Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Am Soc Nephrol ; 33(6): 1137-1153, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35545301

RESUMO

BACKGROUND: C3 glomerulopathy (C3G) is a heterogeneous group of chronic renal diseases characterized predominantly by glomerular C3 deposition and complement dysregulation. Mutations in factor H-related (FHR) proteins resulting in duplicated dimerization domains are prototypical of C3G, although the underlying pathogenic mechanism is unclear. METHODS: Using in vitro and in vivo assays, we performed extensive characterization of an FHR-1 mutant with a duplicated dimerization domain. To assess the FHR-1 mutant's association with disease susceptibility and renal prognosis, we also analyzed CFHR1 copy number variations and FHR-1 plasma levels in two Spanish C3G cohorts and in a control population. RESULTS: Duplication of the dimerization domain conferred FHR-1 with an increased capacity to interact with C3-opsonized surfaces, which resulted in an excessive activation of the alternative pathway. This activation does not involve C3b binding competition with factor H. These findings support a scenario in which mutant FHR-1 binds to C3-activated fragments and recruits native C3 and C3b; this leads to formation of alternative pathway C3 convertases, which increases deposition of C3b molecules, overcoming FH regulation. This suggests that a balanced FHR-1/FH ratio is crucial to control complement amplification on opsonized surfaces. Consistent with this conceptual framework, we show that the genetic deficiency of FHR-1 or decreased FHR-1 in plasma confers protection against developing C3G and associates with better renal outcome. CONCLUSIONS: Our findings explain how FHR-1 mutants with duplicated dimerization domains result in predisposition to C3G. They also provide a pathogenic mechanism that may be shared by other diseases, such as IgA nephropathy or age-related macular degeneration, and identify FHR-1 as a potential novel therapeutic target in C3G.


Assuntos
Proteínas Inativadoras do Complemento C3b , Glomerulonefrite por IGA , Proteínas Sanguíneas , Complemento C3/genética , Complemento C3/metabolismo , Proteínas Inativadoras do Complemento C3b/genética , Proteínas Inativadoras do Complemento C3b/metabolismo , Fator H do Complemento/genética , Variações do Número de Cópias de DNA , Suscetibilidade a Doenças , Glomerulonefrite por IGA/genética , Glomerulonefrite por IGA/metabolismo , Humanos , Prognóstico
2.
Cancers (Basel) ; 14(1)2021 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-35008241

RESUMO

Fibropolycystic liver disease is characterized by hyperproliferation of the biliary epithelium and the formation of multiple dilated cysts, a process associated with unfolded protein response (UPR). In the present study, we aimed to understand the mechanisms of cyst formation and UPR activation in hepatocytic c-Jun N-terminal kinase 1/2 (Jnk1/2) knockout mice. Floxed JNK1/2 (Jnkf/f) and Jnk∆hepa animals were sacrificed at different time points during progression of liver disease. Histological examination of specimens evidenced the presence of collagen fiber deposition, increased α-smooth muscle actin (αSMA), infiltration of CD45, CD11b and F4/80 cells and proinflammatory cytokines (Tnf, Tgfß1) and liver injury (e.g., ALT, apoptosis and Ki67-positive cells) in Jnk∆hepa compared with Jnkf/f livers from 32 weeks of age. This was associated with activation of effectors of the UPR, including BiP/GRP78, CHOP and spliced XBP1. Tunicamycin (TM) challenge strongly induced ER stress and fibrosis in Jnk∆hepa animals compared with Jnkf/f littermates. Finally, thioacetamide (TAA) administration to Jnk∆hepa mice induced UPR activation, peribiliary fibrosis, liver injury and markers of biliary proliferation and cholangiocarcinoma (CCA). Orthoallografts of DEN/CCl4-treated Jnk∆hepa liver tissue triggered malignant CCA. Altogether, these results suggest that activation of the UPR in conjunction with fibrogenesis might trigger hepatic cystogenesis and early stages of CCA.

3.
Mol Immunol ; 114: 123-132, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31351413

RESUMO

IgA nephropathy (IgAN) is common and often progresses to end stage renal disease. IgAN encompasses a wide range of histology and clinical features. IgAN pathogenesis is incompletely understood; the current multi-hit hypothesis of IgAN pathogenesis does not explain the range of glomerular inflammation and renal injury associated with mesangial IgA deposition. Although associations between IgAN and glomerular and circulating markers of complement activation are established, the mechanism of complement activation and contribution to glomerular inflammation and injury are not defined. Recent identification of specific complement pathways and proteins in severe IgAN cases had advanced our understanding of complement in IgAN pathogenesis. In particular, a growing body of evidence implicates the complement factor H related proteins 1 and 5 and lectin pathway as pathogenic in a subset of patients with severe disease. These data suggest complement deregulation and activity may be dominant drivers of renal injury in IgAN. Thereby, markers of complement activation may identify IgAN patients likely to progress to significant renal impairment and complement inhibition may emerge as an effective method of preventing and reducing glomerular injury in IgAN.


Assuntos
Ativação do Complemento/imunologia , Proteínas do Sistema Complemento/imunologia , Glomerulonefrite por IGA/imunologia , Imunoglobulina A/imunologia , Animais , Humanos
4.
Semin Immunopathol ; 40(1): 3-14, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28808775

RESUMO

During the last decade, the complement field has experienced outstanding advancements in the mechanistic understanding of how complement activators are recognized, what C3 activation means, how protein complexes like the C3 convertases and the membrane attack complex are assembled, and how positive and negative complement regulators perform their function. All of this has been made possible mostly because of the contributions of structural biology to the study of the complement components. The wealth of novel structural data has frequently provided support to previously held knowledge, but often has added alternative and unexpected insights into complement function. Here, we will review some of these findings focusing in the alternative and terminal complement pathways.


Assuntos
Proteínas do Sistema Complemento/química , Proteínas do Sistema Complemento/fisiologia , Animais , Proteínas de Transporte , Ativação do Complemento/imunologia , Convertases de Complemento C3-C5/química , Convertases de Complemento C3-C5/imunologia , Convertases de Complemento C3-C5/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Ligação Proteica , Conformação Proteica , Transdução de Sinais , Relação Estrutura-Atividade , Especificidade por Substrato
5.
J Am Soc Nephrol ; 29(1): 240-249, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28993505

RESUMO

Atypical hemolytic uremic syndrome (aHUS), a rare form of thrombotic microangiopathy caused by complement pathogenic variants, mainly affects the kidney microvasculature. A retrospective genetic analysis in our aHUS cohort (n=513) using multiple ligation probe amplification uncovered nine unrelated patients carrying a genetic abnormality in the complement factor H related 1 gene (CFHR1) that originates by recurrent gene conversion events between the CFH and CFHR1 genes. The novel CFHR1 mutants encode an FHR-1 protein with two amino acid substitutions, L290S and A296V, converting the FHR-1 C terminus into that of factor H (FH). Next-generation massive-parallel DNA sequencing (NGS) analysis did not detect these genetic abnormalities. In addition to the CFHR1 mutant, six patients carried the previously uncharacterized CFH-411T variant. In functional analyses, the mutant FHR-1 protein strongly competed the binding of FH to cell surfaces, impairing complement regulation, whereas the CFH-411T polymorphism lacked functional consequences. Carriers of the CFHR1 mutation presented with severe aHUS during adulthood; 57% of affected women in this cohort presented during the postpartum period. Analyses in patients and unaffected carriers showed that FH plasma levels determined by the nonmutated chromosome modulate disease penetrance. Crucially, in the activated endothelial (HMEC-1) cell assay, reduced FH plasma levels produced by the nonmutated chromosome correlated inversely with impairment of complement regulation, measured as C5b-9 deposition. Our data advance understanding of the genetic complexities underlying aHUS, illustrate the importance of performing functional analysis, and support the use of complementary assays to disclose genetic abnormalities not revealed by current NGS analysis.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/genética , Proteínas Sanguíneas/genética , Proteínas Inativadoras do Complemento C3b/genética , Fator H do Complemento/genética , Conversão Gênica , Adulto , Ligação Competitiva , Linhagem Celular , Fator H do Complemento/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Linhagem , Penetrância , Estudos Retrospectivos , Análise de Sequência de DNA/métodos , Adulto Jovem
6.
Kidney Int ; 93(2): 470-481, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28941939

RESUMO

Genetic analyses in atypical hemolytic uremic syndrome (aHUS) and C3-glomerulopathy (C3G) patients have provided an excellent understanding of the genetic component of the disease and informed genotype-phenotype correlations supporting an individualized approach to patient management and treatment. In this context, a correct categorization of the disease-associated gene variants is critical to avoid detrimental consequences for patients and their relatives. Here we describe a comprehensive procedure to measure levels and functional activity of complement regulator factor H (FH) encoded by CFH, the commonest genetic factor associated with aHUS and C3G, and present the results of the analysis of 28 uncharacterized, disease-associated, FH variants. Sixteen variants were not expressed in plasma and eight had significantly reduced functional activities that impact on complement regulation. In total, 24 of 28 CFH variants were unambiguously categorized as pathogenic and the nature of the pathogenicity fully documented for each. The data also reinforce the genotype-phenotype correlations that associate specific FH functional alterations with either aHUS or C3G and illustrate important drawbacks of the prediction algorithms dealing with variants located in FH functional regions. We also report that the novel aHUS-associated M823T variant is functionally impaired. This was unexpected and uncovered the important contribution of regions outside the N-terminal and C-terminal functional domains to FH regulatory activities on surfaces. Thus, our work significantly advances knowledge towards a complete functional understanding of the CFH genetic variability and highlights the importance of functional analysis of the disease-associated CFH variants.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/genética , Ativação do Complemento/genética , Variação Genética , Glomerulonefrite/genética , Adolescente , Adulto , Síndrome Hemolítico-Urêmica Atípica/sangue , Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Síndrome Hemolítico-Urêmica Atípica/imunologia , Criança , Pré-Escolar , Complemento C3/imunologia , Fator H do Complemento/genética , Fator H do Complemento/imunologia , Fator H do Complemento/metabolismo , Feminino , Estudos de Associação Genética , Predisposição Genética para Doença , Glomerulonefrite/sangue , Glomerulonefrite/diagnóstico , Glomerulonefrite/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Domínios Proteicos , Sistema de Registros , Fatores de Risco , Espanha , Relação Estrutura-Atividade , Adulto Jovem
7.
Kidney Int ; 92(4): 953-963, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28637589

RESUMO

IgA nephropathy (IgAN), a frequent cause of chronic kidney disease worldwide, is characterized by mesangial deposition of galactose-deficient IgA1-containing immune complexes. Complement involvement in IgAN pathogenesis is suggested by the glomerular deposition of complement components and the strong protection from IgAN development conferred by the deletion of the CFHR3 and CFHR1 genes (ΔCFHR3-CFHR1). Here we searched for correlations between clinical progression and levels of factor H (FH) and FH-related protein 1 (FHR-1) using well-characterized patient cohorts consisting of 112 patients with IgAN, 46 with non-complement-related autosomal dominant polycystic kidney disease (ADPKD), and 76 control individuals. Patients with either IgAN or ADPKD presented normal FH but abnormally elevated FHR-1 levels and FHR-1/FH ratios compared to control individuals. Highest FHR-1 levels and FHR-1/FH ratios are found in patients with IgAN with disease progression and in patients with ADPKD who have reached chronic kidney disease, suggesting that renal function impairment elevates the FHR-1/FH ratio, which may increase FHR-1/FH competition for activated C3 fragments. Interestingly, ΔCFHR3-CFHR1 homozygotes are protected from IgAN, but not from ADPKD, and we found five IgAN patients with low FH carrying CFH or CFI pathogenic variants. These data support a decreased FH activity in IgAN due to increased FHR-1/FH competition or pathogenic CFH variants. They also suggest that alternative pathway complement activation in patients with IgAN, initially triggered by galactose-deficient IgA1-containing immune complexes, may exacerbate in a vicious circle as renal function deterioration increase FHR-1 levels. Thus, a role of FHR-1 in IgAN pathogenesis is to compete with complement regulation by FH.


Assuntos
Proteínas Inativadoras do Complemento C3b/análise , Via Alternativa do Complemento/genética , Glomerulonefrite por IGA/sangue , Rim Policístico Autossômico Dominante/sangue , Insuficiência Renal Crônica/sangue , Adulto , Proteínas Sanguíneas/genética , Estudos de Coortes , Proteínas Inativadoras do Complemento C3b/genética , Fator H do Complemento/análise , Fator H do Complemento/genética , Progressão da Doença , Feminino , Taxa de Filtração Glomerular , Glomerulonefrite por IGA/genética , Humanos , Masculino , Pessoa de Meia-Idade , Rim Policístico Autossômico Dominante/genética , Insuficiência Renal Crônica/genética , Adulto Jovem
8.
J Immunol ; 199(3): 1021-1040, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28652401

RESUMO

von Willebrand factor (VWF), a multimeric protein with a central role in hemostasis, has been shown to interact with complement components. However, results are contrasting and inconclusive. By studying 20 patients with congenital thrombotic thrombocytopenic purpura (cTTP) who cannot cleave VWF multimers because of genetic ADAMTS13 deficiency, we investigated the mechanism through which VWF modulates complement and its pathophysiological implications for human diseases. Using assays of ex vivo serum-induced C3 and C5b-9 deposits on endothelial cells, we documented that in cTTP, complement is activated via the alternative pathway (AP) on the cell surface. This abnormality was corrected by restoring ADAMTS13 activity in cTTP serum, which prevented VWF multimer accumulation on endothelial cells, or by an anti-VWF Ab. In mechanistic studies we found that VWF interacts with C3b through its three type A domains and initiates AP activation, although assembly of active C5 convertase and formation of the terminal complement products C5a and C5b-9 occur only on the VWF-A2 domain. Finally, we documented that in the condition of ADAMTS13 deficiency, VWF-mediated formation of terminal complement products, particularly C5a, alters the endothelial antithrombogenic properties and induces microvascular thrombosis in a perfusion system. Altogether, the results demonstrated that VWF provides a platform for the activation of the AP of complement, which profoundly alters the phenotype of microvascular endothelial cells. These findings link hemostasis-thrombosis with the AP of complement and open new therapeutic perspectives in cTTP and in general in thrombotic and inflammatory disorders associated with endothelium perturbation, VWF release, and complement activation.


Assuntos
Complemento C3b/metabolismo , Via Alternativa do Complemento , Células Endoteliais/imunologia , Microvasos/patologia , Trombose/fisiopatologia , Fator de von Willebrand/metabolismo , Proteína ADAMTS13/sangue , Proteína ADAMTS13/deficiência , Proteína ADAMTS13/imunologia , Proteína ADAMTS13/metabolismo , Adolescente , Adulto , Criança , Pré-Escolar , Convertases de Complemento C3-C5/metabolismo , Complemento C3b/imunologia , Complemento C5a/imunologia , Complemento C5a/metabolismo , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Complexo de Ataque à Membrana do Sistema Complemento/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Humanos , Recém-Nascido , Masculino , Microvasos/imunologia , Púrpura Trombocitopênica Trombótica/congênito , Púrpura Trombocitopênica Trombótica/imunologia , Púrpura Trombocitopênica Trombótica/fisiopatologia , Trombose/imunologia , Adulto Jovem , Fator de von Willebrand/imunologia
9.
J Immunol ; 199(1): 292-303, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28533443

RESUMO

Factor H-related protein (FHR) 1 is one of the five human FHRs that share sequence and structural homology with the alternative pathway complement inhibitor FH. Genetic studies on disease associations and functional analyses indicate that FHR-1 enhances complement activation by competitive inhibition of FH binding to some surfaces and immune proteins. We have recently shown that FHR-1 binds to pentraxin 3. In this study, our aim was to investigate whether FHR-1 binds to another pentraxin, C-reactive protein (CRP), analyze the functional relevance of this interaction, and study the role of FHR-1 in complement activation and regulation. FHR-1 did not bind to native, pentameric CRP, but it bound strongly to monomeric CRP via its C-terminal domains. FHR-1 at high concentration competed with FH for CRP binding, indicating possible complement deregulation also on this ligand. FHR-1 did not inhibit regulation of solid-phase C3 convertase by FH and did not inhibit terminal complement complex formation induced by zymosan. On the contrary, by binding C3b, FHR-1 allowed C3 convertase formation and thereby enhanced complement activation. FHR-1/CRP interactions increased complement activation via the classical and alternative pathways on surfaces such as the extracellular matrix and necrotic cells. Altogether, these results identify CRP as a ligand for FHR-1 and suggest that FHR-1 enhances, rather than inhibits, complement activation, which may explain the protective effect of FHR-1 deficiency in age-related macular degeneration.


Assuntos
Proteína C-Reativa/imunologia , Proteína C-Reativa/metabolismo , Ativação do Complemento , Proteínas Inativadoras do Complemento C3b/imunologia , Proteínas Inativadoras do Complemento C3b/metabolismo , Sítios de Ligação , Proteína C-Reativa/química , Proteína C-Reativa/farmacologia , Convertases de Complemento C3-C5 , Complemento C3b/imunologia , Complemento C3b/farmacologia , Proteínas Inativadoras do Complemento C3b/farmacologia , Fator H do Complemento , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/imunologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/imunologia , Humanos , Ligantes , Degeneração Macular/imunologia , Ligação Proteica , Componente Amiloide P Sérico/imunologia , Componente Amiloide P Sérico/metabolismo
10.
Eur J Immunol ; 47(3): 504-515, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28083930

RESUMO

C3 is the central component of the complement system. Upon activation, C3 sequentially generates various proteolytic fragments, C3a, C3b, iC3b, C3dg, each of them exposing novel surfaces, which are sites of interaction with other proteins. C3 and its fragments are therapeutic targets and markers of complement activation. We report the structural and functional characterization of four monoclonal antibodies (mAbs) generated by immunizing C3-deficient mice with a mixture of human C3b, iC3b and C3dg fragments, and discuss their potential applications. This collection includes three mAbs interacting with native C3 and inhibiting AP complement activation; two of them by blocking the cleavage of C3 by the AP C3-converase and one by impeding formation of the AP C3-convertase. The interaction sites of these mAbs in the target molecules were determined by resolving the structures of Fab fragments bound to C3b and/or iC3b using electron microscopy. A fourth mAb specifically recognizes the iC3b, C3dg, and C3d fragments. It binds to an evolutionary-conserved neoepitope generated after C3b cleavage by FI, detecting iC3b/C3dg deposition over opsonized surfaces by flow cytometry and immunohistochemistry in human and other species. Because well-characterized anti-complement mAbs are uncommon, the mAbs reported here may offer interesting therapeutic and diagnostic opportunities.


Assuntos
Anticorpos Monoclonais/metabolismo , Complexo Antígeno-Anticorpo/metabolismo , Convertases de Complemento C3-C5/metabolismo , Complemento C3/metabolismo , Via Alternativa do Complemento , Animais , Anticorpos Monoclonais/genética , Complemento C3/genética , Complemento C3/imunologia , Engenharia Genética , Técnica de Placa Hemolítica , Humanos , Hibridomas , Fragmentos Fab das Imunoglobulinas/genética , Camundongos , Camundongos Knockout , Ligação Proteica , Conformação Proteica
11.
Mol Immunol ; 77: 89-96, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27490940

RESUMO

C3 glomerulopathy (C3G) is an ultra-rare complement-mediated renal disease characterized histologically by the predominance of C3 deposition within in the glomerulus. Familial cases of C3G are extremely uncommon and offer unique insight into the genetic drivers of complement dysregulation. In this report, we describe a patient who presented with C3G. Because a relative carried the same diagnosis, we sought an underlying genetic commonality to explain the phenotype. As part of a comprehension genetic screen, we completed multiplex ligation-dependent probe amplification across the complement factor H related region and identified amplification alterations consistent with a genomic rearrangement. Using comparative genomic hybridization, we narrowed and then cloned the rearrangement breakpoints thereby defining a novel fusion gene that is translated into a serum protein comprised of factor H related-5 (short consensus repeats 1 and 2) and factor H-related-2 (short consensus repeats 1-4). These data highlight the role of factor H related proteins in the control of complement activity and illustrate how perturbation of that control leads to C3G.


Assuntos
Proteínas Inativadoras do Complemento C3b/genética , Proteínas do Sistema Complemento/genética , Glomerulonefrite Membranoproliferativa/genética , Adulto , Western Blotting , Hibridização Genômica Comparativa , Feminino , Humanos , Masculino , Reação em Cadeia da Polimerase Multiplex , Linhagem
12.
J Am Soc Nephrol ; 27(5): 1305-11, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26376859

RESUMO

The complement factor H (FH) mutation R1210C, which was described in association with atypical hemolytic uremic syndrome (aHUS), also confers high risk of age-related macular degeneration (AMD) and associates with C3 glomerulopathy (C3G). To reveal the molecular basis of these associations and to provide insight into what determines the disease phenotype in FH-R1210C carriers, we identified FH-R1210C carriers in our aHUS, C3G, and AMD cohorts. Disease status, determined in patients and relatives, revealed an absence of AMD phenotypes in the aHUS cohort and, vice versa, a lack of renal disease in the AMD cohort. These findings were consistent with differences in the R1210C-independent overall risk for aHUS and AMD between mutation carriers developing one pathology or the other. R1210C is an unusual mutation that generates covalent complexes between FH and HSA. Using purified FH proteins and surface plasmon resonance analyses, we demonstrated that formation of these FH-HSA complexes impairs accessibility to all FH functional domains. These data suggest that R1210C is a unique C-terminal FH mutation that behaves as a partial FH deficiency, predisposing individuals to diverse pathologies with distinct underlying pathogenic mechanisms; the final disease outcome is then determined by R1210C-independent genetic risk factors.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/genética , Complemento C3 , Nefropatias/genética , Glomérulos Renais , Degeneração Macular/genética , Mutação , Fator H do Complemento/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem
13.
Trends Immunol ; 36(6): 374-84, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25979655

RESUMO

Complement factor H-related proteins (FHRs) are strongly associated with different diseases involving complement dysregulation, which suggests a major role for these proteins regulating complement activation. Because FHRs are evolutionarily and structurally related to complement inhibitor factor H (FH), the initial assumption was that the FHRs are also negative complement regulators. Whereas weak complement inhibiting activities were originally reported for these molecules, recent developments indicate that FHRs may enhance complement activation, with important implications for the role of these proteins in health and disease. We review these findings here, and propose that FHRs represent a complex set of surface recognition molecules that, by competing with FH, provide improved discrimination of self and non-self surfaces and play a central role in determining appropriate activation of the complement pathway.


Assuntos
Apolipoproteínas/imunologia , Proteínas Sanguíneas/imunologia , Ativação do Complemento/imunologia , Proteínas Inativadoras do Complemento C3b/imunologia , Proteínas do Sistema Complemento/imunologia , Apolipoproteínas/genética , Proteínas Sanguíneas/genética , Proteínas Inativadoras do Complemento C3b/genética , Fator H do Complemento/genética , Fator H do Complemento/imunologia , Proteínas do Sistema Complemento/genética , Predisposição Genética para Doença/genética , Humanos , Modelos Imunológicos
14.
Mol Immunol ; 66(2): 263-73, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25879158

RESUMO

Atypical hemolytic uremic syndrome (aHUS) associates with complement dysregulation caused by mutations and polymorphisms in complement activators and regulators. However, the reasons why some mutations in complement proteins predispose to aHUS are poorly understood. Here, we have investigated the functional consequences of three aHUS-associated mutations in C3, R592W, R161W and I1157T. First, we provide evidence that penetrance and disease severity for these mutations is modulated by inheritance of documented "risk" haplotypes as has been observed with mutations in other complement genes. Next, we show that all three mutations markedly reduce the efficiency of factor I-mediated C3b cleavage when catalyzed by membrane cofactor protein (MCP), but not when catalyzed by factor H. Biacore analysis showed that each mutant C3b bound sMCP (recombinant soluble MCP; CD46) at reduced affinity, providing a molecular basis for its reduced cofactor activity. Lastly, we show by electron microscopy structural analysis a displacement of the TED domain from the MG ring in C3b in two of the C3 mutants that explains these defects in regulation. As a whole our data suggest that aHUS-associated mutations in C3 selectively affect regulation of complement on surfaces and provide a structural framework to predict the functional consequences of the C3 genetic variants found in patients.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/genética , Complemento C3/genética , Predisposição Genética para Doença , Proteína Cofatora de Membrana/genética , Mutação , Adolescente , Adulto , Síndrome Hemolítico-Urêmica Atípica/imunologia , Síndrome Hemolítico-Urêmica Atípica/patologia , Sequência de Bases , Sítios de Ligação , Criança , Complemento C3/química , Complemento C3/imunologia , Fator H do Complemento/genética , Fator H do Complemento/imunologia , Feminino , Fibrinogênio/genética , Fibrinogênio/imunologia , Expressão Gênica , Haplótipos , Humanos , Masculino , Proteína Cofatora de Membrana/imunologia , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Estrutura Terciária de Proteína , Proteólise , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Índice de Gravidade de Doença
15.
J Am Soc Nephrol ; 26(1): 209-19, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24904082

RESUMO

Genomic aberrations affecting the genes encoding factor H (FH) and the five FH-related proteins (FHRs) have been described in patients with atypical hemolytic uremic syndrome (aHUS), a rare condition characterized by microangiopathic hemolytic anemia, thrombocytopenia, and ARF. These genomic rearrangements occur through nonallelic homologous recombinations caused by the presence of repeated homologous sequences in CFH and CFHR1-R5 genes. In this study, we found heterozygous genomic rearrangements among CFH and CFHR genes in 4.5% of patients with aHUS. CFH/CFHR rearrangements were associated with poor clinical prognosis and high risk of post-transplant recurrence. Five patients carried known CFH/CFHR1 genes, but we found a duplication leading to a novel CFHR1/CFH hybrid gene in a family with two affected subjects. The resulting fusion protein contains the first four short consensus repeats of FHR1 and the terminal short consensus repeat 20 of FH. In an FH-dependent hemolysis assay, we showed that the hybrid protein causes sheep erythrocyte lysis. Functional analysis of the FHR1 fraction purified from serum of heterozygous carriers of the CFHR1/CFH hybrid gene indicated that the FHR1/FH hybrid protein acts as a competitive antagonist of FH. Furthermore, sera from carriers of the hybrid CFHR1/CFH gene induced more C5b-9 deposition on endothelial cells than control serum. These results suggest that this novel genomic hybrid mediates disease pathogenesis through dysregulation of complement at the endothelial cell surface. We recommend that genetic screening of aHUS includes analysis of CFH and CFHR rearrangements, particularly before a kidney transplant.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/genética , Proteínas Inativadoras do Complemento C3b/genética , Fator H do Complemento/genética , Proteínas Mutantes Quiméricas/genética , Animais , Ligação Competitiva , Membrana Celular/metabolismo , Proteínas do Sistema Complemento/genética , Eritrócitos/citologia , Éxons , Feminino , Hemólise , Heterozigoto , Humanos , Nefropatias/genética , Transplante de Rim , Masculino , Linhagem , Ovinos
16.
J Immunol ; 193(11): 5567-75, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25355917

RESUMO

The alternative pathway (AP) is critical for the efficient activation of complement regardless of the trigger. It is also a major player in pathogenesis, as illustrated by the long list of diseases in which AP activation contributes to pathology. Its relevance to human disease is further emphasized by the high prevalence of pathogenic inherited defects and acquired autoantibodies disrupting components and regulators of the AP C3-convertase. Because pharmacological downmodulation of the AP emerges as a broad-spectrum treatment alternative, there is a powerful interest in developing new molecules to block formation and/or activity of the AP C3-convertase. In this paper, we describe the generation of a novel mAb targeting human factor B (FB). mAb FB48.4.2, recognizing with high affinity an evolutionary-conserved epitope in the Ba fragment of FB, very efficiently inhibited formation of the AP C3-proconvertase by blocking the interaction between FB and C3b. In vitro assays using rabbit and sheep erythrocytes demonstrated that FB28.4.2 was a potent AP inhibitor that blocked complement-mediated hemolysis in several species. Using ex vivo models of disease we demonstrated that FB28.4.2 protected paroxysmal nocturnal hemoglobinuria erythrocytes from complement-mediated hemolysis and inhibited both C3 fragment and C5b-9 deposition on ADP-activated HMEC-1 cells, an experimental model for atypical hemolytic uremic syndrome. Moreover, i.v. injection of FB28.4.2 in rats blocked complement activation in rat serum and prevented the passive induction of experimental autoimmune Myasthenia gravis. As a whole, these data demonstrate the potential value of FB28.4.2 for the treatment of disorders associated with AP complement dysregulation in man and animal models.


Assuntos
Anticorpos Bloqueadores/imunologia , Anticorpos Monoclonais/imunologia , Síndrome Hemolítico-Urêmica Atípica/terapia , Complemento C3b/metabolismo , Fator B do Complemento/metabolismo , Hemoglobinúria Paroxística/terapia , Miastenia Gravis Autoimune Experimental/terapia , Animais , Anticorpos Bloqueadores/isolamento & purificação , Anticorpos Monoclonais/isolamento & purificação , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Síndrome Hemolítico-Urêmica Atípica/imunologia , Bovinos , Linhagem Celular , C3 Convertase da Via Alternativa do Complemento/metabolismo , Fator B do Complemento/genética , Fator B do Complemento/imunologia , Via Alternativa do Complemento/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Hemoglobinúria Paroxística/imunologia , Humanos , Camundongos , Camundongos Knockout , Miastenia Gravis Autoimune Experimental/imunologia , Ligação Proteica/efeitos dos fármacos , Coelhos , Ratos , Ratos Endogâmicos Lew , Ovinos
17.
Semin Thromb Hemost ; 40(4): 422-30, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24799305

RESUMO

Hemolytic uremic syndrome (HUS) is a rare, life-threatening disease characterized by thrombocytopenia, microangiopathic hemolytic anemia, and acute renal failure. The atypical form of HUS (aHUS), representing 5 to 10% of cases, lacks the association with infection by Shiga toxin producing Escherichia coli strains that characterizes the commonest clinical presentation of HUS. In the majority of aHUS cases, the disease results from the complement-mediated damage to the microvascular endothelium because of inherited defects in complement genes or autoantibodies against complement regulatory proteins. Incomplete penetrance of aHUS in carriers of mutations is common to all aHUS-associated complement genes and it is now established that the overall genetic predisposition to aHUS of an individual results from the combination of different inherited factors. Moreover, the patient's genotype influences the clinical evolution, the response to plasma therapies, and the recurrence after transplantation. Here, we describe the genetic component of aHUS, the lessons that we have learned from the functional characterization of the aHUS-associated mutations, and the benefits of a comprehensive genetic analysis of the patients.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/genética , Proteínas do Sistema Complemento/genética , Mutação , Autoanticorpos/imunologia , Complemento C3/genética , Fator B do Complemento/genética , Fator H do Complemento/genética , Diacilglicerol Quinase/genética , Estudos de Associação Genética , Predisposição Genética para Doença , Heterozigoto , Humanos , Íntrons , Splicing de RNA , Trombomodulina/genética , Microangiopatias Trombóticas/genética
18.
Proc Natl Acad Sci U S A ; 110(33): 13504-9, 2013 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-23901101

RESUMO

Complement is an essential component of innate immunity. Its activation results in the assembly of unstable protease complexes, denominated C3/C5 convertases, leading to inflammation and lysis. Regulatory proteins inactivate C3/C5 convertases on host surfaces to avoid collateral tissue damage. On pathogen surfaces, properdin stabilizes C3/C5 convertases to efficiently fight infection. How properdin performs this function is, however, unclear. Using electron microscopy we show that the N- and C-terminal ends of adjacent monomers in properdin oligomers conform a curly vertex that holds together the AP convertase, interacting with both the C345C and vWA domains of C3b and Bb, respectively. Properdin also promotes a large displacement of the TED (thioester-containing domain) and CUB (complement protein subcomponents C1r/C1s, urchin embryonic growth factor and bone morphogenetic protein 1) domains of C3b, which likely impairs C3-convertase inactivation by regulatory proteins. The combined effect of molecular cross-linking and structural reorganization increases stability of the C3 convertase and facilitates recruitment of fluid-phase C3 convertase to the cell surfaces. Our model explains how properdin mediates the assembly of stabilized C3/C5-convertase clusters, which helps to localize complement amplification to pathogen surfaces.


Assuntos
Convertases de Complemento C3-C5/imunologia , Complemento C3b/imunologia , Fator B do Complemento/imunologia , Via Alternativa do Complemento/imunologia , Imunidade Inata/imunologia , Modelos Imunológicos , Properdina/farmacologia , Animais , Western Blotting , Células CHO , Cricetinae , Cricetulus , Microscopia Eletrônica de Transmissão , Properdina/imunologia
19.
J Clin Invest ; 123(6): 2434-46, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23728178

RESUMO

C3 glomerulopathies (C3G) are a group of severe renal diseases with distinct patterns of glomerular inflammation and C3 deposition caused by complement dysregulation. Here we report the identification of a familial C3G-associated genomic mutation in the gene complement factor H­related 1 (CFHR1), which encodes FHR1. The mutation resulted in the duplication of the N-terminal short consensus repeats (SCRs) that are conserved in FHR2 and FHR5. We determined that native FHR1, FHR2, and FHR5 circulate in plasma as homo- and hetero-oligomeric complexes, the formation of which is likely mediated by the conserved N-terminal domain. In mutant FHR1, duplication of the N-terminal domain resulted in the formation of unusually large multimeric FHR complexes that exhibited increased avidity for the FHR1 ligands C3b, iC3b, and C3dg and enhanced competition with complement factor H (FH) in surface plasmon resonance (SPR) studies and hemolytic assays. These data revealed that FHR1, FHR2, and FHR5 organize a combinatorial repertoire of oligomeric complexes and demonstrated that changes in FHR oligomerization influence the regulation of complement activation. In summary, our identification and characterization of a unique CFHR1 mutation provides insights into the biology of the FHRs and contributes to our understanding of the pathogenic mechanisms underlying C3G.


Assuntos
Complemento C3/metabolismo , Proteínas Inativadoras do Complemento C3b/genética , Nefropatias/genética , Criança , Complemento C3/química , Proteínas Inativadoras do Complemento C3b/química , Proteínas Inativadoras do Complemento C3b/metabolismo , Proteínas do Sistema Complemento/isolamento & purificação , Proteínas do Sistema Complemento/metabolismo , Feminino , Duplicação Gênica , Hemólise , Humanos , Proteínas Imobilizadas/química , Proteínas Imobilizadas/metabolismo , Rim/patologia , Masculino , Pessoa de Meia-Idade , Mutagênese Insercional , Linhagem , Ligação Proteica , Multimerização Proteica , Estrutura Quaternária de Proteína , Análise de Sequência de DNA
20.
Immunobiology ; 217(11): 1034-46, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22964229

RESUMO

During the last decade, numerous studies have associated genetic variations in complement components and regulators with a number of chronic and infectious diseases. The functional characterization of these complement protein variants, in addition to recent structural advances in understanding of the assembly, activation and regulation of the AP C3 convertase, have provided important insights into the pathogenic mechanisms involved in some of these complement related disorders. This knowledge has identified potential targets for complement inhibitory therapies which are demonstrating efficacy and generating considerable expectation in changing the natural history of these diseases. Comprehensive understanding of the genetic and non-genetic risk factors contributing to these disorders will also result in targeting of the right patient groups in a stratified medicine approach through better diagnostics and individually tailored treatments, thereby improving management of patients.


Assuntos
Ativação do Complemento/genética , Proteínas do Sistema Complemento/genética , Proteínas do Sistema Complemento/metabolismo , Inflamação/genética , Convertases de Complemento C3-C5/genética , Convertases de Complemento C3-C5/metabolismo , Proteínas Inativadoras do Complemento/genética , Variação Genética , Glomerulonefrite Membranoproliferativa/genética , Glomerulonefrite Membranoproliferativa/imunologia , Síndrome Hemolítico-Urêmica/genética , Síndrome Hemolítico-Urêmica/imunologia , Humanos , Inflamação/imunologia , Degeneração Macular/genética , Degeneração Macular/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...