Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Kidney Int ; 105(2): 328-337, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38008161

ABSTRACT

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.


Subject(s)
Complement Activation , Complement C3 , Kidney Diseases , Renin , Humans , Amides , Atypical Hemolytic Uremic Syndrome , Complement C3/metabolism , Complement C3-C5 Convertases/metabolism , Complement Pathway, Alternative , Fumarates , Renin/antagonists & inhibitors , Renin/blood , Renin/metabolism
2.
Am J Kidney Dis ; 79(4): 570-581, 2022 04.
Article in English | MEDLINE | ID: mdl-34571062

ABSTRACT

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.


Subject(s)
Complement Inactivator Proteins , Kidney Diseases , Complement Inactivator Proteins/therapeutic use , Complement System Proteins , Humans , Kidney
3.
Kidney Int ; 100(4): 753-779, 2021 10.
Article in English | MEDLINE | ID: mdl-34556300

ABSTRACT

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.


Subject(s)
Glomerulonephritis, IGA , Glomerulonephritis, Membranous , Glomerulonephritis , Nephrosis, Lipoid , Adult , Child , Glomerulonephritis/diagnosis , Glomerulonephritis/therapy , Glomerulonephritis, IGA/diagnosis , Glomerulonephritis, IGA/therapy , Glomerulonephritis, Membranous/diagnosis , Glomerulonephritis, Membranous/drug therapy , Humans , Kidney
4.
Pediatr Nephrol ; 36(8): 2349-2360, 2021 08.
Article in English | MEDLINE | ID: mdl-33693990

ABSTRACT

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.


Subject(s)
Nephrology , Adolescent , Antibodies, Monoclonal, Humanized/adverse effects , Atypical Hemolytic Uremic Syndrome/drug therapy , Atypical Hemolytic Uremic Syndrome/genetics , Child , Humans , Prospective Studies , Retrospective Studies
5.
Kidney Int ; 98(5): 1265-1274, 2020 11.
Article in English | MEDLINE | ID: mdl-32540405

ABSTRACT

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.


Subject(s)
Atypical Hemolytic Uremic Syndrome , Adult , Atypical Hemolytic Uremic Syndrome/diagnosis , Atypical Hemolytic Uremic Syndrome/genetics , Cesarean Section , Child, Preschool , Complement Factor B/genetics , Complement Factor H/genetics , Complement Pathway, Alternative/genetics , Female , Humans , Middle Aged , Mutation , Pregnancy
6.
Semin Immunol ; 28(3): 241-9, 2016 06.
Article in English | MEDLINE | ID: mdl-27402056

ABSTRACT

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.


Subject(s)
Biomarkers/metabolism , Complement C3/immunology , Complement Inactivating Agents/therapeutic use , Complement Pathway, Alternative , Glomerulonephritis, Membranous/therapy , Immunotherapy/methods , Kidney/metabolism , Animals , Complement C3/genetics , Gene Frequency , Genotype , Glomerulonephritis, Membranous/genetics , Glomerulonephritis, Membranous/immunology , Humans , Kidney/pathology , Polymorphism, Genetic
7.
Kidney Int ; 91(3): 539-551, 2017 03.
Article in English | MEDLINE | ID: mdl-27989322

ABSTRACT

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.


Subject(s)
Atypical Hemolytic Uremic Syndrome/immunology , Complement Activation , Complement C3/immunology , Glomerulonephritis/immunology , Kidney/immunology , Animals , Atypical Hemolytic Uremic Syndrome/drug therapy , Atypical Hemolytic Uremic Syndrome/genetics , Atypical Hemolytic Uremic Syndrome/pathology , Complement Activation/drug effects , Complement C3/genetics , Complement Inactivating Agents/therapeutic use , Genetic Predisposition to Disease , Glomerulonephritis/drug therapy , Glomerulonephritis/genetics , Glomerulonephritis/pathology , Humans , Kidney/drug effects , Kidney/pathology , Phenotype , Risk Factors , Treatment Outcome
8.
N Engl J Med ; 371(7): 654-66, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25119611

ABSTRACT

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.


Subject(s)
Thrombotic Microangiopathies , Humans , Purpura, Thrombotic Thrombocytopenic/diagnosis , Purpura, Thrombotic Thrombocytopenic/therapy , Thrombotic Microangiopathies/diagnosis , Thrombotic Microangiopathies/etiology , Thrombotic Microangiopathies/therapy
9.
Am J Kidney Dis ; 70(6): 834-843, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28838767

ABSTRACT

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.


Subject(s)
Autoantibodies/immunology , Complement C3/immunology , Complement System Proteins/immunology , Glomerulonephritis, Membranoproliferative/immunology , Adolescent , Adult , Child , Complement C3 Convertase, Alternative Pathway/immunology , Complement C3 Nephritic Factor/immunology , Complement C5 Convertase, Classical Pathway , Complement Factor H/immunology , Female , Humans , Kidney Diseases/immunology , Male , Young Adult
10.
Kidney Int ; 87(5): 882-4, 2015 May.
Article in English | MEDLINE | ID: mdl-25951068

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Atypical Hemolytic Uremic Syndrome/drug therapy , Female , Humans , Male
12.
Blood ; 122(8): 1487-93, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23847193

ABSTRACT

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.


Subject(s)
ADAM Proteins/deficiency , ADAM Proteins/genetics , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/genetics , ADAMTS13 Protein , Adolescent , Adult , Alleles , Atypical Hemolytic Uremic Syndrome , Autoantibodies/immunology , Child , Child, Preschool , Cohort Studies , Complement Factor H/immunology , DNA/genetics , Female , Humans , Infant , Male , Mutation , Polymorphism, Genetic , Recombinant Fusion Proteins/metabolism , Young Adult
13.
J Am Soc Nephrol ; 25(1): 55-64, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24029428

ABSTRACT

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.


Subject(s)
Blood Coagulation/genetics , Complement System Proteins/genetics , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/genetics , Mutation , Plasminogen/deficiency , Plasminogen/genetics , Adolescent , Adult , Amino Acid Sequence , Atypical Hemolytic Uremic Syndrome , Child , Child, Preschool , Complement Pathway, Alternative/genetics , Conserved Sequence , DNA Copy Number Variations , Female , Genetic Testing , Genetic Variation , Hemolytic-Uremic Syndrome/etiology , Humans , Male , Molecular Sequence Data , Sequence Homology, Amino Acid , Thrombotic Microangiopathies/blood , Thrombotic Microangiopathies/genetics , Thrombotic Microangiopathies/immunology , Young Adult
14.
J Am Soc Nephrol ; 24(11): 1820-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23907509

ABSTRACT

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.


Subject(s)
Complement C3/analysis , Glomerulonephritis, Membranoproliferative/drug therapy , Receptors, Complement 3b/therapeutic use , Animals , Child , Complement Factor H/physiology , Complement Pathway, Alternative , Glomerulonephritis, Membranoproliferative/immunology , Humans , Mice
15.
Adv Kidney Dis Health ; 31(3): 223-233, 2024 May.
Article in English | MEDLINE | ID: mdl-39004462

ABSTRACT

C3 glomerulopathy (C3G) is a rare disorder marked by deposition of C3 in the glomerulus, resulting in damage to the glomerular filtration unit and presenting with features of the nephritic and nephrotic syndromes. Fundamentally, C3G is caused by dysregulation of the alternative pathway of the complement cascade, either due to genetic variants or acquired humoral factors. Despite significant advances in recent years in the understanding of the underlying mechanisms and culprit lesions that result in the development of C3G, treatment options remain severely limited, and the prognosis is often poor. Fortunately, a number of anticomplement therapies are emerging from the drug development pipeline, with several in late-stage testing in patients with C3G, and there is hope that we will soon have more targeted options for managing patients with this devastating disease. In this review, we provide an overview of C3G, as well as summarizing the evidence for current treatments and detailing the clinical trials that are currently underway.


Subject(s)
Complement C3 , Humans , Complement C3/metabolism , Complement C3/genetics , Complement C3/immunology , Kidney Glomerulus/pathology , Kidney Glomerulus/metabolism , Complement Pathway, Alternative/genetics , Complement Pathway, Alternative/drug effects , Glomerulonephritis/pathology , Glomerulonephritis/metabolism , Glomerulonephritis/genetics , Glomerulonephritis/immunology , Glomerulonephritis/therapy , Glomerulonephritis/diagnosis , Nephrotic Syndrome/pathology , Nephrotic Syndrome/genetics , Nephrotic Syndrome/metabolism , Nephrotic Syndrome/diagnosis , Nephrotic Syndrome/therapy
16.
Kidney Int Rep ; 9(2): 464-477, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38344720

ABSTRACT

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.

17.
Kidney Int ; 84(6): 1079-89, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24172683

ABSTRACT

C3 glomerulopathy is a recently introduced pathological entity whose original definition was glomerular pathology characterized by C3 accumulation with absent or scanty immunoglobulin deposition. In August 2012, an invited group of experts (comprising the authors of this document) in renal pathology, nephrology, complement biology, and complement therapeutics met to discuss C3 glomerulopathy in the first C3 Glomerulopathy Meeting. The objectives were to reach a consensus on: the definition of C3 glomerulopathy, appropriate complement investigations that should be performed in these patients, and how complement therapeutics should be explored in the condition. This meeting report represents the current consensus view of the group.


Subject(s)
Complement C3/analysis , Glomerulonephritis/immunology , Kidney Glomerulus/immunology , Biomedical Research , Biopsy , Cooperative Behavior , Glomerulonephritis/diagnosis , Glomerulonephritis/therapy , Humans , International Cooperation , Kidney Glomerulus/pathology , Predictive Value of Tests , Prognosis
18.
Curr Opin Nephrol Hypertens ; 22(2): 231-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23318699

ABSTRACT

PURPOSE OF REVIEW: The purpose of this review is to discuss emerging nomenclature, review the salient clinicopathological features and describe the therapeutic options available for the treatment of C3 glomerulopathy (C3G). RECENT FINDINGS: C3G is minimally responsive to traditional immune suppression and randomized controlled trials to support therapy are absent. The burgeoning understanding of the role of the alternative complement pathway in C3G combined with animal data supporting the use of terminal complement blockade and a few reports suggesting that the anticomplement drug eculizumab may offer a therapeutic advantage have triggered great interest in the field of complement-mediated renal disease. SUMMARY: Anticellular immune suppression and plasma therapy have limited efficacy in C3G. Data suggest that eculizumab may ameliorate disease in some C3G patients. The limited, recently published cohort data highlight crucial aspects of this group of diseases and support the need for extensive genetic and biomarker research to validate the pathologic mechanisms, delineate the spectrum of disease and guide the design of the rigorous trials to identify effective therapies for the treatment of C3G.


Subject(s)
Complement Activation/drug effects , Complement C3/metabolism , Glomerulonephritis, Membranoproliferative/immunology , Immunosuppressive Agents/therapeutic use , Kidney/drug effects , Adolescent , Adult , Animals , Complement Pathway, Alternative/drug effects , Female , Glomerulonephritis, Membranoproliferative/diagnosis , Glomerulonephritis, Membranoproliferative/drug therapy , Humans , Immunosuppressive Agents/adverse effects , Kidney/immunology , Kidney/pathology , Treatment Outcome , Young Adult
19.
Curr Opin Pediatr ; 25(2): 225-31, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23486421

ABSTRACT

PURPOSE OF REVIEW: This review considers the use of eculizumab in the treatment of atypical haemolytic uraemic syndrome (aHUS) as well as the other complement-mediated renal diseases, including dense deposit disease (DDD) and C3 glomerulonephritis (C3GN). In addition, a brief discussion of the effectiveness of eculizumab for the prevention of antibody-mediated rejection (AMR) in the setting of renal transplant and the treatment of shiga toxin associated haemolytic uraemic syndrome (STEC HUS) is also provided. RECENT FINDINGS: No randomized controlled trials exist to support the use of eculizumab in renal disease. The results of two unpublished, prospective adult and adolescent trials support its utility in aHUS, whereas retrospective data support the effectiveness in paediatric aHUS. These two data sets form the basis of the sole renal indication for eculizumab. One small, single-centre trial and a growing number of case reports support the use of eculizumab in C3 glomerulopathy (C3G). There are limited trial data in AMR and renal transplant. Finally, there are conflicting data for the use of eculizumab in STEC HUS. SUMMARY: The cumulative published data establish the effectiveness of eculizumab in a select group of renal diseases that have at the centre of their disease either abnormal complement control or maladaptive complement activation.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Complement Inactivating Agents/therapeutic use , Hemolytic-Uremic Syndrome/drug therapy , Atypical Hemolytic Uremic Syndrome , Complement Activation/immunology , Complement C3/immunology , Glomerulonephritis/drug therapy , Glomerulonephritis/immunology , Graft Rejection/prevention & control , Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/surgery , Humans , Kidney Transplantation
20.
Pediatr Nephrol ; 28(11): 2221-5, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23880784

ABSTRACT

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.


Subject(s)
Complement C3b Inactivator Proteins/genetics , Complement Factor H/genetics , Hemolytic-Uremic Syndrome/genetics , Antibodies, Monoclonal, Humanized/therapeutic use , Autoantibodies/analysis , Blotting, Western , Child , Creatinine/blood , DNA/genetics , Female , Gene Amplification , Humans , Immunosuppressive Agents/therapeutic use , Kidney Transplantation , Polymerase Chain Reaction , Renal Dialysis
SELECTION OF CITATIONS
SEARCH DETAIL