Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Immunol Lett ; 77(1): 55-62, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11348670

ABSTRACT

The human factor H protein family comprises six plasma glycoproteins. Earlier we described a membranal factor H-related (mFHR) molecule that is expressed by human B lymphoblastoid cell lines and exerts cofactor activity. In our present study we screened human blood cells for the presence of mFHR proteins and further characterized these molecules. By cytofluorimetry it is shown that the factor H-specific rabbit antiserum reacts strongly with B cells and neutrophil granulocytes, but not with T cells and monocytes. On B lymphocytes mFHR is shown to be down-regulated upon activation of the cells via sIg. In experiments studying which short consensus repeat (SCR) domains are part of the cell membrane proteins we found that antibodies raised against SCRs 1-4, 19-20 and FHR-3 bound to neutrophils but not to B cells. While mFHRs derived both from B cells and granulocytes are shown to bind heparin, their size and structure are different as revealed by Western blotting. A further characteristic of the granulocyte-derived mFHR is its sensitivity to the PI-specific PLCgamma enzyme. These data demonstrate the existence of new members of the FHR protein family, as two distinct, membranal forms are identified. Based on the differences, the B cell derived molecule is termed mFHR-1 and the neutrophil derived protein mFHR-2.


Subject(s)
B-Lymphocytes/metabolism , Complement Factor H/biosynthesis , Neutrophils/metabolism , B-Lymphocytes/cytology , Cell Membrane/metabolism , Cells, Cultured , Down-Regulation , Heparin/metabolism , Humans , Isoenzymes/metabolism , Lymphocyte Activation , Neutrophils/cytology , Phosphatidylinositol Diacylglycerol-Lyase , Phospholipase C gamma , Type C Phospholipases/metabolism
10.
J Leukoc Biol ; 69(1): 69-74, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11200070

ABSTRACT

The adjuvant effect of gamma-inulin, a strong activator of the alternative complement pathway, is well-known, but its exact mechanism is not revealed yet. Here, we show that macrophages, isolated from the peritoneal cavity of gamma-inulin-injected mice and used as antigen-presenting cells, enhance the proliferation of antigen-specific T-cells up to 2.5-fold when compared with macrophages of non-treated animals. This effect is abrogated by the presence of anti-C3 F(ab')2 fragments and by prior decomplementation of the donor animals with CVF. It is demonstrated that treatment of mice with the adjuvant results in deposition of C3-fragments onto the surface of peritoneal macrophages, as does in vitro incubation of the cells with gamma-inulin in the presence of fresh autologous serum. Prior incubation of macrophages with gamma-inulin plus serum in vitro enhances subsequent C3 production. Because it has been shown earlier that CR1/2 expressed on activated T-cells and interacting with covalently bound C3-fragments plays an important role in the augmentation of the adaptive response, our present results reveal a mechanism that contributes to the adjuvant effect of gamma-inulin and point to a further link between innate and adaptive immunity.


Subject(s)
Antigen Presentation/immunology , Complement C3/immunology , Inulin/immunology , Macrophages, Peritoneal/immunology , Animals , Lymphocyte Activation , Mice , T-Lymphocytes/immunology
11.
Eur J Immunol ; 30(6): 1706-13, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10898508

ABSTRACT

While the interaction of complement component C1q with cellular proteins is extensively studied, much less is known about the binding of the structurally related molecule, mannan-binding lectin (MBL) to various cells. Here we show by cytofluorimetry that the interaction of MBL with immunocompetent cells is much more restricted than that of C1q. It is shown that under conditions of physiological ionic strength MBL binds to human monocyte-derived macrophages (Mphi) and monocytoid cell lines, but not to T and B lymphocytes, in contrast to C1q, which interacts with all these cells under the same conditions. As opposed to the binding of C1q, low ionic strength does not improve the interaction of MBL with Mphi. No competition for cellular binding sites was found when MBL and C1q were added simultaneously to the cells. Studying the functional consequences of the interaction, we found that the release of TNF-alpha from Mphi is induced by C1q but not by MBL. Production of complement C3 by Mphi is stimulated by C1q strongly, while the effect of MBL is much weaker. C3 produced upon C1q-mediated triggering is shown to opsonize RBC, resulting in enhanced phagocytosis. These results suggest that cell membrane molecules binding MBL and C1 q are not identical; moreover, biological functions exerted by these proteins are also markedly different.


Subject(s)
Carrier Proteins/immunology , Complement C1q/immunology , Macrophages/immunology , Binding Sites , Binding, Competitive , Cell Differentiation , Cells, Cultured , Collectins , Complement C3/biosynthesis , Humans , Immunoglobulin Fab Fragments/immunology , Jurkat Cells , Macrophages/cytology , Phagocytosis/immunology , Protein Binding , Tumor Necrosis Factor-alpha/biosynthesis , U937 Cells
12.
Int Immunol ; 10(12): 1923-30, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9885914

ABSTRACT

Murine cells of the B lymphoblastoid line A20 and concanavalin A-elicited peritoneal macrophages are shown to activate and fix C3 fragments covalently when incubated in fresh, autologous serum under conditions allowing the initiation of the alternative complement pathway. For the detection of cell-bound C3, cytofluorimetry was performed using FITC-labeled F(ab')2 fragments of anti-mouse C3. Cell-bound C3 fragments are not internalized or shed by the cells under culture conditions for at least two hours. When the antigen-presenting capacity of serum-treated cells was tested using various antigens and experimental systems, augmentation of the proliferation of antigen-specific T cells was found. This enhancing effect was particularly pronounced at suboptimal antigen doses. The elevation of T cell proliferation induced by C3-opsonized antigen-presenting cells (APC) could be abrogated by F(ab')2 fragments of goat anti-mouse C3, suggesting the involvement of C3 receptors expressed by T cells in the process. Using the 7G6 mAb recognizing murine CR1/CR2, the presence of these complement receptors on activated T cells is demonstrated by cytofluorimetry and immunoprecipitation, as well. These results point to the role of C3 bound to acceptor sites on APC in the facilitation of antigen presentation, providing a further link between innate and adaptive immunity.


Subject(s)
Antigen-Presenting Cells/metabolism , Complement C3/metabolism , Epitopes, T-Lymphocyte/immunology , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , Animals , Antigen Presentation , Antigen-Presenting Cells/immunology , Complement C3/immunology , Complement C3/physiology , Complement Pathway, Alternative , Hybridomas/immunology , Hybridomas/metabolism , Immune Sera/pharmacology , Immunity, Innate , Immunosuppressive Agents/pharmacology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred BALB C , Receptors, Complement 3b/biosynthesis , Receptors, Complement 3d/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...