Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Nat Commun ; 12(1): 7293, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34911940

RESUMO

Age-related Macular Degeneration (AMD), a blinding eye disease, is characterized by pathological protein- and lipid-rich drusen deposits underneath the retinal pigment epithelium (RPE) and atrophy of the RPE monolayer in advanced disease stages - leading to photoreceptor cell death and vision loss. Currently, there are no drugs that stop drusen formation or RPE atrophy in AMD. Here we provide an iPSC-RPE AMD model that recapitulates drusen and RPE atrophy. Drusen deposition is dependent on AMD-risk-allele CFH(H/H) and anaphylatoxin triggered alternate complement signaling via the activation of NF-κB and downregulation of autophagy pathways. Through high-throughput screening we identify two drugs, L-745,870, a dopamine receptor antagonist, and aminocaproic acid, a protease inhibitor that reduce drusen deposits and restore RPE epithelial phenotype in anaphylatoxin challenged iPSC-RPE with or without the CFH(H/H) genotype. This comprehensive iPSC-RPE model replicates key AMD phenotypes, provides molecular insight into the role of CFH(H/H) risk-allele in AMD, and discovers two candidate drugs to treat AMD.


Assuntos
Ácido Aminocaproico/farmacologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Degeneração Macular/tratamento farmacológico , Piridinas/farmacologia , Pirróis/farmacologia , Epitélio Pigmentado da Retina/efeitos dos fármacos , Alelos , Fator H do Complemento/genética , Fator H do Complemento/metabolismo , Avaliação Pré-Clínica de Medicamentos , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Degeneração Macular/genética , Degeneração Macular/metabolismo , Modelos Biológicos , Fenótipo , Epitélio Pigmentado da Retina/metabolismo
2.
Ophthalmology ; 125(3): 391-397, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29032853

RESUMO

PURPOSE: Considerable controversy has erupted in recent years regarding whether genotyping should be part of standard care for patients with age-related macular degeneration (AMD) who are being considered for treatment with antioxidants and zinc. We aimed to determine whether genotype predicts response to supplements in AMD. DESIGN: Three separate statistical teams reanalyzed data derived from the Age-Related Eye Disease Study (AREDS), receiving data prepared by the AREDS investigators and, separately, data from investigators reporting findings that support the use of genotyping. PARTICIPANTS: The population of interest was AREDS participants with AMD worse than category 1 and genotyping data available. Data from the 2 groups overlap imperfectly with respect to measurements made: the largest common set involved 879 participants for whom the same CFH and ARMS2 single nucleotide polymorphisms were measured by both groups. METHODS: Each team took a separate but complementary approach. One team focused on data concordance between conflicting studies. A second team focused on replicating the key claim of an interaction between genotype and treatment. The third team took a blank slate approach in attempting to find baseline predictors of treatment response. MAIN OUTCOME MEASURES: Progression to advanced AMD. RESULTS: We found errors in the data used to support the initial claim of genotype-treatment interaction. Although we found evidence that high-risk patients had more to gain from treatment, we were unable to replicate any genotype-treatment interactions after adjusting for multiple testing. We tested 1 genotype claim on an independent set of data, with negative results. Even if we assumed that interactions in fact did exist, we did not find evidence to support the claim that supplementation leads to a large increase in the risk of advanced AMD in some genotype subgroups. CONCLUSIONS: Patients who meet criteria for supplements to prevent AMD progression should be offered zinc and antioxidants without consideration of genotype.


Assuntos
Antioxidantes/uso terapêutico , DNA/genética , Degeneração Macular/genética , Polimorfismo de Nucleotídeo Único , Proteínas/genética , Zinco/uso terapêutico , Idoso , Alelos , Fator H do Complemento/genética , Fator H do Complemento/metabolismo , Suplementos Nutricionais , Progressão da Doença , Feminino , Genótipo , Humanos , Degeneração Macular/tratamento farmacológico , Degeneração Macular/metabolismo , Masculino , Prognóstico , Proteínas/metabolismo
3.
Br J Ophthalmol ; 100(12): 1731-1737, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27471039

RESUMO

BACKGROUND/AIMS: The Age-Related Eye Disease Study (AREDS) reported the beneficial impact of antioxidant and zinc supplements on the risk of progression to advanced stages of age-related macular degeneration (AMD). We evaluated the role of genetic variants in modifying the relationship between supplementation and progression to advanced AMD. METHODS: Among 4124 eyes (2317 subjects with a genetic specimen), 882 progressed from no AMD, early or intermediate AMD to overall advanced disease, including geographic atrophy (GA) and neovascular disease (NV) over the course of the clinical trial. Survival analysis using individual eyes as the unit of analysis was used to assess the effect of supplementation on AMD outcomes, with adjustment for demographic, environmental, ocular and genetic covariates. Interaction effects between supplement groups and individual complement factor H (CFH) Y402H and age-related maculopathy susceptibility 2 (ARMS2) genotypes, and composite genetic risk groups combining the number of risk alleles for both loci, were evaluated for their association with progression. RESULTS: Among antioxidant and zinc supplement users compared with the placebo group, subjects with a non-risk genotype for CFH (TT) had a lower risk of progression to advanced AMD (HR: 0.55, 95% CI 0.32 to 0.95, p=0.033). No significant treatment effect was apparent among subjects who were homozygous for the CFH risk allele (CC). A protective effect was observed among high-risk ARMS2 (TT) carriers (HR: 0.52, 95% CI 0.33 to 0.82, p=0.005). Similar results were seen for the NV subtype but not GA. CONCLUSIONS: The effectiveness of antioxidant and zinc supplementation appears to differ by genotype. Further study is needed to determine the biological basis for this interaction. TRIAL REGISTRATION NUMBER: NCT00594672, pre-results.


Assuntos
Antioxidantes/administração & dosagem , DNA/genética , Atrofia Geográfica/genética , Polimorfismo de Nucleotídeo Único , Proteínas/genética , Compostos de Zinco/administração & dosagem , Idoso , Alelos , Fator H do Complemento/genética , Fator H do Complemento/metabolismo , Suplementos Nutricionais , Progressão da Doença , Método Duplo-Cego , Feminino , Seguimentos , Genótipo , Atrofia Geográfica/terapia , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Proteínas/metabolismo , Fatores de Risco , Análise de Sobrevida
4.
Antimicrob Agents Chemother ; 60(7): 4023-7, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27090179

RESUMO

Antibiotic susceptibility testing (AST) in Neisseria meningitidis is an important part of the management of invasive meningococcal disease. It defines MICs of antibiotics that are used in treatment and/or prophylaxis and that mainly belong to the beta-lactams. The interpretation of the AST results requires breakpoints to classify the isolates into susceptible, intermediate, or resistant. The resistance to penicillin G is defined by a MIC of >0.25 mg/liter, and that of amoxicillin is defined by a MIC of >1 mg/liter. We provide data that may support revision of resistance breakpoints for beta-lactams in meningococci. We used experimental intraperitoneal infection in 8-week-old transgenic female mice expressing human transferrin and human factor H. Dynamic bioluminescence imaging was performed to follow the infection by bioluminescent meningococcus strains with different MICs. Three hours later, infected mice were treated intramuscularly using several doses of amoxicillin or penicillin G. Signal decreased during infection with a meningococcus strain showing a penicillin G MIC of 0.064 mg/liter at all doses. Signals decreased for the strain with a penicillin G MIC of 0.5 mg/liter only after treatment with the highest doses, corresponding to 250,000 units/kg of penicillin G or 200 mg/kg of amoxicillin, although this decrease was at a lower rate than that of the strain with a MIC of 0.064 mg/liter. The decrease in bioluminescent signals was associated with a decrease in the levels of the proinflammatory cytokine interleukin-6 (IL-6). Our data suggest that a high dose of amoxicillin or penicillin G can reduce growth during infection by isolates showing penicillin G MICs of >0.25 mg/liter and ≤1 mg/liter.


Assuntos
Infecções Meningocócicas/tratamento farmacológico , Neisseria meningitidis/efeitos dos fármacos , Neisseria meningitidis/patogenicidade , Amoxicilina/uso terapêutico , Animais , Fator H do Complemento/genética , Fator H do Complemento/metabolismo , Feminino , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Infecções Meningocócicas/genética , Infecções Meningocócicas/metabolismo , Camundongos , Camundongos Transgênicos , Testes de Sensibilidade Microbiana , Penicilina G/uso terapêutico , Transferrina/genética , Transferrina/metabolismo
5.
PLoS One ; 9(11): e112682, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25393287

RESUMO

UNLABELLED: Age-related macular degeneration (AMD) is the leading cause of blindness in the Western world. AMD is a multifactorial disorder but complement-mediated inflammation at the level of the retina plays a pivotal role. Oral zinc supplementation can reduce the progression of AMD but the precise mechanism of this protective effect is as yet unclear. We investigated whether zinc supplementation directly affects the degree of complement activation in AMD and whether there is a relation between serum complement catabolism during zinc administration and the complement factor H (CFH) gene or the Age-Related Maculopathy susceptibility 2 (ARMS2) genotype. In this open-label clinical study, 72 randomly selected AMD patients in various stages of AMD received a daily supplement of 50 mg zinc sulphate and 1 mg cupric sulphate for three months. Serum complement catabolism-defined as the C3d/C3 ratio-was measured at baseline, throughout the three months of supplementation and after discontinuation of zinc administration. Additionally, downstream inhibition of complement catabolism was evaluated by measurement of anaphylatoxin C5a. Furthermore, we investigated the effect of zinc on complement activation in vitro. AMD patients with high levels of complement catabolism at baseline exhibited a steeper decline in serum complement activation (p<0.001) during the three month zinc supplementation period compared to patients with low complement levels. There was no significant association of change in complement catabolism and CFH and ARMS2 genotype. In vitro zinc sulphate directly inhibits complement catabolism in hemolytic assays and membrane attack complex (MAC) deposition on RPE cells. This study provides evidence that daily administration of 50 mg zinc sulphate can inhibit complement catabolism in AMD patients with increased complement activation. This could explain part of the mechanism by which zinc slows AMD progression. TRIAL REGISTRATION: The Netherlands National Trial Register NTR2605.


Assuntos
Ativação do Complemento/efeitos dos fármacos , Complemento C3/metabolismo , Complemento C3d/metabolismo , Suplementos Nutricionais , Degeneração Macular/dietoterapia , Sulfato de Zinco/administração & dosagem , Idoso , Idoso de 80 Anos ou mais , Células Cultivadas , Complemento C3/imunologia , Complemento C3d/imunologia , Complemento C5a/imunologia , Complemento C5a/metabolismo , Fator B do Complemento/imunologia , Fator B do Complemento/metabolismo , Fator H do Complemento/imunologia , Fator H do Complemento/metabolismo , Sulfato de Cobre/administração & dosagem , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Feminino , Expressão Gênica , Humanos , Degeneração Macular/sangue , Degeneração Macular/imunologia , Degeneração Macular/patologia , Masculino , Mutação , Proteínas/genética , Proteínas/imunologia , Retina/efeitos dos fármacos , Retina/imunologia , Retina/patologia , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/imunologia
6.
PLoS One ; 8(1): e54339, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23365660

RESUMO

BACKGROUND: Mitochondrial dysfunction is associated with the development and progression of age-related macular degeneration (AMD). Recent studies using populations from the United States and Australia have demonstrated that AMD is associated with mitochondrial (mt) DNA haplogroups (as defined by combinations of mtDNA polymorphisms) that represent Northern European Caucasians. The aim of this study was to use the cytoplasmic hybrid (cybrid) model to investigate the molecular and biological functional consequences that occur when comparing the mtDNA H haplogroup (protective for AMD) versus J haplogroup (high risk for AMD). METHODOLOGY/PRINCIPAL FINDINGS: Cybrids were created by introducing mitochondria from individuals with either H or J haplogroups into a human retinal epithelial cell line (ARPE-19) that was devoid of mitochondrial DNA (Rho0). In cybrid lines, all of the cells carry the same nuclear genes but vary in mtDNA content. The J cybrids had significantly lower levels of ATP and reactive oxygen/nitrogen species production, but increased lactate levels and rates of growth. Q-PCR analyses showed J cybrids had decreased expressions for CFH, C3, and EFEMP1 genes, high risk genes for AMD, and higher expression for MYO7A, a gene associated with retinal degeneration in Usher type IB syndrome. The H and J cybrids also have comparatively altered expression of nuclear genes involved in pathways for cell signaling, inflammation, and metabolism. CONCLUSION/SIGNIFICANCE: Our findings demonstrate that mtDNA haplogroup variants mediate not only energy production and cell growth, but also cell signaling for major molecular pathways. These data support the hypothesis that mtDNA variants play important roles in numerous cellular functions and disease processes, including AMD.


Assuntos
DNA Mitocondrial/genética , Células Epiteliais/metabolismo , Expressão Gênica , Células Híbridas/metabolismo , Degeneração Macular/genética , Mitocôndrias/genética , Transdução de Sinais/genética , Trifosfato de Adenosina/biossíntese , Células Cultivadas , Complemento C3/genética , Complemento C3/metabolismo , Fator H do Complemento/genética , Fator H do Complemento/metabolismo , DNA Mitocondrial/metabolismo , Células Epiteliais/citologia , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Haplótipos , Humanos , Células Híbridas/patologia , Ácido Láctico/metabolismo , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Mitocôndrias/metabolismo , Modelos Biológicos , Miosina VIIa , Miosinas/genética , Miosinas/metabolismo , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/metabolismo
7.
Free Radic Biol Med ; 53(6): 1298-307, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22732187

RESUMO

Oxidative damage and inflammation are related to the pathogenesis of age-related macular degeneration (AMD). Epidemiologic studies suggest that insufficient dietary lutein and zeaxanthin intake or lower serum zeaxanthin levels are associated with increased risk for AMD. The objective of this work is to test the protective effects of lutein and zeaxanthin against photooxidative damage to retinal pigment epithelial cells (RPE) and oxidation-induced changes in expression of inflammation-related genes. To mimic lipofuscin-mediated photooxidation in vivo, we used ARPE-19 cells that accumulated A2E, a lipofuscin fluorophore and photosensitizer, as a model system to investigate the effects of lutein and zeaxanthin supplementation. The data show that supplementation with lutein or zeaxanthin in the medium resulted in accumulation of lutein or zeaxanthin in the RPE cells. The concentrations of lutein and zeaxanthin in the cells were 2- to 14-fold of that detected in the medium, indicating that ARPE-19 cells actively take up lutein or zeaxanthin. As compared with untreated cells, exposure of A2E-containing RPE to blue light resulted in a 40-60% decrease in proteasome activity, a 50-80% decrease in expression of CFH and MCP-1, and an~20-fold increase in expression of IL-8. The photooxidation-induced changes in expression of MCP-1, IL-8, and CFH were similar to those caused by chemical inhibition of the proteasome, suggesting that inactivation of the proteasome is involved in the photooxidation-induced alteration in expression of these inflammation-related genes. Incubation of the A2E-containing RPE with lutein or zeaxanthin prior to blue light exposure significantly attenuated the photooxidation-induced inactivation of the proteasome and photooxidation-induced changes in expression of MCP-1, IL-8, and CFH. Together, these data indicate that lutein or zeaxanthin modulates inflammatory responses in cultured RPE in response to photooxidation. Protecting the proteasome from oxidative inactivation appears to be one of the mechanisms by which lutein and zeaxanthin modulate the inflammatory response. Similar mechanisms may explain salutary effects of lutein and zeaxanthin in reducing the risk for AMD.


Assuntos
Mediadores da Inflamação/metabolismo , Luteína/farmacologia , Estresse Oxidativo/efeitos da radiação , Protetores contra Radiação/farmacologia , Raios Ultravioleta , Xantofilas/farmacologia , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Fator H do Complemento/genética , Fator H do Complemento/metabolismo , Meios de Cultura , Suplementos Nutricionais , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/efeitos da radiação , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/efeitos da radiação , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Luteína/metabolismo , Degeneração Macular/tratamento farmacológico , Degeneração Macular/patologia , Oxirredução , Processos Fotoquímicos , Complexo de Endopeptidases do Proteassoma/metabolismo , Protetores contra Radiação/metabolismo , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Xantofilas/metabolismo , Zeaxantinas
8.
Blood ; 119(26): 6307-16, 2012 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-22577173

RESUMO

Paroxysmal nocturnal hemoglobinuria (PNH) is characterized by complement-mediated intravascular hemolysis because of the lack from erythrocyte surface of the complement regulators CD55 and CD59, with subsequent uncontrolled continuous spontaneous activation of the complement alternative pathway (CAP), and at times of the complement classic pathway. Here we investigate in an in vitro model the effect on PNH erythrocytes of a novel therapeutic strategy for membrane-targeted delivery of a CAP inhibitor. TT30 is a 65 kDa recombinant human fusion protein consisting of the iC3b/C3d-binding region of complement receptor 2 (CR2) and the inhibitory domain of the CAP regulator factor H (fH). TT30 completely inhibits in a dose-dependent manner hemolysis of PNH erythrocytes in a modified extended acidified serum assay, and also prevents C3 fragment deposition on surviving PNH erythrocytes. The efficacy of TT30 derives from its direct binding to PNH erythrocytes; if binding to the erythrocytes is disrupted, only partial inhibition of hemolysis is mediated by TT30 in solution, which is similar to that produced by the fH moiety of TT30 alone, or by intact human fH. TT30 is a membrane-targeted selective CAP inhibitor that may prevent both intravascular and C3-mediated extravascular hemolysis of PNH erythrocytes and warrants consideration for the treatment of PNH patients.


Assuntos
Fator H do Complemento/química , Eritrócitos/efeitos dos fármacos , Hemoglobinúria Paroxística/sangue , Hemólise/efeitos dos fármacos , Proteínas de Fusão Oncogênica/farmacologia , Receptores de Complemento 3d/química , Proteínas Recombinantes de Fusão/farmacologia , Estudos de Casos e Controles , Células Cultivadas , Complemento C3/efeitos adversos , Complemento C3/antagonistas & inibidores , Complemento C3/farmacologia , Fator H do Complemento/metabolismo , Fator H do Complemento/farmacologia , Proteínas do Sistema Complemento/efeitos adversos , Proteínas do Sistema Complemento/fisiologia , Citoproteção/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Eritrócitos/fisiologia , Hemoglobinúria Paroxística/patologia , Humanos , Proteínas de Fusão Oncogênica/metabolismo , Ligação Proteica , Receptores de Complemento 3d/metabolismo , Receptores de Complemento 3d/fisiologia , Proteínas Recombinantes de Fusão/metabolismo
9.
Mol Med ; 14(7-8): 443-50, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18496585

RESUMO

We recently discovered that the vascular responsiveness to adrenomedullin (AM), a potent vasoactive peptide, decreased during sepsis and hemorrhage in the rat and was markedly improved by its novel binding protein (AMBP-1). Moreover, AM/AMBP-1 appears to be one of the leading candidates for further development to treat sepsis and hemorrhage. However, the extremely high cost of commercial AMBP-1 limits the development of human AM and AMBP-1 as therapeutic agents. The purpose of this study was to isolate and purify AMBP-1 from normal human serum and test its stability and biological activity under in vitro and in vivo conditions. AMBP-1 was isolated and purified from normal human serum with a yield of about 3.0 mg per 100 mL and purity of >99%. The purified AMBP-1 has a AM-binding capacity similar to that of the commercial AMBP-1. Human AM and human AMBP-1 in combination significantly inhibited lipopolysaccharide-induced tumor necrosis factor (TNF)-alpha and interleukin (IL)-6 production from macrophages. The biological activity of the purified human AMBP-1 was well preserved when stored at 45 degrees C for 5 d in solution or at 100 degrees C for 1 h in powder. Moreover, administration of AM and purified AMBP-1 to hemorrhaged rats attenuated tissue injury and neutrophil accumulation. Purified AMBP-1 in combination with AM also suppressed the hemorrhage-induced rise in serum cytokines TNF-alpha and IL-6. Thus, we have successfully purified biologically active AMBP-1 from human normal serum and demonstrated the stability of purified human AMBP-1. This technique will enable us to further develop human AM/AMBP-1 as a novel treatment for safe and effective therapy of patients with hemorrhagic shock, sepsis, and ischemic injury.


Assuntos
Fator H do Complemento/química , Fator H do Complemento/isolamento & purificação , Animais , Células Cultivadas , Quimiocina CCL2/metabolismo , Fator H do Complemento/metabolismo , Fator H do Complemento/farmacologia , Avaliação Pré-Clínica de Medicamentos , Hemorragia/complicações , Hemorragia/patologia , Hemorragia/prevenção & controle , Humanos , Interleucina-6/metabolismo , Isquemia/patologia , Lipopolissacarídeos/farmacologia , Hepatopatias/etiologia , Hepatopatias/patologia , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley , Soro/química , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA