Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
J Rheumatol ; 50(6): 730-740, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36642429

RESUMEN

The complement system is a tightly regulated, cascading protein network representing a key component linking the innate and humoral immune systems. However, if misdirected or dysregulated, it can be similarly damaging to host-tissue. The role of complement dysregulation on vascular endothelial cells has been well established in atypical hemolytic uremic syndrome (aHUS), a thrombotic microangiopathy (TMA) characterized by microangiopathic hemolytic anemia, thrombocytopenia, and target organ injury. Yet, a great deal of complexity exists around the role of complement in TMA associated with other diseases. A further complicating factor is the cross-talk between complement, neutrophils, and coagulation pathways in the pathophysiology of TMA. Advancements in the understanding of the etiopathogenesis of aHUS paved the way for the successful development of anticomplement therapies (complement C5 inhibitors), which have revolutionized the treatment of aHUS. Therefore, a clearer understanding of the role of the complement system in TMA associated with other conditions will help to identify patients who would benefit from these therapies. This review aims to provide an assessment of the nature and extent of complement involvement in TMA associated with autoimmune diseases such as systemic lupus erythematosus, antiphospholipid syndrome, and scleroderma renal crisis. Defining the role of complement in TMA in these conditions will help to guide timely diagnosis and management.


Asunto(s)
Síndrome Hemolítico Urémico Atípico , Lupus Eritematoso Sistémico , Microangiopatías Trombóticas , Humanos , Células Endoteliales/patología , Microangiopatías Trombóticas/complicaciones , Proteínas del Sistema Complemento , Lupus Eritematoso Sistémico/complicaciones , Síndrome Hemolítico Urémico Atípico/complicaciones , Síndrome Hemolítico Urémico Atípico/patología , Complemento C5
2.
Immunol Rev ; 313(1): 327-338, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36369971

RESUMEN

The glomerulus is often the prime target of dysregulated alternative pathway (AP) activation. In particular, AP activation is the key driver of two severe kidney diseases: atypical hemolytic uremic syndrome and C3 glomerulopathy. Both conditions are associated with a variety of predisposing molecular defects in AP regulation, such as genetic variants in complement regulators, autoantibodies targeting AP proteins, or autoantibodies that stabilize the AP convertases (C3- and C5-activating enzymes). It is noteworthy that these are systemic AP defects, yet in both diseases pathologic complement activation primarily affects the kidneys. In particular, AP activation is often limited to the glomerular capillaries. This tropism of AP-mediated inflammation for the glomerulus points to a unique interaction between AP proteins in plasma and this particular anatomic structure. In this review, we discuss the pre-clinical and clinical data linking the molecular causes of aberrant control of the AP with activation in the glomerulus, and the possible causes of this tropism. Based on these data, we propose a model for why the kidney is so uniquely and frequently targeted in patients with AP defects. Finally, we discuss possible strategies for preventing pathologic AP activation in the kidney.


Asunto(s)
Síndrome Hemolítico Urémico Atípico , Vía Alternativa del Complemento , Humanos , Vía Alternativa del Complemento/genética , Complemento C3/genética , Complemento C3/metabolismo , Riñón , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Síndrome Hemolítico Urémico Atípico/genética , Síndrome Hemolítico Urémico Atípico/patología , Autoanticuerpos
3.
Immunol Rev ; 313(1): 239-261, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36369988

RESUMEN

Dysregulation and accelerated activation of the alternative pathway (AP) of complement is known to cause or accentuate several pathologic conditions in which kidney injury leads to the appearance of hematuria and proteinuria and ultimately to the development of chronic renal failure. Multiple genetic and acquired defects involving plasma- and membrane-associated proteins are probably necessary to impair the protection of host tissues and to confer a significant predisposition to AP-mediated kidney diseases. This review aims to explore how our current understanding will make it possible to identify the mechanisms that underlie AP-mediated kidney diseases and to discuss the available clinical evidence that supports complement-directed therapies. Although the value of limiting uncontrolled complement activation has long been recognized, incorporating complement-targeted treatments into clinical use has proved challenging. Availability of anti-complement therapy has dramatically transformed the outcome of atypical hemolytic uremic syndrome, one of the most severe kidney diseases. Innovative drugs that directly counteract AP dysregulation have also opened new perspectives for the management of other kidney diseases in which complement activation is involved. However, gained experience indicates that the choice of drug should be tailored to each patient's characteristics, including clinical, histologic, genetic, and biochemical parameters. Successfully treating patients requires further research in the field and close collaboration between clinicians and researchers who have special expertise in the complement system.


Asunto(s)
Síndrome Hemolítico Urémico Atípico , Enfermedades Renales , Humanos , Riñón/patología , Enfermedades Renales/terapia , Enfermedades Renales/patología , Proteínas del Sistema Complemento , Síndrome Hemolítico Urémico Atípico/tratamiento farmacológico , Síndrome Hemolítico Urémico Atípico/patología , Activación de Complemento
4.
PLoS One ; 16(11): e0258319, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34748552

RESUMEN

BACKGROUND: Atypical Hemolytic Uremic Syndrome (aHUS) is an ultra-rare disease that potentially leads to kidney graft failure due to ongoing Thrombotic Microangiopathy (TMA). The aim was evaluating the frequency of TMA after kidney transplantation in patients with aHUS in a Brazilian cohort stratified by the use of the specific complement-inhibitor eculizumab. METHODS: This was a multicenter retrospective cohort study including kidney transplant patients diagnosed with aHUS. We collected data from 118 transplant centers in Brazil concerning aHUS transplanted patients between 01/01/2007 and 12/31/2019. Patients were stratified into three groups: no use of eculizumab (No Eculizumab Group), use of eculizumab for treatment of after transplantation TMA (Therapeutic Group), and use of eculizumab for prophylaxis of aHUS recurrence (Prophylactic Group). RESULTS: Thirty-eight patients with aHUS who received kidney transplantation were enrolled in the study. Patients' mean age was 30 years (24-40), and the majority of participants was women (63% of cases). In the No Eculizumab Group (n = 11), there was a 91% graft loss due to the TMA. The hazard ratio of TMA graft loss was 0.07 [0.01-0.55], p = 0.012 in the eculizumab Prophylactic Group and 0.04 [0.00-0.28], p = 0.002 in the eculizumab Therapeutic Group. CONCLUSION: The TMA graft loss in the absence of a specific complement-inhibitor was higher among the Brazilian cohort of kidney transplant patients. This finding reinforces the need of eculizumab use for treatment of aHUS kidney transplant patients. Cost optimization analysis and the early access to C5 inhibitors are suggested, especially in low-medium income countries.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Síndrome Hemolítico Urémico Atípico/tratamiento farmacológico , Trasplante de Riñón/efectos adversos , Microangiopatías Trombóticas/tratamiento farmacológico , Adulto , Síndrome Hemolítico Urémico Atípico/complicaciones , Síndrome Hemolítico Urémico Atípico/patología , Brasil/epidemiología , Inactivadores del Complemento/administración & dosificación , Femenino , Rechazo de Injerto/complicaciones , Rechazo de Injerto/patología , Humanos , Masculino , Estudios Retrospectivos , Microangiopatías Trombóticas/inducido químicamente , Microangiopatías Trombóticas/complicaciones , Microangiopatías Trombóticas/patología , Adulto Joven
6.
Blood ; 138(22): 2185-2201, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34189567

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) is a life-threatening thrombotic microangiopathy that can progress, when untreated, to end-stage renal disease. Most frequently, aHUS is caused by complement dysregulation due to pathogenic variants in genes that encode complement components and regulators. Among these genes, the factor H (FH) gene, CFH, presents with the highest frequency (15% to 20%) of variants and is associated with the poorest prognosis. Correct classification of CFH variants as pathogenic or benign is essential to clinical care but remains challenging owing to the dearth of functional studies. As a result, significant numbers of variants are reported as variants of uncertain significance. To address this knowledge gap, we expressed and functionally characterized 105 aHUS-associated FH variants. All FH variants were categorized as pathogenic or benign and, for each, we fully documented the nature of the pathogenicity. Twenty-six previously characterized FH variants were used as controls to validate and confirm the robustness of the functional assays used. Of the remaining 79 uncharacterized variants, only 29 (36.7%) alter FH expression or function in vitro and, therefore, are proposed to be pathogenic. We show that rarity in control databases is not informative for variant classification, and we identify important limitations in applying prediction algorithms to FH variants. Based on structural and functional data, we suggest ways to circumvent these difficulties and, thereby, improve variant classification. Our work highlights the need for functional assays to interpret FH variants accurately if clinical care of patients with aHUS is to be individualized and optimized.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/genética , Factor H de Complemento/genética , Síndrome Hemolítico Urémico Atípico/metabolismo , Síndrome Hemolítico Urémico Atípico/patología , Factor H de Complemento/química , Factor H de Complemento/metabolismo , Expresión Génica , Predisposición Genética a la Enfermedad , Variación Genética , Humanos , Modelos Moleculares , Mutación Puntual , Polimorfismo de Nucleótido Simple , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
7.
Nephron ; 145(4): 415-427, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33873197

RESUMEN

BACKGROUND: Atypical hemolytic uremic syndrome (aHUS) is a rare but critical illness. To this date, few studies have reported on the disease in Chinese children. METHODS: We studied a Chinese pediatric cohort to delineate the clinical characteristics, genotypes, and prognosis. Ninety-one patients with aHUS were enrolled in this study. RESULTS: Fifty-nine children (64.8%) had anti-complement-factor-H autoantibody-associated aHUS (anti-CFH aHUS). Of these children, 21 (46.7%) had complement factor-H-related protein 1 (CFHR1) homozygous deletion, and most patients with CFHR1 homozygous deletion also had complement factor-H-related protein 3 (CFHR3) homozygous deletions (76.2%). Using gene sequencing of 15 candidate genes, we identified 14 genetic variants in 46 aHUS patients, including 5 pathogenic or like pathogenic variants and 9 variants of uncertain significance. The average follow-up time was 46.1 ± 28 months. Among patients with anti-CFH aHUS, there was a correlation between CFHR1 homozygous deletion and patients with persistent proteinuria (odds ratio [OR] 6.954, 95% confidence interval [CI] 1.033-46.821, p = 0.046). As of the last follow-up, ESRD or deaths occurred in 3.6% of the children with anti-CFH aHUS and 26.7% of children with aHUS who were negative for anti-CFH. CONCLUSIONS: Anti-complement-factor-H antibody positivity is the main cause of morbidity in Chinese children with aHUS. There may be a correlation between CFHR1 homozygous deletion and persistent proteinuria. Comprehensive assessment of anti-CFH antibodies and genetic variants is essential for the management of aHUS children.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/genética , Síndrome Hemolítico Urémico Atípico/patología , Pueblo Asiatico , Autoanticuerpos/análisis , Proteínas Sanguíneas/genética , Niño , Preescolar , China , Estudios de Cohortes , Proteínas Inactivadoras del Complemento C3b/genética , Femenino , Estudios de Seguimiento , Eliminación de Gen , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Lactante , Masculino , Pronóstico , Proteinuria/genética
8.
Blood ; 137(18): 2438-2449, 2021 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-33270832

RESUMEN

The optimal duration of eculizumab treatment in patients with atypical hemolytic uremic syndrome (aHUS) remains poorly defined. We conducted a prospective national multicenter open-label study to assess eculizumab discontinuation in children and adults with aHUS. Fifty-five patients (including 19 children) discontinued eculizumab (mean treatment duration, 16.5 months). Twenty-eight patients (51%) had rare variants in complement genes, mostly in MCP (n = 12; 22%), CFH (n = 6; 11%), and CFI (n = 6; 10%). At eculizumab discontinuation, 17 (30%) and 4 patients (7%) had stage 3 and 4 chronic kidney disease, respectively. During follow-up, 13 patients (23%; 6 children and 7 adults) experienced aHUS relapse. In multivariable analysis, female sex and presence of a rare variant in a complement gene were associated with an increased risk of aHUS relapse, whereas requirement for dialysis during a previous episode of acute aHUS was not. In addition, increased sC5b-9 plasma level at eculizumab discontinuation was associated with a higher risk of aHUS relapse in all patients and in the subset of carriers with a complement gene rare variant, both by log-rank test and in multivariable analysis. Of the 13 relapsing patients, all of whom restarted eculizumab, 11 regained their baseline renal function and 2 had a worsening of their preexisting chronic kidney disease, including 1 patient who progressed to end-stage renal disease. A strategy of eculizumab discontinuation in aHUS patients based on complement genetics is reasonable and safe. It improves the management and quality of life of a sizeable proportion of aHUS patients while reducing the cost of treatment. This trial was registered at www.clinicaltrials.gov as #NCT02574403.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Síndrome Hemolítico Urémico Atípico/tratamiento farmacológico , Inactivadores del Complemento/uso terapéutico , Privación de Tratamiento/estadística & datos numéricos , Adolescente , Adulto , Síndrome Hemolítico Urémico Atípico/metabolismo , Síndrome Hemolítico Urémico Atípico/patología , Niño , Preescolar , Femenino , Estudios de Seguimiento , Humanos , Lactante , Recién Nacido , Masculino , Pronóstico , Estudios Prospectivos , Tasa de Supervivencia , Adulto Joven
10.
Hum Mol Genet ; 29(14): 2313-2324, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32510551

RESUMEN

Factor I (FI) is one of the main inhibitors of complement activity, and numerous rare coding variants have been reported in patients with age-related macular degeneration, atypical hemolytic uremic syndrome and C3 glomerulopathy. Since many of these variants are of unknown clinical significance, this study aimed to determine the effect of rare coding variants in the complement factor I (CFI) gene on FI expression. We measured FI levels in plasma samples of carriers of rare coding variants and in vitro in the supernatants of epithelial cells expressing recombinant FI. FI levels were measured in 177 plasma samples of 155 individuals, carrying 24 different rare coding variants in CFI. In carriers of the variants p.Gly119Arg, p.Leu131Arg, p.Gly188Ala and c.772G>A (r.685_773del), significantly reduced FI plasma levels were detected. Furthermore, recombinant FI expression levels were determined for 126 rare coding variants. Of these variants 68 (54%) resulted in significantly reduced FI expression in supernatant compared to wildtype (WT). The recombinant protein expression levels correlated significantly with the FI level in plasma of carriers of CFI variants. In this study, we performed the most comprehensive FI expression level analysis of rare coding variants in CFI to date. More than half of CFI variants lead to reduced FI expression, which might impair complement regulation in vivo. Our study will aid the interpretation of rare coding CFI variants identified in clinical practice, which is in particular important in light of patient inclusion in ongoing clinical trials for CFI gene supplementation in AMD.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/genética , Factor I de Complemento/genética , Fibrinógeno/genética , Degeneración Macular/genética , Anciano , Anciano de 80 o más Años , Alelos , Síndrome Hemolítico Urémico Atípico/sangre , Síndrome Hemolítico Urémico Atípico/patología , Femenino , Regulación de la Expresión Génica/genética , Predisposición Genética a la Enfermedad , Genotipo , Heterocigoto , Humanos , Degeneración Macular/sangre , Degeneración Macular/patología , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/genética
11.
Pediatr Dev Pathol ; 23(5): 362-371, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32406813

RESUMEN

BACKGROUND: The aim of this study was to investigate the histopathological findings in kidney biopsies in children with atypical hemolytic uremic syndrome (aHUS) and to determine whether specific pathological findings in aHUS have a prognostic value. METHODS: Renal biopsy specimens of 29 patients who were recorded in the national Turkish aHUS registry database were available for review. Histopathological findings were compared with the clinical and laboratory features at the presentation and the final outcome. RESULTS: The mean age at presentation and follow-up period was 4.9 ± 3.9 and 3.9 ± 3.0 years, respectively. The median time interval from the first symptom to biopsy was 10 days. Vascular thrombosis and interstitial fibrosis were significantly related to chronic kidney disease (CKD) requiring dialysis or kidney transplantation during follow-up (5.6-fold, for both). Glomerular necrosis, cortical necrosis, and glomerular sclerosis were markedly associated with CKD without dialysis (6.2-fold, 13.3-fold, and 8.8-fold, respectively). However, presence of endothelial swelling, subendothelial widening, and fragmented erythrocytes was found to be correlated with a favorable final outcome. CONCLUSIONS: Presence of vascular thrombosis, cortical necrosis, and glomerular sclerosis in histopathological evaluation correlated with developing CKD. Chronic changes in the interstitial compartment were also related to poor prognosis, a finding that has been shown for the first time in pediatric aHUS cases.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/diagnóstico , Síndrome Hemolítico Urémico Atípico/patología , Riñón/patología , Síndrome Hemolítico Urémico Atípico/mortalidad , Síndrome Hemolítico Urémico Atípico/fisiopatología , Biopsia , Niño , Preescolar , Femenino , Estudios de Seguimiento , Humanos , Lactante , Modelos Logísticos , Masculino , Pronóstico , Sistema de Registros , Insuficiencia Renal Crónica/diagnóstico , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/mortalidad , Insuficiencia Renal Crónica/terapia , Estudios Retrospectivos
12.
J Med Chem ; 63(11): 5697-5722, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32073845

RESUMEN

The alternative pathway (AP) of the complement system is a key contributor to the pathogenesis of several human diseases including age-related macular degeneration, paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS), and various glomerular diseases. The serine protease factor B (FB) is a key node in the AP and is integral to the formation of C3 and C5 convertase. Despite the prominent role of FB in the AP, selective orally bioavailable inhibitors, beyond our own efforts, have not been reported previously. Herein we describe in more detail our efforts to identify FB inhibitors by high-throughput screening (HTS) and leveraging insights from several X-ray cocrystal structures during optimization efforts. This work culminated in the discovery of LNP023 (41), which is currently being evaluated clinically in several diverse AP mediated indications.


Asunto(s)
Ácido Benzoico/química , Factor B del Complemento/antagonistas & inhibidores , Indoles/química , Síndrome Hemolítico Urémico Atípico/metabolismo , Síndrome Hemolítico Urémico Atípico/patología , Ácido Benzoico/metabolismo , Ácido Benzoico/farmacocinética , Sitios de Unión , Dominio Catalítico , Factor B del Complemento/metabolismo , Cristalografía por Rayos X , Evaluación Preclínica de Medicamentos , Semivida , Humanos , Indoles/metabolismo , Indoles/farmacocinética , Concentración 50 Inhibidora , Degeneración Macular/metabolismo , Degeneración Macular/patología , Simulación de Dinámica Molecular , Relación Estructura-Actividad
13.
FEBS Lett ; 594(16): 2543-2555, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31943152

RESUMEN

Dengue disease is an inflammatory-driven pathology, and complement overactivation is linked to disease severity and vascular leakage. Additionally, dysregulation of complement alternative pathway (AP) components has been described, such as upregulation of complement factor D and downregulation of complement factor H (FH), which activate and inhibit the AP, respectively. Thus, the pathology of severe dengue could in part result from AP dysfunction, even though complement and AP activation usually provide protection against viral infections. In dengue virus-infected macrophages and endothelial cells (ECs), the site of replication and target for vascular pathology, respectively, the AP is activated. The AP activation, reduced FH and vascular leakage seen in dengue disease in part parallels other complement AP pathologies associated with FH deficiency, such as atypical haemolytic uraemic syndrome (aHUS). aHUS can be therapeutically targeted with inhibitors of complement terminal activity, raising the idea that strategies such as inhibition of complement or delivery of FH or other complement regulatory components to EC may be beneficial to combat the vascular leakage seen in severe dengue.


Asunto(s)
Factor D del Complemento/inmunología , Factor H de Complemento/inmunología , Vía Alternativa del Complemento , Virus del Dengue/inmunología , Dengue/inmunología , Animales , Síndrome Hemolítico Urémico Atípico/inmunología , Síndrome Hemolítico Urémico Atípico/patología , Factor H de Complemento/deficiencia , Dengue/patología , Células Endoteliales/inmunología , Células Endoteliales/patología , Enfermedades por Deficiencia de Complemento Hereditario/inmunología , Enfermedades por Deficiencia de Complemento Hereditario/patología , Humanos , Enfermedades Renales/inmunología , Enfermedades Renales/patología , Macrófagos/inmunología , Macrófagos/patología
14.
Front Immunol ; 11: 604759, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33643292

RESUMEN

Objective: To first describe and estimate the potential pathogenic role of Ig4 autoantibodies in complement-mediated thrombotic microangiopathy (TMA) in a patient with IgG4-related disease (IgG4-RD). Methods: This study is a case report presenting a retrospective review of the patient's medical chart. Plasma complement C3 and C4 levels, immunoglobulin isotypes and subclasses were determined by nephelometry, the complement pathways' activity (CH50, AP50, MBL) using WIESLAB® Complement System assays. Human complement factor H levels, anti-complement factor H auto-antibodies were analyzed by ELISA, using HRP-labeled secondary antibodies specific for human IgG, IgG4, and IgA, respectively. Genetic analyses were performed by exome sequencing of 14 gens implicated in complement disorders, as well as multiplex ligation-dependent probe amplification looking specifically for CFH, CFHR1-2-3, and 5. Results: Our brief report presents the first case of IgG4-RD with complement-mediated TMA originating from both pathogenic CFHR 1 and CFHR 4 genes deletions, and inhibitory anti-complement factor H autoantibodies of the IgG4 subclass. Remission was achieved with plasmaphereses, corticosteroids, and cyclophosphamide. Following remission, the patient was diagnosed with lymphocytic meningitis and SARS-CoV-2 pneumonia with an uneventful recovery. Conclusion: IgG4-RD can be associated with pathogenic IgG4 autoantibodies. Genetic predisposition such as CFHR1 and CFHR4 gene deletions enhance the susceptibility to the formation of inhibitory anti-Factor H IgG4 antibodies.


Asunto(s)
Apolipoproteínas/genética , Síndrome Hemolítico Urémico Atípico/genética , Autoanticuerpos/inmunología , Proteínas Inactivadoras del Complemento C3b/genética , Factor H de Complemento/inmunología , Enfermedad Relacionada con Inmunoglobulina G4/genética , Síndrome Hemolítico Urémico Atípico/inmunología , Síndrome Hemolítico Urémico Atípico/patología , Femenino , Eliminación de Gen , Predisposición Genética a la Enfermedad/genética , Humanos , Inmunoglobulina G/inmunología , Enfermedad Relacionada con Inmunoglobulina G4/inmunología , Enfermedad Relacionada con Inmunoglobulina G4/patología , Persona de Mediana Edad , Microangiopatías Trombóticas/inmunología , Microangiopatías Trombóticas/patología
15.
Br J Haematol ; 189(1): 182-186, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31879952

RESUMEN

Atypical haemolytic uremic syndrome (aHUS) is associated with complement system abnormality, such as production of complement factor H (CFH) autoantibodies. The growing evidence indicates complement overactivation on platelets is intimately involved in aHUS pathogenesis, besides endothelial injury. We here showed plasma from patients with anti-CFH antibodies induced aggregation of washed platelets, while purified anti-CFH antibodies suppressed aggregation. This suggested anti-CFH antibody itself suppressed thrombosis, while other plasma factor including complement factors could overactivate the platelets, leading to aggregation, which augmented the notion the state of complement activation influenced by anti-CFH antibodies is important in the aggregation of platelets in aHUS.


Asunto(s)
Síndrome Hemolítico Urémico Atípico , Autoanticuerpos , Plaquetas , Factor H de Complemento/inmunología , Agregación Plaquetaria/inmunología , Síndrome Hemolítico Urémico Atípico/sangre , Síndrome Hemolítico Urémico Atípico/inmunología , Síndrome Hemolítico Urémico Atípico/patología , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Plaquetas/inmunología , Plaquetas/metabolismo , Plaquetas/patología , Niño , Preescolar , Femenino , Humanos , Masculino
16.
Front Immunol ; 10: 2166, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31611870

RESUMEN

Defective complement action is a cause of several human glomerular diseases including atypical hemolytic uremic syndrome (aHUS), anti-neutrophil cytoplasmic antibody mediated vasculitis (ANCA), C3 glomerulopathy, IgA nephropathy, immune complex membranoproliferative glomerulonephritis, ischemic reperfusion injury, lupus nephritis, membranous nephropathy, and chronic transplant mediated glomerulopathy. Here we summarize ongoing clinical trials of complement inhibitors in nine glomerular diseases and show which inhibitors are used in trials for these renal disorders (http://clinicaltrials.gov).


Asunto(s)
Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos , Síndrome Hemolítico Urémico Atípico , Inactivadores del Complemento/uso terapéutico , Glomerulonefritis , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/tratamiento farmacológico , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/inmunología , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/patología , Síndrome Hemolítico Urémico Atípico/tratamiento farmacológico , Síndrome Hemolítico Urémico Atípico/inmunología , Síndrome Hemolítico Urémico Atípico/patología , Ensayos Clínicos como Asunto , Activación de Complemento/efectos de los fármacos , Activación de Complemento/inmunología , Glomerulonefritis/tratamiento farmacológico , Glomerulonefritis/inmunología , Glomerulonefritis/patología , Humanos
18.
Front Immunol ; 10: 1282, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31231391

RESUMEN

Background: Atypical hemolytic uremic syndrome (aHUS), an important cause of acute kidney injury (AKI), is characterized by dysregulation of the alternative complement pathway. Autoantibodies to factor H (FH), a chief regulator of this pathway, account for a distinct subgroup. While high anti-FH titers predict relapse, they do not correlate well with disease activity and their functional characterization is required. Methods: Of 781 patients <18-year-old of aHUS in the nationwide database from 2007 to 2018, 436 (55.8%) had anti-FH antibodies. Clinical features and outcome of patients managed in the last 6-year (n = 317) were compared to before (n = 119). In plasma samples of 44 patients, levels of serial circulating FH immune complexes (CIC), free FH, soluble terminal complement complex (sC5b-9), sheep red blood cell (SRBC) lysis and epitope specificity (n = 8) were examined. Functional renal reserve, ambulatory hypertension, left ventricular hypertrophy (LVH), and proteinuria were evaluated in a subset. Results: Patients presented with markedly elevated anti-FH titers (10,633.2 ± 998.5 AU/ml). Management varied by center, comprising plasma exchange (PEX; 77.5%) and immunosuppression (73.9%). Patients managed in the last 6-year showed better renal survival at mean 28.5 ± 27.3 months (log rank P = 0.022). Mean anti-FH titers stayed 700-1,164 AU/ml during prolonged follow-up, correlating with CIC. Patients with relapse had lower free-FH during remission [Generalized estimating equations (GEE), P = 0.001]; anti-FH levels ≥1,330 AU/ml and free FH ≤440 mg/l predicted relapse (hazards ratio, HR 6.3; P = 0.018). Epitope specificity was similar during onset, remission and relapse. Antibody titer ≥8,000 AU/ml (HR 2.23; P = 0.024), time to PEX ≥14 days (HR 2.09; P = 0.071) and PEX for <14 days (HR 2.60; P = 0.017) predicted adverse renal outcomes. Combined PEX and immunosuppression improved long-term outcomes (HR 0.37; P = 0.026); maintenance therapy reduced risk of relapses (HR 0.11; P < 0.001). At 4.4±2.5 year, median renal reserve was 15.9%; severe ambulatory, masked and pre-hypertension were found in 38, 30, and 18%, respectively. Proteinuria and LVH occurred in 58 and 28% patients, respectively. Conclusion: Prompt recognition and therapy with PEX and immunosuppression, is associated with satisfactory outcomes. Free-FH predicts early relapses in patients with high anti-FH titers. A significant proportion of impaired functional reserve, ambulatory hypertension, proteinuria and LVH highlight the need for vigilant long-term follow-up.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/inmunología , Adolescente , Síndrome Hemolítico Urémico Atípico/patología , Síndrome Hemolítico Urémico Atípico/terapia , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Niño , Preescolar , Factor H de Complemento/inmunología , Bases de Datos Factuales , Femenino , Humanos , Inmunosupresores/uso terapéutico , Lactante , Masculino
19.
BMC Nephrol ; 20(1): 157, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-31064333

RESUMEN

BACKGROUND: Haemolytic uraemic syndrome (HUS) is a thrombotic microangiopathy (TMA) characterized by predominant renal involvement. Several types of HUS can be distinguished: the most frequent « typical ¼ HUS, due to shiga toxin producing Escherichia coli (STEC), "atypical" HUS due to complement alternative pathway dysregulation and "secondary" HUS associated with various diseases/conditions, the classification of which is still subject to debate. CASE PRESENTATION: We report a case of HUS following E.coli prostatitis and bacteraemia in an adult male. He presented with severe renal and neurological involvement. Initially considered as a "typical" HUS, the condition was treated by antibiotics. No other specific treatment for HUS was administered. The outcome was favorable. We eventually identified a non shiga toxin producing E.coli. Genetic testing of the complement alternative pathway revealed a rare - potentially pathogenic - variant of factor H. This constitutes a possible factor of susceptibility for atypical HUS, suggesting that E.coli infection may be the trigger. CONCLUSION: This case raises the question of complement exploration for HUS associated with infections, in order to classify such cases of HUS in accordance with their underlying pathophysiological mechanisms.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/microbiología , Bacteriemia/microbiología , Infecciones por Escherichia coli/microbiología , Escherichia coli , Complicaciones Posoperatorias/microbiología , Lesión Renal Aguda/etiología , Síndrome Hemolítico Urémico Atípico/patología , Síndrome Hemolítico Urémico Atípico/terapia , Biopsia , Factor H de Complemento/genética , Confusión/etiología , Escherichia coli/genética , Escherichia coli/aislamiento & purificación , Alucinaciones/etiología , Hemorroides/cirugía , Humanos , Riñón/patología , Masculino , Persona de Mediana Edad , Diálisis Renal , Convulsiones/etiología
20.
Kidney Int ; 95(6): 1443-1452, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30982675

RESUMEN

Secondary hemolytic uremic syndrome (HUS) is a heterogeneous group of thrombotic microangiopathies associated with various underlying conditions. Whether it belongs to the spectrum of complement-mediated HUS remains controversial. We analysed the presentation, outcome, and frequency of complement gene rare variants in a cohort of 110 patients with secondary HUS attributed to drugs (29%), autoimmune diseases (24%), infections (17%), malignancies (10%), glomerulopathies (9%), extra-renal organ transplantation (8%), and pancreatitis (3%). The frequency of complement gene rare variants was similar in patients with secondary HUS (5%) and in healthy individuals (6% and 8% in French and European controls, respectively). At diagnosis, 40% of patients required dialysis and 18% had neurological manifestations. Fifty percent of patients received plasmatherapy and 35% were treated with eculizumab. Haematological and complete renal remission was achieved in 80% and 24% of patients, respectively. Thirty-nine percent of patients progressed to chronic kidney disease (stages 3-4) and an additional 37% reached end-stage renal disease. Eleven percent of patients died, most often from complications of the underlying cause of HUS. Only one patient experienced an HUS relapse. Patients treated with eculizumab presented with more severe HUS and were more likely to require dialysis at the time of diagnosis as compared to patients not treated with eculizumab. Rates of hematological remission, chronic kidney disease (stages 3-4), and end-stage renal disease were similar in the two groups. Secondary HUS is an acute nonrelapsing form of HUS, not related to complement dysregulation. The efficacy of eculizumab in this setting is not yet established.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/genética , Proteínas del Sistema Complemento/genética , Síndrome Hemolítico-Urémico/etiología , Fallo Renal Crónico/epidemiología , Insuficiencia Renal Crónica/epidemiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales Humanizados/uso terapéutico , Síndrome Hemolítico Urémico Atípico/mortalidad , Síndrome Hemolítico Urémico Atípico/patología , Síndrome Hemolítico Urémico Atípico/terapia , Niño , Preescolar , Activación de Complemento/genética , Inactivadores del Complemento/uso terapéutico , Proteínas del Sistema Complemento/inmunología , Progresión de la Enfermedad , Femenino , Francia/epidemiología , Síndrome Hemolítico-Urémico/mortalidad , Síndrome Hemolítico-Urémico/patología , Síndrome Hemolítico-Urémico/terapia , Humanos , Riñón/inmunología , Riñón/patología , Fallo Renal Crónico/patología , Masculino , Persona de Mediana Edad , Plasmaféresis/estadística & datos numéricos , Sistema de Registros/estadística & datos numéricos , Diálisis Renal/estadística & datos numéricos , Insuficiencia Renal Crónica/patología , Estudios Retrospectivos , Factores de Riesgo , Resultado del Tratamiento , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...