Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 341
Filtrar
1.
J Clin Invest ; 134(9)2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38690727

RESUMO

Careful regulation of the complement system is critical for enabling complement proteins to titrate immune defense while also preventing collateral tissue damage from poorly controlled inflammation. In the eye, this balance between complement activity and inhibition is crucial, as a low level of basal complement activity is necessary to support ocular immune privilege, a prerequisite for maintaining vision. Dysregulated complement activation contributes to parainflammation, a low level of inflammation triggered by cellular damage that functions to reestablish homeostasis, or outright inflammation that disrupts the visual axis. Complement dysregulation has been implicated in many ocular diseases, including glaucoma, diabetic retinopathy, and age-related macular degeneration (AMD). In the last two decades, complement activity has been the focus of intense investigation in AMD pathogenesis, leading to the development of novel therapeutics for the treatment of atrophic AMD. This Review outlines recent advances and challenges, highlighting therapeutic approaches that have advanced to clinical trials, as well as providing a general overview of the complement system in the posterior segment of the eye and selected ocular diseases.


Assuntos
Ativação do Complemento , Proteínas do Sistema Complemento , Degeneração Macular , Humanos , Degeneração Macular/imunologia , Degeneração Macular/patologia , Proteínas do Sistema Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Ativação do Complemento/imunologia , Animais , Olho/imunologia , Olho/patologia
2.
Science ; 379(6627): 45-62, 2023 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-36603072

RESUMO

Age-related macular degeneration is a prevalent neuroinflammatory condition and a major cause of blindness driven by genetic and environmental factors such as obesity. In diseases of aging, modifiable factors can be compounded over the life span. We report that diet-induced obesity earlier in life triggers persistent reprogramming of the innate immune system, lasting long after normalization of metabolic abnormalities. Stearic acid, acting through Toll-like receptor 4 (TLR4), is sufficient to remodel chromatin landscapes and selectively enhance accessibility at binding sites for activator protein-1 (AP-1). Myeloid cells show less oxidative phosphorylation and shift to glycolysis, ultimately leading to proinflammatory cytokine transcription, aggravation of pathological retinal angiogenesis, and neuronal degeneration associated with loss of visual function. Thus, a past history of obesity reprograms mononuclear phagocytes and predisposes to neuroinflammation.


Assuntos
Memória Epigenética , Imunidade Inata , Degeneração Macular , Doenças Neuroinflamatórias , Obesidade , Animais , Camundongos , Citocinas/genética , Imunidade Inata/genética , Doenças Neuroinflamatórias/genética , Doenças Neuroinflamatórias/imunologia , Obesidade/genética , Fagócitos/imunologia , Transcrição Gênica , Degeneração Macular/genética , Degeneração Macular/imunologia , Reprogramação Celular/genética , Receptor 4 Toll-Like/genética
4.
Front Immunol ; 12: 769242, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34819935

RESUMO

Complement Factor H-Related 3 (FHR-3) is a major regulator of the complement system, which is associated with different diseases, such as age-related macular degeneration (AMD). However, the non-canonical local, cellular functions of FHR-3 remained poorly understood. Here, we report that FHR-3 bound to oxidative stress epitopes and competed with FH for interaction. Furthermore, FHR-3 was internalized by viable RPE cells and modulated time-dependently complement component (C3, FB) and receptor (C3aR, CR3) expression of human RPE cells. Independently of any external blood-derived proteins, complement activation products were detected. Anaphylatoxin C3a was visualized in treated cells and showed a translocation from the cytoplasm to the cell membrane after FHR-3 exposure. Subsequently, FHR-3 induced a RPE cell dependent pro-inflammatory microenvironment. Inflammasome NLRP3 activation and pro-inflammatory cytokine secretion of IL-1ß, IL-18, IL-6 and TNF-α were induced after FHR-3-RPE interaction. Our previously published monoclonal anti-FHR-3 antibody, which was chimerized to reduce immunogenicity, RETC-2-ximab, ameliorated the effect of FHR-3 on ARPE-19 cells. Our studies suggest FHR-3 as an exogenous trigger molecule for the RPE cell "complosome" and as a putative target for a therapeutic approach for associated degenerative diseases.


Assuntos
Proteínas Sanguíneas/imunologia , Ativação do Complemento/imunologia , Fator H do Complemento/imunologia , Células Epiteliais/imunologia , Epitélio Pigmentado da Retina/citologia , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Linhagem Celular , Ativação do Complemento/genética , Complemento C3/genética , Complemento C3/imunologia , Complemento C3/metabolismo , Fator H do Complemento/genética , Fator H do Complemento/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/genética , Transição Epitelial-Mesenquimal/imunologia , Expressão Gênica/genética , Expressão Gênica/imunologia , Células HEK293 , Humanos , Inflamassomos/genética , Inflamassomos/imunologia , Inflamassomos/metabolismo , Antígeno de Macrófago 1/genética , Antígeno de Macrófago 1/imunologia , Antígeno de Macrófago 1/metabolismo , Degeneração Macular/genética , Degeneração Macular/imunologia , Degeneração Macular/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Polimorfismo de Nucleotídeo Único/genética , Polimorfismo de Nucleotídeo Único/imunologia
5.
Front Immunol ; 12: 738521, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34721402

RESUMO

Background: Neovascular age-related macular degeneration (nAMD) is a leading cause of blindness in older people. Low-grade inflammation is well-known as one of the pathogenic mechanisms in nAMD. Anti-vascular endothelial growth factor (VEGF) therapy is the first-line treatment for nAMD, although macula atrophy (MA) developed under anti-VEGF therapy causes irreversible visual function impairment and is recognized as a serious disorder. Here, we show specific expression patterns of aqueous humor (AH) cytokines in nAMD eyes developing MA under intravitreal injection of aflibercept (IVA) as an anti-VEGF antibody and present predictive cytokines as biomarkers for the incidence of MA in nAMD eyes under IVA treatment. Methods: Twenty-eight nAMD patients received three consecutive monthly IVA, followed by a pro re nata regimen for 2 years. AH specimens were collected before first IVA (pre-IVA) and before third IVA (post-IVA). AH cytokine levels, visual acuity (VA), and central retinal thickness (CRT) were measured. Results: Two-year incidence of MA was 21.4%. In nAMD eyes developing MA [MA (+) group], pre-IVA levels of monocyte chemoattractant protein-1 (MCP-1), macrophage inflammatory protein (MIP)-1ß, VEGF and post-IVA level of MCP-1 were higher than those in nAMD eyes without MA [MA (-) group]. In hierarchical cluster analysis, pre-IVA MCP-1 and VEGF were grouped into the same subcluster, as were post-IVA MCP-1 and CRT. In principal component analysis, principal component loading (PCL) of pre-IVA interferon-γ-inducible protein 10 (IP-10) was 0.61, but PCL of post-IVA IP-10 decreased to -0.09. In receiver operating characteristic analysis and Kaplan-Meier curves, pre-IVA MCP-1, MIP-1ß, and VEGF and post-IVA interleukin-6, MCP-1, and MIP-1ß were detected as predictive factors for MA incidence. In 2-year clinical course, changes of VA in groups with high levels of pre-IVA MIP-1ß (over 39.9 pg/ml) and VEGF (over 150.4 pg/ml) were comparable to those in MA (+) group. Conclusion: Substantial loss of IP-10 effects and persistent inflammation contribute to incidence of MA, and screening of AH cytokine levels could be a useful method to predict MA incidence in nAMD eyes under anti-VEGF therapy.


Assuntos
Inibidores da Angiogênese/efeitos adversos , Humor Aquoso/metabolismo , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Macula Lutea/efeitos dos fármacos , Degeneração Macular/tratamento farmacológico , Proteínas Recombinantes de Fusão/efeitos adversos , Neovascularização Retiniana , Adulto , Idoso , Idoso de 80 Anos ou mais , Inibidores da Angiogênese/administração & dosagem , Humor Aquoso/imunologia , Atrofia , Biomarcadores/metabolismo , Feminino , Humanos , Injeções Intravítreas , Macula Lutea/imunologia , Macula Lutea/metabolismo , Macula Lutea/patologia , Degeneração Macular/imunologia , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Receptores de Fatores de Crescimento do Endotélio Vascular/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem , Fatores de Tempo , Resultado do Tratamento , Acuidade Visual/efeitos dos fármacos
6.
Aging (Albany NY) ; 13(18): 22040-22058, 2021 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-34544906

RESUMO

N-retinylidene-N-retinylethanolamine (A2E) plays a central role in age-related macular degeneration (AMD) by inducing angiogenesis and inflammation. A2E effects are mediated at least partly via the retinoic acid receptor (RAR)-α. Here we show that A2E binds and transactivates also peroxisome proliferator-activated receptors (PPAR) and retinoid X receptors (RXR). 9'-cis-norbixin, a di-apocarotenoid is also a ligand of these nuclear receptors (NR). Norbixin inhibits PPAR and RXR transactivation induced by A2E. Moreover, norbixin reduces protein kinase B (AKT) phosphorylation, NF-κB and AP-1 transactivation and mRNA expression of the inflammatory interleukins (IL) -6 and -8 and of vascular endothelial growth factor (VEGF) enhanced by A2E. By contrast, norbixin increases matrix metalloproteinase 9 (MMP9) and C-C motif chemokine ligand 2 (CCL2) mRNA expression in response to A2E. Selective PPAR-α, -ß/δ and -γ antagonists inhibit the expression of IL-6 and IL-8 while only the antagonist of PPAR-γ inhibits the transactivation of NF-κB following A2E exposure. In addition, a cocktail of all three PPARs antagonists and also HX531, an antagonist of RXR reproduce norbixin effects on inflammation. Altogether, A2E's deleterious biological effects could be inhibited through PPAR and RXR regulation. Moreover, the modulation of these NR by norbixin may open new avenues for the treatment of AMD.


Assuntos
Carotenoides/administração & dosagem , Degeneração Macular/tratamento farmacológico , PPAR alfa/imunologia , PPAR delta/imunologia , PPAR gama/imunologia , PPAR beta/imunologia , Epitélio Pigmentado da Retina/efeitos dos fármacos , Retinoides/imunologia , Inibidores da Angiogênese/administração & dosagem , Animais , Humanos , Degeneração Macular/induzido quimicamente , Degeneração Macular/genética , Degeneração Macular/imunologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/etiologia , Neovascularização Patológica/genética , Neovascularização Patológica/imunologia , PPAR alfa/genética , PPAR delta/genética , PPAR gama/genética , PPAR beta/genética , Epitélio Pigmentado da Retina/imunologia , Receptores X de Retinoides/agonistas , Receptores X de Retinoides/genética , Receptores X de Retinoides/imunologia , Retinoides/efeitos adversos , Suínos , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/imunologia
7.
Int J Mol Sci ; 22(16)2021 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-34445430

RESUMO

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.


Assuntos
Citocinas/metabolismo , Inativação Gênica , Degeneração Macular/genética , Epitélio Pigmentado da Retina/imunologia , Linhagem Celular , Fator H do Complemento/genética , Proteínas do Sistema Complemento/metabolismo , Citocinas/genética , Regulação da Expressão Gênica , Humanos , Degeneração Macular/imunologia , Modelos Biológicos , NF-kappa B/genética , NF-kappa B/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Transdução de Sinais
8.
J Immunol Res ; 2021: 9975628, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34239945

RESUMO

Age-related macular degeneration (AMD), a degenerative disease of the outer retina, is the leading cause of blindness among the elderly. A hallmark of geographic atrophy (GA), an advanced type of nonneovascular AMD (dry AMD), is photoreceptor and retinal pigment epithelium (RPE) cell death. Currently, there are no FDA-approved therapies for GA due to a lack of understanding of the disease-causing mechanisms. Increasing evidence suggests that chronic inflammation plays a predominant role in the pathogenesis of dry AMD. Dead or stressed cells release danger signals and inflammatory factors, which causes further damage to neighboring cells. It has been reported that type I interferon (IFN) response is activated in RPE cells in patients with AMD. However, how RPE cells sense stress to initiate IFN response and cause further damage to the retina are still unknown. Although it has been reported that RPE can respond to extracellularly added dsRNA, it is unknown whether and how RPE detects and senses internally generated or internalized nucleic acids. Here, we elucidated the molecular mechanism by which RPE cells sense intracellular nucleic acids. Our data demonstrate that RPE cells can respond to intracellular RNA and induce type I IFN responses via the RIG-I (DExD/H-box helicase 58, DDX58) RNA helicase. In contrast, we showed that RPE cells were unable to directly sense and respond to DNA through the cGAS-STING pathway. We demonstrated that this was due to the absence of the cyclic GMP-AMP synthase (cGAS) DNA sensor in these cells. The activation of IFN response via RIG-I induced expression of cell death effectors and caused barrier function loss in RPE cells. These data suggested that RPE-intrinsic pathways of nucleic acid sensing are biased toward RNA sensing.


Assuntos
Proteína DEAD-box 58/metabolismo , Degeneração Macular/imunologia , RNA de Cadeia Dupla/metabolismo , Receptores Imunológicos/metabolismo , Epitélio Pigmentado da Retina/patologia , Apoptose/imunologia , Linhagem Celular , Proteína DEAD-box 58/genética , Técnicas de Inativação de Genes , Humanos , Interferon Tipo I/metabolismo , Degeneração Macular/patologia , Estresse Oxidativo , Receptores Imunológicos/genética , Epitélio Pigmentado da Retina/imunologia , Epitélio Pigmentado da Retina/metabolismo
9.
Mediators Inflamm ; 2021: 6622934, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33953642

RESUMO

Considering the immunological impairment in age-related macular degeneration (AMD), we aimed to determine the associations of IL-9 rs1859430, rs2069870, rs11741137, rs2069885, and rs2069884 and IL-10 rs1800871, rs1800872, and rs1800896 polymorphisms and their haplotypes, as well as the serum levels of IL-9 and IL-10 with AMD. 1209 participants were enrolled in our study. SNPs were genotyped using TaqMan SNP genotyping assays by real-time PCR method. IL-9 and IL-10 serum levels were evaluated using ELISA kits. Our study results have shown that haplotypes A-G-C-G-G and G-A-T-A-T of IL-9 SNPs are associated with the decreased odds of early AMD occurrence (p = 0.035 and p = 0.015, respectively). A set of rare haplotypes was associated with the decreased odds of exudative AMD occurrence (p = 0.033). Also, IL-10 serum levels were lower in exudative AMD than in controls (p = 0.049), patients with early AMD (p = 0.017), and atrophic AMD (p = 0.008). Furthermore, exudative AMD patients with IL-10 rs1800896 CT and TT genotypes had lower IL-10 serum concentrations than those with wild-type (CC) genotype (p = 0.048). In conclusion, our study suggests that IL-10 serum levels can be associated with a minor allele at IL-10 rs1800896 and exudative AMD. The haplotypes of IL-9 SNPs were also associated with the decreased odds of early and exudative AMD.


Assuntos
Interleucina-10/genética , Interleucina-9/genética , Degeneração Macular/genética , Polimorfismo de Nucleotídeo Único , Idoso , Idoso de 80 Anos ou mais , Feminino , Haplótipos , Humanos , Interleucina-10/sangue , Interleucina-9/sangue , Degeneração Macular/etiologia , Degeneração Macular/imunologia , Masculino , Pessoa de Meia-Idade , População Branca
10.
Front Immunol ; 12: 660194, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33868311

RESUMO

Inflammation is a common denominator of diseases. The complement system, an intrinsic part of the innate immune system, is a key driver of inflammation in numerous disorders. Recently, a family of proteins has been suggested to be of vital importance in conditions characterized by complement dysregulation: the human Factor H (FH) family. This group of proteins consists of FH, Factor H-like protein 1 and five Factor H-related proteins. The FH family has been linked to infectious, vascular, eye, kidney and autoimmune diseases. In contrast to FH, the functions of the other highly homologous proteins are largely unknown and, hence, their role in the different disease-specific pathogenic mechanisms remains elusive. In this perspective review, we address the major challenges ahead in this emerging area, including 1) the controversies about the functional roles of the FH protein family, 2) the discrepancies in quantification of the FH protein family, 3) the unmet needs for validated tools and 4) limitations of animal models. Next, we also discuss the opportunities that exist for the immunology community. A strong multidisciplinary approach is required to solve these obstacles and is only possible through interdisciplinary collaboration between biologists, chemists, geneticists and physicians. We position this review in light of our own perspective, as principal investigators of the SciFiMed Consortium, a consortium aiming to create a comprehensive analytical system for the quantitative and functional assessment of the entire FH protein family.


Assuntos
Ativação do Complemento/imunologia , Proteínas do Sistema Complemento/imunologia , Inflamação/imunologia , Degeneração Macular/imunologia , Receptores de Complemento/imunologia , Animais , Fator H do Complemento/análise , Fator H do Complemento/imunologia , Fator H do Complemento/metabolismo , Proteínas do Sistema Complemento/análise , Proteínas do Sistema Complemento/metabolismo , Ensaio de Imunoadsorção Enzimática/métodos , Humanos , Inflamação/sangue , Inflamação/metabolismo , Degeneração Macular/metabolismo , Receptores de Complemento/metabolismo , Reprodutibilidade dos Testes
11.
Methods Mol Biol ; 2227: 1-20, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33847926

RESUMO

The complement cascade is an evolutionary ancient innate immune defense system, playing a major role in the defense against infections. Its function in maintaining host homeostasis on activated cells has been emphasized by the crucial role of its overactivation in ever growing number of diseases, such as atypical hemolytic uremic syndrome (aHUS), autoimmune diseases as systemic lupus erythematosus (SLE), C3 glomerulopathies (C3GN), age-related macular degeneration (AMD), graft rejection, Alzheimer disease, and cancer, to name just a few. The last decade of research on complement has extended its implication in many pathological processes, offering new insights to potential therapeutic targets and asserting the necessity of reliable, sensitive, specific, accurate, and reproducible biomarkers to decipher complement role in pathology. We need to evaluate accurately which pathway or role should be targeted pharmacologically, and optimize treatment efficacy versus toxicity. This chapter is an introduction to the role of complement in human diseases and the use of complement-related biomarkers in the clinical practice. It is a part of a book intending to give reliable and standardized methods to evaluate complement according to nowadays needs and knowledge.


Assuntos
Proteínas do Sistema Complemento/análise , Testes Diagnósticos de Rotina/métodos , Padrões de Prática Médica , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Síndrome Hemolítico-Urêmica Atípica/imunologia , Síndrome Hemolítico-Urêmica Atípica/metabolismo , Doenças Autoimunes/diagnóstico , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Ativação do Complemento/fisiologia , Proteínas do Sistema Complemento/metabolismo , Desenvolvimento de Medicamentos , Rejeição de Enxerto/diagnóstico , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/metabolismo , Humanos , Nefropatias/diagnóstico , Nefropatias/imunologia , Nefropatias/metabolismo , Degeneração Macular/diagnóstico , Degeneração Macular/imunologia , Degeneração Macular/metabolismo , Neoplasias/diagnóstico , Neoplasias/imunologia , Neoplasias/metabolismo
12.
Cell Mol Life Sci ; 78(10): 4487-4505, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33751148

RESUMO

Age-related macular degeneration (AMD) is a chronic and progressive degenerative disease of the retina, which culminates in blindness and affects mainly the elderly population. AMD pathogenesis and pathophysiology are incredibly complex due to the structural and cellular complexity of the retina, and the variety of risk factors and molecular mechanisms that contribute to disease onset and progression. AMD is driven by a combination of genetic predisposition, natural ageing changes and lifestyle factors, such as smoking or nutritional intake. The mechanism by which these risk factors interact and converge towards AMD are not fully understood and therefore drug discovery is challenging, where no therapeutic attempt has been fully effective thus far. Genetic and molecular studies have identified the complement system as an important player in AMD. Indeed, many of the genetic risk variants cluster in genes of the alternative pathway of the complement system and complement activation products are elevated in AMD patients. Nevertheless, attempts in treating AMD via complement regulators have not yet been successful, suggesting a level of complexity that could not be predicted only from a genetic point of view. In this review, we will explore the role of complement system in AMD development and in the main molecular and cellular features of AMD, including complement activation itself, inflammation, ECM stability, energy metabolism and oxidative stress.


Assuntos
Envelhecimento , Ativação do Complemento , Proteínas do Sistema Complemento/metabolismo , Inflamação/fisiopatologia , Degeneração Macular/patologia , Estresse Oxidativo , Animais , Predisposição Genética para Doença , Humanos , Degeneração Macular/imunologia , Degeneração Macular/metabolismo , Fatores de Risco
13.
Neuropharmacology ; 188: 108513, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33662390

RESUMO

The prevalence of age-related macular degeneration (AMD) has increased in the last years. Although anti-VEGF agents have improved the prognosis of exudative AMD, dry AMD has still devastating effects on elderly people vision. Oxidative stress and inflammation are mechanisms involved in AMD pathogenesis and its progression. Molecular pathways involving epidermal growth factor receptor (EGFR), bone morphogenetic protein (BMP4) and the nuclear erythroid related factor 2 (Nrf2) are behind oxidative stress in AMD due to their participation in antioxidant cellular pathways. As a consequence of the disbalance produced in the antioxidant mechanisms, there is an activation of innate and adaptative immune response with cell recruitment, changes in complement factors expression, and modification of cellular milieu. Different therapies are being studied to treat dry AMD based on the possible effects on antioxidant molecular pathways or their action on the immune response. There is a wide range of treatments presented in this review, from natural antioxidant compounds to cell and gene therapy, based on their mechanisms. Finally, we hypothesize that alpha-1-antitrypsin (AAT), an anti-inflammatory and immunomodulatory molecule that can also modulate antioxidant cellular defenses, could be a good candidate for testing in AMD. This article is part of the special ssue on 'The Quest for Disease-Modifying Therapies for Neurodegenerative Disorders'.


Assuntos
Degeneração Macular/fisiopatologia , Degeneração Macular/terapia , Estresse Oxidativo , Envelhecimento , Animais , Antioxidantes , Humanos , Degeneração Macular/imunologia , Degeneração Macular/metabolismo
14.
Front Immunol ; 12: 789897, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35069568

RESUMO

Complement factor I (FI) is a central inhibitor of the complement system, and impaired FI function increases complement activation, contributing to diseases such as age-related macular degeneration (AMD) and atypical hemolytic uremic syndrome (aHUS). Genetic variation in complement factor I (CFI) has been identified in both AMD and aHUS, with more than half of these variants leading to reduced FI secretion levels. For many of the variants with normal FI secretion, however, functional implications are not yet known. Here we studied 11 rare missense variants, with FI secretion levels comparable to wildtype, but a predicted damaging effects based on the Combined Annotation Dependent Depletion (CADD) score. Three variants (p.Pro50Ala, p.Arg339Gln, and p.Ser570Thr) were analyzed in plasma and serum samples of carriers affected by AMD. All 11 variants (nine for the first time in this study) were recombinantly expressed and the ability to degrade C3b was studied with the C3b degradation assay. The amount of degradation was determined by measuring the degradation product iC3b with ELISA. Eight of 11 (73%) mutant proteins (p.Pro50Ala, p.Arg339Gln, p.Ile340Thr, p.Gly342Glu, p.Gly349Arg, p.Arg474Gln, p.Gly487Cys, and p.Gly512Ser) showed significantly impaired C3b degradation, and were therefore classified as likely pathogenic. Our data indicate that genetic variants in CFI with a CADD score >20 are likely to affect FI function, and that monitoring iC3b in a degradation assay is a useful tool to establish the pathogenicity of CFI variants in functional studies.


Assuntos
Síndrome Hemolítico-Urêmica Atípica , Fator I do Complemento , Degeneração Macular , Mutação de Sentido Incorreto , Substituição de Aminoácidos , Síndrome Hemolítico-Urêmica Atípica/sangue , Síndrome Hemolítico-Urêmica Atípica/genética , Síndrome Hemolítico-Urêmica Atípica/imunologia , Complemento C3b/imunologia , Complemento C3b/metabolismo , Fator I do Complemento/genética , Fator I do Complemento/imunologia , Fator I do Complemento/metabolismo , Feminino , Humanos , Degeneração Macular/sangue , Degeneração Macular/genética , Degeneração Macular/imunologia , Masculino
15.
Elife ; 92020 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-33305736

RESUMO

NLRP3 inflammasome activation and complement-mediated inflammation have been implicated in promoting choroidal neovascularization (CNV) in age-related macular degeneration (AMD), but central questions regarding their contributions to AMD pathogenesis remain unanswered. Key open questions are (1) whether NLRP3 inflammasome activation mainly in retinal pigment epithelium (RPE) or rather in non-RPE cells promotes CNV, (2) whether inflammasome activation in CNV occurs via NLRP3 or also through NLRP3-independent mechanisms, and (3) whether complement activation induces inflammasome activation in CNV. Here we show in a neovascular AMD mouse model that NLRP3 inflammasome activation in non-RPE cells but not in RPE cells promotes CNV. We demonstrate that both NLRP3-dependent and NLRP3-independent inflammasome activation mechanisms induce CNV. Finally, we find that complement and inflammasomes promote CNV through independent mechanisms. Our findings uncover an unexpected role of non-NLRP3 inflammasomes for CNV and suggest that combination therapies targeting inflammasomes and complement may offer synergistic benefits to inhibit CNV.


Assuntos
Neovascularização de Coroide/metabolismo , Ativação do Complemento , Proteínas do Sistema Complemento/metabolismo , Inflamassomos/metabolismo , Degeneração Macular/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Animais , Caspase 1/genética , Caspase 1/metabolismo , Caspases Iniciadoras/genética , Caspases Iniciadoras/metabolismo , Neovascularização de Coroide/genética , Neovascularização de Coroide/imunologia , Neovascularização de Coroide/patologia , Modelos Animais de Doenças , Inflamassomos/genética , Degeneração Macular/genética , Degeneração Macular/imunologia , Degeneração Macular/patologia , Camundongos Knockout , Camundongos Transgênicos , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Epitélio Pigmentado da Retina/imunologia , Epitélio Pigmentado da Retina/patologia , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
16.
Asia Pac J Ophthalmol (Phila) ; 10(1): 114-120, 2020 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-33290288

RESUMO

ABSTRACT: Many of the risk factors for developing severe coronavirus disease 2019 (COVID-19) are also risk factors for eye diseases such as age-related macular degeneration (AMD). During the past decades, macrophages and the complement pathway (as a part of the innate immune system) have been identified as important contributors to the development of AMD, and we suggest that these mechanisms are of similar importance for the clinical course of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections. Based on the experience with AMD, we discuss how behavioral factors such as diet, smoking and higher body mass index, as well as genetic determinants such as the complement and immune pathway genes may lead to the overactive inflammatory phenotypes seen in some patients with COVID-19, and may in part explain the heterogeneity of disease manifestations and outcomes. Based on this experience, we discuss potential genetic research projects and elaborate on preventive and treatment approaches related to COVID-19.


Assuntos
COVID-19/imunologia , Proteínas do Sistema Complemento/fisiologia , Inflamação/imunologia , Macrófagos/fisiologia , Degeneração Macular/imunologia , SARS-CoV-2 , Envelhecimento/fisiologia , Animais , COVID-19/fisiopatologia , Comportamentos de Risco à Saúde , Humanos , Imunidade Inata , Degeneração Macular/fisiopatologia , Fatores de Risco
17.
Front Immunol ; 11: 611830, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33363547

RESUMO

Complement-mediated inflammation or dysregulation in lipid metabolism are associated with the pathogenesis of several diseases. These include age-related macular degeneration (AMD), C3 glomerulonephritis (C3GN), dense deposit disease (DDD), atherosclerosis, and Alzheimer's disease (AD). In all these diseases, formation of characteristic lipid-rich deposits is evident. Here, we will discuss molecular mechanisms whereby dysfunction of complement, and especially of its key regulator factor H, could be involved in lipid accumulation and related inflammation. The genetic associations to factor H polymorphisms, the role of factor H in the resolution of inflammation in lipid-rich deposits, modification of macrophage functions, and complement-mediated clearance of apoptotic and damaged cells indicate that the function of factor H is crucial in limiting inflammation in these diseases.


Assuntos
Via Alternativa do Complemento , Inflamação/metabolismo , Metabolismo dos Lipídeos , Doença de Alzheimer/genética , Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Animais , Aterosclerose/genética , Aterosclerose/imunologia , Aterosclerose/metabolismo , Fator H do Complemento/química , Fator H do Complemento/genética , Fator H do Complemento/metabolismo , Glomerulonefrite/genética , Glomerulonefrite/imunologia , Glomerulonefrite/metabolismo , Humanos , Inflamação/genética , Inflamação/imunologia , Degeneração Macular/genética , Degeneração Macular/imunologia , Degeneração Macular/metabolismo , Polimorfismo Genético , Conformação Proteica , Transdução de Sinais , Relação Estrutura-Atividade
18.
J Neuroinflammation ; 17(1): 355, 2020 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-33239022

RESUMO

BACKGROUND: Macular fibrosis causes irreparable vision loss in neovascular age-related macular degeneration (nAMD) even with anti-vascular endothelial growth factor (VEGF) therapy. Inflammation is known to play an important role in macular fibrosis although the underlying mechanism remains poorly defined. The aim of this study was to understand how infiltrating macrophages and complement proteins may contribute to macular fibrosis. METHODS: Subretinal fibrosis was induced in C57BL/6J mice using the two-stage laser protocol developed by our group. The eyes were collected at 10, 20, 30 and 40 days after the second laser and processed for immunohistochemistry for infiltrating macrophages (F4/80 and Iba-1), complement components (C3a and C3aR) and fibrovascular lesions (collagen-1, Isolectin B4 and α-SMA). Human retinal sections with macular fibrosis were also used in the study. Bone marrow-derived macrophages (BMDMs) from C57BL/6J mice were treated with recombinant C3a, C5a or TGF-ß for 48 and 96 h. qPCR, Western blot and immunohistochemistry were used to examine the expression of myofibroblast markers. The involvement of C3a-C3aR pathway in macrophage to myofibroblast transition (MMT) and subretinal fibrosis was further investigated using a C3aR antagonist (C3aRA) and a C3a blocking antibody in vitro and in vivo. RESULTS: Approximately 20~30% of F4/80+ (or Iba-1+) infiltrating macrophages co-expressed α-SMA in subretinal fibrotic lesions both in human nAMD eyes and in the mouse model. TGF-ß and C3a, but not C5a treatment, significantly upregulated expression of α-SMA, fibronectin and collagen-1 in BMDMs. C3a-induced upregulation of α-SMA, fibronectin and collagen-1 in BMDMs was prevented by C3aRA treatment. In the two-stage laser model of induced subretinal fibrosis, treatment with C3a blocking antibody but not C3aRA significantly reduced vascular leakage and Isolectin B4+ lesions. The treatment did not significantly alter collagen-1+ fibrotic lesions. CONCLUSIONS: MMT plays a role in macular fibrosis secondary to nAMD. MMT can be induced by TGF-ß and C3a but not C5a. Further research is required to fully understand the role of MMT in macular fibrosis. Macrophage to myofibroblast transition (MMT) contributes to subretinal fibrosis. Subretinal fibrosis lesions contain various cell types, including macrophages and myofibroblasts, and are fibrovascular. Myofibroblasts are key cells driving pathogenic fibrosis, and they do so by producing excessive amount of extracellular matrix proteins. We have found that infiltrating macrophages can transdifferentiate into myofibroblasts, a phenomenon termed macrophage to myofibroblast transition (MMT) in macular fibrosis. In addition to TGF-ß1, C3a generated during complement activation in CNV can also induce MMT contributing to macular fibrosis. RPE = retinal pigment epithelium. BM = Bruch's membrane. MMT = macrophage to myofibroblast transition. TGFB = transforming growth factor ß. a-SMA = alpha smooth muscle actin. C3a = complement C3a.


Assuntos
Macrófagos/patologia , Degeneração Macular/patologia , Miofibroblastos/patologia , Neovascularização Patológica/patologia , Retina/patologia , Animais , Células Cultivadas , Complemento C3a/toxicidade , Feminino , Fibrose , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Degeneração Macular/induzido quimicamente , Degeneração Macular/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/imunologia , Neovascularização Patológica/induzido quimicamente , Neovascularização Patológica/imunologia , Retina/efeitos dos fármacos , Retina/imunologia
19.
Med Sci (Paris) ; 36(10): 886-892, 2020 Oct.
Artigo em Francês | MEDLINE | ID: mdl-33026331

RESUMO

Age-related macular degeneration (AMD) is a complex, highly heritable, multifactorial disease caused by the interplay of age and genetic and environmental risk factors. No treatment has yet been found to treat the slowly progressing atrophic form of AMD. All forms of AMD are invariably associated with an accumulation of mononuclear phagocytes (MP) in the subretinal space, a family of cells that include inflammatory and resident macrophages. We here present an overview of the inflammatory process occurring in AMD and discuss the origin of MPs and the consequences of their accumulation in the subretinal space. Finally, we will review the role played by the established risk factors for AMD to promote the switch from beneficial inflammation in early stage to a deleterious inflammation in the advanced stage of the disease.


TITLE: Sur les origines inflammatoires de la DMLA. ABSTRACT: La dégénérescence maculaire liée à l'âge (DMLA) est une maladie multifactorielle hautement héréditaire qui survient chez le sujet âgé et est causée par une combinaison de facteurs de risques génétiques et environnementaux. Les formes atrophiques de la maladie constituent aujourd'hui une impasse thérapeutique. La physiopathologie de la DMLA est invariablement associée à une accumulation dans l'espace sous-rétinien, de phagocytes mononucléés (PM), une famille de cellules qui inclue des macrophages résidents et inflammatoires. Nous aborderons dans cette revue l'ensemble des mécanismes de cette inflammation spécifique, de l'origine des PM aux conséquences de leur accumulation dans l'espace sous-rétinien. Finalement, nous discuterons de l'impact des facteurs de risques génétiques et environnementaux établis de la DMLA sur le passage d'une inflammation bénéfique aux stades précoces de la maladie à une inflammation délétère aux stades avancés.


Assuntos
Inflamação/complicações , Degeneração Macular/etiologia , Olho/imunologia , Olho/metabolismo , Olho/patologia , Humanos , Privilégio Imunológico/fisiologia , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/fisiologia , Degeneração Macular/epidemiologia , Degeneração Macular/imunologia , Degeneração Macular/metabolismo , Fatores de Risco
20.
Proc Natl Acad Sci U S A ; 117(18): 9952-9963, 2020 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-32345717

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Anticorpos Anti-Idiotípicos/farmacologia , Atrofia Geográfica/tratamento farmacológico , Serina Peptidase 1 de Requerimento de Alta Temperatura A/genética , Degeneração Macular/tratamento farmacológico , Proteínas Adaptadoras de Transdução de Sinal/isolamento & purificação , Idoso , Animais , Anticorpos Anti-Idiotípicos/genética , Anticorpos Anti-Idiotípicos/imunologia , Biomarcadores/sangue , Progressão da Doença , Feminino , Predisposição Genética para Doença , Genótipo , Atrofia Geográfica/sangue , Atrofia Geográfica/genética , Atrofia Geográfica/imunologia , Serina Peptidase 1 de Requerimento de Alta Temperatura A/antagonistas & inibidores , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Fragmentos Fab das Imunoglobulinas/farmacologia , Degeneração Macular/sangue , Degeneração Macular/genética , Degeneração Macular/imunologia , Masculino , Polimorfismo de Nucleotídeo Único/genética , Proteoma/genética , Proteoma/imunologia , Ratos , Retina/efeitos dos fármacos , Retina/imunologia , Retina/patologia , Bibliotecas de Moléculas Pequenas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA