Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
1.
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
2.
Int J Immunogenet ; 39(4): 328-37, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22296677

ABSTRACT

Invasive infection caused by Neisseria meningitidis is a worldwide public health problem. Previous reports have indicated that carriage of common 'defective' structural polymorphisms of the host mannose-binding lectin gene (MBL2) greatly increases an individual's risk of developing the disease. We report the largest case-control study so far to investigate the effect of these polymorphisms in meningococcal disease (296 PCR-positive cases and 5196 population controls, all of European ancestry) and demonstrate that no change in risk is associated with the polymorphisms overall or in any age-defined subgroup. This finding contrasts with two smaller studies that reported an increase in risk. A systematic review of all studies of MBL2 polymorphisms in people of European ancestry published since 1999, including 24,693 individuals, revealed a population frequency of the combined 'defective'MBL2 allele of 0.230 (95% confidence limits: 0.226-0.234). The past reported associations of increased risk of meningococcal disease were because of low 'defective' allele frequencies in their study control populations (0.13 and 0.04) that indicate systematic problems with the studies. The data from our study and all other available evidence indicate that MBL2 structural polymorphisms do not predispose children or adults to invasive meningococcal disease.


Subject(s)
Genetic Predisposition to Disease , Mannose-Binding Lectin/genetics , Meningococcal Infections/genetics , Polymorphism, Single Nucleotide , Adolescent , Adult , Aged , Alleles , Case-Control Studies , Child , Child, Preschool , Confidence Intervals , Gene Frequency , Genetic Testing , HapMap Project , Humans , Infant , Meningococcal Infections/epidemiology , Meningococcal Infections/microbiology , Middle Aged , Neisseria meningitidis/pathogenicity , Odds Ratio , Polymerase Chain Reaction , Principal Component Analysis , Risk Factors , White People/genetics , Young Adult
3.
Eye (Lond) ; 25(6): 683-93, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21394116

ABSTRACT

Age-related macular degeneration (AMD) is an inflammatory disease, which causes visual impairment and blindness in older people. The proteins of the complement system are central to the development of this disease. Local and systemic inflammation in AMD are mediated by the deregulated action of the alternative pathway of the complement system. Variants in complement system genes alter an individual's risk of developing AMD. Recent studies have shown how some risk-associated genetic variants alter the function of the complement system. In this review, we describe the evolution of the complement system and bring together recent research to form a picture of how changes in complement system genes and proteins affect the function of the complement cascade, and how this affects the development of AMD. We discuss the application of this knowledge to prevention and possible future treatments of AMD.


Subject(s)
Complement Pathway, Alternative , Complement Pathway, Classical , Complement System Proteins/physiology , Macular Degeneration/immunology , Complement System Proteins/genetics , Humans , Macular Degeneration/therapy , Risk Factors
4.
Infect Immun ; 78(6): 2677-90, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20308293

ABSTRACT

Yersinia adhesin A (YadA) is a trimeric autotransporter adhesin with multiple functions in host-pathogen interactions. The aim of this study was to dissect the virulence functions promoted by YadA in vitro and in vivo. To accomplish this, we generated Yersinia enterocolitica O:8 mutants expressing point mutations in YadA G389, a highly conserved residue in the membrane anchor of YadA, and analyzed their impact on YadA expression and virulence functions. We found that point mutations of YadA G389 led to impaired transport, stability, and surface display of YadA. YadA G389A and G389S mutants showed comparable YadA surface expression, autoagglutination, and adhesion to those of wild-type YadA but displayed reduced trimer stability and complement resistance in vitro and were 10- to 1,000-fold attenuated in experimental Y. enterocolitica infection in mice. The G389T, G389N, and G389H mutants lost trimer stability, exhibited strongly reduced surface display, autoagglutination, adhesion properties, and complement resistance, and were avirulent (>10,000-fold attenuation) in mice. Our data demonstrate that G389 is a critical residue of YadA, required for optimal trimer stability, transport, surface display, and serum resistance. We also show that stable trimeric YadA protein is essential for virulence of Y. enterocolitica.


Subject(s)
Adhesins, Bacterial/chemistry , Protein Multimerization , Virulence Factors/chemistry , Yersinia enterocolitica/chemistry , Yersinia enterocolitica/pathogenicity , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Animals , Bacterial Adhesion , Blood Bactericidal Activity , Colony Count, Microbial , Complement System Proteins/immunology , Female , HeLa Cells , Humans , Lymph Nodes/microbiology , Membrane Proteins/analysis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Mutation, Missense , Peyer's Patches/microbiology , Point Mutation , Protein Stability , Spleen/microbiology , Spleen/pathology , Survival Analysis , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism , Yersinia Infections/mortality , Yersinia Infections/pathology , Yersinia enterocolitica/genetics
5.
Cell Death Differ ; 16(12): 1630-40, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19680263

ABSTRACT

Complement forms the first defense line of innate immunity and has an important role in the non-inflammatory clearance of apoptotic and necrotic cells. Factor H is one essential complement inhibitor that binds to the acute phase reactant C-reactive protein (CRP). By using recombinant proteins, calcium-independent binding of Factor H to monomeric CRP (mCRP), but not to pentameric CRP (pCRP), was shown. In addition to the two known CRP-binding sites, a novel third site was localized within the C-terminus. This region is frequently mutated in the hemolytic uremic syndrome and the mutant proteins show reduced mCRP binding. In this study, we show that mCRP directs Factor H to the surface of apoptotic and necrotic endothelial cells and identify phosphocholine as one binding moiety for this complex. Factor H-mCRP complexes enhance C3b inactivation both in the fluid phase and on the surface of damaged cells and inhibit the production of pro-inflammatory cytokines. By recruiting the soluble complement inhibitor Factor H to the surface of damaged cells, mCRP blocks the progression of the complement cascade beyond the step of the C3 convertase, prevents the formation of inflammatory activation products, and thus contributes to the safe removal of opsonized damaged cells and particles.


Subject(s)
C-Reactive Protein/metabolism , Cell Membrane/metabolism , Complement C3b/metabolism , Complement Factor H/metabolism , Phagocytosis , Apoptosis , Cells, Cultured , Complement Factor H/genetics , Humans , Mutation , Phosphorylcholine/metabolism , Protein Binding , Protein Multimerization
6.
Mol Immunol ; 45(5): 1485-93, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17915330

ABSTRACT

Pathogenic fungi represent a major threat particularly to immunocompromised hosts, leading to severe, and often lethal, systemic opportunistic infections. Although the impaired immune status of the host is clearly the most important factor leading to disease, virulence factors of the fungus also play a role. Factor H (FH) and its splice product FHL-1 represent the major fluid phase inhibitors of the alternative pathway of complement, whereas C4b-binding protein (C4bp) is the main fluid phase inhibitor of the classical and lectin pathways. Both proteins can bind to the surface of various human pathogens conveying resistance to complement destruction and thus contribute to their pathogenic potential. We have recently shown that Candida albicans evades complement by binding both Factor H and C4bp. Here we show that moulds such as Aspergillus spp. bind Factor H, the splicing variant FHL-1 and also C4bp. Immunofluorescence and flow cytometry studies show that the binding of Factor H and C4bp to Aspergillus spp. appears to be even stronger than to Candida spp. and that different, albeit possibly nearby, binding moieties mediate this surface attachment.


Subject(s)
Complement C4b-Binding Protein/metabolism , Complement Factor H/metabolism , Complement Inactivator Proteins/physiology , Immunity , Aspergillus/immunology , Complement C3b Inactivator Proteins , Humans , Protein Binding
7.
Clin Exp Immunol ; 144(2): 342-52, 2006 May.
Article in English | MEDLINE | ID: mdl-16634809

ABSTRACT

The complement inhibitor Factor H has three distinct binding sites for C3b and for heparin, but in solution uses specifically the most C-terminal domain, i.e. short consensus repeats (SCR) 20 for ligand interaction. Two novel monoclonal antibodies (mABs C14 and C18) that bind to the most C-terminal domain SCR 20 completely blocked interaction of Factor H with the ligands C3b, C3d, heparin and binding to endothelial cells. In contrast, several mAbs that bind to the N-terminus and to the middle regions of the molecule showed no or minor inhibitory effects when assayed by enzyme-linked immunosorbent assay (ELISA) and ligand interaction assays. This paradox between a single functional binding site identified for native Factor H versus multiple interaction sites reported for deletion constructs is explained by a compact conformation of the fluid phase protein with one accessible binding site. On zymosan particles mAbs C14 and C18 blocked alternative pathway activation completely. Thus demonstrating that native Factor H makes the first and initial contact with the C terminus, which is followed by N terminally mediated complement regulation. These results are explained by a conformational hypothetical model: the native Factor H protein has a compact structure and only one binding site accessible. Upon the first contact the protein unfolds and exposes the additional binding sites. This model does explain how Factor H mediates recognition functions during complement control and the clustering of disease associated mutations in patients with haemolytic uraemic syndrome that have been reported in the C-terminal recognition domain of Factor H.


Subject(s)
Antibodies, Monoclonal/immunology , Binding Sites, Antibody/immunology , Complement C3b/immunology , Complement C3d/immunology , Complement Factor H/genetics , Complement Factor H/immunology , Complement Pathway, Alternative/immunology , Endothelial Cells/immunology , Epitopes/immunology , Heparin/immunology , Humans , Ligands , Models, Biological , Mutation , Protein Conformation , Zymosan/immunology
8.
Kidney Int ; 70(1): 42-50, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16612335

ABSTRACT

We report a novel pathomechanism for membranoproliferative glomerulonephritis type II (MPGN II) caused by a mutant Factor H protein expressed in the plasma. Genetic analyses of two patients revealed deletion of a single Lys residue (K224) located within the complement regulatory region in domain 4 of Factor H. This deletion resulted in defective complement control: mutant protein purified from the plasma of patients showed severely reduced cofactor and decay-accelerating activity, as well as reduced binding to the central complement component C3b. However, cell-binding activity of the mutant protein was normal and comparable to wild-type Factor H. The patients are daughters of consanguineous parents. As both patients but also their healthy mother were positive for C3 nephritic factor, the mutant Factor H protein is considered relevant for unrestricted activation of the disease-causing activation of the alternative complement pathway. Replacement of functional Factor H by fresh frozen plasma (10-15 ml/kg/14 days) was well tolerated, prevented so far disease progression in both patients, and is in the long run expected to preserve kidney function.


Subject(s)
Complement Factor H/genetics , Complement Factor H/metabolism , Complement Pathway, Alternative , Glomerulonephritis, Membranoproliferative/genetics , Glomerulonephritis, Membranoproliferative/immunology , Amino Acid Sequence , Child , Complement C3 Nephritic Factor/analysis , Complement C3 Nephritic Factor/metabolism , Complement Factor H/analysis , Consanguinity , Female , Humans , Lysine/chemistry , Lysine/genetics , Molecular Sequence Data , Pedigree , Plasma/chemistry , Plasma/metabolism , Protein Structure, Tertiary/genetics , Sequence Deletion
9.
J Med Genet ; 43(7): 582-9, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16299065

ABSTRACT

INTRODUCTION: Membranoproliferative glomerulonephritis type II or dense deposit disease (MPGN II/DDD) causes chronic renal dysfunction that progresses to end stage renal disease in about half of patients within 10 years of diagnosis. Deficiency of and mutations in the complement factor H (CFH) gene are associated with the development of MPGN II/DDD, suggesting that dysregulation of the alternative pathway of the complement cascade is important in disease pathophysiology. SUBJECTS: Patients with MPGN II/DDD were studied to determine whether specific allele variants of CFH and CFHR5 segregate preferentially with the MPGN II/DDD disease phenotype. The control group was compromised of 131 people in whom age related macular degeneration had been excluded. RESULTS: Allele frequencies of four single nucleotide polymorphisms in CFH and three in CFHR5 were significantly different between MPGN II/DDD patients and controls. CONCLUSION: We have identified specific allele variants of CFH and CFHR5 associated with the MPGN II/DDD disease phenotype. While our data can be interpreted to further implicate complement in the pathogenesis of MPGN II/DDD, these associations could also be unrelated to disease pathophysiology. Functional studies are required to resolve this question.


Subject(s)
Blood Proteins/genetics , Complement Factor H/genetics , Genetic Variation , Glomerulonephritis, Membranoproliferative/genetics , Biopsy , Complement System Proteins , DNA Primers , Gene Deletion , Gene Frequency , Glomerulonephritis, Membranoproliferative/classification , Glomerulonephritis, Membranoproliferative/pathology , Humans , Mutation , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Reference Values
10.
J Thromb Haemost ; 3(1): 154-62, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15634279

ABSTRACT

BACKGROUND: Factor H regulates the alternative pathway of complement. The protein has three heparin-binding sites, is synthesized primarily in the liver and copurifies from platelets with thrombospondin-1. Factor H mutations at the C-terminus are associated with atypical hemolytic uremic syndrome, a condition in which platelets are consumed. Objectives The aim of this study was to investigate if factor H interacts with platelets. METHODS: Binding of factor H, recombinant C- or N-terminus constructs and a C-terminus mutant to washed (plasma and complement-free) platelets was analyzed by flow cytometry. Binding of factor H and constructs to thrombospondin-1 was measured by surface plasmon resonance. RESULTS: Factor H bound to platelets in a dose-dependent manner. The major binding site was localized to the C-terminus. The interaction was partially blocked by heparin. Inhibition with anti-GPIIb/IIIa, or with fibrinogen, suggested that the platelet GPIIb/IIIa receptor is involved in factor H binding. Factor H binds to thrombospondin-1. Addition of thrombospondin-1 increased factor H binding to platelets. Factor H mutated at the C-terminus also bound to platelets, albeit to a significantly lesser degree. CONCLUSIONS: This study reports a novel property of factor H, i.e. binding to platelets, either directly via the GPIIb/IIIa receptor or indirectly via thrombospondin-1, in the absence of complement. Binding to platelets was mostly mediated by the C-terminal region of factor H and factor H mutated at the C-terminus exhibited reduced binding.


Subject(s)
Blood Platelets/metabolism , Complement Factor H/chemistry , Complement Factor H/metabolism , Binding Sites , Blood Platelets/cytology , Complement System Proteins/chemistry , Dose-Response Relationship, Drug , Female , Flow Cytometry , Hemolytic-Uremic Syndrome/genetics , Heparin/chemistry , Humans , Kinetics , Liver/metabolism , Male , Mutation , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Surface Plasmon Resonance , Thrombospondin 1/metabolism , Time Factors
11.
Infect Immun ; 72(11): 6633-41, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15501796

ABSTRACT

Candida albicans, an important pathogenic yeast, activates all three pathways of the complement system. To understand how this yeast evades the effects of the activated system, we have analyzed the binding of the classical pathway inhibitor C4b-binding protein (C4BP) by C. albicans. Purified native as well as recombinant C4BP bound dose dependently to the yeast and hyphal forms, as shown by multiple methods, such as confocal microscopy, flow cytometry, a novel enzyme-linked immunosorbent assay, absorption from human serum, and direct binding assays with purified proteins. A prominent binding site was identified at the tip of the germ tube, a structure that is considered important for tissue penetration and pathogenesis. The binding site in C4BP was localized to the two N-terminal complement control protein domains by using recombinant deletion constructs and site-specific monoclonal antibodies. As the alternative pathway inhibitors factor H and FHL-1 also bind to C. albicans, the binding of all three plasma proteins was compared. Simultaneous binding of the classical regulator C4BP and the alternative pathway regulator factor H was demonstrated by confocal microscopy. In addition, FHL-1 competed for binding with C4BP, suggesting that these two related complement regulators bind to the same structures on the yeast surface. The surface-attached C4BP maintains its complement regulatory activities and inactivates C4b. The surface-attached human C4BP serves multiple functions relevant for immune evasion and likely pathogenicity. It inhibits complement activation at the yeast surface and, in addition, mediates adhesion of C. albicans to host endothelial cells.


Subject(s)
Candida albicans/growth & development , Candida albicans/pathogenicity , Complement Inactivator Proteins/metabolism , Glycoproteins/metabolism , Binding Sites , Binding, Competitive , Blood Proteins/metabolism , Candida albicans/metabolism , Complement C3b Inactivator Proteins , Complement C4b/metabolism , Complement Factor H/metabolism , Complement Inactivator Proteins/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Glycoproteins/genetics , Humans , Microscopy, Confocal
12.
Histol Histopathol ; 19(1): 251-8, 2004 01.
Article in English | MEDLINE | ID: mdl-14702193

ABSTRACT

Complement is a central element of innate immunity and this vital defense system initiates and coordinates immediate immune reactions which attack and eliminate microbes, foreign particles and altered self cells. Newly generated activation products are extremely toxic and consequently, activation is highly restricted in terms of time and space. The initial activation of the alternative complement pathway occurs continuously and the early phase acts indiscriminatoryl and forms on any surface. However, the system discriminates between self and foreign, and therefore allows activation on foreign surfaces e.g. microbes, and restricts activation on host cells. Consequently, self cells and tissues are protected from the harmful activation products. This protection is mediated by specific regulators or inhibitors, which exist in the fluid phase and/or in membrane-bound forms. Here we review a novel mechanism, i.e. the attachment of the soluble complement regulator factor H to the surface of self cells. This attachment, which is demonstrated experimentally by means of immunofluorescense microscopy and by flow cytometry, increases the inhibitory potential at the cell surface and mediates protection by reducing the local formation of toxic inflammatory products. This attachment is highly relevant and has pathophysiological consequences in several human diseases, including Factor H-associated hemolytic uremic syndrome (FH-HUS), membrano-proliferative glomerulonephritis type II, recurrent microbial infections and chronic inflammation, e.g. rheumatoid arthritis and immune evasion of tumor cells. Defects of this safeguard activity have been recently understood in patients with FH-HUS. Point mutations in the Factor H gene occurring in the C-terminus of the protein result in impaired cell binding capacity of Factor H and, consequently, during an inflammatory insult endothelial cells are not properly protected and are damaged.


Subject(s)
Complement Factor H/metabolism , Endothelium, Vascular/metabolism , Pathology , Animals , Binding Sites , Cell Line , Complement Activation , Complement C3b/metabolism , Complement Factor H/genetics , Complement Pathway, Alternative , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/metabolism , Humans , Immunity, Innate , Models, Biological , Point Mutation , Solubility , Surface Properties , Umbilical Veins/cytology
13.
Trop Med Int Health ; 8(11): 1018-24, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14629769

ABSTRACT

Procalcitonin (PCT) plasma levels and the fraction of CTLA-4-positive T cells are both elevated in acute Plasmodium falciparum malaria in human adults and the degree of elevation is positively correlated with other markers of disease severity, for example with parasitaemia. However, the clinical manifestations of malaria are strongly age-dependent and children from endemic areas carry the main disease burden. Therefore, we measured PCT plasma levels and CTLA-4 expression by T cells in four groups of children from the Ashanti Region in Ghana: asymptomatic children with or without parasitaemia, children with uncomplicated P. falciparum malaria and children with severe disease. PCT levels were highly elevated in both groups with acute malaria but they did not discriminate between uncomplicated and severe disease. In contrast, CTLA-4-expression by T cells was increased only in severe malaria. The fraction of CTLA-4 positive T cells in the blood of children with severe disease differed significantly from that in uncomplicated malaria, which was not elevated in spite of the high parasite loads observed in these children. This was unexpected, as in adults uncomplicated malaria is associated with a dramatic sixfold increase of the fraction of CTLA-4-positive T cells. The data from this study support the hypothesis that strong T cell activation as measured here by CTLA-4 expression is not just the by-product of a high parasite burden, but that it contributes to the pathogenesis of P. falciparum malaria.


Subject(s)
Antigens, Differentiation/analysis , Calcitonin/blood , Immunosuppressive Agents/immunology , Malaria, Falciparum/immunology , Protein Precursors/blood , T-Lymphocytes/immunology , Antigens, CD , CTLA-4 Antigen , Calcitonin Gene-Related Peptide , Child , Child, Preschool , Ghana , Humans , Infant , Malaria, Falciparum/blood , Severity of Illness Index
14.
J Med Genet ; 40(9): 676-81, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12960213

ABSTRACT

BACKGROUND: The aetiology of atypical haemolytic uraemic syndrome (aHUS) is, in contrast to classical, Shiga-like toxin induced HUS in children, largely unknown. Deficiency of human complement factor H and familial occurrence led to identification of the factor H gene (FH1) as the susceptibility gene, but the frequency and relevance of FH1 mutations are unknown. METHODS: We established a German registry for aHUS and analysed in all patients and 100 controls the complete FH1 gene by single strand confirmational polymorphism and DNA sequencing. In addition, complement C3 and factor H serum levels were assayed. Demographic data at onset of aHUS and follow up were compared for the mutation positive and negative groups. RESULTS: Of 111 patients with aHUS (68 female, 43 male, mean age 33 years) 14% had FH1 germline mutations, including two of eight patients with familial aHUS. For each of these eight patients, both parents were tested, and we were able to trace the mutation for five cases. In the other three cases (one with the mutation 3749 C/T, one with 3200 T/C, and one with 3566+1 G/A), we could not detect the mutation in either parent, although paternity was proven by genetic fingerprinting, suggesting that these subjects have new mutations. C3 was decreased in five mutation carriers but also in two non-carriers, and factor H was decreased in none of the carriers, but elevated in six carriers and 15 non-carriers. Clinical parameters including associated medications and diseases, and outcome of aHUS and of post-aHUS kidney transplantation were similar in the mutation positive and negative groups. CONCLUSION: FH1 germline mutations occur with considerable frequency in patients with aHUS. Hypocomplementaemia is not regularly associated with a germline mutation, and factor H serum levels can even be elevated. Screening for FH1 mutations contributes to the classification of aHUS.


Subject(s)
Complement Factor H/genetics , Hemolytic-Uremic Syndrome/genetics , Adult , Austria , Complement C3/metabolism , Complement Factor H/metabolism , DNA/chemistry , DNA/genetics , DNA Mutational Analysis , Enzyme-Linked Immunosorbent Assay , Female , Germany , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/complications , Humans , Italy , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/therapy , Kidney Transplantation , Male , Mutation , Polymorphism, Single-Stranded Conformational , Registries/statistics & numerical data , Switzerland
15.
Clin Exp Immunol ; 132(3): 485-95, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12780697

ABSTRACT

Rheumatoid arthritis is a chronic inflammatory disease of unknown aetiology predominantly affecting cells and tissues of synovial joints. Here we show that the two important complement regulators FHL-1 and factor H play a protective anti-inflammatory role in rheumatoid arthritis. Expression analyses at the mRNA- and protein level show in vitro expression and secretion of both regulators by synovial fibroblasts derived from patients with rheumatoid arthritis. Similarly the two regulators are synthesized in vivo in diseased synovial tissue, and in particular synovial lining cells express high levels of FHL-1. The anti-inflammatory role of these regulators in rheumatoid arthritis is highlighted by their induction with IFN-gamma and dexamethasone, whilst the pro-inflammatory cytokine TNF-alpha had no effect. Transient transfection experiments with various FHL-1/factor H promoter-luciferase reporter constructs into cells of distinct origin show independent cell and tissue specific promoter regulated transcription of these two regulators. The inducible expression, specifically of FHL-1 has physiological consequences. By binding directly to surfaces the released proteins protect cells from inflammatory damage and complement-mediated cell lysis. This study shows a novel protective and anti-inflammatory role of the two important complement regulators FHL-1 and factor H in rheumatoid arthritis and suggests a disease controlling role of the two proteins.


Subject(s)
Arthritis, Rheumatoid/metabolism , Blood Proteins/physiology , Complement Factor H/physiology , Fibroblasts/metabolism , Synovial Membrane/metabolism , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Blood Proteins/genetics , Blood Proteins/metabolism , Cell Line , Complement C3b Inactivator Proteins , Complement Factor H/genetics , Complement Factor H/metabolism , Cytotoxicity, Immunologic , Gene Expression , Humans , Promoter Regions, Genetic , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Synovial Membrane/immunology , Synovial Membrane/pathology , Transfection , Tumor Cells, Cultured
16.
Biochem Soc Trans ; 30(Pt 6): 971-8, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12440956

ABSTRACT

At present, the human Factor H protein family represents seven multidomain, multifunctional serum proteins. This group includes the complement and immune regulators Factor H, the Factor H-like protein 1 (FHL-1) and five Factor H-related proteins proteins (FHR-1, -2, -3, -4 and -5). Each is exclusively composed of individually folded protein domains, termed short consensus repeats (SCRs) or complement control modules. Structure-function analyses allowed the localization of the complement regulatory domain of Factor H and FHL-1 in the N-terminal region within SCRs 1-4. In addition, multiple binding sites for C3b, heparin and microbial surface proteins were localized in the N-terminus, within the middle region and also in the C-terminus of Factor H and FHL-1. Recent results show a central role for the C-terminus of Factor H, i.e. SCRs 19-20. These particular domains are conserved in all FHRs identified so far, include contact points for C3b, heparin and microbial surface proteins and represent a 'hot-spot' for gene mutations in patients that suffer from the Factor H-associated form of haemolytic uraemic syndrome.


Subject(s)
Complement Factor H/chemistry , Complement System Proteins/chemistry , Borrelia/pathogenicity , Escherichia coli/pathogenicity , Hemolytic-Uremic Syndrome/genetics , Hemolytic-Uremic Syndrome/microbiology , Humans , Models, Biological , Multigene Family , Mutation , Protein Structure, Tertiary , Structure-Activity Relationship
17.
Br J Cancer ; 87(10): 1119-27, 2002 Nov 04.
Article in English | MEDLINE | ID: mdl-12402151

ABSTRACT

We observed that the soluble complement regulators factor H and factor H-like protein were abundantly present in ascites samples as well as in primary tumours of patients with ovarian cancer. RT-PCR and immunoblotting analyses showed that the two complement inhibitors were constitutively produced by the ovarian tumour cell lines SK-OV-3 and Caov-3, but not PA-1 or SW626 cells. The amounts of factor H-like protein secreted were equal to those of factor H. This is exceptional, because e.g. in normal human serum the concentration of factor H-like protein is below 1/10th of that of factor H. In ascites samples the mean level of factor H-like protein (130+/-55 microg ml(-1)) was 5.5-fold higher than in normal human serum (24+/-3 microg ml(-1)). Ovarian tumour cells thus preferentially synthesise factor H-like protein, the alternatively spliced short variant of factor H. The tumour cells were found to bind both (125)I-labelled factor H and recombinant factor H-like protein to their surfaces. Surprisingly, the culture supernatants of all of the ovarian tumour cell lines studied, including those of PA-1 and SW626 that did not produce factor H/factor H-like protein, promoted factor I-mediated cleavage of C3b to inactive iC3b. Subsequently, the PA-1 and SW626 cell lines were found to secrete a soluble form of the membrane cofactor protein (CD46). Thus, our studies reveal two novel complement resistance mechanisms of ovarian tumour cells: (i) production of factor H-like protein and factor H and (ii) secretion of soluble membrane cofactor protein. Secretion of soluble complement inhibitors could protect ovarian tumour cells against humoral immune attack and pose an obstacle for therapy with monoclonal antibodies.


Subject(s)
Blood Proteins/biosynthesis , Complement Factor H/biosynthesis , Ovarian Neoplasms/metabolism , Ascitic Fluid/chemistry , Blood Proteins/analysis , Complement C3b/metabolism , Complement C3b Inactivator Proteins , Complement Factor H/analysis , Female , Humans , Immunohistochemistry , Ovarian Neoplasms/chemistry , Tumor Cells, Cultured
18.
Infect Immun ; 70(9): 5185-92, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12183569

ABSTRACT

The human facultative pathogenic yeast Candida albicans causes mucocutaneous infections and is the major cause of opportunistic fungal infections in immunocompromised patients. C. albicans activates both the alternative and classical pathway of the complement system. The aim of this study was to assay whether C. albicans binds human complement regulators in order to control complement activation at its surface. We observed binding of two central complement regulators, factor H and FHL-1, from normal human serum to C. albicans by adsorption assays, immunostaining, and fluorescence-activated cell sorter (FACS) analyses. Specificity of acquisition was further confirmed in direct binding assays with purified proteins. The surface-attached regulators maintained their complement regulatory activities and mediated factor I-dependent cleavage of C3b. Adsorption assays with recombinant deletion mutant proteins were used to identify binding domains. Two binding sites were localized. One binding domain common to both factor H and FHL-1 is located in the N-terminal short consensus repeat domains (SCRs) 6 and 7, and the other one located in C-terminal SCRs 19 and 20 is unique to factor H. These data indicate that by surface acquisition of host complement regulators, the human pathogenic yeast C. albicans is able to regulate alternative complement activation at its surface and to inactivate toxic complement activation products.


Subject(s)
Blood Proteins/metabolism , Candida albicans/immunology , Candida albicans/pathogenicity , Complement Factor H/metabolism , Binding Sites/genetics , Blood Proteins/genetics , Complement Activation , Complement C3b Inactivator Proteins , Complement Factor H/genetics , Heparin/pharmacology , Humans , In Vitro Techniques , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Deletion
19.
Infect Immun ; 69(12): 7800-9, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11705962

ABSTRACT

The three genospecies Borrelia burgdorferi, Borrelia garinii, and Borrelia afzelii, all causative agents of Lyme disease, differ in their susceptibilities to human complement-mediated lysis. We recently reported that serum resistance of borrelias correlates largely with their ability to bind the human complement regulators FHL-1/reconectin and factor H. To date, two complement regulator-acquiring-proteins (CRASP-1 and CRASP-2) have been identified in serum-resistant B. afzelii isolates (P. Kraiczy, C. Skerka, M. Kirschfink, V. Brade, and P. F. Zipfel, Eur. J. Immunol. 31:1674-1684, 2001). Here, we present a comprehensive study of the CRASPs detectable in both serum-resistant and intermediate serum-sensitive B. afzelii and B. burgdorferi isolates. These CRASPs were designated according to the genospecies either as BaCRASPs, when derived from B. afzelii, or as BbCRASPs, for proteins identified in B. burgdorferi isolates. Each borrelial isolate expresses distinct CRASPs that can be differentiated by their mobility and binding phenotypes. A detailed comparison reveals overlapping and even identical binding profiles for BaCRASP-1 (27.5 kDa), BbCRASP-1 (25.9 kDa), and BbCRASP-2 (23.2 kDa), which bind FHL-1/reconectin strongly and interact weakly with factor H. In contrast, two B. afzelii proteins (BaCRASP-4 [19.2 kDa] and BaCRASP-5 [22.5 kDa]) and three B. burgdorferi proteins (BbCRASP-3 [19.8 kDa], BbCRASP-4 [18.5 kDa], and BbCRASP-5 [17.7 kDa]) bind factor H but not FHL-1/reconectin. Most CRASPs bind both human immune regulators at their C-terminal ends. Temperature-dependent up-regulation of CRASPs (BaCRASP-1, BaCRASP-2, and BaCRASP-5) is detected in low-passage borrelias cultured at 33 or 37 degrees C compared with those cultured at 20 degrees C. The characterization of the individual CRASPs on the molecular level is expected to identify new virulence factors and potential vaccine candidates.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi Group/immunology , Complement System Proteins/metabolism , Membrane Proteins/metabolism , Binding Sites , Blood Proteins/metabolism , Borrelia burgdorferi/immunology , Complement C3b Inactivator Proteins , Complement Factor H/metabolism , Gene Expression Regulation, Bacterial , Humans , Immunity, Innate , Lyme Disease/etiology , Protein Binding , Temperature
20.
Mol Microbiol ; 41(4): 817-26, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11532146

ABSTRACT

Factor H (FH) and factor H-like protein 1 (FHL-1) regulate complement activation through the alternative pathway. Several extracellular bacterial pathogens, prime targets for the complement system, bind FH and FHL-1, thereby acquiring a potential mechanism for minimizing complement deposition on their surface. For group A streptococci (GAS), surface-bound antiphagocytic M proteins mediate the interaction. To study the role of the FH-FHL-1 interaction for complement deposition and opsonophagocytosis of GAS, we first constructed a set of truncated M5 protein variants and expressed them on the surface of a homologous M-negative GAS strain. Binding experiments with the resulting strains demonstrated that the major FH-FHL-1 binding is located in a 42-amino-acid region within the N-terminal third of M5. Measurement of bacteria-bound complement factor C3 after incubation in plasma showed that the presence of this region had little impact upon complement deposition through the alternative pathway. Moreover, streptococci expressing M5 proteins lacking the major FH and FHL-1 binding sequence resisted phagocytosis in human blood as efficiently as bacteria expressing the wild-type protein. Consequently, the data suggest that the binding of the regulators of the alternative pathway is of limited importance for GAS phagocytosis resistance.


Subject(s)
Antigens, Bacterial , Blood Proteins/metabolism , Complement Factor H/metabolism , Phagocytosis , Streptococcus/metabolism , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Binding Sites , Blood Proteins/immunology , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Division , Complement C3b/immunology , Complement C3b/metabolism , Complement C3b Inactivator Proteins/metabolism , Complement Factor H/immunology , Complement Pathway, Alternative/immunology , DNA Primers/genetics , Protein Binding , Sequence Deletion , Streptococcus/classification , Streptococcus/growth & development , Streptococcus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...