Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Exp Dermatol ; 32(5): 632-640, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36704908

RESUMO

Bullous pemphigoid (BP) is characterized by deposition of immunoglobulins and complement along the epidermal basement membrane (BM). In humans, there is a lack of functional studies targeting the complement system (CS). This study investigates activation of all complement pathways in BP skin biopsies. Moreover, pharmacological inhibition at different levels of the CS was investigated using anti-complement compounds in a complement fixation BP assay. In this retrospective study, 21 frozen biopsies from BP patients were stained by direct immunofluorescence for C1q, MBL, ficolin-2, C4d, properdin, C3c and C5b-9. Sera from 10 patients were analysed in a complement fixation assay in the presence of C1 inhibitor, anti-factor B monoclonal antibody (mAb), anti-C3 mAb and anti-C5 mAb and compared with dexamethasone. The two readouts were the quantity of complement deposited along the BM and the release of sC5b-9 in the supernatant. Our results show classical and alternative complement pathway activation in BP skin biopsies, but could not demonstrate significant lectin pathway activation. In contrast to dexamethasone, complement deposition along the BM could be selectively inhibited by anti-C1 and anti- factor B. More downstream, selective intervention at the level of C3 and C5 could effectively reduce complement deposition along the BM and the release of sC5b-9 in the supernatant. This study shows that selective intervention in either the classical, alternative or terminal pathway prevented deposition of complement along the BM in an in vitro BP model. The results of our study greatly encourage the clinical development of complement inhibitors for the treatment of BP.


Assuntos
Penfigoide Bolhoso , Humanos , Estudos Retrospectivos , Proteínas do Sistema Complemento , Anticorpos , Dexametasona
2.
Pediatr Transplant ; 25(3): e13903, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33217135

RESUMO

aHUS is a rare disease characterized by episodes of TMA that frequently progresses to CKD and often recurs after KT. The most frequent cause of aHUS is defective regulation of complement activation because of genetic anomalies. Eculizumab interrupts the process of TMA and improves renal function. We describe one female patient with aHUS who debuted in 2005 at 3-mo-old with extrarenal manifestations and progressed to end-stage kidney disease (ESKD) within a year. Her family history included several affected members with similar bad outcomes. Our patient carries a strong aHUS genetic predisposition consisting in a pathogenic gain-of-function mutation in complement factor B concurrent with the MCP aHUS risk haplotype MCPggaac. She received a kidney transplant in 2011 without eculizumab prophylaxis. The graft, which was negative for the MCPggaac risk haplotype, had an unexpected excellent evolution without aHUS recurrence. Different retrospective studies have shown that the risk of aHUS recurrence after KT correlates well with the genetic load of aHUS risk factors. Knowing important contribution of the MCPggaac risk haplotype to the risk of developing aHUS in Factor B mutations carriers, we speculate whether the absence of this polymorphism in the graft that our patient received may have decreased the risk of aHUS recurrence after KT.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/genética , Transplante de Rim , Proteína Cofatora de Membrana/genética , Polimorfismo Genético , Feminino , Humanos , Lactente , Linhagem , Medição de Risco
4.
Kidney Int ; 88(5): 1153-60, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26221755

RESUMO

C3 glomerulonephritis is a clinicopathologic entity defined by the presence of isolated or dominant deposits of C3 on immunofluorescence. To explore the effect of immunosuppression on C3 glomerulonephritis, we studied a series of 60 patients in whom a complete registry of treatments was available over a median follow-up of 47 months. Twenty patients had not received immunosuppressive treatments. In the remaining 40 patients, 22 had been treated with corticosteroids plus mycophenolate mofetil while 18 were treated with other immunosuppressive regimens (corticosteroids alone or corticosteroids plus cyclophosphamide). The number of patients developing end-stage renal disease was significantly lower among treated compared with untreated patients (3 vs. 7 patients, respectively). No patient in the corticosteroids plus mycophenolate mofetil group doubled serum creatinine nor developed end-stage renal disease, as compared with 7 (significant) and 3 (not significant), respectively, in patients treated with other immunosuppressive regimens. Renal survival (100, 80, and 72% at 5 years) and the number of patients achieving clinical remission (86, 50, and 25%) were significantly higher in patients treated with corticosteroids plus mycophenolate mofetil as compared with patients treated with other immunosuppressive regimens and untreated patients, respectively. Thus, immunosuppressive treatments, particularly corticosteroids plus mycophenolate mofetil, can be beneficial in C3 glomerulonephritis.


Assuntos
Complemento C3 , Glomerulonefrite/tratamento farmacológico , Imunossupressores/uso terapêutico , Ácido Micofenólico/análogos & derivados , Adolescente , Corticosteroides/uso terapêutico , Adulto , Idoso , Anti-Inflamatórios/uso terapêutico , Creatinina/sangue , Ciclofosfamida/uso terapêutico , Progressão da Doença , Quimioterapia Combinada , Feminino , Seguimentos , Glomerulonefrite/complicações , Glomerulonefrite/imunologia , Humanos , Falência Renal Crônica/etiologia , Masculino , Pessoa de Meia-Idade , Ácido Micofenólico/uso terapêutico , Estudos Retrospectivos , Taxa de Sobrevida , Adulto Jovem
5.
BMC Nephrol ; 16: 40, 2015 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-25886501

RESUMO

BACKGROUND: Glomerulonephritis is one of the most severe complications of lupus, a systemic disease with multi-organ involvement, with tissue damage produced mainly by complement activation. As a result of this activation, patients with active lupus present hypocomplementemia during disease flares, but C3 and C4 levels are recovered between episodes. CASE PRESENTATION: We present a patient who suffered two lupus nephritis episodes in 5 years, achieving complete remission with treatment after both of them, but with C3 levels persistently below normal range. Genetic study revealed that the patient carried a mutation in heterozygosis in the C3 gene. Serial sera samples were analyzed, and autoantibodies to complement alternative pathway proteins (Factor I, Factor B, C3 and Properdin) were found. Functional assays showed that these autoantibodies cause alternative pathway activation. CONCLUSION: This case is the first reported of a heterozygous C3 mutation associated with lupus nephritis and autoantibodies against complement alternative pathway proteins (Factor I, Factor B, C3 and Properdin).These autoantibodies cause activation of this pathway and this fact could explain that the tissue damage is restricted to the kidney.


Assuntos
Autoanticorpos/imunologia , Complemento C3/genética , Rim/patologia , Nefrite Lúpica/genética , Complemento C3/imunologia , Fator B do Complemento/imunologia , Feminino , Fibrinogênio/imunologia , Heterozigoto , Humanos , Nefrite Lúpica/imunologia , Nefrite Lúpica/patologia , Mutação , Properdina/imunologia , Adulto Jovem
6.
Proc Natl Acad Sci U S A ; 108(32): 13236-40, 2011 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-21788512

RESUMO

Activation of C3, deposition of C3b on the target surface, and subsequent amplification by formation of a C3-cleaving enzyme (C3-convertase; C3bBb) triggers the effector functions of complement that result in inflammation and cell lysis. Concurrently, surface-bound C3b is proteolyzed to iC3b by factor I and appropriate cofactors. iC3b then interacts with the complement receptors (CR) of the Ig superfamily, CR2 (CD21), CR3 (CD11b/CD18), and CR4 (CD11c/CD18) on leukocytes, down-modulating inflammation, enhancing B cell-mediated immunity, and targeting pathogens for clearance by phagocytosis. Using EM and small-angle X-ray scattering, we now present a medium-resolution structure of iC3b (24 Å). iC3b displays a unique conformation with structural features distinct from any other C3 fragment. The macroglobulin ring in iC3b is similar to that in C3b, whereas the TED (thioester-containing domain) domain and the remnants of the CUB (complement protein subcomponents C1r/C1s, urchin embryonic growth factor and bone morphogenetic protein 1) domain have moved to locations more similar to where they were in native C3. A consequence of this large conformational change is the disruption of the factor B binding site, which renders iC3b unable to assemble a C3-convertase. This structural model also justifies the decreased interaction between iC3b and complement regulators and the recognition of iC3b by the CR of the Ig superfamily, CR2, CR3, and CR4. These data further illustrate the extraordinary conformational versatility of C3 to accommodate a great diversity of functional activities.


Assuntos
Complemento C3b/química , Complemento C3b/ultraestrutura , Microscopia Eletrônica , Complemento C3b/isolamento & purificação , Humanos , Modelos Moleculares , Estrutura Terciária de Proteína , Receptores de Complemento 3b/química , Espalhamento a Baixo Ângulo , Soluções , Difração de Raios X
7.
J Exp Med ; 204(6): 1249-56, 2007 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-17517971

RESUMO

Factor H (FH) is an abundant serum glycoprotein that regulates the alternative pathway of complement-preventing uncontrolled plasma C3 activation and nonspecific damage to host tissues. Age-related macular degeneration (AMD), atypical hemolytic uremic syndrome (aHUS), and membranoproliferative glomerulonephritis type II (MPGN2) are associated with polymorphisms or mutations in the FH gene (Cfh), suggesting the existence of a genotype-phenotype relationship. Although AMD and MPGN2 share pathological similarities with the accumulation of complement-containing debris within the eye and kidney, respectively, aHUS is characterized by renal endothelial injury. This pathological distinction was reflected in our Cfh association analysis, which demonstrated that although AMD and MPGN2 share a Cfh at-risk haplotype, the haplotype for aHUS was unique. FH-deficient mice have uncontrolled plasma C3 activation and spontaneously develop MPGN2 but not aHUS. We show that these mice, transgenically expressing a mouse FH protein functionally equivalent to aHUS-associated human FH mutants, regulate C3 activation in plasma and spontaneously develop aHUS but not MPGN2. These animals represent the first model of aHUS and provide in vivo evidence that effective plasma C3 regulation and the defective control of complement activation on renal endothelium are the critical events in the molecular pathogenesis of FH-associated aHUS.


Assuntos
Fator H do Complemento/genética , Glomerulonefrite Membranoproliferativa/genética , Síndrome Hemolítico-Urêmica/genética , Degeneração Macular/genética , Idoso , Animais , Western Blotting , Complemento C3/metabolismo , Primers do DNA , Olho/patologia , Haplótipos/genética , Síndrome Hemolítico-Urêmica/patologia , Humanos , Rim/patologia , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Estrutura Terciária de Proteína
8.
Kidney Int ; 81(1): 56-63, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21881555

RESUMO

Mutations and polymorphisms in the gene-encoding factor H (CFH) are associated with atypical hemolytic uremic syndrome, dense deposit disease, and age-related macular degeneration. Many of these CFH genetic variations disrupt the regulatory role of factor H, supporting the concept that dysregulation of complement is a unifying pathogenic feature of these disorders. Evidence of a causal relationship with the disease is, however, not available for all CFH genetic variations found in patients, which is a potential cause of misinterpretations with important consequences for the patients and their relatives. CFH I890 and L1007 are two genetic variations repeatedly associated with atypical hemolytic uremic syndrome and also found in patients with dense deposit disease and age-related macular degeneration. Here we report an extensive genetic and functional analysis of these CFH variants. Our results indicate that I890 and L1007 segregate together as part of a distinct and relatively infrequent CFH haplotype in Caucasians. Extensive analysis of the S890/V1007 (control) and I890/L1007 (disease-associated) factor H protein variants failed to provide evidence that these amino acid changes have functional implications. Thus, the presence of the I890 and L1007 variants in healthy individuals and their high frequency in sub-Saharan African and African-American populations strongly suggest that I890 and L1007 are rare factor H polymorphisms unrelated to disease.


Assuntos
Variação Genética , Síndrome Hemolítico-Urêmica/genética , Adulto , África Subsaariana , Negro ou Afro-Americano/genética , Substituição de Aminoácidos , Síndrome Hemolítico-Urêmica Atípica , População Negra/genética , Pré-Escolar , Fator H do Complemento/genética , Frequência do Gene , Predisposição Genética para Doença , Glomerulonefrite Membranoproliferativa/genética , Haplótipos , Síndrome Hemolítico-Urêmica/sangue , Humanos , Lactente , Degeneração Macular/genética , Masculino , Pessoa de Meia-Idade , Mutação , Polimorfismo Genético , Espanha
9.
Kidney Int ; 82(10): 1084-92, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22854646

RESUMO

C3 nephritic factors are autoantibodies that prolong the half-life or prevent regulation of the alternative pathway C3 convertase, resulting in uncontrolled complement activation. They are strongly associated with renal disease but their role in pathogenesis remains controversial. Here we optimized and compared a panel of assays to identify and interrogate nephritic factor activities. Of 101 patients with histologic or clinically evident disease, 48 were positive in some or all assays. In the presence of properdin, binding of autoantibody was detected in 39 samples and convertase stabilization was detected in 36. Forty-two of 48 nephritic factors tested prevented convertase decay by factor H, and most of these by decay accelerating factor (28) and complement receptor 1 (34). Representative properdin-independent nephritic factors had no effect on C5 cleavage and terminal pathway activity, while properdin-dependent nephritic factors enhanced activity. Biacore analysis of four purified IgG samples confirmed resistance to decay and showed that properdin-independent nephritic factors increased convertase half-life over 50-fold, whereas properdin-dependent nephritic factors increased the half-life 10- to 20-fold and also increased activity of the C3 convertase up to 10-fold. Thus, our study provides a rational approach to detect and characterize nephritic factors in patients.


Assuntos
Ativação do Complemento , Fator Nefrítico do Complemento 3/metabolismo , Glomerulonefrite Membranoproliferativa/diagnóstico , Imunoensaio , Animais , Biomarcadores/sangue , Antígenos CD55/metabolismo , Estudos de Casos e Controles , Complemento C3/metabolismo , Convertases de Complemento C3-C5/metabolismo , Fator D do Complemento/metabolismo , Fator H do Complemento/metabolismo , Ensaio de Atividade Hemolítica de Complemento , Ensaio de Imunoadsorção Enzimática , Glomerulonefrite Membranoproliferativa/sangue , Glomerulonefrite Membranoproliferativa/imunologia , Humanos , Imunoensaio/métodos , Valor Preditivo dos Testes , Properdina/metabolismo , Ligação Proteica , Receptores de Complemento/metabolismo , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Ovinos , Fatores de Tempo
10.
Hum Mol Genet ; 19(14): 2867-76, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20453062

RESUMO

Lafora disease (LD) is an autosomal recessive, progressive myoclonus epilepsy, which is characterized by the accumulation of polyglucosan inclusion bodies, called Lafora bodies, in the cytoplasm of cells in the central nervous system and in many other organs. However, it is unclear at the moment whether Lafora bodies are the cause of the disease, or whether they are secondary consequences of a primary metabolic alteration. Here we describe that the major genetic lesion that causes LD, loss-of-function of the protein laforin, impairs autophagy. This phenomenon is confirmed in cell lines from human patients, mouse embryonic fibroblasts from laforin knockout mice and in tissues from such mice. Conversely, laforin expression stimulates autophagy. Laforin regulates autophagy via the mammalian target of rapamycin kinase-dependent pathway. The changes in autophagy mediated by laforin regulate the accumulation of diverse autophagy substrates and would be predicted to impact on the Lafora body accumulation and the cell stress seen in this disease that may eventually contribute to cell death.


Assuntos
Autofagia/genética , Doença de Lafora/genética , Proteínas Tirosina Fosfatases não Receptoras/genética , Proteínas Tirosina Fosfatases não Receptoras/fisiologia , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Doença de Lafora/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Mutantes/genética , Proteínas Mutantes/fisiologia , Fagossomos/genética , Fagossomos/metabolismo , Processamento de Proteína Pós-Traducional/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR
11.
Hum Mol Genet ; 18(18): 3452-61, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19549636

RESUMO

Mutations and polymorphisms in the gene encoding factor H (CFH) have been associated with atypical haemolytic uraemic syndrome, dense deposit disease and age-related macular degeneration. The disease-predisposing CFH variants show a differential association with pathology that has been very useful to unravel critical events in the pathogenesis of one or other disease. In contrast, the factor H (fH)-Ile(62) polymorphism confers strong protection to all three diseases. Using ELISA-based methods and surface plasmon resonance analyses, we show here that the protective fH-Ile(62) variant binds more efficiently to C3b than fH-Val(62) and competes better with factor B in proconvertase formation. Functional analyses demonstrate an increased cofactor activity for fH-Ile(62) in the factor I-mediated cleavage of fluid phase and surface-bound C3b; however, the two fH variants show no differences in decay accelerating activity. From these data, we conclude that the protective effect of the fH-Ile(62) variant is due to its better capacity to bind C3b, inhibit proconvertase formation and catalyze inactivation of fluid-phase and surface-bound C3b. This demonstration of the functional consequences of the fH-Ile(62) polymorphism provides relevant insights into the complement regulatory activities of fH that will be useful in disease prediction and future development of effective therapeutics for disorders caused by complement dysregulation.


Assuntos
Complemento C3b/metabolismo , Polimorfismo Genético , Fator B do Complemento/metabolismo , Fator H do Complemento/genética , Fator H do Complemento/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Isoleucina/genética , Ligação Proteica , Ressonância de Plasmônio de Superfície
12.
Lancet ; 376(9743): 794-801, 2010 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-20800271

RESUMO

BACKGROUND: Complement is a key component of the innate immune system, and variation in genes that regulate its activation is associated with renal and other disease. We aimed to establish the genetic basis for a familial disorder of complement regulation associated with persistent microscopic haematuria, recurrent macroscopic haematuria, glomerulonephritis, and progressive renal failure. METHODS: We sought patients from the West London Renal and Transplant Centre (London, UK) with unusual renal disease and affected family members as a method of identification of new genetic causes of kidney disease. Two families of Cypriot origin were identified in which renal disease was consistent with autosomal dominant transmission and renal biopsy of at least one individual showed C3 glomerulonephritis. A mutation was identified via a genome-wide linkage study and candidate gene analysis. A PCR-based diagnostic test was then developed and used to screen for the mutation in population-based samples and in individuals and families with renal disease. FINDINGS: Occurrence of familial renal disease cosegregated with the same mutation in the complement factor H-related protein 5 gene (CFHR5). In a cohort of 84 Cypriots with unexplained renal disease, four had mutation in CFHR5. Overall, we identified 26 individuals with the mutation and evidence of renal disease from 11 ostensibly unrelated kindreds, including the original two families. A mutant CFHR5 protein present in patient serum had reduced affinity for surface-bound complement. We term this renal disease CFHR5 nephropathy. INTERPRETATION: CFHR5 nephropathy accounts for a substantial burden of renal disease in patients of Cypriot origin and can be diagnosed with a specific molecular test. The high risk of progressive renal disease in carriers of the CFHR5 mutation implies that isolated microscopic haematuria or recurrent macroscopic haematuria should not be regarded as a benign finding in individuals of Cypriot descent. FUNDING: UK Medical Research Council and Wellcome Trust.


Assuntos
Proteínas Sanguíneas/genética , Complemento C5/genética , Fator H do Complemento/genética , Glomerulonefrite/genética , Glomerulonefrite/patologia , Falência Renal Crônica/etiologia , Mutação , Adulto , Idoso , Proteínas do Sistema Complemento , Chipre/epidemiologia , Chipre/etnologia , Doenças Endêmicas , Feminino , Estudo de Associação Genômica Ampla , Glomerulonefrite/sangue , Glomerulonefrite/complicações , Glomerulonefrite/epidemiologia , Humanos , Falência Renal Crônica/genética , Masculino , Pessoa de Meia-Idade , Linhagem , Polimorfismo de Nucleotídeo Único
13.
Blood ; 114(19): 4261-71, 2009 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-19745068

RESUMO

The factor H-related protein family (CFHR) is a group of minor plasma proteins genetically and structurally related to complement factor H (fH). Notably, deficiency of CFHR1/CFHR3 associates with protection against age-related macular degeneration and with the presence of anti-fH autoantibodies in atypical hemolytic uremic syndrome (aHUS). We have developed a proteomics strategy to analyze the CFHR proteins in plasma samples from controls, patients with aHUS, and patients with type II membranoproliferative glomerulonephritis. Here, we report on the identification of persons carrying novel deficiencies of CFHR1, CFHR3, and CFHR1/CFHR4A, resulting from point mutations in CFHR1 and CFHR3 or from a rearrangement involving CFHR1 and CFHR4. Remarkably, patients with aHUS lacking CFHR1, but not those lacking CFHR3, present anti-fH autoantibodies, suggesting that generation of these antibodies is specifically related to CFHR1 deficiency. We also report the characterization of a novel CFHR1 polymorphism, resulting from a gene conversion event between CFH and CFHR1, which strongly associates with aHUS. The risk allotype CFHR1*B, with greater sequence similarity to fH, may compete with fH, decreasing protection of cellular surfaces against complement damage. In summary, our comprehensive analyses of the CFHR proteins have improved our understanding of these proteins and provided further insights into aHUS pathogenesis.


Assuntos
Proteínas Inativadoras do Complemento C3b/genética , Síndrome Hemolítico-Urêmica/sangue , Síndrome Hemolítico-Urêmica/genética , Mutação , Adolescente , Adulto , Idoso , Alelos , Autoanticorpos/sangue , Sequência de Bases , Proteínas Sanguíneas/genética , Estudos de Casos e Controles , Criança , Pré-Escolar , Proteínas Inativadoras do Complemento C3b/deficiência , Fator H do Complemento/genética , Fator H do Complemento/imunologia , Feminino , Conversão Gênica , Glomerulonefrite Membranoproliferativa/sangue , Glomerulonefrite Membranoproliferativa/genética , Síndrome Hemolítico-Urêmica/imunologia , Humanos , Degeneração Macular/sangue , Degeneração Macular/genética , Masculino , Pessoa de Meia-Idade , Sondas de Oligonucleotídeos/genética , Polimorfismo de Nucleotídeo Único , Isoformas de Proteínas/genética , Proteômica , Adulto Jovem
14.
J Am Soc Nephrol ; 19(3): 639-46, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18235085

RESUMO

Mutations in the gene encoding complement factor H (CFH) that alter the C3b/polyanions-binding site in the C-terminal region impair the capacity of factor H to protect host cells. These mutations are also strongly associated with atypical hemolytic uremic syndrome (aHUS). Although most of the aHUS-associated CFH mutations seem "unique" to an individual patient or family, the R1210C mutation has been reported in several unrelated aHUS patients from distinct geographic origins. Five aHUS pedigrees and 7 individual aHUS patients were analyzed to identify potential correlations between the R1210C mutation and clinical phenotype and to characterize the origins of this mutation. The clinical phenotype of aHUS patients carrying the R1210C mutation was heterogeneous. Interestingly, 12 of the 13 affected patients carried at least one additional known genetic risk factor for aHUS. These data are in accord with the 30% penetrance of aHUS in R1210C mutation carriers, as it seems that the presence of other genetic or environmental risk factors significantly contribute to the manifestation and severity of aHUS in these subjects. Genotype analysis of CFH and CFHR3 polymorphisms in the 12 unrelated carriers suggested that the R1210C mutation has a single origin. In conclusion, the R1210C mutation of complement factor H is a prototypical aHUS mutation that is present as a rare polymorphism in geographically separated human populations.


Assuntos
Fator H do Complemento/genética , Síndrome Hemolítico-Urêmica/genética , Adolescente , Adulto , Estudos de Casos e Controles , Criança , Pré-Escolar , Fator I do Complemento/genética , Feminino , Heterozigoto , Humanos , Lactente , Masculino , Proteína Cofatora de Membrana/genética , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Fenótipo , Polimorfismo Genético
15.
Nat Rev Nephrol ; 15(3): 129-143, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30692664

RESUMO

The C3 glomerulopathies are a group of rare kidney diseases characterized by complement dysregulation occurring in the fluid phase and in the glomerular microenvironment, which results in prominent complement C3 deposition in kidney biopsy samples. The two major subgroups of C3 glomerulopathy - dense deposit disease (DDD) and C3 glomerulonephritis (C3GN) - have overlapping clinical and pathological features suggestive of a disease continuum. Dysregulation of the complement alternative pathway is fundamental to the manifestations of C3 glomerulopathy, although terminal pathway dysregulation is also common. Disease is driven by acquired factors in most patients - namely, autoantibodies that target the C3 or C5 convertases. These autoantibodies drive complement dysregulation by increasing the half-life of these vital but normally short-lived enzymes. Genetic variation in complement-related genes is a less frequent cause. No disease-specific treatments are available, although immunosuppressive agents and terminal complement pathway blockers are helpful in some patients. Unfortunately, no treatment is universally effective or curative. In aggregate, the limited data on renal transplantation point to a high risk of disease recurrence (both DDD and C3GN) in allograft recipients. Clinical trials are underway to test the efficacy of several first-generation drugs that target the alternative complement pathway.


Assuntos
Autoimunidade , Complemento C3/imunologia , Nefropatias/imunologia , Glomérulos Renais/patologia , Autoanticorpos/imunologia , Biópsia , Complemento C3/metabolismo , Humanos , Nefropatias/metabolismo , Glomérulos Renais/metabolismo , Doenças Raras
16.
Am J Kidney Dis ; 52(1): 171-80, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18423815

RESUMO

Atypical hemolytic uremic syndrome is associated with mutations in the complement proteins factor H, factor I, factor B, C3, or membrane cofactor protein in about 50% of patients. The evolution and prognosis of the disease in patients carrying mutations in factor H is particularly poor, and renal transplantation most often fails because of recurrence of the disease in the graft. The risk of rapid loss of renal function in patients with functional mutations in factor H requires that effective treatment be initiated as soon as possible, but identification of these patients relies on genetic studies that are time consuming. We describe a case in which an in vitro hemolytic assay proved useful for rapidly assessing factor H dysfunction and for testing whether this dysfunction could be corrected with fresh frozen plasma. In the context of this case, we summarize recent advances in understanding the molecular mechanisms contributing to atypical hemolytic uremic syndrome, including descriptions of DNA- and protein-based analysis. We conclude that functional analysis of factor H should help rationalize the plasma treatment of patients with atypical hemolytic uremic syndrome.


Assuntos
Fator H do Complemento/genética , Síndrome Hemolítico-Urêmica/genética , Síndrome Hemolítico-Urêmica/fisiopatologia , Mutação , Ativação do Complemento , Análise Mutacional de DNA , Progressão da Doença , Evolução Fatal , Feminino , Síndrome Hemolítico-Urêmica/terapia , Humanos , Lactente , Plasmaferese , Diálise Renal/métodos , Insuficiência Respiratória , Índice de Gravidade de Doença
17.
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
18.
Mol Immunol ; 43(11): 1769-75, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16386793

RESUMO

Mutations in the complement regulators factor H, membrane cofactor protein (MCP), and factor I are associated with atypical hemolytic uremic syndrome (aHUS, MIM 235400), suggesting that the disease develops as a consequence of the inefficient protection of the renal endothelium from damage by the complement system. Incomplete penetrance of the disease in individuals carrying these mutations is, however, relatively frequent. Here, we report the identification of a large, multiple affected aHUS pedigree in which there is independent segregation of three different aHUS risk factors: a MCP missense mutation (c.-598C>T; Pro165Ser) that decreases MCP expression on the cell surface, a dinucleotide insertion in the coding sequence of factor I (c.-1610insAT) that introduces a premature stop codon in the factor I protein, and the MCPggaac SNP haplotype block that was previously shown to decrease the transcription activity from the MCP promoter. Interestingly, individuals affected by aHUS in the pedigree are only those who have inherited the three aHUS risk factors. These data show an additive effect for mutations in MCP and factor I and provide definitive support to the conclusion that aHUS results from a defective protection of cellular surfaces from complement activation. Furthermore, they help to explain the incomplete penetrance of the disease, illustrating that concurrence of multiple hits in complement regulatory proteins may be necessary to significantly impair host tissue protection and to confer susceptibility to aHUS.


Assuntos
Fator H do Complemento/genética , Fator I do Complemento/genética , Ligação Genética/genética , Predisposição Genética para Doença/genética , Síndrome Hemolítico-Urêmica/genética , Proteína Cofatora de Membrana/genética , Linhagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Sequência de Aminoácidos , Segregação de Cromossomos/genética , Fator I do Complemento/química , Éxons/genética , Feminino , Genótipo , Humanos , Proteína Cofatora de Membrana/química , Pessoa de Meia-Idade , Dados de Sequência Molecular , Mutação/genética
19.
Immunobiology ; 221(6): 709-14, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26004345

RESUMO

Different genome-wide linkage and association studies performed during the last 15 years have associated mutations and polymorphisms in complement genes with different diseases characterized by tissue damage and inflammation. These are complex disorders in which genetically susceptible individuals usually develop the pathology as a consequence of environmental triggers. Although complement dysregulation is a common feature of these pathologies, how the disease phenotype is determined is only partly understood. One way to advance understanding is to focus the research in the analysis of the peculiar genotype-phenotype correlations that characterize some of these diseases. I will review here how understanding the functional consequences of these disease-associated complement genetic variants is providing us with novel insights into the underpinning complement biology and a better knowledge of the pathogenic mechanisms underlying each of these pathologies. These advances have important therapeutic and diagnostic implications.


Assuntos
Ativação do Complemento/genética , Proteínas do Sistema Complemento/genética , Inflamação/genética , Animais , Interação Gene-Ambiente , Predisposição Genética para Doença , Genótipo , Humanos , Fenótipo , Polimorfismo Genético
20.
J Nephrol ; 28(5): 641-5, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25712233

RESUMO

Pregnancy-associated atypical hemolytic uremic syndrome is a systemic disease associated with high morbidity and mortality rates, caused by dysregulation of the alternative complement pathway, leading to uncontrolled complement activation resulting in thrombotic microangiopathy. This condition can be effectively treated by anti-C5 therapy, which controls complement activation. Treatment can be safely discontinued after complete remission and resolution of the precipitating cause, especially in patients with a low-risk genetic profile.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Síndrome Hemolítico-Urêmica Atípica/tratamento farmacológico , Gerenciamento Clínico , Complicações na Gravidez/tratamento farmacológico , Adulto , Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Biópsia , Diagnóstico Diferencial , Feminino , Seguimentos , Humanos , Rim/patologia , Gravidez , Indução de Remissão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA