE cells from a mouse model of inherited MD due to a p.R345W mutation in EGF-containing fibulin-like extracellular matrix protein 1 (EFEMP1) to investigate the role of the RPE in early MD pathogenesis. Efemp1(R345W) RPE cells recapitulate the basal deposit formation observed in vivo by producing sub-RPE deposits in vitro. The deposits share features with basal deposits, and their formation was mediated by EFEMP1(R345W) or complement component 3a (C3a), but not by complement component 5a (C5a). Increased activation of complement appears to occur in response to an abnormal extracellular matrix (ECM), generated by the mutant EFEMP1(R345W) protein and reduced ECM turnover due to inhibition of matrix metalloproteinase 2 by EFEMP1(R345W) and C3a. Increased production of C3a also stimulated the release of cytokines such as interleukin (IL)-6 and IL-1B, which appear to have a role in deposit formation, albeit downstream of C3a. These studies provide the first direct indication that complement components produced locally by the RPE are involved in the formation of basal deposits. Furthermore, these results suggest that C3a generated by RPE is a potential therapeutic target for the treatment of EFEMP1-associated MD as well as AMD.
Subject(s)
Complement C3a/metabolism , Extracellular Matrix Proteins/genetics , Macular Degeneration/genetics , Macular Degeneration/pathology , Retinal Pigment Epithelium/immunology , Animals , Cells, Cultured , Complement C5a/metabolism , Disease Models, Animal , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Macular Degeneration/immunology , Matrix Metalloproteinases/metabolism , Mice , MutationABSTRACT
Activation of the innate immune system plays a major role in retinal degenerative diseases including age-related macular degeneration (AMD). In this study, we investigated whether reactive microglia trigger and sustain NLRP3 inflammasome activation in human retinal pigment epithelium (ARPE-19) cells. Specifically, we analyzed the potential of cell culture supernatants from lipopolysaccharide (LPS)-stimulated human microglia in combination with the lysosomal destabilization agent Leu-Leu-O-Me (LLOMe) to activate the inflammasome in ARPE-19 cells. We found disorganization of ARPE-19 cytoskeletal structure after incubation with conditioned media of LPS-stimulated microglia and LLOMe and accumulation of lipid deposits in these cells using Nile Red staining. LC3-II, the active form of the autophagy marker microtubule-associated protein 1 light chain 3 beta (LC3B), was also elevated in ARPE-19 cells after inducing inflammasome activation. Finally, a significant increase of transcripts for IL-6, IL-8, IL-1ß, GM-CSF and CCL-2 was detected in ARPE-19 stimulated with both microglia-conditioned medium and LLOMe. Our findings highlight a potential role of microglia in RPE inflammasome activation.
Subject(s)
Inflammasomes/immunology , Lysosomes/immunology , Microglia/immunology , Microglia/pathology , Retinal Pigment Epithelium/immunology , Retinal Pigment Epithelium/pathology , Cell Line , Cytoskeleton/immunology , Cytoskeleton/pathology , Humans , Immunologic Factors/immunologyABSTRACT
Diabetic retinopathy (DR) is a major microvascular complication of diabetes, resulting in neuronal dysfunction, retinal vascular leakage, and apoptosis within the retina. Innate immunity plays an important role in the pathogenesis of type 2 diabetes (T2D) and related complications. The toll-like receptors (TLRs), central to innate immunity, are essential participants in the progression and pathogenesis of the disease and its complications. In the study, streptozotocin (STZ) was combined with whole-body hypoxia for quicker induction of early-stage diabetic retinopathy (DR) in the wild type (WT) and TLR7-knockout (KO) C57BL/6 mice. The effects of TLR7 were also investigated in fructose-treated retinal pigment epithelial (RPE) cells. In the retinas of WT/DR mice, abnormal a-wave and b-wave activity, hyperfluorescence, and reduced retinal thickness were observed. DR development was associated with enhanced TLR7 expression, whose deletion dramatically reduced VEGF expression levels. And the secretion of pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α), interleukin (IL)-1ß, IL-6, IL-18 and IL-12, was highly reduced by TLR7-deficiency in DR mice. Consistently, WT/DR mice exhibited higher phosphorylation of IκB kinase α (IKKα), inhibitor of NF-κB α (IκBα) and nuclear factor κB (NF-κB), which were found to be down-regulated in KO/DR mice. Similarly, DR-induced mitogen-activated protein kinases (MAPKs) activation was blocked by TLR7-knockout. In vitro, fructose incubation-triggered inflammation was reversed by TLR7 knockdown, accompanied with inactivated NF-κB and MAPKs pathways. And reduced reactive oxygen species (ROS) generation was observed in TLR7-knockdown cells with fructose treatment. Together, inhibiting TLR7 suppressed diabetic retinopathy by reducing inflammation and suggested a potential application in clinics.
Subject(s)
Diabetes Mellitus, Type 2/complications , Diabetic Retinopathy/genetics , Gene Deletion , Inflammation/genetics , Membrane Glycoproteins/genetics , Retina/pathology , Toll-Like Receptor 7/genetics , Animals , Cell Line , Cytokines/analysis , Cytokines/immunology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/pathology , Diabetic Retinopathy/etiology , Diabetic Retinopathy/immunology , Diabetic Retinopathy/pathology , Inflammation/etiology , Inflammation/immunology , Inflammation/pathology , Male , Membrane Glycoproteins/immunology , Mice, Inbred C57BL , Mice, Knockout , Retina/immunology , Retina/metabolism , Retinal Pigment Epithelium/immunology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Toll-Like Receptor 7/immunologyABSTRACT
The discovery that genetic abnormalities in complement factor H (FH) are associated with an increased risk for age-related macular degeneration (AMD), the most common cause of blindness among the elderly, raised hope of new treatments for this vision-threatening disease. Nonetheless, over a decade after the identification of this important association, how innate immunity contributes to AMD remains unresolved. Pentraxin 3 (PTX3), an essential component of the innate immunity system that plays a non-redundant role in controlling inflammation, regulates complement by interacting with complement components. Here, we show that PTX3 is induced by oxidative stress, a known cause of AMD, in the retinal pigmented epithelium (RPE). PTX3 deficiency in vitro and in vivo magnified complement activation induced by oxidative stress, leading to increased C3a, FB, and C3d, but not C5b-9 complex formation. Increased C3a levels, resulting from PTX3 deficiency, raised the levels of Il1b mRNA and secretion of activated interleukin (IL)-1ß by interacting with C3aR. Importantly, PTX3 deficiency augmented NLRP3 inflammasome activation, resulting in enhanced IL-1ß, but not IL-18, production by the RPE. Thus, in the presence of PTX3 deficiency, the complement and inflammasome pathways worked in concert to produce IL-1ß in sufficient abundance to, importantly, result in macrophages accumulating in the choroid. These results demonstrate that PTX3 acts as an essential brake for complement and inflammasome activation by regulating the abundance of FH in the RPE, and provide critical insights into the complex interplay between oxidative stress and innate immunity in the early stages of AMD development. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.