Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Infect Immun ; 87(2)2019 02.
Article in English | MEDLINE | ID: mdl-30510105

ABSTRACT

Neisseria gonorrhoeae, the causative agent of gonorrhea, has evolved several mechanisms to subvert complement, including binding of the complement inhibitor factor H (FH). We previously reported FH binding to N. gonorrhoeae independently of lipooligosaccharide (LOS) sialylation. Here we report that factor H-like protein 1 (FHL-1), which contains FH domains 1 through 7 and possesses complement-inhibitory activity, also binds to N. gonorrhoeae The ligand for both FH and FHL-1 was identified as neisserial surface protein A (NspA), which has previously been identified as a ligand for these molecules on Neisseria meningitidis As with N. meningitidis NspA (Nm-NspA), N. gonorrhoeae NspA (Ng-NspA) bound FH/FHL-1 through FH domains 6 and 7. Binding of FH/FHL-1 to NspA was human specific; the histidine (H) at position 337 of domain 6 contributed to human-specific FH binding to both Ng- and Nm-NspA. FH/FHL-1 bound Nm-NspA better than Ng-NspA; introducing Q at position 73 (loop 2, present in Ng-NspA) or replacing V and D at positions 112 and 113 in Nm-NspA loop 3 with A and H (Ng-NspA), respectively, reduced FH/FHL-1 binding. The converse Ng-NspA to Nm-NspA mutations increased FH/FHL-1 binding. Binding of FH/FHL-1 through domains 6 and 7 to N. gonorrhoeae increased with truncation of the heptose I (HepI) chain of LOS and decreased with LOS sialylation. Loss of NspA significantly decreased serum resistance of N. gonorrhoeae with either wild-type or truncated LOS. This report highlights the role for NspA in enabling N. gonorrhoeae to subvert complement despite LOS phase variation. Knowledge of FH-NspA interactions will inform the design of vaccines and immunotherapies against the global threat of multidrug-resistant gonorrhea.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Complement C3b Inactivator Proteins/immunology , Complement Factor H/immunology , Gonorrhea/immunology , Neisseria gonorrhoeae/immunology , Neisseria meningitidis/immunology , Bacterial Adhesion/immunology , Cells, Cultured , Humans , Neisseria gonorrhoeae/pathogenicity
2.
Front Immunol ; 9: 1635, 2018.
Article in English | MEDLINE | ID: mdl-30166981

ABSTRACT

The opportunistic fungal pathogen Aspergillus fumigatus can cause life-threatening infections, particularly in immunocompromised patients. Most pathogenic microbes control host innate immune responses at the earliest time, already before infiltrating host immune cells arrive at the site of infection. Here, we identify Aspf2 as the first A. fumigatus Factor H-binding protein. Aspf2 recruits several human plasma regulators, Factor H, factor-H-like protein 1 (FHL-1), FHR1, and plasminogen. Factor H contacts Aspf2 via two regions located in SCRs6-7 and SCR20. FHL-1 binds via SCRs6-7, and FHR1 via SCRs3-5. Factor H and FHL-1 attached to Aspf2-maintained cofactor activity and assisted in C3b inactivation. A Δaspf2 knockout strain was generated which bound Factor H with 28% and FHL-1 with 42% lower intensity. In agreement with less immune regulator acquisition, when challenged with complement-active normal human serum, Δaspf2 conidia had substantially more C3b (>57%) deposited on their surface. Consequently, Δaspf2 conidia were more efficiently phagocytosed (>20%) and killed (44%) by human neutrophils as wild-type conidia. Furthermore, Aspf2 recruited human plasminogen and, when activated by tissue-type plasminogen activator, newly generated plasmin cleaved the chromogenic substrate S2251 and degraded fibrinogen. Furthermore, plasmin attached to conidia damaged human lung epithelial cells, induced cell retraction, and caused matrix exposure. Thus, Aspf2 is a central immune evasion protein and plasminogen ligand of A. fumigatus. By blocking host innate immune attack and by disrupting human lung epithelial cell layers, Aspf2 assists in early steps of fungal infection and likely allows tissue penetration.


Subject(s)
Aspergillosis/immunology , Aspergillus fumigatus/immunology , Fungal Proteins/immunology , Aspergillosis/microbiology , Complement C3b Inactivator Proteins/immunology , Complement C3b Inactivator Proteins/metabolism , Complement Factor H/immunology , Complement Factor H/metabolism , Fungal Proteins/genetics , Fungal Proteins/isolation & purification , Fungal Proteins/metabolism , Humans , Immune Evasion , Immunity, Innate , Plasminogen/immunology , Plasminogen/metabolism , Protein Binding
3.
Semin Immunopathol ; 40(1): 15-35, 2018 01.
Article in English | MEDLINE | ID: mdl-29167939

ABSTRACT

This review is not intended to cover in detail all aspects of the discovery and evolution of our understanding of the "alternative pathway" of complement activation, there are many excellent reviews that do this (see Fearon (CRC Crit Rev Immunol 1:1-32, 1979), Pangburn and Müller-Eberhard (Springer Semin Immunopathol 7:163-192, 1984)), but instead to give sufficient background for current concepts to be put in context. The prevailing textbook view, of components having a primary role as an alternative "pathway" for C3 activation, is challenged, with an argument developed for the primary role of the system being that of providing a surface-dependent amplification loop for both C3 and C5 activation. Whatever the mechanism by which the initial C3b molecule is generated, deposition onto a surface has the potential to target that surface for elimination. Elimination or escape from initial targeting is determined by a sophisticated and highly regulated amplification loop for C3 activation. This viewpoint of the system is then briefly developed to provide a context for therapeutic treatment of disease caused, at least in part, by dysregulated amplification of C3 activation, and to highlight some of the challenges that such therapies will face and need to address.


Subject(s)
Complement Pathway, Alternative , Properdin/metabolism , Signal Transduction , Animals , Carrier Proteins/metabolism , Cell Membrane/immunology , Cell Membrane/metabolism , Complement Activation/immunology , Complement C3 Nephritic Factor/immunology , Complement C3 Nephritic Factor/metabolism , Complement C3-C5 Convertases/chemistry , Complement C3-C5 Convertases/immunology , Complement C3-C5 Convertases/metabolism , Complement C3b Inactivator Proteins/immunology , Complement C3b Inactivator Proteins/metabolism , Elapid Venoms/immunology , Elapid Venoms/metabolism , Host-Pathogen Interactions/immunology , Humans , Properdin/immunology , Protein Binding
4.
J Immunol ; 199(1): 292-303, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28533443

ABSTRACT

Factor H-related protein (FHR) 1 is one of the five human FHRs that share sequence and structural homology with the alternative pathway complement inhibitor FH. Genetic studies on disease associations and functional analyses indicate that FHR-1 enhances complement activation by competitive inhibition of FH binding to some surfaces and immune proteins. We have recently shown that FHR-1 binds to pentraxin 3. In this study, our aim was to investigate whether FHR-1 binds to another pentraxin, C-reactive protein (CRP), analyze the functional relevance of this interaction, and study the role of FHR-1 in complement activation and regulation. FHR-1 did not bind to native, pentameric CRP, but it bound strongly to monomeric CRP via its C-terminal domains. FHR-1 at high concentration competed with FH for CRP binding, indicating possible complement deregulation also on this ligand. FHR-1 did not inhibit regulation of solid-phase C3 convertase by FH and did not inhibit terminal complement complex formation induced by zymosan. On the contrary, by binding C3b, FHR-1 allowed C3 convertase formation and thereby enhanced complement activation. FHR-1/CRP interactions increased complement activation via the classical and alternative pathways on surfaces such as the extracellular matrix and necrotic cells. Altogether, these results identify CRP as a ligand for FHR-1 and suggest that FHR-1 enhances, rather than inhibits, complement activation, which may explain the protective effect of FHR-1 deficiency in age-related macular degeneration.


Subject(s)
C-Reactive Protein/immunology , C-Reactive Protein/metabolism , Complement Activation , Complement C3b Inactivator Proteins/immunology , Complement C3b Inactivator Proteins/metabolism , Binding Sites , C-Reactive Protein/chemistry , C-Reactive Protein/pharmacology , Complement C3-C5 Convertases , Complement C3b/immunology , Complement C3b/pharmacology , Complement C3b Inactivator Proteins/pharmacology , Complement Factor H , Extracellular Matrix/drug effects , Extracellular Matrix/immunology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Humans , Ligands , Macular Degeneration/immunology , Protein Binding , Serum Amyloid P-Component/immunology , Serum Amyloid P-Component/metabolism
5.
J Immunol ; 196(2): 866-76, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26643478

ABSTRACT

The serum proteins factor H (FH), consisting of 20 complement control protein modules (CCPs), and its splice product FH-like protein 1 (FHL-1; consisting of CCPs 1-7) are major regulators of the alternative pathway (AP) of complement activation. The engineered version of FH, miniFH, contains only the N- and C-terminal portions of FH linked by an optimized peptide and shows ∼ 10-fold higher ex vivo potency. We explored the hypothesis that regulatory potency is enhanced by unmasking of a ligand-binding site in the C-terminal CCPs 19-20 that is cryptic in full-length native FH. Therefore, we produced an FH variant lacking the central domains 10-15 (FHΔ10-15). To explore how avidity affects regulatory strength, we generated a duplicated version of miniFH, termed midiFH. We compared activities of FHΔ10-15 and midiFH to miniFH, FH, and FHL-1. Relative to FH, FHΔ10-15 exhibited an altered binding profile toward C3 activation products and a 5-fold-enhanced complement regulation on a paroxysmal nocturnal hemoglobinuria patient's erythrocytes. Contrary to dogma, FHL-1 and FH exhibited equal regulatory activity, suggesting that the role of FHL-1 in AP regulation has been underestimated. Unexpectedly, a substantially increased avidity for complement opsonins, as seen in midiFH, did not potentiate the inhibitory potential on host cells. In conclusion, comparisons of engineered and native FH-based regulators have identified features that determine high AP regulatory activity on host cells. Unrestricted availability of FH CCPs 19-20 and an optimal spatial orientation between the N- and C-terminal FH regions are key.


Subject(s)
Complement C3b Inactivator Proteins/immunology , Complement Factor H/immunology , Complement Inactivating Agents/pharmacology , Complement Pathway, Alternative/immunology , Recombinant Proteins/pharmacology , Amino Acid Sequence , Complement Factor H/chemistry , Complement Inactivating Agents/chemical synthesis , Complement Inactivating Agents/immunology , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Molecular Sequence Data , Polymerase Chain Reaction , Protein Binding , Recombinant Proteins/chemical synthesis , Recombinant Proteins/immunology
6.
Trends Immunol ; 36(6): 374-84, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25979655

ABSTRACT

Complement factor H-related proteins (FHRs) are strongly associated with different diseases involving complement dysregulation, which suggests a major role for these proteins regulating complement activation. Because FHRs are evolutionarily and structurally related to complement inhibitor factor H (FH), the initial assumption was that the FHRs are also negative complement regulators. Whereas weak complement inhibiting activities were originally reported for these molecules, recent developments indicate that FHRs may enhance complement activation, with important implications for the role of these proteins in health and disease. We review these findings here, and propose that FHRs represent a complex set of surface recognition molecules that, by competing with FH, provide improved discrimination of self and non-self surfaces and play a central role in determining appropriate activation of the complement pathway.


Subject(s)
Apolipoproteins/immunology , Blood Proteins/immunology , Complement Activation/immunology , Complement C3b Inactivator Proteins/immunology , Complement System Proteins/immunology , Apolipoproteins/genetics , Blood Proteins/genetics , Complement C3b Inactivator Proteins/genetics , Complement Factor H/genetics , Complement Factor H/immunology , Complement System Proteins/genetics , Genetic Predisposition to Disease/genetics , Humans , Models, Immunological
7.
Mol Immunol ; 67(1): 31-42, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25843230

ABSTRACT

Tremendous advances in our understanding of the thrombotic microangiopathies (TMAs) have revealed distinct disease mechanisms within this heterogeneous group of diseases. As a direct result of this knowledge, both children and adults with complement-mediated TMA now enjoy higher expectations for long-term health. In this update on atypical hemolytic uremic syndrome, we review the clinical characteristics; the genetic and acquired drivers of disease; the broad spectrum of environmental triggers; and current diagnosis and treatment options. Many questions remain to be addressed if additional improvements in patient care and outcome are to be achieved in the coming decade.


Subject(s)
Atypical Hemolytic Uremic Syndrome/pathology , Communicable Diseases/pathology , Gene Expression Regulation/immunology , Kidney Neoplasms/pathology , Kidney/pathology , Adult , Atypical Hemolytic Uremic Syndrome/etiology , Atypical Hemolytic Uremic Syndrome/genetics , Atypical Hemolytic Uremic Syndrome/immunology , Autoantibodies/biosynthesis , Child , Communicable Diseases/complications , Communicable Diseases/genetics , Communicable Diseases/immunology , Complement Activation , Complement C3b/genetics , Complement C3b/immunology , Complement C3b Inactivator Proteins/genetics , Complement C3b Inactivator Proteins/immunology , Complement Factor H/genetics , Complement Factor H/immunology , Genetic Predisposition to Disease , Humans , Kidney/immunology , Kidney Neoplasms/complications , Kidney Neoplasms/genetics , Kidney Neoplasms/immunology , Kidney Transplantation
8.
Mol Immunol ; 67(1): 108-18, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25886718

ABSTRACT

In recent years, the view of platelets has changed from mere elements of hemostasis to immunological multitaskers. They are connected in manifold ways to other cellular and humoral components of the immune network, one of which is the complement system, a potent player in soluble innate immunity. Our article reviews the crucial and complex interplay between platelets and complement, focusing on mutual regulation of these two interaction partners by their respective molecular mechanisms. Furthermore, the putative relevance of these processes to infectious diseases, inflammatory conditions, and autoimmune disorders, as well as the treatment of patients with biomaterials is highlighted.


Subject(s)
Autoimmune Diseases/immunology , Bacterial Infections/immunology , Blood Platelets/immunology , Complement System Proteins/immunology , Mycoses/immunology , Virus Diseases/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Bacterial Infections/genetics , Bacterial Infections/microbiology , Bacterial Infections/pathology , Blood Platelets/pathology , Complement Activation , Complement C3b Inactivator Proteins/genetics , Complement C3b Inactivator Proteins/immunology , Complement System Proteins/genetics , Gene Expression Regulation , Humans , Immunity, Innate , Mycoses/genetics , Mycoses/microbiology , Mycoses/pathology , Receptors, Complement/genetics , Receptors, Complement/immunology , Signal Transduction , Virus Diseases/genetics , Virus Diseases/pathology , Virus Diseases/virology
9.
Mol Immunol ; 67(1): 71-84, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25816986

ABSTRACT

Malaria remains one of the world's deadliest diseases. Plasmodium falciparum is responsible for the most severe and lethal form of human malaria. P. falciparum's life cycle involves two obligate hosts: human and mosquito. From initial entry into these hosts, malaria parasites face the onslaught of the first line of host defence, the complement system. In this review, we discuss the complex interaction between complement and malaria infection in terms of hosts immune responses, parasite survival and pathogenesis of severe forms of malaria. We will focus on the role of complement receptor 1 and its associated polymorphisms in malaria immune complex clearance, as a mediator of parasite rosetting and as an entry receptor for P. falciparum invasion. Complement evasion strategies of P. falciparum parasites will also be highlighted. The sexual forms of the malaria parasites recruit the soluble human complement regulator Factor H to evade complement-mediated killing within the mosquito host. A novel evasion strategy is the deployment of parasite organelles to divert complement attack from infective blood stage parasites. Finally we outline the future challenge to understand the implications of these exploitation mechanisms in the interplay between successful infection of the host and pathogenesis observed in severe malaria.


Subject(s)
Complement C3b Inactivator Proteins/immunology , Life Cycle Stages/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Receptors, Complement/immunology , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/genetics , Complement Activation , Complement C3b Inactivator Proteins/genetics , Complement Factor H/genetics , Complement Factor H/immunology , Gene Expression , Humans , Immune Evasion , Life Cycle Stages/genetics , Malaria, Falciparum/genetics , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Polymorphism, Genetic , Receptors, Complement/genetics
10.
Mol Immunol ; 67(1): 43-50, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25804937

ABSTRACT

Age-related macular degeneration (AMD) is a leading cause of visual impairment. It is characterised by damage to a tissue complex composed of the retinal pigment epithelium, Bruch's membrane and choriocapillaris. In early AMD extracellular debris including drusen accumulates in Bruch's membrane and then in late AMD geographic atrophy and/or neovascularisation develop. Variants in genes encoding components of the alternative pathway of the complement cascade have a major influence on AMD risk, especially at the RCA locus on chromosome 1, which contains CFH and the CFHR genes. Immunohistochemical studies have demonstrated complement components in unaffected and AMD macular tissue. Whilst other factors, including oxidative stress, play important roles in AMD pathogenesis, evidence for the central role played by complement dysregulation is discussed in this review.


Subject(s)
Bruch Membrane/pathology , Complement C3b Inactivator Proteins/immunology , Macular Degeneration/pathology , Retinal Pigment Epithelium/pathology , Bruch Membrane/chemistry , Bruch Membrane/immunology , Chromosomes, Human, Pair 1 , Complement Activation , Complement C3b Inactivator Proteins/genetics , Complement Factor H/genetics , Complement Factor H/immunology , Gene Expression Regulation , Genetic Loci , Humans , Macular Degeneration/genetics , Macular Degeneration/immunology , Oxidative Stress , Retinal Pigment Epithelium/chemistry , Retinal Pigment Epithelium/immunology
11.
J Biol Chem ; 290(15): 9500-10, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25659429

ABSTRACT

Atypical hemolytic uremic syndrome (aHUS) is characterized by complement attack against host cells due to mutations in complement proteins or autoantibodies against complement factor H (CFH). It is unknown why nearly all patients with autoimmune aHUS lack CFHR1 (CFH-related protein-1). These patients have autoantibodies against CFH domains 19 and 20 (CFH19-20), which are nearly identical to CFHR1 domains 4 and 5 (CFHR14-5). Here, binding site mapping of autoantibodies from 17 patients using mutant CFH19-20 constructs revealed an autoantibody epitope cluster within a loop on domain 20, next to the two buried residues that are different in CFH19-20 and CFHR14-5. The crystal structure of CFHR14-5 revealed a difference in conformation of the autoantigenic loop in the C-terminal domains of CFH and CFHR1, explaining the variation in binding of autoantibodies from some aHUS patients to CFH19-20 and CFHR14-5. The autoantigenic loop on CFH seems to be generally flexible, as its conformation in previously published structures of CFH19-20 bound to the microbial protein OspE and a sialic acid glycan is somewhat altered. Cumulatively, our data suggest that association of CFHR1 deficiency with autoimmune aHUS could be due to the structural difference between CFHR1 and the autoantigenic CFH epitope, suggesting a novel explanation for CFHR1 deficiency in the pathogenesis of autoimmune aHUS.


Subject(s)
Autoantibodies/chemistry , Complement C3b Inactivator Proteins/chemistry , Complement Factor H/chemistry , Epitopes/chemistry , Atypical Hemolytic Uremic Syndrome/genetics , Atypical Hemolytic Uremic Syndrome/immunology , Atypical Hemolytic Uremic Syndrome/metabolism , Autoantibodies/immunology , Autoimmunity/genetics , Autoimmunity/immunology , Binding Sites/genetics , Binding Sites/immunology , Complement C3b Inactivator Proteins/genetics , Complement C3b Inactivator Proteins/immunology , Complement Factor H/genetics , Complement Factor H/immunology , Crystallography, X-Ray , Epitope Mapping , Epitopes/immunology , Humans , Models, Molecular , Mutation , Peptide Fragments/chemistry , Peptide Fragments/immunology , Protein Binding/immunology , Protein Structure, Tertiary
12.
J Immunol ; 193(10): 4962-70, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25305316

ABSTRACT

The tight regulation of innate immunity on extracellular matrix (ECM) is a vital part of immune homeostasis throughout the human body, and disruption to this regulation in the eye is thought to contribute directly to the progression of age-related macular degeneration (AMD). The plasma complement regulator factor H (FH) is thought to be the main regulator that protects ECM against damaging complement activation. However, in the present study we demonstrate that a truncated form of FH, called FH-like protein 1 (FHL-1), is the main regulatory protein in the layer of ECM under human retina, called Bruch's membrane. Bruch's membrane is a major site of AMD disease pathogenesis and where drusen, the hallmark lesions of AMD, form. We show that FHL-1 can passively diffuse through Bruch's membrane, whereas the full sized, glycosylated, FH cannot. FHL-1 is largely bound to Bruch's membrane through interactions with heparan sulfate, and we show that the common Y402H polymorphism in the CFH gene, associated with an increased risk of AMD, reduces the binding of FHL-1 to this heparan sulfate. We also show that FHL-1 is retained in drusen whereas FH coats the periphery of the lesions, perhaps inhibiting their clearance. Our results identify a novel mechanism of complement regulation in the human eye, which highlights potential new avenues for therapeutic strategies.


Subject(s)
Bruch Membrane/metabolism , Complement C3b Inactivator Proteins/metabolism , Complement Factor H/metabolism , Macular Degeneration/metabolism , Retina/metabolism , Retinal Drusen/metabolism , Bruch Membrane/immunology , Bruch Membrane/pathology , Complement Activation , Complement C3b Inactivator Proteins/genetics , Complement C3b Inactivator Proteins/immunology , Complement Factor H/genetics , Complement Factor H/immunology , Extracellular Matrix/immunology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Gene Expression Regulation , Glycosylation , Heparitin Sulfate/immunology , Heparitin Sulfate/metabolism , Homeostasis , Humans , Immunity, Innate , Macular Degeneration/genetics , Macular Degeneration/immunology , Macular Degeneration/pathology , Protein Binding , Protein Transport , Retina/immunology , Retina/pathology , Retinal Drusen/genetics , Retinal Drusen/immunology , Retinal Drusen/pathology , Signal Transduction
13.
Semin Thromb Hemost ; 40(4): 431-43, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24799303

ABSTRACT

The presence of circulating autoantibodies, primarily to complement factor H antibodies (CFH-Abs) in plasma characterizes the autoimmune form of atypical hemolytic uremic syndrome (aHUS). This acquired form of aHUS defines a distinct subgroup of aHUS patients, which requires diagnostic and treatment approaches in part different from those of the genetically defined forms. The mechanisms leading to CFH-Ab production and disease onset are not completely understood, but CFH-Ab HUS seems to be secondary to a combination of genetic predisposition and environmental factors. Early diagnosis of this specific aHUS entity is important, as prompt induction of plasma exchange and concomitant immunosuppression leads to a favorable outcome. Nevertheless, information on clinical features and outcome in children is limited. Here, we review the literature on the biological and clinical features of CFH-Ab HUS and discuss therapeutic options.


Subject(s)
Atypical Hemolytic Uremic Syndrome/diagnosis , Atypical Hemolytic Uremic Syndrome/therapy , Complement Factor H/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Atypical Hemolytic Uremic Syndrome/immunology , Autoantibodies/blood , Binding Sites , Clinical Trials as Topic , Complement C3b Inactivator Proteins/immunology , Genetic Predisposition to Disease , Humans , Kidney Transplantation , Protein Structure, Tertiary , Treatment Outcome
14.
Kidney Int ; 85(5): 1019-22, 2014 May.
Article in English | MEDLINE | ID: mdl-24786877

ABSTRACT

Atypical hemolytic uremic syndrome (HUS) secondary to anti-factor H autoantibodies has a poor prognosis. The study by Sinha et al. of a large cohort of Indian children makes a substantial contribution to improved management of this form of HUS by showing that standardized titration of anti-factor H autoantibodies is applicable worldwide and that early treatment initiation and guidance of maintenance treatment by autoantibody titer monitoring significantly improve outcomes.


Subject(s)
Autoantibodies/blood , Blood Proteins/immunology , Complement C3b Inactivator Proteins/immunology , Hemolytic-Uremic Syndrome/therapy , Immunosuppressive Agents/therapeutic use , Plasma Exchange , Time-to-Treatment , Female , Humans , Male
15.
Mol Immunol ; 58(1): 77-84, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24317278

ABSTRACT

Shiga toxin 2 (Stx2) is believed to be a major virulence factor of enterohemorrhagic Escherichia coli (EHEC) contributing to hemolytic uremic syndrome (HUS). The complement system has recently been found to be involved in the pathogenesis of EHEC-associated HUS. Stx2 was shown to activate complement via the alternative pathway, to bind factor H (FH) at short consensus repeats (SCRs) 6-8 and 18-20 and to delay and reduce FH cofactor activity on the cell surface. We now show that complement factor H-related protein 1 (FHR-1) and factor H-like protein 1 (FHL-1), proteins of the FH protein family that show amino acid sequence and regulatory function similarities with FH, also bind to Stx2. The FHR-1 binding site for Stx2 was located at SCRs 3-5 and the binding capacity of FHR-1*A allotype was higher than that of FHR-1*B. FHR-1 and FHL-1 competed with FH for Stx2 binding, and in the case of FHR-1 this competition resulted in a reduction of FH cofactor activity. FHL-1 retained its cofactor activity in the fluid phase when bound to Stx2. In conclusion, multiple interactions of key complement inhibitors FH, FHR-1 and FHL-1 with Stx2 corroborate our hypothesis of a direct role of complement in EHEC-associated HUS.


Subject(s)
Blood Proteins/immunology , Complement C3b Inactivator Proteins/immunology , Complement Factor H/immunology , Hemolytic-Uremic Syndrome/immunology , Shiga Toxin 2/immunology , Amino Acid Sequence , Binding Sites , Blood Proteins/genetics , Complement C3b Inactivator Proteins/genetics , Complement Factor H/genetics , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Hemolytic-Uremic Syndrome/microbiology , Hemolytic-Uremic Syndrome/pathology , Humans , Protein Binding/immunology
16.
Kidney Int ; 85(5): 1151-60, 2014 May.
Article in English | MEDLINE | ID: mdl-24088957

ABSTRACT

Antibodies to complement factor H are an uncommon cause of hemolytic uremic syndrome (HUS). Information on clinical features and outcomes in children is limited. In order to explore this we studied a multicenter cohort of 138 Indian children with anti-complement factor H antibody associated HUS, constituting 56% of patients with HUS. Antibody titers were high (mean 7054 AU/ml) and correlated inversely with levels of complement C3, but not complement factor H. Homozygous deletion of the CFHR1 gene was found in 60 of 68 patients. Therapies included dialysis in 119 children, 105 receiving plasma exchanges and 26 intravenous immunoglobulin. Induction immunosuppression consisted of 87 children receiving prednisolone with or without intravenous cyclophosphamide or rituximab. Antibody titers fell significantly following plasma exchanges and increased during relapses. Adverse outcome (stage 4-5 CKD or death) was seen in 36 at 3 months and 41 by last follow up, with relapse in 14 of 122 available children. Significant independent risk factors for adverse outcome were an antibody titer over 8000 AU/ml, low C3 and delay in plasma exchange. Combined plasma exchanges and induction immunosuppression resulted in significantly improved renal survival: one adverse outcome prevented for every 2.6 patients treated. Maintenance immunosuppressive therapy, of prednisolone with either mycophenolate mofetil or azathioprine, significantly reduced the risk of relapses. Thus, prompt use of immunosuppressive agents and plasma exchanges are useful for improving outcomes in pediatric patients with anti-complement factor H-associated HUS.


Subject(s)
Autoantibodies/blood , Blood Proteins/immunology , Complement C3b Inactivator Proteins/immunology , Hemolytic-Uremic Syndrome/therapy , Immunosuppressive Agents/therapeutic use , Plasma Exchange , Time-to-Treatment , Age Factors , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Azathioprine/therapeutic use , Biomarkers/blood , Blood Proteins/genetics , Case-Control Studies , Child , Child, Preschool , Combined Modality Therapy , Complement C3b Inactivator Proteins/genetics , Cyclophosphamide/therapeutic use , Drug Therapy, Combination , Female , Gene Deletion , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/diagnosis , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/immunology , Homozygote , Humans , Immunosuppressive Agents/adverse effects , India , Infant , Male , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/therapeutic use , Plasma Exchange/adverse effects , Prednisolone/therapeutic use , Recurrence , Risk Factors , Rituximab , Time Factors , Treatment Outcome
17.
Mol Immunol ; 56(3): 170-80, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23830046

ABSTRACT

Factor H related proteins comprise a group of five plasma proteins: CFHR1, CFHR2, CFHR3, CFHR4 and CFHR5, and each member of this group binds to the central complement component C3b. Mutations, genetic deletions, duplications or rearrangements in the individual CFHR genes are associated with a number of diseases including atypical hemolytic uremic syndrome (aHUS), C3 glomerulopathies (C3 glomerulonephritis (C3GN), dense deposit disease (DDD) and CFHR5 nephropathy), IgA nephropathy, age related macular degeneration (AMD) and systemic lupus erythematosus (SLE). Although complement regulatory functions were attributed to most of the members of the CFHR protein family, the precise role of each CFHR protein in complement activation and the exact contribution to disease pathology is still unclear. Recent publications show that CFHR proteins form homo- as well as heterodimers. Genetic abnormalities within the CFHR gene locus can result in hybrid proteins with affected dimerization or recognition domains which cause defective functions. Here we summarize the recent data about CFHR genes and proteins in order to better understand the role of CFHR proteins in complement activation and in complement associated diseases.


Subject(s)
Apolipoproteins/immunology , Complement C3b Inactivator Proteins/immunology , Complement Factor H/immunology , Complement System Proteins/immunology , Apolipoproteins/genetics , Apolipoproteins/metabolism , Complement C3b Inactivator Proteins/genetics , Complement C3b Inactivator Proteins/metabolism , Complement Factor H/genetics , Complement Factor H/metabolism , Complement System Proteins/metabolism , Glomerulonephritis, Membranoproliferative/genetics , Glomerulonephritis, Membranoproliferative/immunology , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/immunology , Hereditary Complement Deficiency Diseases , Humans , Macular Degeneration/genetics , Macular Degeneration/immunology , Multigene Family
18.
J Immunol ; 189(4): 1858-67, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22786770

ABSTRACT

Atypical hemolytic uremic syndrome (aHUS) is a renal disease associated with complement alternative pathway dysregulation and is characterized by endothelial injury. Pentraxin 3 (PTX3) is a soluble pattern recognition molecule expressed by endothelial cells and upregulated under inflammatory conditions. PTX3 activates complement, but it also binds the complement inhibitor factor H. In this study, we show that native factor H, factor H-like protein 1, and factor H-related protein 1 (CFHR1) bind to PTX3 and that PTX3-bound factor H and factor H-like protein 1 maintain their complement regulatory activities. PTX3, when bound to extracellular matrix, recruited functionally active factor H. Residues within short consensus repeat 20 of factor H that are relevant for PTX3 binding were identified using a peptide array. aHUS-associated factor H mutations within this binding site caused a reduced factor H binding to PTX3. Similarly, seven of nine analyzed anti-factor H autoantibodies isolated from aHUS patients inhibited the interaction between factor H and PTX3, and five autoantibodies also inhibited PTX3 binding to CFHR1. Moreover, the aHUS-associated CFHR1*B variant showed reduced binding to PTX3 in comparison with CFHR1*A. Thus, the interactions of PTX3 with complement regulators are impaired by certain mutations and autoantibodies affecting factor H and CFHR1, which could result in an enhanced local complement-mediated inflammation, endothelial cell activation, and damage in aHUS.


Subject(s)
Autoantibodies/immunology , C-Reactive Protein/metabolism , Complement C3b Inactivator Proteins/metabolism , Complement Factor H/metabolism , Hemolytic-Uremic Syndrome/metabolism , Serum Amyloid P-Component/metabolism , Atypical Hemolytic Uremic Syndrome , Autoantigens/immunology , C-Reactive Protein/immunology , Complement C3b Inactivator Proteins/immunology , Complement Factor H/immunology , Extracellular Matrix/immunology , Extracellular Matrix/metabolism , Hemolytic-Uremic Syndrome/immunology , Humans , Serum Amyloid P-Component/immunology
19.
Am J Transplant ; 12(9): 2546-53, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22681773

ABSTRACT

Antibody mediated rejection (AMR) activates the classical complement pathway and can be detrimental to graft survival. AMR can be accompanied by thrombotic microangiopathy (TMA). Eculizumab, a monoclonal C5 antibody prevents induction of the terminal complement cascade (TCC) and has recently emerged as a therapeutic option for AMR. We present a highly sensitized 13-year-old female with end-stage kidney disease secondary to spina bifida-associated reflux nephropathy, who developed severe steroid-, ATG- and plasmapheresis-resistant AMR with TMA 1 week post second kidney transplant despite previous desensitization therapy with immunoglobulin infusions. Eculizumab rescue therapy resulted in a dramatic improvement in biochemical (C3; creatinine) and hematological (platelets) parameters within 6 days. The patient was proven to be deficient in complement Factor H-related protein 3/1 (CFHR3/1), a plasma protein that regulates the complement cascade at the level of C5 conversion and has been involved in the pathogenesis of atypical hemolytic uremic syndrome caused by CFH autoantibodies (DEAP-HUS). CFHR1 deficiency may have worsened the severe clinical progression of AMR and possibly contributed to the development of donor-specific antibodies. Thus, screening for CFHR3/1 deficiency should be considered in patients with severe AMR associated with TMA.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies/immunology , Blood Proteins/immunology , Complement C3b Inactivator Proteins/immunology , Graft Rejection/drug therapy , Graft Rejection/immunology , Adolescent , Female , Humans
20.
Clin Dev Immunol ; 2012: 349657, 2012.
Article in English | MEDLINE | ID: mdl-22400034

ABSTRACT

Borrelia burgdorferi evades complement-mediated killing by interacting with complement regulators through distinct complement regulator-acquiring surface proteins (CRASPs). Here, we extend our analyses to the contribution of CRASP-4 in mediating complement resistance of B. burgdorferi and its interaction with human complement regulators. CRASP-4 (also known as ErpC) was immobilized onto magnetic beads and used to capture proteins from human serum. Following Western blotting, factor H (CFH), CFH-related protein 1 (CFHR1), CFHR2, and CFHR5 were identified as ligands of CRASP-4. To analyze the impact of native CRASP-4 on mediating survival of serum-sensitive cells in human serum, a B. garinii strain was generated that ectopically expresses CRASP-4. CRASP-4-producing bacteria bound CFHR1, CFHR2, and CFHR5 but not CFH. In addition, transformed spirochetes deposited significant amounts of lethal complement components on their surface and were susceptible to human serum, thus indicating that CRASP-4 plays a subordinate role in complement resistance of B. burgdorferi.


Subject(s)
Bacterial Proteins/immunology , Borrelia burgdorferi/immunology , Immune Evasion , Lyme Disease/immunology , Membrane Proteins/immunology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Blotting, Western , Borrelia burgdorferi/genetics , Complement C3b Inactivator Proteins/immunology , Complement C3b Inactivator Proteins/metabolism , Complement Factor H/immunology , Complement Factor H/metabolism , Complement System Proteins/immunology , Complement System Proteins/metabolism , Drug Resistance, Bacterial , Humans , Immobilized Proteins/genetics , Immobilized Proteins/immunology , Immobilized Proteins/metabolism , Lyme Disease/metabolism , Lyme Disease/microbiology , Magnetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Plasmids , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Transformation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...