Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Kidney Int ; 105(2): 328-337, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38008161

RESUMEN

Renin, an aspartate protease, regulates the renin-angiotensin system by cleaving its only known substrate angiotensinogen to angiotensin. Recent studies have suggested that renin may also cleave complement component C3 to activate complement or contribute to its dysregulation. Typically, C3 is cleaved by C3 convertase, a serine protease that uses the hydroxyl group of a serine residue as a nucleophile. Here, we provide seven lines of evidence to show that renin does not cleave C3. First, there is no association between renin plasma levels and C3 levels in patients with C3 Glomerulopathies (C3G) and atypical Hemolytic Uremic Syndrome (aHUS), implying that serum C3 consumption is not increased in the presence of high renin. Second, in vitro tests of C3 conversion to C3b do not detect differences when sera from patients with high renin levels are compared to sera from patients with normal/low renin levels. Third, aliskiren, a renin inhibitor, does not block abnormal complement activity introduced by nephritic factors in the fluid phase. Fourth, aliskiren does not block dysregulated complement activity on cell surfaces. Fifth, recombinant renin from different sources does not cleave C3 even after 24 hours of incubation at 37 °C. Sixth, direct spiking of recombinant renin into sera samples of patients with C3G and aHUS does not enhance complement activity in either the fluid phase or on cell surfaces. And seventh, molecular modeling and docking place C3 in the active site of renin in a position that is not consistent with a productive ground state complex for catalytic hydrolysis. Thus, our study does not support a role for renin in the activation of complement.


Asunto(s)
Activación de Complemento , Complemento C3 , Enfermedades Renales , Renina , Humanos , Amidas , Síndrome Hemolítico Urémico Atípico , Complemento C3/metabolismo , Convertasas de Complemento C3-C5/metabolismo , Vía Alternativa del Complemento , Fumaratos , Renina/antagonistas & inhibidores , Renina/sangre , Renina/metabolismo
2.
Am J Kidney Dis ; 79(4): 570-581, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34571062

RESUMEN

Blocking the complement system as a therapeutic strategy has been proposed for numerous glomerular diseases but presents myriad questions and challenges, not the least of which is demonstrating efficacy and safety. In light of these potential issues and because there are an increasing number of anticomplement therapy trials either planned or under way, the National Kidney Foundation facilitated an all-virtual scientific workshop entitled "Improving Clinical Trials for Anti-Complement Therapies in Complement-Mediated Glomerulopathies." Attended by patient representatives and experts in glomerular diseases, complement physiology, and clinical trial design, the aim of this workshop was to develop standards applicable for designing and conducting clinical trials for anticomplement therapies across a wide spectrum of complement-mediated glomerulopathies. Discussions focused on study design, participant risk assessment and mitigation, laboratory measurements and biomarkers to support these studies, and identification of optimal outcome measures to detect benefit, specifically for trials in complement-mediated diseases. This report summarizes the discussions from this workshop and outlines consensus recommendations.


Asunto(s)
Proteínas Inactivadoras de Complemento , Enfermedades Renales , Proteínas Inactivadoras de Complemento/uso terapéutico , Proteínas del Sistema Complemento , Humanos , Riñón
3.
Am J Nephrol ; 53(10): 675-686, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36404708

RESUMEN

INTRODUCTION: C3 glomerulopathy (C3G) is a rare, progressive kidney disease resulting from dysregulation of the alternative pathway (AP) of complement. Biomarkers at baseline were investigated in patients with C3G who participated in two phase 2 studies with the factor D (FD) inhibitor, danicopan. METHODS: Patients with biopsy-confirmed C3G, proteinuria ≥500 mg/day, and estimated glomerular filtration rate (eGFR) ≥30 mL/min/1.73 m2 were enrolled into two studies (NCT03369236 and NCT03459443). Biomarker analysis was performed for patients with C3G confirmed by central pathology laboratory re-evaluation. Complement and clinical biomarkers, biopsy composite score, and activity and chronicity indices were assessed at baseline and analyzed by pairwise Spearman correlation analysis. RESULTS: Twenty-nine patients were included in the analysis (median [interquartile range] age: 24.0 [10.0] years). Systemic complement AP activation was evident by reduced median concentrations of C3 and C5, elevated sC5b-9, and normal C4, relative to reference ranges. C3 showed strong pairwise correlations with C5 and sC5b-9 (r = 0.80 and -0.73, respectively; p < 0.0001). Baseline Ba and FD concentrations were inversely correlated with eGFR (r = -0.83 and -0.87, respectively; p < 0.0001). Urinary concentrations of sC5b-9 were correlated with both plasma sC5b-9 and proteinuria (r = 0.69 and r = 0.83, respectively; p < 0.0001). Biopsy activity indices correlated strongly with biomarkers of systemic AP activation, including C3 (r = -0.76, p < 0.0001), whereas chronicity indices aligned more closely with eGFR (r = -0.57, p = 0.0021). CONCLUSION: Associations among complement biomarkers, kidney function, and kidney histology may add to the current understanding of C3G and assist with the characterization of patients with this heterogenous disease.


Asunto(s)
Glomerulonefritis Membranoproliferativa , Enfermedades Renales , Humanos , Adulto Joven , Adulto , Complemento C3/metabolismo , Factor D del Complemento , Glomerulonefritis Membranoproliferativa/patología , Biomarcadores , Proteinuria
4.
Am J Nephrol ; 53(10): 687-700, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36423588

RESUMEN

INTRODUCTION: C3 glomerulopathy (C3G) is an ultrarare, chronic and progressive nephropathy mediated by dysregulation of the alternative pathway of complement (AP), with poor prognosis and limited treatment options. Targeted inhibition of proximal AP through factor D (FD) blockade represents a rational treatment approach. We present two phase 2 proof-of-concept clinical studies of the orally active FD inhibitor danicopan in patients with C3G and immune complex-mediated membranoproliferative glomerulonephritis (IC-MPGN) (NCT03369236 and NCT03459443). METHODS: A double-blind, placebo-controlled study in patients with C3G and a single-arm, open-label study in patients with C3G or IC-MPGN treated with danicopan are reported. The studies evaluated pharmacokinetic/pharmacodynamic (PK/PD), efficacy, and safety outcomes. The co-primary endpoints were change from baseline in composite biopsy score and the proportion of patients with a 30% reduction in proteinuria relative to baseline at 6 or 12 months. RESULTS: Optimal systemic concentrations of danicopan were not achieved for complete and sustained inhibition of AP, although there was evidence that blockade of FD reduced AP activity shortly after drug administration. Consequently, limited clinical response was observed in key efficacy endpoints. While stable disease or improvement from baseline was seen in some patients, response was not consistent. The data confirmed the favorable safety profile of danicopan. CONCLUSION: While demonstrating a favorable safety profile, danicopan resulted in incomplete and inadequately sustained inhibition of AP, probably due to limitations in its PK/PD profile in C3G, leading to lack of efficacy. Complete and sustained AP inhibition is required for a clinical response in patients with C3G.


Asunto(s)
Glomerulonefritis Membranoproliferativa , Enfermedades Renales , Humanos , Factor D del Complemento/uso terapéutico , Glomerulonefritis Membranoproliferativa/tratamiento farmacológico , Glomerulonefritis Membranoproliferativa/patología , Proteínas del Sistema Complemento
5.
Kidney Int ; 100(4): 753-779, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34556300

RESUMEN

The Kidney Disease: Improving Global Outcomes (KDIGO) Clinical Practice Guideline for the Management of Glomerular Diseases is an update to the KDIGO 2012 guideline. The aim is to assist clinicians caring for individuals with glomerulonephritis (GN), both adults and children. The scope includes various glomerular diseases, including IgA nephropathy and IgA vasculitis, membranous nephropathy, nephrotic syndrome, minimal change disease (MCD), focal segmental glomerulosclerosis (FSGS), infection-related GN, antineutrophil cytoplasmic antibody (ANCA) vasculitis, lupus nephritis, and anti-glomerular basement membrane antibody GN. In addition, this guideline will be the first to address the subtype of complement-mediated diseases. Each chapter follows the same format providing guidance related to diagnosis, prognosis, treatment, and special situations. The goal of the guideline is to generate a useful resource for clinicians and patients by providing actionable recommendations based on evidence syntheses, with useful infographics incorporating views from experts in the field. Another aim is to propose research recommendations for areas where there are gaps in knowledge. The guideline targets a broad global audience of clinicians treating GN while being mindful of implications for policy and cost. Development of this guideline update followed an explicit process whereby treatment approaches and guideline recommendations are based on systematic reviews of relevant studies, and appraisal of the quality of the evidence and the strength of recommendations followed the "Grading of Recommendations Assessment, Development and Evaluation" (GRADE) approach. Limitations of the evidence are discussed, with areas of future research also presented.


Asunto(s)
Glomerulonefritis por IGA , Glomerulonefritis Membranosa , Glomerulonefritis , Nefrosis Lipoidea , Adulto , Niño , Glomerulonefritis/diagnóstico , Glomerulonefritis/terapia , Glomerulonefritis por IGA/diagnóstico , Glomerulonefritis por IGA/terapia , Glomerulonefritis Membranosa/diagnóstico , Glomerulonefritis Membranosa/tratamiento farmacológico , Humanos , Riñón
6.
Pediatr Nephrol ; 36(8): 2349-2360, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33693990

RESUMEN

BACKGROUND: Eculizumab is approved for the treatment of atypical hemolytic uremic syndrome (aHUS). Its use off-label is frequently reported. The aim of this study was to describe the broader use and outcomes of a cohort of pediatric patients exposed to eculizumab. METHODS: A retrospective, cohort analysis was performed on the clinical and biomarker characteristics of eculizumab-exposed patients < 25 years of age seen across 21 centers of the Pediatric Nephrology Research Consortium. Patients were included if they received at least one dose of eculizumab between 2008 and 2015. Traditional summary statistics were applied to demographic and clinical data. RESULTS: A total of 152 patients were identified, mean age 9.1 (+/-6.8) years. Eculizumab was used "off-label" in 44% of cases. The most common diagnoses were aHUS (47.4%), Shiga toxin-producing Escherichia coli HUS (12%), unspecified thrombotic microangiopathies (9%), and glomerulonephritis (9%). Genetic testing was available for 60% of patients; 20% had gene variants. Dosing regimens were variable. Kidney outcomes tended to vary according to diagnosis. Infectious adverse events were the most common adverse event (33.5%). No cases of meningitis were reported. Nine patients died of noninfectious causes while on therapy. CONCLUSIONS: This multi-center retrospective cohort analysis indicates that a significant number of children and young adults are being exposed to C5 blockade for off-label indications. Dosing schedules were highly variable, limiting outcome conclusions. Attributable adverse events appeared to be low. Cohort mortality (6.6%) was not insignificant. Prospective studies in homogenous disease cohorts are needed to support the role of C5 blockade in kidney outcomes.


Asunto(s)
Nefrología , Adolescente , Anticuerpos Monoclonales Humanizados/efectos adversos , Síndrome Hemolítico Urémico Atípico/tratamiento farmacológico , Síndrome Hemolítico Urémico Atípico/genética , Niño , Humanos , Estudios Prospectivos , Estudios Retrospectivos
7.
Kidney Int ; 98(5): 1265-1274, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32540405

RESUMEN

Atypical hemolytic uremic syndrome is an ultra-rare disease characterized by microangiopathic hemolytic anemia, thrombocytopenia and acute kidney injury. Its pathogenesis is driven most frequently by dysregulated cell-surface control of the alternative pathway of complement secondary to inherited and/or acquired factors. Here we evaluated two unrelated patients with atypical hemolytic uremic syndrome. The first, a five-year-old Caucasian female, presented at 10 months with schistocytes, thrombocytopenia and kidney injury. The second, a 55-year-old Caucasian female, presented at age 31 following caesarean section for preeclampsia. Complement biomarker testing was remarkable for undetectable levels of C3 in both. Circulating levels of C5 and properdin were also low consistent with over-activity of the alternative and terminal pathways of complement. Genetic testing identified a heterozygous novel variant in CFB (c.1101 C>A, p.Ser367Arg) in both patients. Functional studies found strong fluid-phase C3 cleavage when normal and proband sera were mixed. Cell-surface C3b deposition was strongly positive when patient serum was supplemented with C3. In vitro control of C3 convertase activity could be restored with increased concentrations of factor H. Thus, CFB p.Ser367Arg is a gain-of-function pathogenic variant that leads to dysregulation of the alternative pathway in the fluid-phase and increased C3b deposition on cell surfaces. Our study highlights the complexities of complement-mediated diseases like atypical hemolytic uremic syndrome and illustrates the importance of functional studies at the variant level to gain insight into the disease phenotype.


Asunto(s)
Síndrome Hemolítico Urémico Atípico , Adulto , Síndrome Hemolítico Urémico Atípico/diagnóstico , Síndrome Hemolítico Urémico Atípico/genética , Cesárea , Preescolar , Factor B del Complemento/genética , Factor H de Complemento/genética , Vía Alternativa del Complemento/genética , Femenino , Humanos , Persona de Mediana Edad , Mutación , Embarazo
8.
Semin Immunol ; 28(3): 241-9, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27402056

RESUMEN

C3 glomerulopathy (C3G) describes a spectrum of glomerular diseases defined by shared renal biopsy pathology: a predominance of C3 deposition on immunofluorescence with electron microscopy permitting disease sub-classification. Complement dysregulation underlies the observed pathology, a causal relationship that is supported by well described studies of genetic and acquired drivers of disease. In this article, we provide an overview of the features of C3G, including a discussion of disease definition and a review of the causal role of complement. We discuss molecular markers of disease and how biomarkers are informing our evolving understanding of underlying pathology. Research advances are laying the foundation for complement inhibition as a targeted approach to treatment of C3G.


Asunto(s)
Biomarcadores/metabolismo , Complemento C3/inmunología , Inactivadores del Complemento/uso terapéutico , Vía Alternativa del Complemento , Glomerulonefritis Membranosa/terapia , Inmunoterapia/métodos , Riñón/metabolismo , Animales , Complemento C3/genética , Frecuencia de los Genes , Genotipo , Glomerulonefritis Membranosa/genética , Glomerulonefritis Membranosa/inmunología , Humanos , Riñón/patología , Polimorfismo Genético
9.
J Am Soc Nephrol ; 29(12): 2809-2819, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30377230

RESUMEN

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.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/genética , Adolescente , Adulto , Anciano , Síndrome Hemolítico Urémico Atípico/sangre , Factores de Coagulación Sanguínea/genética , Niño , Preescolar , Proteínas del Sistema Complemento/genética , Bases de Datos Genéticas , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Variación Genética , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Mutación , Vitronectina/genética , Adulto Joven
10.
Kidney Int ; 91(3): 539-551, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27989322

RESUMEN

In both atypical hemolytic uremic syndrome (aHUS) and C3 glomerulopathy (C3G) complement plays a primary role in disease pathogenesis. Herein we report the outcome of a 2015 Kidney Disease: Improving Global Outcomes (KDIGO) Controversies Conference where key issues in the management of these 2 diseases were considered by a global panel of experts. Areas addressed included renal pathology, clinical phenotype and assessment, genetic drivers of disease, acquired drivers of disease, and treatment strategies. In order to help guide clinicians who are caring for such patients, recommendations for best treatment strategies were discussed at length, providing the evidence base underpinning current treatment options. Knowledge gaps were identified and a prioritized research agenda was proposed to resolve outstanding controversial issues.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/inmunología , Activación de Complemento , Complemento C3/inmunología , Glomerulonefritis/inmunología , Riñón/inmunología , Animales , Síndrome Hemolítico Urémico Atípico/tratamiento farmacológico , Síndrome Hemolítico Urémico Atípico/genética , Síndrome Hemolítico Urémico Atípico/patología , Activación de Complemento/efectos de los fármacos , Complemento C3/genética , Inactivadores del Complemento/uso terapéutico , Predisposición Genética a la Enfermedad , Glomerulonefritis/tratamiento farmacológico , Glomerulonefritis/genética , Glomerulonefritis/patología , Humanos , Riñón/efectos de los fármacos , Riñón/patología , Fenotipo , Factores de Riesgo , Resultado del Tratamiento
11.
N Engl J Med ; 371(7): 654-66, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-25119611

RESUMEN

This review article covers the diverse pathophysiological pathways that can lead to microangiopathic hemolytic anemia and a procoagulant state with or without damage to the kidneys and other organs.


Asunto(s)
Microangiopatías Trombóticas , Humanos , Púrpura Trombocitopénica Trombótica/diagnóstico , Púrpura Trombocitopénica Trombótica/terapia , Microangiopatías Trombóticas/diagnóstico , Microangiopatías Trombóticas/etiología , Microangiopatías Trombóticas/terapia
12.
Am J Kidney Dis ; 70(6): 834-843, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28838767

RESUMEN

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.


Asunto(s)
Autoanticuerpos/inmunología , Complemento C3/inmunología , Proteínas del Sistema Complemento/inmunología , Glomerulonefritis Membranoproliferativa/inmunología , Adolescente , Adulto , Niño , C3 Convertasa de la Vía Alternativa del Complemento/inmunología , Factor Nefrítico del Complemento 3/inmunología , C5 Convertasa de la Vía Clásica del Complemento , Factor H de Complemento/inmunología , Femenino , Humanos , Enfermedades Renales/inmunología , Masculino , Adulto Joven
13.
J Am Soc Nephrol ; 27(4): 1245-53, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26283675

RESUMEN

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.


Asunto(s)
Enfermedades Renales/diagnóstico , Enfermedades Renales/genética , Glomérulos Renales , Microangiopatías Trombóticas/diagnóstico , Microangiopatías Trombóticas/genética , Adolescente , Niño , Preescolar , Complemento C3 , Femenino , Pruebas Genéticas/métodos , Humanos , Enfermedades Renales/inmunología , Masculino
14.
Pediatr Nephrol ; 31(1): 15-39, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25859752

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) emerged during the last decade as a disease largely of complement dysregulation. This advance facilitated the development of novel, rational treatment options targeting terminal complement activation, e.g., using an anti-C5 antibody (eculizumab). We review treatment and patient management issues related to this therapeutic approach. We present consensus clinical practice recommendations generated by HUS International, an international expert group of clinicians and basic scientists with a focused interest in HUS. We aim to address the following questions of high relevance to daily clinical practice: Which complement investigations should be done and when? What is the importance of anti-factor H antibody detection? Who should be treated with eculizumab? Is plasma exchange therapy still needed? When should eculizumab therapy be initiated? How and when should complement blockade be monitored? Can the approved treatment schedule be modified? What approach should be taken to kidney and/or combined liver-kidney transplantation? How should we limit the risk of meningococcal infection under complement blockade therapy? A pressing question today regards the treatment duration. We discuss the need for prospective studies to establish evidence-based criteria for the continuation or cessation of anticomplement therapy in patients with and without identified complement mutations.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/terapia , Nefrología/normas , Adolescente , Factores de Edad , Anticuerpos Monoclonales Humanizados/uso terapéutico , Síndrome Hemolítico Urémico Atípico/diagnóstico , Síndrome Hemolítico Urémico Atípico/epidemiología , Síndrome Hemolítico Urémico Atípico/inmunología , Niño , Preescolar , Terapia Combinada , Activación de Complemento/efectos de los fármacos , Consenso , Conducta Cooperativa , Monitoreo de Drogas , Humanos , Factores Inmunológicos/uso terapéutico , Lactante , Recién Nacido , Cooperación Internacional , Trasplante de Riñón , Trasplante de Hígado , Monitorización Inmunológica , Selección de Paciente , Intercambio Plasmático , Valor Predictivo de las Pruebas , Factores de Riesgo , Resultado del Tratamiento
15.
Kidney Int ; 87(5): 882-4, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25951068

RESUMEN

Licht et al. present the 2-year follow-up data of the landmark trials studying the efficacy of eculizumab in the treatment of atypical hemolytic uremic syndrome (aHUS). They report sustained improvements in hematologic parameters, continued safety, and additional improvements in kidney function with extended treatment. This report adds a layer of comfort to our care of patients with this rare disease; however, it is unlikely to be the final chapter in the treatment of aHUS.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Síndrome Hemolítico Urémico Atípico/tratamiento farmacológico , Femenino , Humanos , Masculino
17.
Blood ; 122(8): 1487-93, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23847193

RESUMEN

Complement dysregulation leads to atypical hemolytic uremic syndrome (aHUS), while ADAMTS13 deficiency causes thrombotic thrombocytopenic purpura. We investigated whether genetic variations in the ADAMTS13 gene partially explain the reduced activity known to occur in some patients with aHUS. We measured complement activity and ADAMTS13 function, and completed mutation screening of multiple complement genes and ADAMTS13 in a large cohort of aHUS patients. In over 50% of patients we identified complement gene mutations. Surprisingly, 80% of patients also carried at least 1 nonsynonymous change in ADAMTS13, and in 38% of patients, multiple ADAMTS13 variations were found. Six of the 9 amino acid substitutions in ADAMTS13 were common single nucleotide polymorphisms; however, 3 variants-A747V, V832M, and R1096H- were rare, with minor allele frequencies of 0.0094%, 0.5%, and 0.32%, respectively. Reduced complement and ADAMTS13 activity (<60% of normal activity) were found in over 60% and 50% of patients, respectively. We concluded that partial ADAMTS13 deficiency is a common finding in aHUS patients and that genetic screening and functional tests of ADAMTS13 should be considered in these patients.


Asunto(s)
Proteínas ADAM/deficiencia , Proteínas ADAM/genética , Síndrome Hemolítico-Urémico/sangre , Síndrome Hemolítico-Urémico/genética , Proteína ADAMTS13 , Adolescente , Adulto , Alelos , Síndrome Hemolítico Urémico Atípico , Autoanticuerpos/inmunología , Niño , Preescolar , Estudios de Cohortes , Factor H de Complemento/inmunología , ADN/genética , Femenino , Humanos , Lactante , Masculino , Mutación , Polimorfismo Genético , Proteínas Recombinantes de Fusión/metabolismo , Adulto Joven
18.
J Am Soc Nephrol ; 25(1): 55-64, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24029428

RESUMEN

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.


Asunto(s)
Coagulación Sanguínea/genética , Proteínas del Sistema Complemento/genética , Síndrome Hemolítico-Urémico/sangre , Síndrome Hemolítico-Urémico/genética , Mutación , Plasminógeno/deficiencia , Plasminógeno/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Síndrome Hemolítico Urémico Atípico , Niño , Preescolar , Vía Alternativa del Complemento/genética , Secuencia Conservada , Variaciones en el Número de Copia de ADN , Femenino , Pruebas Genéticas , Variación Genética , Síndrome Hemolítico-Urémico/etiología , Humanos , Masculino , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Microangiopatías Trombóticas/sangre , Microangiopatías Trombóticas/genética , Microangiopatías Trombóticas/inmunología , Adulto Joven
19.
J Am Soc Nephrol ; 24(11): 1820-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23907509

RESUMEN

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.


Asunto(s)
Complemento C3/análisis , Glomerulonefritis Membranoproliferativa/tratamiento farmacológico , Receptores de Complemento 3b/uso terapéutico , Animales , Niño , Factor H de Complemento/fisiología , Vía Alternativa del Complemento , Glomerulonefritis Membranoproliferativa/inmunología , Humanos , Ratones
20.
Kidney Int Rep ; 9(2): 464-477, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38344720

RESUMEN

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.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA