Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Kidney Int Rep ; 9(2): 464-477, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38344720

RESUMO

Introduction: C3 glomerulopathy (C3G) is an ultrarare renal disease characterized by deposition of complement component C3 in the glomerular basement membrane (GBM). Rare and novel genetic variation in complement genes and autoantibodies to complement proteins are commonly identified in the C3G population and thought to drive the underlying complement dysregulation that results in renal damage. However, disease heterogeneity and rarity make accurately defining characteristics of the C3G population difficult. Methods: Here, we present a retrospective analysis of the Molecular Otolaryngology and Renal Research Laboratories C3G cohort. This study integrated complement biomarker testing and in vitro tests of autoantibody function to achieve the following 3 primary goals: (i) define disease profiles of C3G based on disease drivers, complement biomarkers, and age; (ii) determine the relationship between in vitro autoantibody tests and in vivo complement dysregulation; and (iii) evaluate the association between autoantibody function and disease progression. Results: The largest disease profiles of C3G included patients with autoantibodies to complement proteins (48%) and patients for whom no genetic and/or acquired drivers of disease could be identified (43%). The correlation between the stabilization of convertases by complement autoantibodies as measured by in vitro modified hemolytic assays and systemic biomarkers that reflect in vivo complement dysregulation was remarkably strong. In patients positive for autoantibodies, the degree of stabilization capacity predicted worse renal function. Conclusion: This study implicates complement autoantibodies as robust drivers of systemic complement dysregulation in approximately 50% of C3G but also highlights the need for continued discovery-based research to identify novel drivers of disease.

2.
Front Immunol ; 13: 866330, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35619721

RESUMO

C3 glomerulopathy (C3G) and atypical hemolytic uremic syndrome (aHUS) are two rare diseases caused by dysregulated activity of the alternative pathway of complement secondary to the presence of genetic and/or acquired factors. Complement factor I (FI) is a serine protease that downregulates complement activity in the fluid phase and/or on cell surfaces in conjunction with one of its cofactors, factor H (FH), complement receptor 1 (CR1/CD35), C4 binding protein (C4BP) or membrane cofactor protein (MCP/CD46). Because altered FI activity is causally related to the pathogenesis of C3G and aHUS, we sought to test functional activity of select CFI missense variants in these two patient cohorts. We identified 65 patients (16, C3G; 48, aHUS; 1 with both) with at least one rare variant in CFI (defined as a MAF < 0.1%). Eight C3G and eleven aHUS patients also carried rare variants in either another complement gene, ADAMTS13 or THBD. We performed comprehensive complement analyses including biomarker profiling, pathway activity and autoantibody testing, and developed a novel FI functional assay, which we completed on 40 patients. Seventy-eight percent of rare CFI variants (31/40) were associated with FI protein levels below the 25th percentile; in 22 cases, FI levels were below the lower limit of normal (type 1 variants). Of the remaining nine variants, which associated with normal FI levels, two variants reduced FI activity (type 2 variants). No patients carried currently known autoantibodies (including FH autoantibodies and nephritic factors). We noted that while rare variants in CFI predispose to complement-mediated diseases, phenotypes are strongly contingent on the associated genetic background. As a general rule, in isolation, a rare CFI variant most frequently leads to aHUS, with the co-inheritance of a CD46 loss-of-function variant driving the onset of aHUS to the younger age group. In comparison, co-inheritance of a gain-of-function variant in C3 alters the phenotype to C3G. Defects in CFH (variants or fusion genes) are seen with both C3G and aHUS. This variability underscores the complexity and multifactorial nature of these two complement-mediated renal diseases.


Assuntos
Síndrome Hemolítico-Urêmica Atípica , Fator I do Complemento , Síndrome Hemolítico-Urêmica Atípica/genética , Autoanticorpos/genética , Fator I do Complemento/genética , Proteínas do Sistema Complemento/genética , Proteínas do Sistema Complemento/metabolismo , Humanos , Fenótipo
3.
Front Immunol ; 13: 1073802, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36846022

RESUMO

Introduction: C3 glomerulopathies (C3G) are ultra-rare complement-mediated diseases that lead to end-stage renal disease (ESRD) within 10 years of diagnosis in ~50% of patients. Overactivation of the alternative pathway (AP) of complement in the fluid phase and on the surface of the glomerular endothelial glycomatrix is the underlying cause of C3G. Although there are animal models for C3G that focus on genetic drivers of disease, in vivo studies of the impact of acquired drivers are not yet possible. Methods: Here we present an in vitro model of AP activation and regulation on a glycomatrix surface. We use an extracellular matrix substitute (MaxGel) as a base upon which we reconstitute AP C3 convertase. We validated this method using properdin and Factor H (FH) and then assessed the effects of genetic and acquired drivers of C3G on C3 convertase. Results: We show that C3 convertase readily forms on MaxGel and that this formation was positively regulated by properdin and negatively regulated by FH. Additionally, Factor B (FB) and FH mutants impaired complement regulation when compared to wild type counterparts. We also show the effects of C3 nephritic factors (C3Nefs) on convertase stability over time and provide evidence for a novel mechanism of C3Nef-mediated C3G pathogenesis. Discussion: We conclude that this ECM-based model of C3G offers a replicable method by which to evaluate the variable activity of the complement system in C3G, thereby offering an improved understanding of the different factors driving this disease process.


Assuntos
Complemento C3 , Nefropatias , Animais , Complemento C3/genética , Complemento C3/metabolismo , Via Alternativa do Complemento/genética , Properdina/genética , Properdina/metabolismo , Convertases de Complemento C3-C5/metabolismo , Fator Nefrítico do Complemento 3/metabolismo , Matriz Extracelular/metabolismo
4.
Front Immunol ; 11: 607211, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33384694

RESUMO

Factor H (FH), a member of the regulators-of-complement-activation (RCA) family of proteins, circulates in human plasma at concentrations of 180-420 mg/L where it controls the alternative pathway (AP) of complement in the fluid phase and on cell surfaces. When the regulatory function of FH is impaired, complement-mediated tissue injury and inflammation occur, leading to diseases such as atypical hemolytic uremic syndrome (a thrombotic microangiopathy or TMA), C3 glomerulopathy (C3G) and monoclonal gammopathy of renal significance (MGRS). A pathophysiological cause of compromised FH function is the development of autoantibodies to various domains of the FH protein. FH autoantibodies (FHAAs) are identified in 10.9% of patients with aHUS, 3.2% of patients with C3G, and rarely in patients with MGRS. The phenotypic variability of FHAA-mediated disease reflects both the complexity of FH and the epitope specificity of FHAA for select regions of the native protein. In this paper, we have characterized FHAA epitopes in a large cohort of patients diagnosed with TMA, C3G or MGRS. We explore the epitopes recognized by FHAAs in these diseases and the association of FHAAs with the genetic deletion of both copies of the CFHR1 gene to show how these disease phenotypes are associated with this diverse spectrum of autoantibodies.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/imunologia , Autoanticorpos/sangue , Glomerulonefrite/imunologia , Paraproteinemias/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Síndrome Hemolítico-Urêmica Atípica/sangue , Síndrome Hemolítico-Urêmica Atípica/epidemiologia , Síndrome Hemolítico-Urêmica Atípica/genética , Biomarcadores/sangue , Criança , Pré-Escolar , Proteínas Inativadoras do Complemento C3b/genética , Fator H do Complemento/imunologia , Epitopos , Feminino , Deleção de Genes , Predisposição Genética para Doença , Glomerulonefrite/sangue , Glomerulonefrite/epidemiologia , Glomerulonefrite/genética , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Paraproteinemias/sangue , Paraproteinemias/epidemiologia , Paraproteinemias/genética , Fenótipo , Prevalência , Estudos Retrospectivos , Estados Unidos/epidemiologia , Adulto Jovem
5.
J Am Soc Nephrol ; 29(12): 2809-2819, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30377230

RESUMO

BACKGROUND: Genetic variation in complement genes is a predisposing factor for atypical hemolytic uremic syndrome (aHUS), a life-threatening thrombotic microangiopathy, however interpreting the effects of genetic variants is challenging and often ambiguous. METHODS: We analyzed 93 complement and coagulation genes in 400 patients with aHUS, using as controls 600 healthy individuals from Iowa and 63,345 non-Finnish European individuals from the Genome Aggregation Database. After adjusting for population stratification, we then applied the Fisher exact, modified Poisson exact, and optimal unified sequence kernel association tests to assess gene-based variant burden. We also applied a sliding-window analysis to define the frequency range over which variant burden was significant. RESULTS: We found that patients with aHUS are enriched for ultrarare coding variants in the CFH, C3, CD46, CFI, DGKE, and VTN genes. The majority of the significance is contributed by variants with a minor allele frequency of <0.1%. Disease-related variants tend to occur in specific complement protein domains of FH, CD46, and C3. We observed no enrichment for multiple rare coding variants in gene-gene combinations. CONCLUSIONS: In known aHUS-associated genes, variants with a minor allele frequency >0.1% should not be considered pathogenic unless valid enrichment and/or functional evidence are available. VTN, which encodes vitronectin, an inhibitor of the terminal complement pathway, is implicated as a novel aHUS-associated gene. Patients with aHUS are not enriched for multiple rare variants in complement genes. In aggregate, these data may help in directing clinical management of aHUS.


Assuntos
Síndrome Hemolítico-Urêmica Atípica/genética , Adolescente , Adulto , Idoso , Síndrome Hemolítico-Urêmica Atípica/sangue , Fatores de Coagulação Sanguínea/genética , Criança , Pré-Escolar , Proteínas do Sistema Complemento/genética , Bases de Dados Genéticas , Feminino , Frequência do Gene , Predisposição Genética para Doença , Testes Genéticos , Variação Genética , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Mutação , Vitronectina/genética , Adulto Jovem
6.
Adv Med Educ Pract ; 9: 85-91, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29445308

RESUMO

INTRODUCTION: There are over a dozen medical shows airing on television, many of which are during prime time. Researchers have recently become more interested in the role of these shows, and the awareness on cardiopulmonary resuscitation. Several cases have been reported where a lay person resuscitated a family member using medical TV shows as a reference. The purpose of this study is to examine and evaluate college students' perception on cardiopulmonary resuscitation and when to shock using an automated external defibrillator based on their experience of watching medical TV shows. METHODS: A total of 170 students (nonmedical major) were surveyed in four different colleges in the United States. The survey consisted of questions that reflect their perception and knowledge acquired from watching medical TV shows. A stepwise regression was used to determine the significant predictors of "How often do you watch medical drama TV shows" in addition to chi-square analysis for nominal variables. RESULTS: Regression model showed significant effect that TV shows did change students' perception positively (p<0.001), and they would select shock on asystole as the frequency of watching increases (p=0.023). CONCLUSION: The findings of this study show that high percentage of nonmedical college students are influenced significantly by medical shows. One particular influence is the false belief about when a shock using the automated external defibrillator (AED) is appropriate as it is portrayed falsely in most medical shows. This finding raises a concern about how these shows portray basic life support, especially when not following American Heart Association (AHA) guidelines. We recommend the medical advisors in these shows to use AHA guidelines and AHA to expand its expenditures to include medical shows to educate the public on the appropriate action to rescue an out-of-hospital cardiac arrest patient.

7.
Nephrol Dial Transplant ; 33(12): 2260-2265, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29370420

RESUMO

Background: C3 glomerulonephritis (C3GN) is caused by alternate complement pathway over-activation. It frequently progresses to end-stage renal disease, recurs in two-thirds of transplants and in half of these cases progresses to allograft loss. There is currently no proven treatment for C3GN. Case Presentation: We describe a family segregating pathogenic alleles of complement factor H and I (CFH and CFI). The only member carrying both mutations developed C3GN. Prolonged delayed graft function after deceased donor transplantation, heavy proteinuria and isolated C3 hypocomplementemia prompted an allograft biopsy confirming diagnosis of recurrent C3GN. Discussion: This is the first report of early recurrence of C3GN in an allograft in a patient with known mutations in complement regulatory genes and no preexisting para-proteinemia. Complement activation resulting from ischemia-reperfusion injury from prolonged cold ischemia time unabated in the setting of deficiency of two major complement regulators likely led to the early and severe recurrence. In atypical hemolytic uremic syndrome, the terminal complement cascade activation in the sentinel event initiating endothelial injury; blockade at the level of C5 convertase with eculizumab is uniformly highly effective in management. C3 glomerulopathies (C3GN and dense deposit disease) are a more complex and heterogeneous group. The relative degree of dysregulation at the levels of C3 and C5 convertases and therefore response to eculizumab varies among patients. In our patient, the clinical response to eculizumab was dramatic with recovery of allograft function and complete resolution of proteinuria. We review all cases of recurrent C3 glomerulopathy treated with eculizumab and discuss how complement biomarkers may aid in predicting response to therapy.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Fator H do Complemento/genética , Fibrinogênio/genética , Glomerulonefrite/genética , Transplante de Rim/efeitos adversos , Mutação , Doadores de Tecidos , Aloenxertos , Biomarcadores/metabolismo , Biópsia , Ativação do Complemento , Fator H do Complemento/metabolismo , Via Alternativa do Complemento , DNA/genética , Análise Mutacional de DNA , Fibrinogênio/metabolismo , Glomerulonefrite/tratamento farmacológico , Glomerulonefrite/metabolismo , Humanos , Falência Renal Crônica/cirurgia , Glomérulos Renais/patologia , Masculino , Pessoa de Meia-Idade , Linhagem , Recidiva
8.
Am J Kidney Dis ; 70(6): 834-843, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28838767

RESUMO

BACKGROUND: C3 glomerulopathy (C3G) defines a group of rare complement-mediated kidney diseases with a shared underlying pathophysiology: dysregulation of complement in the fluid phase and glomerular microenvironment. Dysregulation can be driven by autoantibodies to C3 and C5 convertases. STUDY DESIGN: Case series. SETTING & PARTICIPANTS: 168 patients with C3G (dense deposit disease, 68; C3 glumerulonephritis, 100) selected from our C3G biobank. OUTCOMES: Patient-purified immunoglobulin Gs were tested for C4 nephritic factors (C4NeFs). These autoantibodies recognize C4b2a, the C3 convertase of the classical pathway of complement. MEASUREMENTS: C4NeFs were detected using a modified hemolytic assay. RESULTS: C4NeFs were identified in 5 patients, 4 of whom had C3 glomerulonephritis. C4NeFs were associated with dysregulation of C3 and C5 convertases, and they appear to stabilize these convertases in a dose-dependent manner. C4NeFs also appear to protect C4b2a from decay mediated by soluble CR1 and C4 binding protein. The stabilizing activity of the autoantibodies was further demonstrated by using heat treatment to inactivate complement. C4NeFs were not detected in 150 patients with another complement-mediated kidney disease, atypical hemolytic uremic syndrome. They were also absent in 300 apparently healthy controls. LIMITATIONS: In addition to C4NeFs, 2 patients had positive findings for other autoantibodies: one patient also had autoantibodies to factor H; the other patient also had autoantibodies to C3bBb (C3NeFs). CONCLUSIONS: The finding of C4NeFs in a small percentage of patients with C3G highlights the challenge in identifying autoantibodies that drive complement dysregulation and underscores the complexity of the autoantibody repertoire that can be identified in these patients.


Assuntos
Autoanticorpos/imunologia , Complemento C3/imunologia , Proteínas do Sistema Complemento/imunologia , Glomerulonefrite Membranoproliferativa/imunologia , Adolescente , Adulto , Criança , C3 Convertase da Via Alternativa do Complemento/imunologia , Fator Nefrítico do Complemento 3/imunologia , C5 Convertase da Via Clássica do Complemento , Fator H do Complemento/imunologia , Feminino , Humanos , Nefropatias/imunologia , Masculino , Adulto Jovem
9.
J Am Soc Nephrol ; 27(4): 1245-53, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26283675

RESUMO

The thrombotic microangiopathies (TMAs) and C3 glomerulopathies (C3Gs) include a spectrum of rare diseases such as atypical hemolytic uremic syndrome, thrombotic thrombocytopenic purpura, C3GN, and dense deposit disease, which share phenotypic similarities and underlying genetic commonalities. Variants in several genes contribute to the pathogenesis of these diseases, and identification of these variants may inform the diagnosis and treatment of affected patients. We have developed and validated a comprehensive genetic panel that screens all exons of all genes implicated in TMA and C3G. The closely integrated pipeline implemented includes targeted genomic enrichment, massively parallel sequencing, bioinformatic analysis, and a multidisciplinary conference to analyze identified variants in the context of each patient's specific phenotype. Herein, we present our 1-year experience with this panel, during which time we studied 193 patients. We identified 17 novel and 74 rare variants, which we classified as pathogenic (11), likely pathogenic (12), and of uncertain significance (68). Compared with controls, patients with C3G had a higher frequency of rare and novel variants in C3 convertase (C3 and CFB) and complement regulator (CFH, CFI, CFHR5, and CD46) genes (P<0.05). In contrast, patients with TMA had an increase in rare and novel variants only in complement regulator genes (P<0.01), a distinction consistent with differing sites of complement dysregulation in these two diseases. In summary, we were able to provide a positive genetic diagnosis in 43% and 41% of patients carrying the clinical diagnosis of C3G and TMA, respectively.


Assuntos
Nefropatias/diagnóstico , Nefropatias/genética , Glomérulos Renais , Microangiopatias Trombóticas/diagnóstico , Microangiopatias Trombóticas/genética , Adolescente , Criança , Pré-Escolar , Complemento C3 , Feminino , Testes Genéticos/métodos , Humanos , Nefropatias/imunologia , Masculino
10.
J Med Genet ; 52(12): 823-9, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26445815

RESUMO

BACKGROUND: Countries with culturally accepted consanguinity provide a unique resource for the study of rare recessively inherited genetic diseases. Although hereditary hearing loss (HHL) is not uncommon, it is genetically heterogeneous, with over 85 genes causally implicated in non-syndromic hearing loss (NSHL). This heterogeneity makes many gene-specific types of NSHL exceedingly rare. We sought to define the spectrum of autosomal recessive HHL in Iran by investigating both common and rarely diagnosed deafness-causing genes. DESIGN: Using a custom targeted genomic enrichment (TGE) panel, we simultaneously interrogated all known genetic causes of NSHL in a cohort of 302 GJB2-negative Iranian families. RESULTS: We established a genetic diagnosis for 67% of probands and their families, with over half of all diagnoses attributable to variants in five genes: SLC26A4, MYO15A, MYO7A, CDH23 and PCDH15. As a reflection of the power of consanguinity mapping, 26 genes were identified as causative for NSHL in the Iranian population for the first time. In total, 179 deafness-causing variants were identified in 40 genes in 201 probands, including 110 novel single nucleotide or small insertion-deletion variants and three novel CNV. Several variants represent founder mutations. CONCLUSION: This study attests to the power of TGE and massively parallel sequencing as a diagnostic tool for the evaluation of hearing loss in Iran, and expands on our understanding of the genetics of HHL in this country. Families negative for variants in the genes represented on this panel represent an excellent cohort for novel gene discovery.


Assuntos
Perda Auditiva/genética , Conexina 26 , Conexinas , Consanguinidade , Efeito Fundador , Frequência do Gene , Genes Recessivos , Estudos de Associação Genética , Predisposição Genética para Doença , Perda Auditiva/patologia , Humanos , Irã (Geográfico)
11.
Am J Med Genet A ; 167A(12): 2957-65, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26416264

RESUMO

Deafness is the most frequent sensory disorder. With over 90 genes and 110 loci causally implicated in non-syndromic hearing loss, it is phenotypically and genetically heterogeneous. Here, we investigate the genetic etiology of deafness in four families of Iranian origin segregating autosomal recessive non-syndromic hearing loss (ARNSHL). We used a combination of linkage analysis, homozygosity mapping, and a targeted genomic enrichment platform to simultaneously screen 90 known deafness-causing genes for pathogenic variants. Variant segregation was confirmed by Sanger sequencing. Linkage analysis and homozygosity mapping showed segregation with the DFNB57 locus on chromosome 10 in two families. Targeted genomic enrichment with massively parallel sequencing identified causal variants in PDZD7: a homozygous missense variant (p.Gly103Arg) in one family and compound heterozygosity for missense (p.Met285Arg) and nonsense (p.Tyr500Ter) variants in the second family. Screening of two additional families identified two more variants: (p.Gly228Arg) and (p.Gln526Ter). Variant segregation with the hearing loss phenotype was confirmed in all families by Sanger sequencing. The missense variants are predicted to be deleterious, and the two nonsense mutations produce null alleles. This report is the first to show that mutations in PDZD7 cause ARNSHL, a finding that offers addition insight into the USH2 interactome. We also describe a novel likely disease-causing mutation in CIB2 and illustrate the complexity associated with gene identification in diseases that exhibit large genetic and phenotypic heterogeneity.


Assuntos
Proteínas de Transporte/genética , Surdez/genética , Perda Auditiva/genética , Mutação , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Cromossomos Humanos Par 10 , Feminino , Genes Recessivos , Heterogeneidade Genética , Ligação Genética , Haplótipos , Heterozigoto , Homozigoto , Humanos , Masculino , Modelos Moleculares , Linhagem
13.
Clin J Am Soc Nephrol ; 9(11): 1876-82, 2014 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-25341722

RESUMO

BACKGROUND AND OBJECTIVES: C3 glomerulopathy (C3G) applies to a group of renal diseases defined by a specific renal biopsy finding: a dominant pattern of C3 fragment deposition on immunofluorescence. The primary pathogenic mechanism involves abnormal control of the alternative complement pathway, although a full description of the disease spectrum remains to be determined. This study sought to validate and define the association of complement dysregulation with C3G and to determine whether specific complement pathway abnormalities could inform disease definition. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: This study included 34 patients with C3G (17 with C3 glomerulonephritis [C3GN] and 17 with dense deposit disease [DDD]) diagnosed between 2008 and 2013 selected from the C3G Registry. Control samples (n=100) were recruited from regional blood drives. Nineteen complement biomarkers were assayed on all samples. Results were compared between C3G disease categories and with normal controls. RESULTS: Assessment of the alternative complement pathway showed that compared with controls, patients with C3G had lower levels of serum C3 (P<0.001 for both DDD and C3GN) and factor B (P<0.001 for both DDD and C3GN) as well as higher levels of complement breakdown products including C3d (P<0.001 for both DDD and C3GN) and Bb (P<0.001 for both DDD and C3GN). A comparison of terminal complement pathway proteins showed that although C5 levels were significantly suppressed (P<0.001 for both DDD and C3GN) its breakdown product C5a was significantly higher only in patients with C3GN (P<0.05). Of the other terminal pathway components (C6-C9), the only significant difference was in C7 levels between patients with C3GN and controls (P<0.01). Soluble C5b-9 was elevated in both diseases but only the difference between patients with C3GN and controls reached statistical significance (P<0.001). Levels of C3 nephritic factor activity were qualitatively higher in patients with DDD compared with patients with C3GN. CONCLUSIONS: Complement biomarkers are significantly abnormal in patients with C3G compared with controls. These data substantiate the link between complement dysregulation and C3G and identify C3G interdisease differences.


Assuntos
Fator Nefrítico do Complemento 3/metabolismo , Complemento C3/metabolismo , Complemento C5/metabolismo , Complemento C7/metabolismo , Fator B do Complemento/metabolismo , Glomerulonefrite Membranoproliferativa/sangue , Adolescente , Adulto , Biomarcadores/sangue , Estudos de Casos e Controles , Criança , Complemento C3d/metabolismo , Complemento C5a/metabolismo , Feminino , Glomerulonefrite Membranoproliferativa/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
14.
Otol Neurotol ; 35(3): 395-400, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24492129

RESUMO

HYPOTHESIS: Genetic variation in BMP2 and BMP4 found in otosclerosis patients result in altered Smad signaling. BACKGROUND: Otosclerosis is a common form of adult-onset conductive hearing loss resulting from abnormal bone remodeling of the bony labyrinth that surrounds the inner ear. Both genetic and environmental factors are implicated in the disease, yet very little is known about its pathogenesis. The evidence for a genetic component has been established through family-based linkage and population-based association studies. Previously, members of the TGF-ß superfamily of genes have been associated with otosclerosis. METHODS: Sequencing of BMP2 and BMP4 coding regions was performed to identify common and rare variation in German otosclerosis patients compared with controls. Functional analyses of rare variation in the patient cohort were conducted by exposing an osteosarcoma cell line to conditioned media containing either wild type or variant forms of BMP2 or BMP4 and analyzing Smad1/5/8 phosphorylation. RESULTS: Although no significant association with common variation in these 2 genes was detected, there were 8 singleton variants identified in the German population. Of the 4 coding variants found solely in otosclerosis patients, two--BMP4(N150K) and BMP2(K357-R396del)--were found to decrease Smad1/5/8 signaling. CONCLUSION: Rare variants in BMP2 and BMP4 are not a major genetic component in the otosclerosis population. However, those with functional affect showed decreased Smad signaling. Further analysis of Smad signaling molecules should be performed to determine if these pathways in combination are a major contributor to otosclerosis, which could lead to additional treatment options for otosclerosis patients.


Assuntos
Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 4/genética , Otosclerose/genética , Transdução de Sinais/genética , Proteínas Smad/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 4/metabolismo , Feminino , Estudos de Associação Genética , Ligação Genética , Predisposição Genética para Doença , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Otosclerose/metabolismo , Fosforilação , Polimorfismo de Nucleotídeo Único , Adulto Jovem
15.
J Am Soc Nephrol ; 25(1): 55-64, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24029428

RESUMO

Atypical hemolytic uremic syndrome (aHUS) is a thrombotic microangiopathy caused by uncontrolled activation of the alternative pathway of complement at the cell surface level that leads to microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney failure. In approximately one half of affected patients, pathogenic loss-of-function variants in regulators of complement or gain-of-function variants in effectors of complement are identified, clearly implicating complement in aHUS. However, there are strong lines of evidence supporting the presence of additional genetic contributions to this disease. To identify novel aHUS-associated genes, we completed a comprehensive screen of the complement and coagulation pathways in 36 patients with sporadic aHUS using targeted genomic enrichment and massively parallel sequencing. After variant calling, quality control, and hard filtering, we identified 84 reported or novel nonsynonymous variants, 22 of which have been previously associated with disease. Using computational prediction methods, 20 of the remaining 62 variants were predicted to be deleterious. Consistent with published data, nearly one half of these 42 variants (19; 45%) were found in genes implicated in the pathogenesis of aHUS. Several genes in the coagulation pathway were also identified as important in the pathogenesis of aHUS. PLG, in particular, carried more pathogenic variants than any other coagulation gene, including three known plasminogen deficiency mutations and a predicted pathogenic variant. These data suggest that mutation screening in patients with aHUS should be broadened to include genes in the coagulation pathway.


Assuntos
Coagulação Sanguínea/genética , Proteínas do Sistema Complemento/genética , Síndrome Hemolítico-Urêmica/sangue , Síndrome Hemolítico-Urêmica/genética , Mutação , Plasminogênio/deficiência , Plasminogênio/genética , Adolescente , Adulto , Sequência de Aminoácidos , Síndrome Hemolítico-Urêmica Atípica , Criança , Pré-Escolar , Via Alternativa do Complemento/genética , Sequência Conservada , Variações do Número de Cópias de DNA , Feminino , Testes Genéticos , Variação Genética , Síndrome Hemolítico-Urêmica/etiologia , Humanos , Masculino , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos , Microangiopatias Trombóticas/sangue , Microangiopatias Trombóticas/genética , Microangiopatias Trombóticas/imunologia , Adulto Jovem
16.
J Am Soc Nephrol ; 24(11): 1820-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23907509

RESUMO

Dense deposit disease (DDD) and C3 glomerulonephritis (C3GN) are widely recognized subtypes of C3 glomerulopathy. These ultra-rare renal diseases are characterized by fluid-phase dysregulation of the alternative complement pathway that leads to deposition of complement proteins in the renal glomerulus. Disease triggers are unknown and because targeted treatments are lacking, progress to end stage renal failure is a common final outcome. We studied soluble CR1, a potent regulator of complement activity, to test whether it restores complement regulation in C3 glomerulopathy. In vitro studies using sera from patients with DDD showed that soluble CR1 prevents dysregulation of the alternative pathway C3 convertase, even in the presence of C3 nephritic factors. In mice deficient in complement factor H and transgenic for human CR1, soluble CR1 therapy stopped alternative pathway activation, resulting in normalization of serum C3 levels and clearance of iC3b from glomerular basement membranes. Short-term use of soluble CR1 in a pediatric patient with end stage renal failure demonstrated its safety and ability to normalize activity of the terminal complement pathway. Overall, these data indicate that soluble CR1 re-establishes regulation of the alternative complement pathway and provide support for a limited trial to evaluate soluble CR1 as a treatment for DDD and C3GN.


Assuntos
Complemento C3/análise , Glomerulonefrite Membranoproliferativa/tratamento farmacológico , Receptores de Complemento 3b/uso terapêutico , Animais , Criança , Fator H do Complemento/fisiologia , Via Alternativa do Complemento , Glomerulonefrite Membranoproliferativa/imunologia , Humanos , Camundongos
17.
Pediatr Nephrol ; 28(11): 2221-5, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23880784

RESUMO

BACKGROUND: Mutations in complement factor H (CFH) are associated with complement dysregulation and the development of an aggressive form of atypical hemolytic uremic syndrome (aHUS) that progresses to end-stage renal disease (ESRD) and in most patients has a high rate of recurrence following transplantation. Sequence analysis of CFH and its downstream complement factor H-related genes (CFHR1-5) reveals several macrohomologous blocks caused by large genomic duplications. This high degree of sequence identity renders this area susceptible to nonallelic homologous recombination (NAHR) events, resulting in large-scale deletions, duplications, and the generation of hybrid CFH genes. CASE-DIAGNOSIS: Here, we report the finding of a novel CFHR1/CFH hybrid gene created by a de novo NAHR event in a 14-year-old girl with aHUS. The resulting fusion protein contains the first three short consensus repeats (SCRs) of CFHR1 and the terminal two SCRs of CFH. CONCLUSIONS: This finding demonstrates a novel pathogenic mechanism for the development of aHUS. Additionally, since standard Sanger sequencing is unable to detect such rearrangements, all aHUS patients should receive comprehensive genetic screening that includes analysis of copy number variation in order to identify patients with poor clinical prognoses.


Assuntos
Proteínas Inativadoras do Complemento C3b/genética , Fator H do Complemento/genética , Síndrome Hemolítico-Urêmica/genética , Anticorpos Monoclonais Humanizados/uso terapêutico , Autoanticorpos/análise , Western Blotting , Criança , Creatinina/sangue , DNA/genética , Feminino , Amplificação de Genes , Humanos , Imunossupressores/uso terapêutico , Transplante de Rim , Reação em Cadeia da Polimerase , Diálise Renal
18.
Kidney Int ; 83(2): 293-9, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23235567

RESUMO

Postinfectious glomerulonephritis is a common disorder that develops following an infection. In the majority of cases, there is complete recovery of renal function within a few days to weeks following resolution of the infection. In a small percentage of patients, however, the glomerulonephritis takes longer to resolve, resulting in persistent hematuria and proteinuria, or even progression to end-stage kidney disease. In some cases of persistent hematuria and proteinuria, kidney biopsies show findings of a postinfectious glomerulonephritis even in the absence of any evidence of a preceding infection. The cause of such 'atypical' postinfectious glomerulonephritis, with or without evidence of preceding infection, is unknown. Here we show that most patients diagnosed with this 'atypical' postinfectious glomerulonephritis have an underlying defect in the regulation of the alternative pathway of complement. These defects include mutations in complement-regulating proteins and antibodies to the C3 convertase known as C3 nephritic factors. As a result, the activated alternative pathway is not brought under control even after resolution of the infection. Hence, the sequela is continual glomerular deposition of complement factors with resultant inflammation and development of an 'atypical' postinfectious glomerulonephritis.


Assuntos
Via Alternativa do Complemento , Glomerulonefrite/imunologia , Infecções/complicações , Adolescente , Adulto , Idoso , Biópsia , Convertases de Complemento C3-C5/fisiologia , Feminino , Glomerulonefrite/tratamento farmacológico , Glomerulonefrite/etiologia , Glomerulonefrite/patologia , Humanos , Rim/patologia , Masculino , Pessoa de Meia-Idade
19.
Clin J Am Soc Nephrol ; 7(2): 265-74, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22223606

RESUMO

BACKGROUND AND OBJECTIVES: This study was designed to investigate the causes of alternative pathway dysregulation in a cohort of patients with dense deposit disease (DDD). DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: Thirty-two patients with biopsy-proven DDD underwent screening for C3 nephritic factors (C3Nefs), factor H autoantibodies (FHAAs), factor B autoantibodies (FBAAs), and genetic variants in CFH. C3Nefs were detected by: ELISA, C3 convertase surface assay (C3CSA), C3CSA with properdin (C3CSAP), two-dimensional immunoelectrophoresis (2DIEP), and immunofixation electrophoresis (IFE). FHAAs and FBAAs were detected by ELISA, and CFH variants were identified by Sanger sequencing. RESULTS: Twenty-five patients (78%) were positive for C3Nefs. Three C3Nef-positive patients were also positive for FBAAs and one of these patients additionally carried two novel missense variants in CFH. Of the seven C3Nef-negative patients, one patient was positive for FHAAs and two patients carried CFH variants that may be causally related to their DDD phenotype. C3CASP was the most sensitive C3Nef-detection assay. C3CASP and IFE are complementary because C3CSAP measures the stabilizing properties of C3Nefs, whereas IFE measures their expected consequence-breakdown of C3b. CONCLUSIONS: A test panel that includes C3CSAP, IFE, FHAAs, FBAAs, and genetic testing for CFH variants will identify a probable cause for alternative pathway dysregulation in approximately 90% of DDD patients. Dysregulation is most frequently due to C3Nefs, although some patients test positive for FHAAs, FBAAs, and CFH mutations. Defining the pathophysiology of DDD should facilitate the development of mechanism-directed therapies.


Assuntos
Via Alternativa do Complemento , Glomerulonefrite Membranoproliferativa/imunologia , Rim/imunologia , Adolescente , Adulto , Animais , Autoanticorpos/sangue , Biomarcadores/sangue , Biópsia , Criança , Fator Nefrítico do Complemento 3/análise , Convertases de Complemento C3-C5/imunologia , Fator B do Complemento/imunologia , Fator H do Complemento/genética , Fator H do Complemento/imunologia , Análise Mutacional de DNA , Ensaio de Imunoadsorção Enzimática , Eritrócitos/imunologia , Feminino , Genótipo , Glomerulonefrite Membranoproliferativa/sangue , Glomerulonefrite Membranoproliferativa/diagnóstico , Glomerulonefrite Membranoproliferativa/genética , Humanos , Imunoeletroforese Bidimensional , Iowa , Estimativa de Kaplan-Meier , Rim/patologia , Masculino , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Fenótipo , Prognóstico , Properdina/imunologia , Ovinos , Fatores de Tempo , Adulto Jovem
20.
DNA Cell Biol ; 30(9): 699-708, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21612410

RESUMO

Hearing loss in Ménière's disease (MD) is associated with loss of spiral ganglion neurons and hair cells. In a guinea pig model of endolymphatic hydrops, nitric oxide synthases (NOS) and oxidative stress mediate loss of spiral ganglion neurons. To test the hypothesis that functional variants of NOS1 and NOS2A are associated with MD, we genotyped three functional variants of NOS1 (rs41279104, rs2682826, and a cytosine-adenosine microsatellite repeat in exon 1f) and the CCTTT repeat in the promoter of NOS2A gene (rs3833912) in two independent MD sets (273 patients in total) and 550 controls. A third cohort of American patients was genotyped as replication cohort for the CCTTT repeat. Neither allele nor genotype frequencies of rs41279104 and rs2682826 were associated with MD, although longer alleles of the cytosine-adenosine microsatellite repeat were marginally significant (corrected p = 0.05) in the Mediterranean cohort but not in a second Galicia cohort. Shorter numbers of the CCTTT repeat in NOS2A were significantly more frequent in Galicia controls (OR = 0.37 [CI, 0.18-0.76], corrected p = 0.04), but this finding could not be replicated in Mediterranean or American case-control populations. Meta-analysis did not support an association between CCTTT repeats and risk for MD. Severe hearing loss (>75 dB) was also not associated with any functional variants studied. Functional variants of NOS1 and NOS2A do not confer susceptibility for MD.


Assuntos
Variação Genética , Perda Auditiva Neurossensorial/genética , Doença de Meniere/genética , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo I/genética , População Branca/genética , Sequência de Bases , Sítios de Ligação/genética , Frequência do Gene , Genótipo , Perda Auditiva Neurossensorial/patologia , Humanos , Doença de Meniere/patologia , Repetições de Microssatélites/genética , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética , Análise de Sequência de DNA , Espanha , Estados Unidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...