Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
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.
Semin Cell Dev Biol ; 85: 86-97, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29292221

RESUMO

The complement system is a complex and autoregulated multistep cascade at the interface of innate and adaptive immunity. It is activated by immune complexes or apoptotic cells (classical pathway), pathogen-associated glycoproteins (lectin pathway) or a variety of molecular and cellular surfaces (alternative pathway). Upon activation, complement triggers the generation of proteolytic fragments that allow the elimination of the activating surface by enhancing inflammation, opsonization, phagocytosis, and cellular lysis. Moreover, complement efficiently discriminates self from non-self surfaces by means of soluble and membrane-bound complement regulators which are critical for innate self-tolerance. Complement deficiency or dysfunction disturb complement homeostasis and give rise to diseases as diverse as bacterial infections, autoimmunity, or renal and neurological disorders. Research on complement-targeted therapies is an expanding field that has already improved the prognosis of severe diseases such as atypical Haemolytic Uremic syndrome or Paroxysmal Nocturnal Haemoglobinuria. Therefore, complement analysis and monitoring provides valuable information with deep implications for diagnosis and therapy. In addition to its important role as an extracellular defense system, it has now become evident that complement is also present intracellularly, and its activation has profound implications for leukocyte survival and function. In this review, we summarize the essential, up-to-date information on the use of complement as a diagnostic and therapeutic tool in the clinics.


Assuntos
Doenças Autoimunes/diagnóstico , Doenças Transmissíveis/diagnóstico , Doenças Transmissíveis/imunologia , Proteínas do Sistema Complemento/imunologia , Doenças Neurodegenerativas/diagnóstico , Doenças Neurodegenerativas/imunologia , Sepse/diagnóstico , Sepse/imunologia , Doenças Autoimunes/imunologia , Proteínas do Sistema Complemento/metabolismo , Humanos , Tolerância Imunológica , Imunidade Inata
3.
J Immunol ; 191(2): 912-21, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23772024

RESUMO

Complement is an essential humoral component of innate immunity; however, its inappropriate activation leads to pathology. Polymorphisms, mutations, and autoantibodies affecting factor H (FH), a major regulator of the alternative complement pathway, are associated with various diseases, including age-related macular degeneration, atypical hemolytic uremic syndrome, and C3 glomerulopathies. Restoring FH function could be a treatment option for such pathologies. In this article, we report on an engineered FH construct that directly combines the two major functional regions of FH: the N-terminal complement regulatory domains and the C-terminal surface-recognition domains. This minimal-size FH (mini-FH) binds C3b and has complement regulatory functions similar to those of the full-length protein. In addition, we demonstrate that mini-FH binds to the FH ligands C-reactive protein, pentraxin 3, and malondialdehyde epitopes. Mini-FH was functionally active when bound to the extracellular matrix and endothelial cells in vitro, and it inhibited C3 deposition on the cells. Furthermore, mini-FH efficiently inhibited complement-mediated lysis of host-like cells caused by a disease-associated FH mutation or by anti-FH autoantibodies. Therefore, mini-FH could potentially be used as a complement inhibitor targeting host surfaces, as well as to replace compromised FH in diseases associated with FH dysfunction.


Assuntos
Complemento C3b/imunologia , Fator H do Complemento/genética , Fator H do Complemento/imunologia , Sequência de Aminoácidos , Sequência de Bases , Proteína C-Reativa/metabolismo , Ativação do Complemento , Complemento C3b/antagonistas & inibidores , Complemento C3b/metabolismo , Via Alternativa do Complemento , Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , Humanos , Malondialdeído/metabolismo , Ligação Proteica , Engenharia de Proteínas , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Análise de Sequência de DNA , Componente Amiloide P Sérico/metabolismo
4.
J Immunol ; 189(4): 1858-67, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22786770

RESUMO

Atypical hemolytic uremic syndrome (aHUS) is a renal disease associated with complement alternative pathway dysregulation and is characterized by endothelial injury. Pentraxin 3 (PTX3) is a soluble pattern recognition molecule expressed by endothelial cells and upregulated under inflammatory conditions. PTX3 activates complement, but it also binds the complement inhibitor factor H. In this study, we show that native factor H, factor H-like protein 1, and factor H-related protein 1 (CFHR1) bind to PTX3 and that PTX3-bound factor H and factor H-like protein 1 maintain their complement regulatory activities. PTX3, when bound to extracellular matrix, recruited functionally active factor H. Residues within short consensus repeat 20 of factor H that are relevant for PTX3 binding were identified using a peptide array. aHUS-associated factor H mutations within this binding site caused a reduced factor H binding to PTX3. Similarly, seven of nine analyzed anti-factor H autoantibodies isolated from aHUS patients inhibited the interaction between factor H and PTX3, and five autoantibodies also inhibited PTX3 binding to CFHR1. Moreover, the aHUS-associated CFHR1*B variant showed reduced binding to PTX3 in comparison with CFHR1*A. Thus, the interactions of PTX3 with complement regulators are impaired by certain mutations and autoantibodies affecting factor H and CFHR1, which could result in an enhanced local complement-mediated inflammation, endothelial cell activation, and damage in aHUS.


Assuntos
Autoanticorpos/imunologia , Proteína C-Reativa/metabolismo , Proteínas Inativadoras do Complemento C3b/metabolismo , Fator H do Complemento/metabolismo , Síndrome Hemolítico-Urêmica/metabolismo , Componente Amiloide P Sérico/metabolismo , Síndrome Hemolítico-Urêmica Atípica , Autoantígenos/imunologia , Proteína C-Reativa/imunologia , Proteínas Inativadoras do Complemento C3b/imunologia , Fator H do Complemento/imunologia , Matriz Extracelular/imunologia , Matriz Extracelular/metabolismo , Síndrome Hemolítico-Urêmica/imunologia , Humanos , Componente Amiloide P Sérico/imunologia
5.
Pediatr Nephrol ; 29(1): 149-53, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23982707

RESUMO

BACKGROUND: Atypical hemolytic uremic syndrome (aHUS) is a form of thrombotic microangiopathy (TMA) caused by dysregulation of the complement system. Outcomes of kidney transplantation are poor owing to aHUS recurrence and loss of graft. Patients carrying CFH mutations or CFH/CFHR1 hybrid genes present a very high risk of recurrence despite preventive plasmapheresis. Evaluation of recent data suggests that prophylactic eculizumab pretransplant might be the preferred therapy if available. CASE-DIAGNOSIS/TREATMENT: We report 3-year follow-up data in a 9-year-old boy with aHUS and successful renal transplant treated with prophylactic eculizumab without recurrence. He presented with aHUS at age 3, irreversible renal failure and uncontrolled severe hypertension with concentric left ventricular hypertrophy, recurrent acute pulmonary edema, and congestive heart failure despite five hypotensive agents and bilateral nephrectomy. Complement analysis demonstrated the presence of a CFH/CFHR1 hybrid gene inherited from his mother and a SNP risk CFH haplotype inherited from his father. Kidney transplant was performed with prophylactic eculizumab and subsequent fortnightly administration. Three years post-transplant, graft function remains stable (serum creatinine 0.9 mg/dl), hypertension is controlled, no left ventricular hypertrophy, no opportunistic infections, and negative clinical chemistry parameters for hemolysis. CONCLUSION: Eculizumab is a safe and effective therapy for preventing TMA recurrence and provides long-term graft function in aHUS with the CFH/CFHR1 hybrid gene.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Síndrome Hemolítico-Urêmica/terapia , Transplante de Rim , Proteínas Mutantes Quiméricas/genética , Síndrome Hemolítico-Urêmica Atípica , Criança , Síndrome Hemolítico-Urêmica/genética , Humanos , Masculino
6.
J Am Soc Nephrol ; 24(3): 475-86, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23431077

RESUMO

Several abnormalities in complement genes reportedly contribute to atypical hemolytic uremic syndrome (aHUS), but incomplete penetrance suggests that additional factors are necessary for the disease to manifest. Here, we sought to describe genotype-phenotype correlations among patients with combined mutations, defined as mutations in more than one complement gene. We screened 795 patients with aHUS and identified single mutations in 41% and combined mutations in 3%. Only 8%-10% of patients with mutations in CFH, C3, or CFB had combined mutations, whereas approximately 25% of patients with mutations in MCP or CFI had combined mutations. The concomitant presence of CFH and MCP risk haplotypes significantly increased disease penetrance in combined mutated carriers, with 73% penetrance among carriers with two risk haplotypes compared with 36% penetrance among carriers with zero or one risk haplotype. Among patients with CFH or CFI mutations, the presence of mutations in other genes did not modify prognosis; in contrast, 50% of patients with combined MCP mutation developed end stage renal failure within 3 years from onset compared with 19% of patients with an isolated MCP mutation. Patients with combined mutations achieved remission with plasma treatment similar to patients with single mutations. Kidney transplant outcomes were worse, however, for patients with combined MCP mutation compared with an isolated MCP mutation. In summary, these data suggest that genotyping for the risk haplotypes in CFH and MCP may help predict the risk of developing aHUS in unaffected carriers of mutations. Furthermore, screening patients with aHUS for all known disease-associated genes may inform decisions about kidney transplantation.


Assuntos
Proteínas do Sistema Complemento/genética , Síndrome Hemolítico-Urêmica/genética , Síndrome Hemolítico-Urêmica/imunologia , Mutação , Adulto , Síndrome Hemolítico-Urêmica Atípica , Criança , Pré-Escolar , Complemento C3/genética , Fator B do Complemento/genética , Fator H do Complemento/genética , Feminino , Fibrinogênio/genética , Estudos de Associação Genética , Haplótipos , Humanos , Lactente , Masculino , Proteína Cofatora de Membrana/genética , Pessoa de Meia-Idade , Linhagem , Penetrância , Fatores de Risco , Adulto Jovem
7.
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
8.
Nefrologia (Engl Ed) ; 42(3): 280-289, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36154806

RESUMO

The complement system is a first line of defence against infectious, tumoral or autoimmune processes, and it is constitutively regulated to avoid excessive or unspecific activation. Factor H (FH), a most relevant complement regulator, controls complement activation in plasma and on the cellular surfaces of autologous tissues. FH shares evolutionary origin and structural features with a group of plasma proteins known as FH-Related Proteins (FHRs), which could act as FH functional antagonists. Studies in patient cohorts of atypical Haemolytic-Uraemic Syndrome (aHUS), C3 Glomerulopathy (C3G), and IgA nephropathy (IgAN), have identified rare genetic variants that give rise to severe FH and FHRs dysfunctions, and are major genetic predisposing factors. These patients also have a higher frequency of a few polymorphisms whose relevance as disease risk factors is incompletely understood. In the last years, the availability of specific reagents has allowed a more precise quantitation of FH and FHRs in plasma samples from patients and controls. These studies have revealed that some aHUS, C3G or IgAN risk polymorphisms determine mild changes in FH or FHRs levels that could somehow perturb complement regulation and favour disease pathogenesis.


Assuntos
Síndrome Hemolítico-Urêmica Atípica , Glomerulonefrite por IGA , Síndrome Hemolítico-Urêmica Atípica/genética , Ativação do Complemento , Fator H do Complemento/genética , Humanos , Rim/patologia
9.
Kidney Int ; 80(4): 397-404, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21677636

RESUMO

The autoimmune form of atypical hemolytic uremic syndrome (HUS) is characterized by circulating autoantibodies against the complement regulator factor H, and is often associated with deficiency of the factor H-related proteins CFHR1 and CFHR3. Here we studied whether anti-factor H autoantibodies crossreact with CFHR1, and determined functional consequences of this. In ELISA, anti-factor H immunoglobulin G (IgG) autoantibodies from 24 atypical HUS patients bound to the short consensus repeat 20 domain of factor H, 21 antibodies also recognized CFHR1, but none CFHR3. Three patients also had anti-factor H IgA autoantibodies crossreacting with CFHR1. Analysis of the IgG fractions in CFHR1-deficient patients found that CFHR1-IgG complexes were formed during plasma exchange treatment, indicating that autoantibodies recognize CFHR1 in vivo. Recombinant CFHR1 prevented hemolysis of sheep erythrocytes caused by patient plasma containing anti-factor H IgG, but it did not inhibit red cell lysis caused by a factor H mutation (W1183 L) in the short consensus repeat 20 domain. Thus, exogenous CFHR1 provided during plasma exchange therapy may neutralize anti-factor H autoantibodies and help in the treatment of autoimmune atypical HUS.


Assuntos
Autoanticorpos/sangue , Autoantígenos , Autoimunidade , Proteínas Inativadoras do Complemento C3b/imunologia , Síndrome Hemolítico-Urêmica/imunologia , Imunoglobulina G/sangue , Animais , Especificidade de Anticorpos , Complexo Antígeno-Anticorpo , Síndrome Hemolítico-Urêmica Atípica , Autoantígenos/genética , Proteínas Sanguíneas/imunologia , Estudos de Casos e Controles , Proteínas Inativadoras do Complemento C3b/genética , Fator H do Complemento/imunologia , Ensaio de Imunoadsorção Enzimática , Alemanha , Hemólise , Síndrome Hemolítico-Urêmica/sangue , Síndrome Hemolítico-Urêmica/terapia , Humanos , Mutação , Troca Plasmática , Ovinos , Espanha
10.
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
11.
Nefrologia (Engl Ed) ; 2021 Aug 16.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-34412931

RESUMO

The complement system is a first line of defence against infectious, tumoral or autoimmune processes, and it is constitutively regulated to avoid excessive or unspecific activation. Factor H (FH), a most relevant complement regulator, controls complement activation in plasma and on the cellular surfaces of autologous tissues. FH shares evolutionary origin and structural features with a group of plasma proteins known as FH-Related Proteins (FHRs), which could act as FH functional antagonists. Studies in patient cohorts of atypical Haemolytic-Uraemic Syndrome (aHUS), C3 Glomerulopathy (C3G), and IgA nephropathy (IgAN), have identified rare genetic variants that give rise to severe FH and FHRs dysfunctions, and are major genetic predisposing factors. These patients also have a higher frequency of a few polymorphisms whose relevance as disease risk factors is incompletely understood. In the last years, the availability of specific reagents has allowed a more precise quantitation of FH and FHRs in plasma samples from patients and controls. These studies have revealed that some aHUS, C3G or IgAN risk polymorphisms determine mild changes in FH or FHRs levels that could somehow perturb complement regulation and favour disease pathogenesis.

12.
Clin Kidney J ; 14(2): 707-709, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35355886

RESUMO

Dysregulation of the alternative complement pathway is a major pathogenic mechanism in two rare renal diseases: atypical haemolytic uraemic syndrome (aHUS) and membranoproliferative glomerulonephritis (MPGN). We report on a 66-year-old male with chronic hepatitis C virus (HCV) infection and a combined liver-kidney transplant that was diagnosed with MPGN at the age of 63 years and a 5-year-old boy who presented with aHUS at the age of 21 months following a Streptococcus pneumoniae infection. Both patients carried similar frameshift variants in the complement CFHR5 gene that segregate with reduced levels of factor H-related 5 (FHR-5). We conclude that low FHR-5 levels may predispose to viral and bacterial infections that then trigger different renal phenotypes.

13.
Front Immunol ; 12: 751093, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34721423

RESUMO

Pathogenic gain-of-function variants in complement Factor B were identified as causative of atypical Hemolytic Uremic syndrome (aHUS) in 2007. These mutations generate a reduction on the plasma levels of complement C3. A four-month-old boy was diagnosed with hypocomplementemic aHUS in May 2000, and he suffered seven recurrences during the following three years. He developed a severe hypertension which required 6 anti-hypertensive drugs and presented acrocyanosis and several confusional episodes. Plasma infusion or exchange, and immunosuppressive treatments did not improve the clinical evolution, and the patient developed end-stage renal disease at the age of 3 years. Hypertension and vascular symptoms persisted while he was on peritoneal dialysis or hemodialysis, as well as after bilateral nephrectomy. C3 levels remained low, while C4 levels were normal. In 2005, a heterozygous gain-of-function mutation in Factor B (K323E) was found. A combined liver and kidney transplantation (CLKT) was performed in March 2009, since there was not any therapy for complement inhibition in these patients. Kidney and liver functions normalized in the first two weeks, and the C3/C4 ratio immediately after transplantation, indicating that the C3 activation has been corrected. After remaining stable for 4 years, the patient suffered a B-cell non-Hodgkin lymphoma that was cured by chemotherapy and reduction of immunosuppressive drugs. Signs of liver rejection with cholangitis were observed a few months later, and a second liver graft was done 11 years after the CLKT. One year later, the patient maintains normal kidney and liver functions, also C3 and C4 levels are within the normal range. The 12-year follow-up of the patient reveals that, in spite of severe complications, CLKT was an acceptable therapeutic option for this aHUS patient.


Assuntos
Síndrome Hemolítico-Urêmica Atípica , Fator B do Complemento/genética , Transplante de Rim , Transplante de Fígado , Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Síndrome Hemolítico-Urêmica Atípica/genética , Síndrome Hemolítico-Urêmica Atípica/terapia , Mutação com Ganho de Função , Humanos , Lactente , Masculino
14.
Front Immunol ; 12: 641656, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33777036

RESUMO

Haemolytic Uraemic Syndrome associated with Streptococcus pneumoniae infections (SP-HUS) is a clinically well-known entity that generally affects infants, and could have a worse prognosis than HUS associated to E. coli infections. It has been assumed that complement genetic variants associated with primary atypical HUS cases (aHUS) do not contribute to SP-HUS, which is solely attributed to the action of the pneumococcal neuraminidase on the host cellular surfaces. We previously identified complement pathogenic variants and risk polymorphisms in a few Hungarian SP-HUS patients, and have now extended these studies to a cohort of 13 Spanish SP-HUS patients. Five patients presented rare complement variants of unknown significance, but the frequency of the risk haplotypes in the CFH-CFHR3-CFHR1 region was similar to the observed in aHUS. Moreover, we observed desialylation of Factor H (FH) and the FH-Related proteins in plasma samples from 2 Spanish and 4 Hungarian SP-HUS patients. To analyze the functional relevance of this finding, we compared the ability of native and "in vitro" desialylated FH in: (a) binding to C3b-coated microtiter plates; (b) proteolysis of fluid-phase and surface-bound C3b by Factor I; (c) dissociation of surface bound-C3bBb convertase; (d) haemolytic assays on sheep erythrocytes. We found that desialylated FH had reduced capacity to control complement activation on sheep erythrocytes, suggesting a role for FH sialic acids on binding to cellular surfaces. We conclude that aHUS-risk variants in the CFH-CFHR3-CFHR1 region could also contribute to disease-predisposition to SP-HUS, and that transient desialylation of complement FH by the pneumococcal neuraminidase may have a role in disease pathogenesis.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/genética , Proteínas Sanguíneas/genética , Proteínas Inativadoras do Complemento C3b/genética , Fator H do Complemento/genética , Infecções Pneumocócicas/complicações , Síndrome Hemolítico-Urêmica Atípica/microbiologia , Pré-Escolar , Feminino , Predisposição Genética para Doença/genética , Humanos , Lactente , Masculino , Polimorfismo Genético , Streptococcus pneumoniae
15.
Br J Haematol ; 150(5): 529-42, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20629662

RESUMO

Atypical Haemolytic Uraemic Syndrome (aHUS) is a thrombotic microangiopathy that often provokes irreversible renal damage and post-transplantation recurrence. Studies performed during the last decade have shown that 50-60% of aHUS patients present genetic or acquired defects in the complement system that enhance the initial endothelial damage and favour disease development. This review analyses the complement proteins and processes that are disturbed in aHUS patients, and outlines the relevance of a prompt genetic/molecular diagnosis for improving clinical management and prognosis.


Assuntos
Síndrome Hemolítico-Urêmica/etiologia , Via Alternativa do Complemento/genética , Via Alternativa do Complemento/imunologia , Predisposição Genética para Doença , Síndrome Hemolítico-Urêmica/genética , Síndrome Hemolítico-Urêmica/imunologia , Síndrome Hemolítico-Urêmica/terapia , Humanos , Transplante de Rim , Mutação
16.
Methods Mol Biol ; 2110: 73-81, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32002902

RESUMO

In pig-to-primate xenotransplantation, flow cytometry assays allow the examination of antibody reactivity to intact antigens in their natural conformation and location on cell membranes. Here we describe in detail the procedures of two flow cytometry assays to measure the antibody-mediated complement-dependent cytotoxicity (CDC) response or serum levels of IgG and IgM xenoantibodies. This information is key for understanding the rejection process of vascularized xenografts and finding strategies to overcome it.


Assuntos
Anticorpos Heterófilos/imunologia , Ativação do Complemento/imunologia , Citometria de Fluxo , Xenoenxertos/imunologia , Transplante Heterólogo , Animais , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Citotoxicidade Imunológica , Citometria de Fluxo/métodos , Rejeição de Enxerto/diagnóstico , Rejeição de Enxerto/imunologia , Imunoglobulina G/imunologia , Imunoglobulina M/imunologia , Primatas , Suínos , Transplante Heterólogo/efeitos adversos , Transplante Heterólogo/métodos
17.
Mol Immunol ; 45(10): 2897-904, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18336910

RESUMO

Age-related macular degeneration (AMD) and membranoproliferative glomerulonephritis type II (MPGN2) are dense deposit diseases that share a genetic association with complement genes and have complement proteins as important components of the dense deposits. Here, we present the case of a 64-year-old smoker male who developed both AMD and MPGN2 in his late 50s. The patient presented persistent low plasma levels of C3, factor H levels in the lower part of the normal range and C3NeF traces. Genetic analyses of the CFH, CFB, C3, CFHR1-CFHR3 and LOC387715/HTRA1 genes revealed that the patient was heterozygote for a novel missense mutation in exon 9 of CFH (c.1292 G>A) that results in a Cys431Tyr substitution in SCR7 of the factor H protein. In addition, he was homozygote for the His402 CFH allele, heterozygote for the Ser69 LOC387715 allele, homozygote for the Arg32 (BFS) CFB allele, heterozygote for the Gly102 (C3F) C3 allele and carried no deletion of the CFHR1/CFHR3 genes. Proteomic and functional analyses indicate absence in plasma of the factor H allele carrying the Cys431Tyr mutation. As a whole, these data recapitulate a prototypical complement genetic profile, including a partial factor H deficiency and the presence of major risk factors for AMD and MPGN2, which support the hypothesis that these dense deposit diseases have a common pathogenic mechanism involving dysregulation of the alternative pathway of complement activation.


Assuntos
Fator H do Complemento/genética , Glomerulonefrite Membranoproliferativa/complicações , Glomerulonefrite Membranoproliferativa/genética , Degeneração Macular/complicações , Degeneração Macular/genética , Substituição de Aminoácidos , Animais , Sequência de Bases , Western Blotting , Células COS , Chlorocebus aethiops , Fator H do Complemento/química , Cisteína , Olho/patologia , Predisposição Genética para Doença , Humanos , Rim/patologia , Espectrometria de Massas , Pessoa de Meia-Idade , Dados de Sequência Molecular , Mutação/genética , Proteínas Recombinantes , Tirosina
18.
Mol Immunol ; 45(10): 2764-71, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18374984

RESUMO

Complement Factor I (CFI) is a regulator of the classical and alternative pathways. CFI has enzymatic activity and is able to cleave C3b and C4b. Homozygous Factor I deficiency is associated with infectious and/or autoimmune diseases. Here we describe the biochemical and genetic characterization in two Spanish families with complete Factor I deficiency. In Family 1, the propositus suffered from several episodes of meningitis for more than a year. Biochemical complement studies showed undetectable Factor I levels in the propositus and in her sister, while their parents and a brother had partial Factor I deficiency and were healthy. In Family 2, three out of five children were homozygous for Factor I deficiency, two of whom suffered from meningitis and the third one from several infections. The parents and the other two siblings were healthy and heterozygous for Factor I deficiency. Molecular studies showed that the two families had different mutations at exon 5 of the Factor I gene, which codifies for module LDLr1. One mutation corresponds to a 772G>A change at the donor splice site that was originally found in a family from Northern England. The second is a new missense mutation 739T>G, that generates a Cys to Gly change.


Assuntos
Fator I do Complemento/deficiência , Fator I do Complemento/genética , População Branca/genética , Adolescente , Adulto , Sequência de Bases , Western Blotting , Criança , Pré-Escolar , Fator I do Complemento/química , Mapeamento de Sequências Contíguas , Análise Mutacional de DNA , DNA Complementar , Família , Feminino , Genoma Humano/genética , Humanos , Masculino , Dados de Sequência Molecular , Mutação/genética , Linhagem , Estrutura Terciária de Proteína , Espanha
19.
Front Immunol ; 10: 886, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31068950

RESUMO

Nephritic factors comprise a heterogeneous group of autoantibodies against neoepitopes generated in the C3 and C5 convertases of the complement system, causing its dysregulation. Classification of these autoantibodies can be clustered according to their stabilization of different convertases either from the classical or alternative pathway. The first nephritic factor described with the capacity to stabilize C3 convertase of the alternative pathway was C3 nephritic factor (C3NeF). Another nephritic factor has been characterized by the ability to stabilize C5 convertase of the alternative pathway (C5NeF). In addition, there are autoantibodies against assembled C3/C5 convertase of the classical and lectin pathways (C4NeF). These autoantibodies have been mainly associated with kidney diseases, like C3 glomerulopathy and immune complex-associated-membranoproliferative glomerulonephritis. Other clinical situations where these autoantibodies have been observed include infections and autoimmune disorders such as systemic lupus erythematosus and acquired partial lipodystrophy. C3 hypocomplementemia is a common finding in all patients with nephritic factors. The methods to measure nephritic factors are not standardized, technically complex, and lack of an appropriate quality control. This review will be focused in the description of the mechanism of action of the three known nephritic factors (C3NeF, C4NeF, and C5NeF), and their association with human diseases. Moreover, we present an overview regarding the diagnostic tools for its detection, and the main therapeutic approach for the patients with nephritic factors.


Assuntos
Autoanticorpos/imunologia , Proteínas do Sistema Complemento/imunologia , Suscetibilidade a Doenças , Epitopos/imunologia , Animais , Ativação do Complemento/imunologia , Fator Nefrítico do Complemento 3/imunologia , Convertases de Complemento C3-C5/imunologia , Complemento C3a/imunologia , Complemento C3a/metabolismo , Complemento C5a/imunologia , Complemento C5a/metabolismo , Gerenciamento Clínico , Glomerulonefrite/diagnóstico , Glomerulonefrite/etiologia , Glomerulonefrite/metabolismo , Glomerulonefrite/terapia , Humanos , Técnicas de Diagnóstico Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA