Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
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
2.
Contrib Nephrol ; 169: 337-350, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21252531

RESUMO

Thrombotic microangiopathies are a group of microvascular disorders, with reduced organ perfusion and hemolytic anemia. The two most relevant conditions characterized by thrombotic microangiopathic anemia (TMA) are thrombotic thrombocytopenic purpura (TTP) and hemolytic uremic syndrome (HUS). In TTP, systemic microvascular aggregation of platelets causes ischemia in the brain and other organs. In HUS, platelet-fibrin thrombi predominantly occlude the renal circulation. TTP can be inherited due to deficiencies in the activity of von Willebrand factor cleaving protease (ADAMTS13) or acquired due to the presence of autoantibodies directed against ADAMTS13. The majority of HUS cases are secondary to infections by strains of Escherichia coli that produce Shiga-like toxins (Stx-HUS), while about 5- 10% of all cases are classified as atypical HUS (aHUS). Genetically derived impaired regulation of the complement system is associated with aHUS. Infusion or the exchange of fresh frozen plasma have ameliorated the prognosis of TMA; however, no specific therapies aimed at preventing or limiting the microangiopathic process have been proven to affect the course of TMA. Large mammals, small animal models, knockout and transgenic mouse models of TTP and both Stx-HUS and aHUS have been developed and have provided outstanding contributions to nearly all areas of TMA research. A better understanding of the key clinical features of the diseases and of the importance of genetic and/or environmental factors involved in the pathogenesis of the diseases have been obtained. These animal models have also allowed the set up of protocols aimed at ameliorating the clinical approach to patients and for the development of new drugs and vaccines.


Assuntos
Modelos Animais de Doenças , Microangiopatias Trombóticas/etiologia , Microangiopatias Trombóticas/fisiopatologia , Proteínas ADAM/deficiência , Proteínas ADAM/genética , Proteína ADAMTS13 , Animais , Proteínas do Sistema Complemento/genética , Cães , Síndrome Hemolítico-Urêmica/etiologia , Síndrome Hemolítico-Urêmica/genética , Síndrome Hemolítico-Urêmica/fisiopatologia , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Plasma , Púrpura Trombocitopênica Trombótica/etiologia , Púrpura Trombocitopênica Trombótica/genética , Púrpura Trombocitopênica Trombótica/fisiopatologia , Coelhos , Toxinas Shiga/efeitos adversos , Microangiopatias Trombóticas/terapia
3.
Clin J Am Soc Nephrol ; 5(10): 1844-59, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20595690

RESUMO

BACKGROUND AND OBJECTIVES: Hemolytic uremic syndrome (HUS) is characterized by microangiopathic hemolytic anemia, thrombocytopenia, and renal impairment. Most childhood cases are caused by Shiga toxin-producing bacteria. The other form, atypical HUS (aHUS), accounts for 10% of cases and has a poor prognosis. Genetic complement abnormalities have been found in aHUS. DESIGN, SETTING, PARTICIPANTS, AND MEASUREMENTS: We screened 273 consecutive patients with aHUS for complement abnormalities and studied their role in predicting clinical phenotype and response to treatment. We compared mutation frequencies and localization and clinical outcome in familial (82) and sporadic (191) cases. RESULTS: In >70% of sporadic and familial cases, gene mutations, disease-associated factor H (CFH) polymorphisms, or anti-CFH autoantibodies were found. Either mutations or CFH polymorphisms were also found in the majority of patients with secondary aHUS, suggesting a genetic predisposition. Familial cases showed a higher prevalence of mutations in SCR20 of CFH and more severe disease than sporadic cases. Patients with CFH or THBD (thrombomodulin) mutations had the earliest onset and highest mortality. Membrane-cofactor protein (MCP) mutations were associated with the best prognosis. Plasma therapy induced remission in 55 to 80% of episodes in patients with CFH, C3, or THBD mutations or autoantibodies, whereas patients with CFI (factor I) mutations were poor responders. aHUS recurred frequently after kidney transplantation except for patients with MCP mutations. CONCLUSIONS: Results underline the need of genetic screening for all susceptibility factors as part of clinical management of aHUS and for identification of patients who could safely benefit from kidney transplant.


Assuntos
Proteínas do Sistema Complemento/genética , Síndrome Hemolítico-Urêmica/genética , Mutação , Polimorfismo Genético , Adolescente , Adulto , Idoso de 80 Anos ou mais , Autoanticorpos/sangue , Distribuição de Qui-Quadrado , Complemento C3/genética , Fator H do Complemento/genética , Proteínas do Sistema Complemento/imunologia , Análise Mutacional de DNA , Intervalo Livre de Doença , Feminino , Frequência do Gene , Predisposição Genética para Doença , Testes Genéticos , Síndrome Hemolítico-Urêmica/imunologia , Síndrome Hemolítico-Urêmica/mortalidade , Síndrome Hemolítico-Urêmica/terapia , Humanos , Lactente , Recém-Nascido , Estimativa de Kaplan-Meier , Transplante de Rim , Masculino , Proteína Cofatora de Membrana/genética , Linhagem , Fenótipo , Modelos de Riscos Proporcionais , Recidiva , Sistema de Registros , Medição de Risco , Fatores de Risco , Trombomodulina/genética , Fatores de Tempo , Resultado do Tratamento , Adulto Jovem
4.
Can J Physiol Pharmacol ; 86(8): 505-10, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18758497

RESUMO

Almost 50% of hypertensive individuals manifest blood pressure changes in response to salt depletion or repletion and are termed "salt sensitive" (SS). Blunted activity of the endothelin (ET) system and the renin-angiotensin-aldosterone system (RAAS) have been reported as possible mechanisms contributing to salt sensitivity. Data are available that endothelin receptor subtype B (ETBR)-deficient rats develop salt-sensitive hypertension when fed a high-salt diet. Whether the ETBR gene (EDNRB) is involved in genetic predisposition to human salt-sensitive hypertension has not been studied so far. We screened EDNRB in 104 hypertensive patients (49 salt sensitive and 55 salt resistant) and 110 normotensive controls. No new sequence variation was found, but genotype distribution of the common polymorphism G1065A revealed that the AA + GA genotypes were significantly more frequent in salt-resistant than in salt-sensitive individuals (p = 0.007), suggesting a protective role for the A allele. We also screened angiotensinogen gene AGT M235T and angiotensin-converting enzyme insertion/deletion polymorphism ACE I/D and found an association between TT genotype and hypertension. A possible synergistic effect to salt-sensitive hypertension was found by combining EDNRB GG with ACE DD/ID genotypes. In conclusion, our data confirm the role of ET system and RAAS in salt-sensitive hypertension.


Assuntos
Angiotensinogênio/genética , Hipertensão/induzido quimicamente , Hipertensão/genética , Peptidil Dipeptidase A/genética , Receptor de Endotelina B/genética , Cloreto de Sódio , Alelos , Primers do DNA , Éxons/genética , Genótipo , Humanos , Polimorfismo Genético/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Proc Natl Acad Sci U S A ; 105(7): 2538-43, 2008 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-18268355

RESUMO

Glomerulopathy with fibronectin (FN) deposits (GFND) is an autosomal dominant disease with age-related penetrance, characterized by proteinuria, microscopic hematuria, hypertension, and massive glomerular deposits of FN that lead to end-stage renal failure. The genetic abnormality underlying GFND was still unknown. We hypothesized that mutations in FN1, which encodes FN, were the cause of GFND. In a large Italian pedigree with eight affected subjects, we found linkage with GFND at the FN1 locus at 2q32. We sequenced the FN1 in 15 unrelated pedigrees and found three heterozygous missense mutations, the W1925R, L1974R, and Y973C, that cosegregated with the disease in six pedigrees. The mutations affected two domains of FN (Hep-II domain for the W1925R and the L1974R, and Hep-III domain for the Y973C) that play key roles in FN-cell interaction and in FN fibrillogenesis. Mutant recombinant Hep-II fragments were expressed, and functional studies revealed a lower binding to heparin and to endothelial cells and podocytes compared with wild-type Hep-II and an impaired capability to induce endothelial cell spreading and cytoskeletal reorganization. Overall dominant mutations in FN1 accounted for 40% of cases of GFND in our study group. These findings may help understanding the pathogenesis of proteinuria and glomerular FN deposits in GFND and possibly in more common renal diseases such as diabetic nephropathy, IgA nephropathy, and lupus nephritis. To our knowledge no FN1 mutation causing a human disease was previously reported.


Assuntos
Fibronectinas/genética , Fibronectinas/metabolismo , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Mutação/genética , Adolescente , Adulto , Criança , Feminino , Fibronectinas/química , Fibronectinas/classificação , Regulação da Expressão Gênica , Predisposição Genética para Doença , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Linhagem , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
8.
Blood ; 108(4): 1267-79, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16621965

RESUMO

Hemolytic uremic syndrome (HUS) is a thrombotic microangiopathy with manifestations of hemolytic anemia, thrombocytopenia, and renal impairment. Genetic studies have shown that mutations in complement regulatory proteins predispose to non-Shiga toxin-associated HUS (non-Stx-HUS). We undertook genetic analysis on membrane cofactor protein (MCP), complement factor H (CFH), and factor I (IF) in 156 patients with non-Stx-HUS. Fourteen, 11, and 5 new mutational events were found in MCP, CFH, and IF, respectively. Mutation frequencies were 12.8%, 30.1%, and 4.5% for MCP, CFH, and IF, respectively. MCP mutations resulted in either reduced protein expression or impaired C3b binding capability. MCP-mutated patients had a better prognosis than CFH-mutated and nonmutated patients. In MCP-mutated patients, plasma treatment did not impact the outcome significantly: remission was achieved in around 90% of both plasma-treated and plasma-untreated acute episodes. Kidney transplantation outcome was favorable in patients with MCP mutations, whereas the outcome was poor in patients with CFH and IF mutations due to disease recurrence. This study documents that the presentation, the response to therapy, and the outcome of the disease are influenced by the genotype. Hopefully this will translate into improved management and therapy of patients and will provide the way to design tailored treatments.


Assuntos
Fator H do Complemento/genética , Fator I do Complemento/genética , Síndrome Hemolítico-Urêmica/genética , Proteína Cofatora de Membrana/genética , Mutação , Transfusão de Componentes Sanguíneos , Complemento C3b/genética , Complemento C3b/metabolismo , Fator H do Complemento/biossíntese , Fator I do Complemento/biossíntese , Feminino , Frequência do Gene/genética , Genótipo , Síndrome Hemolítico-Urêmica/metabolismo , Síndrome Hemolítico-Urêmica/terapia , Humanos , Transplante de Rim , Masculino , Proteína Cofatora de Membrana/biossíntese , Plasma , Ligação Proteica/genética , Biossíntese de Proteínas/genética , Recidiva , Toxina Shiga , Resultado do Tratamento
9.
Curr Opin Crit Care ; 11(5): 487-92, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16175037

RESUMO

PURPOSE OF REVIEW: Recent studies have provided a better understanding of the molecular mechanisms responsible for hemolytic uremic syndromes. In this review, we summarize biochemical and genetic data that may lead to new clinical approaches. RECENT FINDINGS: The structures and modes of action of Shiga toxins have been deciphered. Patients with non-Shiga-like toxin hemolytic uremic syndrome have been found to carry mutations in three genes that encode for regulators of the complement system (factor H, membrane cofactor protein, and factor I). SUMMARY: Shiga-like toxin-1 and Shiga-like toxin-2 regulate genes that encode for chemokines, cytokines, cell adhesion molecules, and transcription factors involved in immune response and apoptosis. Mutations in factor H, membrane cofactor protein and factor I have recently been identified. Reduced expression of compliment regulators might prevent restriction of complement deposition on glomerular endothelial cells, leading to microvascular cell damage and tissue injury. Shiga-like toxin hemolytic uremic syndrome in children has a favorable prognosis in 90% of cases; kidney transplantation shows a good graft survival rate (80%) in children who progress to end stage renal disease. As for non-Shiga-like toxin hemolytic uremic syndrome, treatment with plasma infusion or exchange has been used with controversial effects. Kidney transplantation is not recommended in those patients with mutations in factor H and factor I; however, a kidney transplant corrects membrane cofactor protein dysfunction. These findings vividly underscore the clinical heterogeneity of outcomes depending upon the nature of the underlying cause of the disease.


Assuntos
Proteínas do Sistema Complemento/genética , Síndrome Hemolítico-Urêmica/genética , Mutação , Toxinas Shiga/genética , Fator H do Complemento/genética , Fibrinogênio/genética , Humanos , Proteína Cofatora de Membrana/genética
10.
J Am Soc Nephrol ; 16(5): 1177-83, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15800115

RESUMO

Thrombotic thrombocytopenic purpura is a rare disorder of small vessels that is associated with deficiency of the von Willebrand factor-cleaving protease ADAMTS13, which favors platelet adhesion and aggregation in the microcirculation. The disease manifests mainly with central nervous system symptoms, but cases of renal insufficiency have been reported. Presented are findings of the genetic basis of phenotype heterogeneity in thrombotic thrombocytopenic purpura in two sisters within one family. The patients had ADAMTS13 deficiency as a result of two heterozygous mutations (causing V88M and G1239V changes). In addition, a heterozygous mutation (causing an S890I change) in factor H of complement was found in the patient who developed chronic renal failure but not in her sister, who presented with exclusive neurologic symptoms.


Assuntos
Fator H do Complemento/genética , Falência Renal Crônica/genética , Metaloendopeptidases/genética , Mutação Puntual , Púrpura Trombocitopênica Trombótica/genética , Proteínas ADAM , Proteína ADAMTS13 , Feminino , Ligação Genética , Humanos , Falência Renal Crônica/complicações , Metaloendopeptidases/deficiência , Pessoa de Meia-Idade , Linhagem , Fenótipo , Púrpura Trombocitopênica Trombótica/complicações
11.
Lancet ; 362(9395): 1542-7, 2003 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-14615110

RESUMO

BACKGROUND: Mutations in factor H (HF1) have been reported in a consistent number of diarrhoea-negative, non-Shiga toxin-associated cases of haemolytic uraemic syndrome (D-HUS). However, most patients with D-HUS have no HF1 mutations, despite decreased serum concentrations of C3. Our aim, therefore, was to assess whether genetic abnormalities in other complement regulatory proteins are involved. METHODS: We screened genes that encode the complement regulatory proteins-ie, factor H related 5, complement receptor 1, and membrane cofactor protein (MCP)-by PCR-single-strand conformation polymorphism (PCR-SSCP) and by direct sequencing, in 25 consecutive patients with D-HUS, an abnormal complement profile, and no HF1 mutation, from our International Registry of Recurrent and Familial HUS/TTP (HUS/thrombotic thrombocytopenic purpura). FINDINGS: We identified a heterozygous mutation in MCP, a surface-bound complement regulator, in two patients with a familial history of HUS. The mutation causes a change in three aminoacids at position 233-35 and insertion of a premature stop-codon, which results in loss of the transmembrane domain of the protein and severely reduced cell-surface expression of MCP. INTERPRETATION: Results of previous studies on HF1 indicate an association between HF1 deficiency and D-HUS. Our findings of an MCP mutation in two related patients suggest that impaired regulation of complement activation might be a factor in the pathogenesis of genetic forms of HUS. MCP could be a second putative candidate gene for D-HUS. The protein is highly expressed in the kidney and plays a major part in regulation of glomerular C3 activation. We propose, therefore, that reduced expression of MCP in response to complement-activating stimuli could prevent restriction of complement deposition on glomerular endothelial cells, leading to microvascular cell damage and tissue injury.


Assuntos
Antígenos CD/genética , Proteínas Inativadoras do Complemento/genética , Proteínas do Sistema Complemento/genética , Síndrome Hemolítico-Urêmica/genética , Glicoproteínas de Membrana/genética , Mutação , Adolescente , Adulto , Idade de Início , Fator H do Complemento/genética , Proteínas do Sistema Complemento/fisiologia , Feminino , Síndrome Hemolítico-Urêmica/epidemiologia , Síndrome Hemolítico-Urêmica/fisiopatologia , Humanos , Glomérulos Renais/fisiopatologia , Masculino , Proteína Cofatora de Membrana , Linhagem , Reação em Cadeia da Polimerase/métodos , Polimorfismo Conformacional de Fita Simples , Receptores de Complemento/genética
12.
Hum Mol Genet ; 12(24): 3385-95, 2003 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-14583443

RESUMO

Mutations in complement factor H (HF1) gene have been reported in non-Shiga toxin-associated and diarrhoea-negative haemolytic uraemic syndrome (D-HUS). We analysed the complete HF1 in 101 patients with HUS, in 32 with thrombotic thrombocytopenic purpura (TTP) and in 106 controls to evaluate the frequency of HF1 mutations, the clinical outcome in mutation and non-mutation carriers and the role of HF1 polymorphisms in the predisposition to HUS. We found 17 HF1 mutations (16 heterozygous, one homozygous) in 33 HUS patients. Thirteen mutations were located in exons XXII and XXIII. No TTP patient carried HF1 mutations. The disease manifested earlier and the mortality rate was higher in mutation carriers than in non-carriers. Kidney transplants invariably failed for disease recurrences in patients with HF1 mutations, while in non-mutated patients half of the grafts were functioning after 1 year. Three HF1 polymorphic variants were strongly associated with D-HUS: -257T (promoter region), 2089G (exonXIV, silent) and 2881T (963Asp, SCR16). The association was stronger in patients without HF1 mutations. Two or three disease-associated variants led to a higher risk of HUS than a single one. Analysis of available relatives of mutated patients revealed a penetrance of 50%. In 5/9 families the proband inherited the mutation from one parent and two disease-associated variants from the other, while unaffected carriers inherited the protective variants. In conclusion HF1 mutations are frequent in patients with D-HUS (24%). Common polymorphisms of HF1 may contribute to D-HUS manifestation in subjects with and without HF1 mutations.


Assuntos
Fator H do Complemento/genética , Síndrome Hemolítico-Urêmica/genética , Mutação , Polimorfismo Genético , Púrpura Trombocitopênica Trombótica/genética , Sequência de Aminoácidos , Sequência de Bases , Análise Mutacional de DNA , Feminino , Frequência do Gene , Predisposição Genética para Doença , Humanos , Masculino , Modelos Moleculares , Dados de Sequência Molecular , Linhagem , Polimorfismo Conformacional de Fita Simples
13.
J Clin Invest ; 111(8): 1181-90, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12697737

RESUMO

Hemolytic uremic syndrome (HUS) is a disease characterized by microangiopathic hemolytic anemia, thrombocytopenia, and acute renal failure. Recent studies have identified a factor H-associated form of HUS, caused by gene mutations that cluster in the C-terminal region of the complement regulator factor H. Here we report how three mutations (E1172Stop, R1210C, and R1215G; each of the latter two identified in three independent cases from different, unrelated families) affect protein function. All three mutations cause reduced binding to the central complement component C3b/C3d to heparin, as well as to endothelial cells. These defective features of the mutant factor H proteins explain progression of endothelial cell and microvascular damage in factor H-associated genetic HUS and indicate a protective role of factor H for tissue integrity during thrombus formation.


Assuntos
Complemento C3b/metabolismo , Fator H do Complemento/genética , Endotélio Vascular/metabolismo , Síndrome Hemolítico-Urêmica/genética , Heparina/metabolismo , Mutação , Animais , Fator H do Complemento/metabolismo , Citometria de Fluxo , Imunofluorescência , Humanos , Isoformas de Proteínas , Proteínas Recombinantes/metabolismo
15.
Clin Biochem ; 35(5): 363-8, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12270765

RESUMO

OBJECTIVES: Conflicting results on the relationship between M235T polymorphism of angiotensinogen (AGT) gene and diabetic nephropathy are reported in the literature, probably due to the small number of subjects, to different inclusion criteria and the different genotype analysis methods used. The aim of the present study was to set up a fast, cheap and reliable method to allow the genotyping of M235T polymorphism in a large number of subjects. DESIGN AND METHODS: We developed in our laboratory a new specifically designed PCR-SSCP method for M235T genotyping whose specificity was compared with that of Allele Specific PCR (ASPCR) and Mutagenically Separated PCR (MS-PCR). The exact M235T genotype was estabilished by direct sequencing. The new PCR-SSCP method was then used to genotype a population of 1171 hypertensive, normoalbuminuric type II diabetes mellitus patients. The patients were also genotyped for ACE I/D polymorphism. For comparison a group of hypertensive non diabetic patients (n = 88) were also screened. RESULTS: The PCR-SSCP method identified the M235T polymorphism with no misinterpretation at variance with ASPCR and MS-PCR methods that showed a preferential amplification of the T allele. The rare Y248C polymorphism of the AGT gene was also detected by PCR-SSCP. In diabetic hypertensive patients the prevalence of TT genotype was higher than in normotensive healthy controls and equivalent to that found in hypertensive non diabetic patients. CONCLUSIONS: The PCR-SSCP method for detection of M235T polymorphism is a powerful and sensitive tool for rapid, cheap and efficient screening of a large number of samples. The results obtained with this method demonstrate an association of the TT genotype of AGT gene with hypertension, both in diabetic and non diabetic patients.


Assuntos
Angiotensinogênio/genética , Testes Genéticos/métodos , Polimorfismo Conformacional de Fita Simples , Adulto , Idoso , Alelos , Sequência de Bases , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/genética , Nefropatias Diabéticas/genética , Feminino , Genótipo , Humanos , Hipertensão/complicações , Hipertensão/genética , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Reprodutibilidade dos Testes , Tamanho da Amostra , Sensibilidade e Especificidade , Fatores de Tempo
16.
Blood ; 100(3): 778-85, 2002 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12130486

RESUMO

Whether measurement of ADAMTS13 activity may enable physicians to distinguish thrombotic thrombocytopenic purpura (TTP) from hemolytic uremic syndrome (HUS) is still a controversial issue. Our aim was to clarify whether patients with normal or deficient ADAMTS13 activity could be distinguished in terms of disease manifestations and multimeric patterns of plasma von Willebrand factor (VWF). ADAMTS13 activity, VWF antigen, and multimeric pattern were evaluated in patients with recurrent and familial TTP (n = 20) and HUS (n = 29). Results of the collagen-binding assay of ADAMTS13 activity were confirmed in selected samples by testing the capacity of plasma to cleave recombinant VWF A1-A2-A3. Most patients with TTP had complete or partial deficiency of ADAMTS13 activity during the acute phase, and in some the defect persisted at remission. However, complete ADAMTS13 deficiency was also found in 5 of 9 patients with HUS during the acute phase and in 5 patients during remission. HUS patients with ADAMTS13 deficiency could not be distinguished clinically from those with normal ADAMTS13. In a subgroup of patients with TTP or HUS, the ADAMTS13 defect was inherited, as documented by half-normal levels of ADAMTS13 in their asymptomatic parents, consistent with the heterozygous carrier state. In patients with TTP and HUS there was indirect evidence of increased VWF fragmentation, and this occurred also in patients with ADAMTS13 deficiency. In conclusion, deficient ADAMTS13 activity does not distinguish TTP from HUS, at least in the recurrent and familial forms, and it is not the only determinant of VWF abnormalities in these conditions.


Assuntos
Síndrome Hemolítico-Urêmica/enzimologia , Metaloendopeptidases/deficiência , Púrpura Trombocitopênica Trombótica/enzimologia , Proteínas ADAM , Proteína ADAMTS13 , Adolescente , Adulto , Diagnóstico Diferencial , Dimerização , Saúde da Família , Feminino , Síndrome Hemolítico-Urêmica/diagnóstico , Humanos , Itália/epidemiologia , Masculino , Metaloendopeptidases/metabolismo , Pessoa de Meia-Idade , Púrpura Trombocitopênica Trombótica/diagnóstico , Recidiva , Fator de von Willebrand/metabolismo
17.
Lancet ; 359(9318): 1671-2, 2002 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-12020532

RESUMO

Recurrent haemolytic uraemic syndrome (HUS) is a genetic form of thrombotic microangiopathy that is mostly associated with low activity of complement factor H. The disorder usually develops in families, leads to end stage renal disease, and invariably recurs after kidney transplantation. We did a simultaneous kidney and liver transplantation in a 2-year-old child with HUS and a mutation in complement factor H to restore the defective factor H, with no recurrence of the disease. The operation was successful, and at discharge, the child had healthy kidney and liver function, with no sign of haemolysis.


Assuntos
Síndrome Hemolítico-Urêmica/genética , Síndrome Hemolítico-Urêmica/cirurgia , Transplante de Rim , Transplante de Fígado , Humanos , Lactente , Masculino , Mutação Puntual
18.
J Am Soc Nephrol ; 12(2): 297-307, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11158219

RESUMO

The aim of the present study was to clarify whether factor H mutations were involved in genetic predisposition to hemolytic uremic syndrome, by performing linkage and mutation studies in a large number of patients from those referred to the Italian Registry for Recurrent and Familial HUS/TTP. PCR and Western blot analyses were conducted to characterize the biochemical consequences of the mutations. Five mutations in the factor H gene were identified. Three, identified in two families and in a sporadic case, are heterozygous point mutations within the most C-terminal short consensus repeat 20 (SCR20) of factor H, resulting in single amino acid substitutions. The other two mutations introduce premature stop codons that interrupt the translation of factor H. A heterozygous nonsense mutation was identified in SCR8 in one family, and a homozygous 24-bp deletion within SCR20 was identified in a Bedouin family with a recessive mode of inheritance. Reverse transcription-PCR analysis of cDNA from peripheral blood leukocytes from the Bedouin family showed that the deletion lowered factor H mRNA levels. Although heterozygous mutations were associated with normal factor H levels and incomplete penetrance of the disease, the homozygous mutation in the Bedouin family resulted in severe reduction of factor H levels accompanied by very early disease onset. These data provide compelling molecular evidence that genetically determined deficiencies in factor H are involved in both autosomal-dominant and autosomal-recessive hemolytic uremic syndrome and identify SCR20 as a hot spot for mutations in the disease. The mutations identified here give an important hint to the relevance of the C-terminus of factor H in the control of the alternative complement activation pathway.


Assuntos
Fator H do Complemento/genética , Síndrome Hemolítico-Urêmica/genética , Mutação , Sequências Repetitivas de Aminoácidos , Sequência de Aminoácidos , Sequência de Bases , Fator H do Complemento/química , Predisposição Genética para Doença , Humanos , Dados de Sequência Molecular , Linhagem , Polimorfismo Conformacional de Fita Simples
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...