Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
Front Immunol ; 12: 712572, 2021.
Article En | MEDLINE | ID: mdl-34566967

The complement system is central to first-line defense against invading pathogens. However, excessive complement activation and/or the loss of complement regulation contributes to the development of autoimmune diseases, systemic inflammation, and thrombosis. One of the three pathways of the complement system, the alternative complement pathway, plays a vital role in amplifying complement activation and pathway signaling. Complement factor D, a serine protease of this pathway that is required for the formation of C3 convertase, is the rate-limiting enzyme. In this review, we discuss the function of factor D within the alternative pathway and its implication in both healthy physiology and disease. Because the alternative pathway has a role in many diseases that are characterized by excessive or poorly mediated complement activation, this pathway is an enticing target for effective therapeutic intervention. Nonetheless, although the underlying disease mechanisms of many of these complement-driven diseases are quite well understood, some of the diseases have limited treatment options or no approved treatments at all. Therefore, in this review we explore factor D as a strategic target for advancing therapeutic control of pathological complement activation.


Complement Factor D/antagonists & inhibitors , Complement Pathway, Alternative/drug effects , Molecular Targeted Therapy , Adipose Tissue/metabolism , Aging/immunology , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/drug therapy , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Complement Factor D/biosynthesis , Complement Factor D/deficiency , Complement Factor D/physiology , Energy Metabolism , Geographic Atrophy/genetics , Geographic Atrophy/immunology , Hemoglobinuria, Paroxysmal/drug therapy , Hemoglobinuria, Paroxysmal/genetics , Hemoglobinuria, Paroxysmal/immunology , Hepatocytes , Humans , Kidney Diseases/immunology , Liver/injuries , Oligonucleotides, Antisense/therapeutic use , Peptides, Cyclic/therapeutic use , Phagocytosis
2.
Proc Natl Acad Sci U S A ; 117(18): 9952-9963, 2020 05 05.
Article En | MEDLINE | ID: mdl-32345717

Genetic polymorphisms in the region of the trimeric serine hydrolase high-temperature requirement 1 (HTRA1) are associated with increased risk of age-related macular degeneration (AMD) and disease progression, but the precise biological function of HtrA1 in the eye and its contribution to disease etiologies remain undefined. In this study, we have developed an HtrA1-blocking Fab fragment to test the therapeutic hypothesis that HtrA1 protease activity is involved in the progression of AMD. Next, we generated an activity-based small-molecule probe (ABP) to track target engagement in vivo. In addition, we used N-terminomic proteomic profiling in preclinical models to elucidate the in vivo repertoire of HtrA1-specific substrates, and identified substrates that can serve as robust pharmacodynamic biomarkers of HtrA1 activity. One of these HtrA1 substrates, Dickkopf-related protein 3 (DKK3), was successfully used as a biomarker to demonstrate the inhibition of HtrA1 activity in patients with AMD who were treated with the HtrA1-blocking Fab fragment. This pharmacodynamic biomarker provides important information on HtrA1 activity and pharmacological inhibition within the ocular compartment.


Adaptor Proteins, Signal Transducing/genetics , Antibodies, Anti-Idiotypic/pharmacology , Geographic Atrophy/drug therapy , High-Temperature Requirement A Serine Peptidase 1/genetics , Macular Degeneration/drug therapy , Adaptor Proteins, Signal Transducing/isolation & purification , Aged , Animals , Antibodies, Anti-Idiotypic/genetics , Antibodies, Anti-Idiotypic/immunology , Biomarkers/blood , Disease Progression , Female , Genetic Predisposition to Disease , Genotype , Geographic Atrophy/blood , Geographic Atrophy/genetics , Geographic Atrophy/immunology , High-Temperature Requirement A Serine Peptidase 1/antagonists & inhibitors , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fab Fragments/pharmacology , Macular Degeneration/blood , Macular Degeneration/genetics , Macular Degeneration/immunology , Male , Polymorphism, Single Nucleotide/genetics , Proteome/genetics , Proteome/immunology , Rats , Retina/drug effects , Retina/immunology , Retina/pathology , Small Molecule Libraries/pharmacology
3.
Invest Ophthalmol Vis Sci ; 61(3): 45, 2020 03 09.
Article En | MEDLINE | ID: mdl-32207814

Purpose: Age-related macular degeneration (AMD) is the leading cause of blindness in Western populations. While an overactive complement system has been linked to pathogenesis, mechanisms contributing to its activation are largely unknown. In aged and AMD eyes, loss of the elastin layer (EL) of Bruch's membrane (BrM) has been reported. Elastin antibodies are elevated in patients with AMD, the pathogenic significance of which is unclear. Here we assess the role of elastin antibodies using a mouse model of smoke-induced ocular pathology (SIOP), which similarly demonstrates EL loss. Methods: C57BL/6J mice were immunized with elastin or elastin peptide oxidatively modified by cigarette smoke (ox-elastin). Mice were then exposed to cigarette smoke or air for 6 months. Visual function was assessed by optokinetic response, retinal morphology by spectral-domain optical coherence tomography and electron microscopy, and complement activation and antibody deposition by Western blot. Results: Ox-elastin IgG and IgM antibodies were elevated in ox-elastin immunized mice following 6 months of smoke, whereas elastin immunization had a smaller effect. Ox-elastin immunization exacerbated smoke-induced vision loss, with thicker BrM and more damaged retinal pigment epithelium (RPE) mitochondria compared with mice immunized with elastin or nonimmunized controls. These changes were correlated with increased levels of IgM, IgG2, IgG3, and complement activation products in RPE/choroid. Conclusions: These data demonstrate that SIOP mice generate elastin-specific antibodies and that immunization with ox-elastin exacerbates ocular pathology. Elastin antibodies represented complement fixing isotypes that, together with the increased presence of complement activation seen in immunized mice, suggest that elastin antibodies exert pathogenic effects through mediating complement activation.


Autoantibodies/blood , Bruch Membrane/pathology , Disease Models, Animal , Elastin/immunology , Geographic Atrophy/etiology , Retinal Pigment Epithelium/pathology , Smoking/adverse effects , Animals , Blotting, Western , Complement Activation/physiology , Complement System Proteins/metabolism , Contrast Sensitivity/physiology , Enzyme-Linked Immunosorbent Assay , Geographic Atrophy/immunology , Geographic Atrophy/pathology , Immunization , Immunoglobulin G/blood , Immunoglobulin M/blood , Mice , Mice, Inbred C57BL , Nystagmus, Optokinetic/physiology , Oxidation-Reduction , Tobacco Products , Visual Acuity/physiology
4.
Invest Ophthalmol Vis Sci ; 59(2): 662-673, 2018 02 01.
Article En | MEDLINE | ID: mdl-29392311

Purpose: A large body of evidence supports a central role for complement activation in the pathobiology of age-related macular degeneration (AMD), including plasma complement component 5a (C5a). Interestingly, C5a is a chemotactic agent for monocytes, a cell type also shown to contribute to AMD. However, the role monocytes play in the pathogenesis of "dry" AMD and the pharmacologic potential of targeting C5a to regulate these cells are unclear. We addressed these questions via C5a blockade in a unique model of early/intermediate dry AMD and large panel flow cytometry to immunophenotype monocytic involvement. Methods: Heterozygous complement factor H (Cfh+/-) mice aged to 90 weeks were fed a high-fat, cholesterol-enriched diet (Cfh+/-∼HFC) for 8 weeks and were given weekly intraperitoneal injections of 30 mg/kg anti-C5a (4C9, Pfizer). Flow cytometry, retinal pigmented epithelium (RPE) flat mounts, and electroretinograms were used to characterize anti-C5a treatment. Results: Aged Cfh+/- mice developed RPE damage, sub-RPE basal laminar deposits, and attenuation of visual function and immune cell recruitment to the choroid that was accompanied by expression of inflammatory and extracellular matrix remodeling genes following 8 weeks of HFC diet. Concomitant systemic administration of an anti-C5a antibody successfully inhibited local recruitment of mononuclear phagocytes to the choroid-RPE interface but did not ameliorate these AMD-like pathologies in this mouse model. Conclusions: These results show that immunotherapy targeting C5a is not sufficient to block the development of the AMD-like pathologies observed in Cfh+/-∼HFC mice and suggest that other complement components or molecules/mechanisms may be driving "early" and "intermediate" AMD pathologies.


Antibodies, Blocking/therapeutic use , Choroidal Neovascularization/therapy , Complement C5a/antagonists & inhibitors , Disease Models, Animal , Geographic Atrophy/therapy , Immunotherapy , Animals , Cholesterol, Dietary/administration & dosage , Choroidal Neovascularization/immunology , Choroidal Neovascularization/pathology , Complement Activation , Complement C5a/immunology , Electroretinography , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Geographic Atrophy/immunology , Geographic Atrophy/pathology , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Retinal Pigment Epithelium/pathology
5.
Ophthalmologica ; 236(1): 53-8, 2016.
Article En | MEDLINE | ID: mdl-27287199

PURPOSE: To evaluate the presence of interleukin-17 (IL-17)-producing cells in patients with geographic atrophy (GA). METHODS: In this short report, we analyzed IL-17, CD3, and IBA-1 expression by immunohistochemistry on paraffin-embedded sections from 13 donors with a known history of GA, confirmed by fundus appearance and histology, and 7 age-matched control donors. RESULTS/CONCLUSION: We showed that IL-17+ cells are found near areas of retinal pigmented epithelium atrophy in the eyes of GA patients. IL-17+ cells mainly localized to CD3+ cells, which identifies T lymphocytes, as well as IBA-1+ cells, which identifies mononuclear phagocytes. Therefore, IL-17 could be involved in the pathological mechanisms that contribute to the degeneration observed in GA.


Choroid/pathology , Geographic Atrophy/metabolism , Immunity, Cellular , Interleukin-17/biosynthesis , Macrophages/metabolism , T-Lymphocytes/metabolism , Aged, 80 and over , Choroid/immunology , Female , Fluorescein Angiography , Fundus Oculi , Geographic Atrophy/immunology , Geographic Atrophy/pathology , Humans , Immunohistochemistry , Macrophages/immunology , Macrophages/pathology , Male , T-Lymphocytes/immunology , T-Lymphocytes/pathology
6.
Discov Med ; 22(123): 337-349, 2016 12.
Article En | MEDLINE | ID: mdl-28147216

It is generally accepted that geographic atrophy (GA), a currently untreatable advanced form of age-related macular degeneration (AMD), is a multifactorial disease resulting in gradual and permanent blindness. Various risk factors are demonstrated to be responsible for its pathogenesis, such as aging, light exposure, and smoking. Molecular components associated with those risk factors form a complex and interwoven network at the confluence of inflammation, highlighting the significance of inflammasome activation in GA progression. Recently, a new type of modification in AMD microenvironment has been discovered, other than extensively-studied complement dysregulation, lipofuscin deposit, and oxidative by-products, to activate inflammasome. The accumulation of Alu RNA, resulting from DICER1 deficiency, is shown capable of triggering the activation of NLRP3 inflammasome and causing caspase-8-activated apoptosis in an IL-18/MyD88-dependent manner, which provides a new source of evidence for the interplay between cell death and inflammasome. In this review, we lay the emphasis on the mechanism by which Alu RNA activates NLRP3 inflammasome and downstream apoptotic proteins, and on its clinical relevance to GA and potential therapeutic approaches. We also point out several possible crosstalks among inflammasome and different acts of cell death which remain to be further investigated in Alu RNA-induced RPE degeneration.


Alu Elements/immunology , DEAD-box RNA Helicases/genetics , Geographic Atrophy/immunology , Inflammasomes/immunology , Molecular Targeted Therapy/methods , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , RNA, Untranslated/immunology , Ribonuclease III/genetics , Apoptosis/immunology , Apoptosis Regulatory Proteins/metabolism , Geographic Atrophy/genetics , Geographic Atrophy/therapy , Humans , Lipofuscin/metabolism , MAP Kinase Signaling System , Reactive Oxygen Species/immunology
7.
Article En | MEDLINE | ID: mdl-26126818

The state-of-the-art of oxygen-ozone therapy is now clarified and all the mechanisms of action of medical ozone are within classical biochemistry and molecular biology. The outcomes of standard treatments in peripheral arterial occlusive disease (PAOD) and dry-form of age-related macular degeneration (AMD) have been compared with the documented therapeutic results achieved with ozonated autohemotherapy (O-AHT). On the other hand, the clinical data of O-AHT on stroke remain indicative. As the cost of O-AHT is almost irrelevant, its application in all public hospitals, especially those of poor Countries, would allow two advantages: the first is for the patient, who will improve her/his conditions, and the second is for Health Authorities burdened with increasing costs. The aim of this paper is to report to clinical scientists that O-AHT is a scientific-based therapeutic approach without side effects. The integration of O-AHT with effective approved drugs is likely to yield the best clinical results in several chronic inflammatory diseases.


Blood Transfusion, Autologous/methods , Cardiovascular Diseases/therapy , Geographic Atrophy/therapy , Ozone/therapeutic use , Pulmonary Disease, Chronic Obstructive/therapy , Cardiovascular Diseases/blood , Cardiovascular Diseases/immunology , Chronic Disease , Geographic Atrophy/blood , Geographic Atrophy/immunology , Humans , Oxygen/blood , Oxygen/therapeutic use , Ozone/blood , Pulmonary Disease, Chronic Obstructive/blood , Pulmonary Disease, Chronic Obstructive/immunology , Treatment Outcome
8.
BMC Ophthalmol ; 14: 154, 2014 Dec 08.
Article En | MEDLINE | ID: mdl-25488058

BACKGROUND: Age-related macular degeneration (AMD) is the leading cause of central vision loss in older adults. Anti-retinal autoantibodies (AAbs) have been found in individuals with AMD. The goal of the study was to determine the AAb specificity in different stages of AMD, and determine whether there is a prevalent AAb signature. METHODS: Sera of 134 participants in the Age-related Eye Disease Study were analyzed for anti-retinal AAbs by western blotting. The subjects were classified by diagnostic subgroups based upon their clinical classification: No AMD, Intermediate AMD, and Late AMD - geographic atrophy (GA) and Late AMD - neovascular (NV). RESULTS: The presence of anti-retinal AAb was detected in 58% patients with Intermediate and Late AMD, and 54% of those with no AMD. AAbs bound to fifteen different retinal antigens. Most individuals had 1 specific AAbs (67%), with the remainder having 2 to 4 different AAbs. Over 40% of patients with Intermediate AMD, and 46% of those with GA had anti-enolase AAbs, compared with 29% of individuals with NV and 29% with no AMD. Different AAbs signatures related to NV as compared to GA and/or Intermediate AMD were distinguished. Anti-40-kDa (10%) and 42-kDa (16%) autoantibodies were associated with Intermediate AMD, while anti-30-kDa AAbs (23%) were primarily present in GA. Anti-32-kDa (12%), 35-kDa (21%), and 60-kDa (8%) AAbs were more frequent in NV AMD. CONCLUSIONS: A unique AAb pattern for each of the disease subgroups was present when AMD progressed from the intermediate to the late forms of severity. Differences in the frequency of specific AAbs between AMD subgroups suggested that they may participate in pathogenicity of AMD. Further studies are necessary to confirm these observations in the larger cohort and individual AMD patients over time.


Autoantibodies/blood , Autoantigens/immunology , Geographic Atrophy/immunology , Retina/immunology , Wet Macular Degeneration/classification , Wet Macular Degeneration/immunology , Aged , Arthritis/immunology , Biomarkers/blood , Blotting, Western , Female , Geographic Atrophy/classification , Humans , Male , Prevalence , Smoking/immunology
9.
Adv Exp Med Biol ; 801: 355-63, 2014.
Article En | MEDLINE | ID: mdl-24664718

We have characterized a naturally-occurring mutation in mice that causes slow, progressive photoreceptor degeneration, white fundus flecks, and late-onset RPE atrophy. These animals predictably lose visual function as photoreceptors degenerate. Genetic studies identified a deletion in the 5' coding sequence of Mfrp, designated Mfrp (174delG) , which essentially results in a complete knockout at the protein level. We have shown in Mfrp (174delG) mice that these white fundus flecks are due to the presence of F4/80+ inflammatory cells in the subretinal space. Here we expand on our initial description of the cells with additional markers and by determining their origin. We have also begun an analysis of complement factors in the RPE and found decreased levels of C3d, suggesting that the alternative complement pathway may be misregulated. Finally, we compare and contrast the characteristics of fundus images in Mfrp (174delG) mice with those of other mutations that cause similar irregularities, including Crb1 (rd8) and RDH5, and discuss the structural differences that may underlie them.


Complement System Proteins/immunology , Geographic Atrophy/pathology , Macular Degeneration/pathology , Monocytes/pathology , Alcohol Oxidoreductases/genetics , Animals , Bone Marrow Transplantation , Complement Pathway, Alternative/immunology , Disease Models, Animal , Eye Proteins/genetics , Geographic Atrophy/genetics , Geographic Atrophy/immunology , Macular Degeneration/genetics , Macular Degeneration/immunology , Membrane Proteins/genetics , Mice , Mice, Knockout , Monocytes/immunology , Nerve Tissue Proteins/genetics , Retina/immunology , Retina/pathology
10.
Vestn Oftalmol ; 129(2): 86-90, 2013.
Article Ru | MEDLINE | ID: mdl-23808188

Visual loss due to age-related macular degeneration (AMD) is caused by one or both forms of advanced disease: "wet" (neovascular) or "dry" (geographic atrophy). Immune system plays a central role in pathogenesis and progression of both AMD forms. Main genetic polymorphisms associated with risk of AMD development and progression were found to be genes that regulate inflammation especially in complement factor H gen (1q31 locus) and 10q26 locus (PLEKHAI/ARMS2/HTRA1). Association of response to treatment and genotype was shown in patients with AMD. Complete characterization of both common and rare alleles that influence AMD risk is necessary for accurate determination of individual genetic risk as well as identification of new targets for therapeutic intervention.


Geographic Atrophy , Glaucoma, Neovascular , Macular Degeneration , Pharmacogenetics/methods , Polymorphism, Genetic , Complement Factor H/genetics , Complement Pathway, Alternative/genetics , Disease Progression , Genetic Predisposition to Disease , Geographic Atrophy/complications , Geographic Atrophy/immunology , Glaucoma, Neovascular/complications , Glaucoma, Neovascular/immunology , Humans , Macular Degeneration/etiology , Macular Degeneration/genetics , Macular Degeneration/physiopathology , Macular Degeneration/therapy , Mutation , Precision Medicine
11.
PLoS One ; 8(6): e67263, 2013.
Article En | MEDLINE | ID: mdl-23840644

AIMS: With ageing extracellular material is deposited in Bruch's membrane, as drusen. Lipofuscin is deposited in retinal pigment epithelial cells. Both of these changes are associated with age related macular degeneration, a disease now believed to involve chronic inflammation at the retinal-choroidal interface. We hypothesise that these molecules may act as danger signals, causing the production of inflammatory chemokines and cytokines by the retinal pigment epithelium, via activation of pattern recognition receptors. METHODS: ARPE-19 cells were stimulated in vitro with the following reported components of drusen: amyloid-ß (1-42), Carboxyethylpyrrole (CEP) modified proteins (CEP-HSA), Nε-(Carboxymethyl)lysine (CML) modified proteins and aggregated vitronectin. The cells were also stimulated with the major fluorophore of lipofuscin: N-retinylidene-N-retinylethanolamine (A2E). Inflammatory chemokine and cytokine production was assessed using Multiplex assays and ELISA. The mechanistic evaluation of the NLRP3 inflammasome pathway was assessed in a stepwise fashion. RESULTS: Of all the molecules tested only A2E induced inflammatory chemokine and cytokine production. 25 µM A2E induced the production of significantly increased levels of the chemokines IL-8, MCP-1, MCG and MIP-1α, the cytokines IL-1ß, IL-2, IL-6, and TNF-α, and the protein VEGF-A. The release of IL-1ß was studied further, and was determined to be due to NLRP3 inflammasome activation. The pathway of activation involved endocytosis of A2E, and the three inflammasome components NLRP3, ASC and activated caspase-1. Immunohistochemical staining of ABCA4 knockout mice, which show progressive accumulation of A2E levels with age, showed increased amounts of IL-1ß proximal to the retinal pigment epithelium. CONCLUSIONS: A2E has the ability to stimulate inflammatory chemokine and cytokine production by RPE cells. The pattern recognition receptor NLRP3 is involved in this process. This provides further evidence for the link between A2E, inflammation, and the pathogenesis of AMD. It also supports the recent discovery of NLRP3 inflammasome activation in AMD.


Carrier Proteins/metabolism , Epithelial Cells/metabolism , Inflammasomes/metabolism , Interleukin-1beta/biosynthesis , Retinoids/physiology , ATP-Binding Cassette Transporters/deficiency , ATP-Binding Cassette Transporters/genetics , Animals , Carrier Proteins/genetics , Cell Line , Cytoplasm/metabolism , Endocytosis , Epithelial Cells/drug effects , Epithelial Cells/immunology , Gene Knockdown Techniques , Geographic Atrophy/immunology , Geographic Atrophy/pathology , Humans , Inflammation Mediators/metabolism , Interleukin-1beta/genetics , Mice , Mice, 129 Strain , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Precursors/biosynthesis , Protein Precursors/genetics , Protein Transport , RNA, Small Interfering/genetics , Retinal Pigment Epithelium/immunology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Signal Transduction , Up-Regulation
12.
Can J Ophthalmol ; 47(3): 264-8, 2012 Jun.
Article En | MEDLINE | ID: mdl-22687304

PURPOSE: To investigate the association of age-related macular degeneration (AMD) with plasma antiphospholipid antibody levels. METHODS: This prospective study included 19 patients diagnosed as having dry-type AMD, 23 patients with exudative-type AMD, and 25 control subjects. Venous blood samples of the participants were obtained. Anticardiolipin antibodies (aCL) isotypes IgG and IgM were measured by means of an enzyme-linked immunosorbent assay. Lupus anticoagulant (LA) antibodies were measured by the dilute Russell viper venom time screen test. RESULTS: The mean aCL IgG concentration in patients with exudative-type AMD was significantly higher than in patients with dry-type AMD and control subjects. The mean ± SE of aCL IgG levels in patients with exudative-type AMD and dry-type AMD and control subjects was 5.46 ± 1.26; 2.55 ± 0.78; and 0.32 ± 0.1, respectively. The mean aCL IgM levels and LA levels in the 3 groups were not statistically different. CONCLUSIONS: Our findings suggest that elevated levels of serum aCL, a risk factor for cardiovascular and cerebrovascular diseases, may be associated with exudative-type AMD.


Antibodies, Anticardiolipin/blood , Geographic Atrophy/immunology , Wet Macular Degeneration/immunology , Aged , Cardiovascular Diseases/immunology , Cerebrovascular Disorders/immunology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoglobulin G/blood , Immunoglobulin Isotypes , Immunoglobulin M/blood , Lupus Coagulation Inhibitor/blood , Male , Prospective Studies , Risk Factors
13.
Cell ; 149(4): 847-59, 2012 May 11.
Article En | MEDLINE | ID: mdl-22541070

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.


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
14.
Exp Mol Pathol ; 93(2): 193-9, 2012 Oct.
Article En | MEDLINE | ID: mdl-22465421

Age-related macular degeneration (AMD) is the leading cause of irreversible blindness in industrial counties. Recent findings indicate that the autoimmunity is involved in the pathogenesis of the disease. However, there is no autoantibody biomarker applied in a clinical setting for diagnosis and prognosis of AMD. In order to reveal retinal antigens targeted by serum IgG from AMD patients, mouse retinal tissue proteins were separated by 2-dimensional electrophoresis and the proteins in the immunoblots that were specific for dry and wet AMD patients IgG were identified by LC-MS/MS. Retinol-binding protein 3 and aldolase C (ALDOC) were mainly recognized by IgG form wet AMD patients. Pyruvate kinase M2 (PKM2) was targeted by both dry and wet AMD and level of anti-PKM2 IgG antibody was correlated best with the stage of AMD. Expression of ALDOC and PKM2 was decreased in mouse retina from aging whereas PKM2 deposit on RPE was increased in aged mice. Our data demonstrate that sera of AMD patients contain autoantibodies against retinal proteins and anti-PKM2 IgG serves as a biomarker for diagnosis and prognosis of AMD. Further investigation of the association of anti-retinal antibody level with expression level of antigens in retina will be needed to reveal the disease pathogenesis.


Autoantibodies/blood , Geographic Atrophy/immunology , Retina/immunology , Wet Macular Degeneration/immunology , Aged , Aged, 80 and over , Animals , Autoantibodies/isolation & purification , Autoantigens/immunology , Biomarkers/blood , Female , Fructose-Bisphosphate Aldolase/immunology , Geographic Atrophy/diagnosis , Humans , Immunoglobulin G/immunology , Male , Mice , Prognosis , Pyruvate Kinase/immunology , Retinol-Binding Proteins/immunology , Wet Macular Degeneration/diagnosis
15.
Adv Exp Med Biol ; 703: 1-7, 2010.
Article En | MEDLINE | ID: mdl-20711703

The complement cascade has been identified as a key factor in the pathogenesis of age-related macular degeneration (AMD). As a result, pharmacological modulation of the complement cascade is being investigated as a therapeutic strategy for AMD. The genetic data point to a triggering of the complement cascade, which subsequently cannot be damped down. Despite promising genetic, preclinical and immunolabeling data, important questions remain to be answered regarding the role of complement in the pathogenesis of AMD. The involvement of the complement cascade in the vision threatening stages of AMD, e.g. geographic atrophy and choroidal neovascularization, remain unknown. Additionally, the optimal component(s) of the complement cascade to be targeted for modulation still need to be identified. Answering these and other questions will provide investigators with a clear framework with which to evaluate progress in the field and help guide the development of future clinical therapeutics.


Complement System Proteins/metabolism , Macular Degeneration/etiology , Macular Degeneration/immunology , Choroidal Neovascularization/immunology , Complement Activation , Complement Inactivating Agents/therapeutic use , Geographic Atrophy/immunology , Humans , Inflammation/immunology , Macular Degeneration/drug therapy , Models, Immunological , Retinal Drusen/immunology
16.
Br J Ophthalmol ; 94(7): 918-25, 2010 Jul.
Article En | MEDLINE | ID: mdl-19965817

AIM: To determine the sub-macular Bruch's membrane (BrM) macrophage count and the choroidal and BrM macrophage immunophenotype in normal eyes and in eyes with early and advanced age-related macular degeneration (AMD). METHODS: BrM macrophages were counted in 125 human eyes (normal, normal aged, early AMD and geographical atrophy), and CD68 and inducible nitric oxide synthase (iNOS) immunohistochemistry was performed on 16 human eyes (normal, normal aged, early AMD, geographical atrophy and disciform scarring). All eyes were examined clinically ante mortem. Results were correlated with histopathological features, including basal laminar deposit and membranous debris, and with clinical fundus appearance. RESULTS: CD68(+) macrophages were found in the choroid of normal human eyes, and did not express iNOS. Expression of iNOS by choroidal macrophages (as well as endothelial cells and pericytes) was associated with: (1) recruitment of macrophages to BrM in early AMD eyes with soft drusen or thick continuous basal laminar deposit, corresponding to clinically detectable soft drusen or pigment changes; and (2) active disciform scarring. iNOS expression was absent in BrM macrophages, suggesting immunomodulatory differences between the choroid and BrM. The highest BrM macrophage counts were found in eyes with subclinical choroidal neovascularisation. CONCLUSION: The presence of extracellular deposits (soft drusen and thick continuous basal laminar deposit) is associated with macrophage recruitment to BrM and alteration in the immunophenotype of resident choroidal macrophages.


Bruch Membrane/pathology , Choroid/pathology , Macrophages/pathology , Macular Degeneration/pathology , Aged , Aged, 80 and over , Aging/pathology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Bruch Membrane/immunology , Cell Count , Choroid/immunology , Choroidal Neovascularization/immunology , Choroidal Neovascularization/pathology , Choroidal Neovascularization/physiopathology , Disease Progression , Geographic Atrophy/immunology , Geographic Atrophy/pathology , Geographic Atrophy/physiopathology , Humans , Immunophenotyping , Macrophages/immunology , Macular Degeneration/immunology , Macular Degeneration/physiopathology , Nitric Oxide Synthase Type II/metabolism , Visual Acuity
...