Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Invest ; 123(6): 2434-46, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23728178

ABSTRACT

C3 glomerulopathies (C3G) are a group of severe renal diseases with distinct patterns of glomerular inflammation and C3 deposition caused by complement dysregulation. Here we report the identification of a familial C3G-associated genomic mutation in the gene complement factor H­related 1 (CFHR1), which encodes FHR1. The mutation resulted in the duplication of the N-terminal short consensus repeats (SCRs) that are conserved in FHR2 and FHR5. We determined that native FHR1, FHR2, and FHR5 circulate in plasma as homo- and hetero-oligomeric complexes, the formation of which is likely mediated by the conserved N-terminal domain. In mutant FHR1, duplication of the N-terminal domain resulted in the formation of unusually large multimeric FHR complexes that exhibited increased avidity for the FHR1 ligands C3b, iC3b, and C3dg and enhanced competition with complement factor H (FH) in surface plasmon resonance (SPR) studies and hemolytic assays. These data revealed that FHR1, FHR2, and FHR5 organize a combinatorial repertoire of oligomeric complexes and demonstrated that changes in FHR oligomerization influence the regulation of complement activation. In summary, our identification and characterization of a unique CFHR1 mutation provides insights into the biology of the FHRs and contributes to our understanding of the pathogenic mechanisms underlying C3G.


Subject(s)
Complement C3/metabolism , Complement C3b Inactivator Proteins/genetics , Kidney Diseases/genetics , Child , Complement C3/chemistry , Complement C3b Inactivator Proteins/chemistry , Complement C3b Inactivator Proteins/metabolism , Complement System Proteins/isolation & purification , Complement System Proteins/metabolism , Female , Gene Duplication , Hemolysis , Humans , Immobilized Proteins/chemistry , Immobilized Proteins/metabolism , Kidney/pathology , Male , Middle Aged , Mutagenesis, Insertional , Pedigree , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Sequence Analysis, DNA
2.
Kidney Int ; 80(4): 397-404, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21677636

ABSTRACT

The autoimmune form of atypical hemolytic uremic syndrome (HUS) is characterized by circulating autoantibodies against the complement regulator factor H, and is often associated with deficiency of the factor H-related proteins CFHR1 and CFHR3. Here we studied whether anti-factor H autoantibodies crossreact with CFHR1, and determined functional consequences of this. In ELISA, anti-factor H immunoglobulin G (IgG) autoantibodies from 24 atypical HUS patients bound to the short consensus repeat 20 domain of factor H, 21 antibodies also recognized CFHR1, but none CFHR3. Three patients also had anti-factor H IgA autoantibodies crossreacting with CFHR1. Analysis of the IgG fractions in CFHR1-deficient patients found that CFHR1-IgG complexes were formed during plasma exchange treatment, indicating that autoantibodies recognize CFHR1 in vivo. Recombinant CFHR1 prevented hemolysis of sheep erythrocytes caused by patient plasma containing anti-factor H IgG, but it did not inhibit red cell lysis caused by a factor H mutation (W1183 L) in the short consensus repeat 20 domain. Thus, exogenous CFHR1 provided during plasma exchange therapy may neutralize anti-factor H autoantibodies and help in the treatment of autoimmune atypical HUS.


Subject(s)
Autoantibodies/blood , Autoantigens , Autoimmunity , Complement C3b Inactivator Proteins/immunology , Hemolytic-Uremic Syndrome/immunology , Immunoglobulin G/blood , Animals , Antibody Specificity , Antigen-Antibody Complex , Atypical Hemolytic Uremic Syndrome , Autoantigens/genetics , Blood Proteins/immunology , Case-Control Studies , Complement C3b Inactivator Proteins/genetics , Complement Factor H/immunology , Enzyme-Linked Immunosorbent Assay , Germany , Hemolysis , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/therapy , Humans , Mutation , Plasma Exchange , Sheep , Spain
3.
Blood ; 114(19): 4261-71, 2009 Nov 05.
Article in English | MEDLINE | ID: mdl-19745068

ABSTRACT

The factor H-related protein family (CFHR) is a group of minor plasma proteins genetically and structurally related to complement factor H (fH). Notably, deficiency of CFHR1/CFHR3 associates with protection against age-related macular degeneration and with the presence of anti-fH autoantibodies in atypical hemolytic uremic syndrome (aHUS). We have developed a proteomics strategy to analyze the CFHR proteins in plasma samples from controls, patients with aHUS, and patients with type II membranoproliferative glomerulonephritis. Here, we report on the identification of persons carrying novel deficiencies of CFHR1, CFHR3, and CFHR1/CFHR4A, resulting from point mutations in CFHR1 and CFHR3 or from a rearrangement involving CFHR1 and CFHR4. Remarkably, patients with aHUS lacking CFHR1, but not those lacking CFHR3, present anti-fH autoantibodies, suggesting that generation of these antibodies is specifically related to CFHR1 deficiency. We also report the characterization of a novel CFHR1 polymorphism, resulting from a gene conversion event between CFH and CFHR1, which strongly associates with aHUS. The risk allotype CFHR1*B, with greater sequence similarity to fH, may compete with fH, decreasing protection of cellular surfaces against complement damage. In summary, our comprehensive analyses of the CFHR proteins have improved our understanding of these proteins and provided further insights into aHUS pathogenesis.


Subject(s)
Complement C3b Inactivator Proteins/genetics , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/genetics , Mutation , Adolescent , Adult , Aged , Alleles , Autoantibodies/blood , Base Sequence , Blood Proteins/genetics , Case-Control Studies , Child , Child, Preschool , Complement C3b Inactivator Proteins/deficiency , Complement Factor H/genetics , Complement Factor H/immunology , Female , Gene Conversion , Glomerulonephritis, Membranoproliferative/blood , Glomerulonephritis, Membranoproliferative/genetics , Hemolytic-Uremic Syndrome/immunology , Humans , Macular Degeneration/blood , Macular Degeneration/genetics , Male , Middle Aged , Oligonucleotide Probes/genetics , Polymorphism, Single Nucleotide , Protein Isoforms/genetics , Proteomics , Young Adult
4.
Mol Immunol ; 45(10): 2764-71, 2008 May.
Article in English | MEDLINE | ID: mdl-18374984

ABSTRACT

Complement Factor I (CFI) is a regulator of the classical and alternative pathways. CFI has enzymatic activity and is able to cleave C3b and C4b. Homozygous Factor I deficiency is associated with infectious and/or autoimmune diseases. Here we describe the biochemical and genetic characterization in two Spanish families with complete Factor I deficiency. In Family 1, the propositus suffered from several episodes of meningitis for more than a year. Biochemical complement studies showed undetectable Factor I levels in the propositus and in her sister, while their parents and a brother had partial Factor I deficiency and were healthy. In Family 2, three out of five children were homozygous for Factor I deficiency, two of whom suffered from meningitis and the third one from several infections. The parents and the other two siblings were healthy and heterozygous for Factor I deficiency. Molecular studies showed that the two families had different mutations at exon 5 of the Factor I gene, which codifies for module LDLr1. One mutation corresponds to a 772G>A change at the donor splice site that was originally found in a family from Northern England. The second is a new missense mutation 739T>G, that generates a Cys to Gly change.


Subject(s)
Complement Factor I/deficiency , Complement Factor I/genetics , White People/genetics , Adolescent , Adult , Base Sequence , Blotting, Western , Child , Child, Preschool , Complement Factor I/chemistry , Contig Mapping , DNA Mutational Analysis , DNA, Complementary , Family , Female , Genome, Human/genetics , Humans , Male , Molecular Sequence Data , Mutation/genetics , Pedigree , Protein Structure, Tertiary , Spain
5.
Am J Kidney Dis ; 52(1): 171-80, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18423815

ABSTRACT

Atypical hemolytic uremic syndrome is associated with mutations in the complement proteins factor H, factor I, factor B, C3, or membrane cofactor protein in about 50% of patients. The evolution and prognosis of the disease in patients carrying mutations in factor H is particularly poor, and renal transplantation most often fails because of recurrence of the disease in the graft. The risk of rapid loss of renal function in patients with functional mutations in factor H requires that effective treatment be initiated as soon as possible, but identification of these patients relies on genetic studies that are time consuming. We describe a case in which an in vitro hemolytic assay proved useful for rapidly assessing factor H dysfunction and for testing whether this dysfunction could be corrected with fresh frozen plasma. In the context of this case, we summarize recent advances in understanding the molecular mechanisms contributing to atypical hemolytic uremic syndrome, including descriptions of DNA- and protein-based analysis. We conclude that functional analysis of factor H should help rationalize the plasma treatment of patients with atypical hemolytic uremic syndrome.


Subject(s)
Complement Factor H/genetics , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/physiopathology , Mutation , Complement Activation , DNA Mutational Analysis , Disease Progression , Fatal Outcome , Female , Hemolytic-Uremic Syndrome/therapy , Humans , Infant , Plasmapheresis , Renal Dialysis/methods , Respiratory Insufficiency , Severity of Illness Index
6.
Hum Mutat ; 28(3): 222-34, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17089378

ABSTRACT

Atypical hemolytic uremic syndrome (aHUS) is a disease of hemolytic anemia, thrombocytopenia, and renal failure associated with defective alternative pathway (AP) complement control. Previously, we presented a database (www.FH-HUS.org) focusing on aHUS mutations in the Factor H gene (CFH). Here, new aHUS mutations are reported for the complement regulatory proteins Factor H (FH), Factor I (FI), and membrane cofactor protein (MCP). Additional mutations or polymorphisms within CFH have been associated with membranoproliferative glomerulonephritis (MPGN) and age-related macular degeneration (AMD). Accordingly, the database now includes substitutions that predispose to aHUS, MPGN, and AMD. For this, structural models for the domains in MCP and FI were developed using homology modeling. With this new database, patients with mutations in more than one gene can be displayed and interpreted in a coherent manner. The database also includes SNP polymorphisms in CFH, MCP, and IF. There are now a total of 167 genetic alterations, including 100 in CFH, 43 in MCP, and 24 in IF. The mutations characterize clinical outcomes that vary from several AMD-associated polymorphisms to those associated with aHUS, MPGN, or FI deficiency. A consensus short complement regulator (SCR) domain structure facilitated the interpretations of aHUS mutations. Specific locations within this consensus domain often correlate with the occurrence of clinical phenotypes. The AMD Tyr402His polymorphism is structurally located at a hotspot for several aHUS mutations. The database emphasizes the causative role of the alternative pathway of complement in disease and provides a repository of knowledge to assist future diagnosis and novel therapeutic approaches.


Subject(s)
Complement Factor H/genetics , Databases, Genetic , Fibrinogen/genetics , Hemolytic-Uremic Syndrome/genetics , Membrane Cofactor Protein/genetics , Amino Acid Sequence , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Protein Structure, Tertiary/genetics , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...