Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Exp Dermatol ; 30(11): 1631-1641, 2021 11.
Article in English | MEDLINE | ID: mdl-33813765

ABSTRACT

The incidence of cutaneous squamous cell carcinoma (cSCC) is increasing globally. Here, we have studied the functional role of complement factor I (CFI) in the progression of cSCC. CFI was knocked down in cSCC cells, and RNA-seq analysis was performed. Significant downregulation of genes in IPA biofunction categories Proliferation of cells and Growth of malignant tumor, in Gene Ontology (GO) terms Metallopeptidase activity and Extracellular matrix component, as well as Reactome Degradation of extracellular matrix was detected after CFI knockdown. Further analysis of the latter three networks, revealed downregulation of several genes coding for invasion-associated matrix metalloproteinases (MMPs) after CFI knockdown. The downregulation of MMP-13 and MMP-2 was confirmed at mRNA, protein and tissue levels by qRT-qPCR, Western blot and immunohistochemistry, respectively. Knockdown of CFI decreased the invasion of cSCC cells through type I collagen. Overexpression of CFI in cSCC cells resulted in enhanced production of MMP-13 and MMP-2 and increased invasion through type I collagen and Matrigel, and in increased ERK1/2 activation and cell proliferation. Altogether, these findings identify a novel mechanism of action of CFI in upregulation of MMP-13 and MMP-2 expression and cSCC invasion. These results identify CFI as a prospective molecular marker for invasion and metastasis of cSCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Complement Factor I/physiology , Gene Expression Regulation, Neoplastic , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 2/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Up-Regulation , Animals , Humans , Mice , Neoplasm Invasiveness , Tumor Cells, Cultured
2.
Immunobiology ; 224(4): 511-517, 2019 07.
Article in English | MEDLINE | ID: mdl-31109748

ABSTRACT

Factor I was first discovered in 1966. Its importance became apparent with the description of the original Factor I deficient patient in Boston in 1967. This patient presented with a hyperactive alternative complement pathway resulting in secondary complement deficiency due to continuous complement consumption. On the basis of these findings, the mechanism of the alternative pathway was worked out. In 1975, the surprise finding was made that elevating levels of Factor I in plasma down-regulated the alternative pathway. Attempts to exploit this finding for clinical use had a long and frustrating history and it was not until 2019 that the first patient was treated with the gene therapy vector for age related macular degeneration by Professor Sir Robert MacLaren in Oxford. This review follows the long and contorted course from initial observations to clinical use of complement Factor I.


Subject(s)
Complement Factor I/physiology , Animals , Complement Activation/genetics , Complement Activation/immunology , Complement Factor I/chemistry , Complement Factor I/therapeutic use , Complement Pathway, Alternative/genetics , Complement Pathway, Alternative/immunology , Complement System Proteins/genetics , Complement System Proteins/immunology , Complement System Proteins/metabolism , Disease Susceptibility , Humans , Immunoconglutinins/immunology , Signal Transduction , Structure-Activity Relationship
3.
J Invest Dermatol ; 135(2): 579-588, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25184960

ABSTRACT

The incidence of cutaneous squamous cell carcinoma (cSCC) is rising worldwide. We have examined the role of complement components in the progression of cSCC. Analysis of cSCC cell lines (n=8) and normal human epidermal keratinocytes (n=11) with whole transcriptome profiling (SOLiD), quantitative real-time reverse transcriptase-PCR, and western blotting revealed marked overexpression of complement factor I (CFI) in cSCC cells. Immunohistochemical analysis for CFI in vivo showed stronger tumor cell-specific labeling intensity in invasive sporadic cSCCs (n=83) and recessive dystrophic epidermolysis bullosa-associated cSCCs (n=7) than in cSCC in situ (n=65), premalignant epidermal lesions (actinic keratoses, n=64), benign epidermal papillomas (seborrheic keratoses, n=39), and normal skin (n=9). The expression of CFI was higher in the aggressive Ha-ras-transformed cell line (RT3) than in less tumorigenic HaCaT cell lines (HaCaT, A5, and II-4). The expression of CFI by cSCC cells was upregulated by IFN-γ and IL-1ß. Knockdown of CFI expression inhibited proliferation and migration of cSCC cells and resulted in inhibition of basal extracellular signal-regulated kinase (ERK) 1/2 activation. Knockdown of CFI expression potently inhibited growth of human cSCC xenograft tumors in vivo. These results provide evidence for the role of CFI in the progression of cSCC and identify it as a potential therapeutic target in this nonmelanoma skin cancer.


Subject(s)
Carcinoma, Squamous Cell/pathology , Complement Factor I/physiology , Skin Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Female , Genes, ras , Humans , Mice , Mice, SCID , Middle Aged
4.
J Clin Immunol ; 33(8): 1293-301, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24142231

ABSTRACT

PURPOSE: Complement regulators control the activated complement system. Defects in this homeostasis can result in tissue damage and autoimmune diseases with a heterogeneity in clinical presentation. Complement factor I (FI), a serine protease, is an important regulator of alternative pathway activation. We report a diagnostic work-up of a patient with relapsing inflammatory mediated meningo-encephalitis. Our work-up revealed a rare genetic factor I (FI) deficiency. So far, all cases of reported complete factor I deficiency have absent serum levels of FI. We present here a unique case of a complete factor I deficiency based on a functional FI defect. METHODS: Complement assays and measurement of FI activity were performed in the patient, her family, factor H-deficient patients, a patient with C3-nephritic factor and 11 healthy controls. Genetic sequencing of the FI coding regions in the patient and her parents was performed. RESULTS: The patient had absent alternative pathway activity with low levels of C3 and normal serum level of FI. The patient's plasma FI did not degrade C3b, with normalisation of C3b degradation after adding purified FI. Mutation analysis of the complement factor I gene revealed two heterozygous mutations (I322T and D506V). CONCLUSION: To our knowledge, this paper describes a complete FI deficiency caused by a defect of FI activity for the first time. Normal FI concentration does not exclude a complete FI defect, additional functional analysis of FI is required in any patient with a defect of complement activation. Recurrent aseptic meningo-encephalitis is a rare clinical presentation of complete FI deficiency.


Subject(s)
Complement Factor I/deficiency , Complement Factor I/genetics , Meningitis, Aseptic/genetics , Meningitis, Aseptic/immunology , Meningoencephalitis/genetics , Meningoencephalitis/immunology , Complement Activation/genetics , Complement Activation/immunology , Complement Factor I/physiology , Hemolysis/genetics , Hemolysis/immunology , Humans , Meningitis, Aseptic/metabolism , Meningoencephalitis/metabolism , Recurrence , Sequence Analysis, DNA
5.
Nat Genet ; 45(7): 813-7, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23685748

ABSTRACT

Up to half of the heritability of age-related macular degeneration (AMD) is explained by common variants. Here, we report the identification of a rare, highly penetrant missense mutation in CFI encoding a p.Gly119Arg substitution that confers high risk of AMD (P = 3.79 × 10⁻6; odds ratio (OR) = 22.20, 95% confidence interval (CI) = 2.98-164.49). Plasma and sera from cases carrying the p.Gly119Arg substitution mediated the degradation of C3b, both in the fluid phase and on the cell surface, to a lesser extent than those from controls. Recombinant protein studies showed that the Gly119Arg mutant protein is both expressed and secreted at lower levels than wild-type protein. Consistent with these findings, human CFI mRNA encoding Arg119 had reduced activity compared to wild-type mRNA encoding Gly119 in regulating vessel thickness and branching in the zebrafish retina. Taken together, these findings demonstrate that rare, highly penetrant mutations contribute to the genetic burden of AMD.


Subject(s)
Complement Factor I/genetics , Macular Degeneration/genetics , Mutation, Missense , Amino Acid Substitution , Animals , Animals, Genetically Modified , Base Sequence , Complement Factor I/physiology , Embryo, Nonmammalian , Genetic Predisposition to Disease , HEK293 Cells , Humans , Macular Degeneration/pathology , Models, Genetic , Models, Molecular , Mutation, Missense/physiology , Retina/embryology , Retina/metabolism , Retina/pathology , Risk Factors , Zebrafish
6.
J Clin Invest ; 118(2): 608-18, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18202746

ABSTRACT

The inflammatory kidney disease membranoproliferative glomerulonephritis type II (MPGN2) is associated with dysregulation of the alternative pathway of complement activation. MPGN2 is characterized by the presence of complement C3 along the glomerular basement membrane (GBM). Spontaneous activation of C3 through the alternative pathway is regulated by 2 plasma proteins, factor H and factor I. Deficiency of either of these regulators results in uncontrolled C3 activation, although the breakdown of activated C3 is dependent on factor I. Deficiency of factor H, but not factor I, is associated with MPGN2 in humans, pigs, and mice. To explain this discordance, mice with single or combined deficiencies of these factors were studied. MPGN2 did not develop in mice with combined factor H and I deficiency or in mice deficient in factor I alone. However, administration of a source of factor I to mice with combined factor H and factor I deficiency triggered both activated C3 fragments in plasma and GBM C3 deposition. Mouse renal transplant studies demonstrated that C3 deposited along the GBM was derived from plasma. Together, these findings provide what we believe to be the first evidence that factor I-mediated generation of activated C3 fragments in the circulation is a critical determinant for the development of MPGN2 associated with factor H deficiency.


Subject(s)
Complement C3/metabolism , Complement Factor H/deficiency , Complement Factor I/physiology , Glomerulonephritis, Membranoproliferative/genetics , Animals , Complement C3/analysis , Complement Factor H/administration & dosage , Complement Factor H/genetics , Complement Factor I/administration & dosage , Complement Factor I/genetics , Glomerulonephritis, Membranoproliferative/blood , Glomerulonephritis, Membranoproliferative/immunology , Mice , Mice, Mutant Strains
7.
J Immunol ; 174(10): 6227-32, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15879120

ABSTRACT

Complement component C5 binds to components C6 and C7 in reversible reactions that are distinct from the essentially nonreversible associations that form during assembly of the complement membrane attack complex (MAC). We previously reported that the approximately 150-aa residue C345C domain (also known as NTR) of C5 mediates these reversible reactions, and that the corresponding recombinant module (rC5-C345C) binds directly to the tandem pair of approximately 75-residue factor I modules from C7 (C7-FIMs). We suggested from these and other observations that binding of the C345C module of C5 to the FIMs of C7, but not C6, is also essential for MAC assembly itself. The present report describes a novel method for assembling a complex that appears to closely resemble the MAC on the sensor chip of a surface plasmon resonance instrument using the complement-reactive lysis mechanism. This method provides the ability to monitor individually the incorporation of C7, C8, and C9 into the complex. Using this method, we found that C7 binds to surface-bound C5b,6 with a K(d) of approximately 3 pM, and that micromolar concentrations of either rC5-C345C or rC7-FIMs inhibit this early step in MAC formation. We also found that similar concentrations of either module inhibited complement-mediated erythrocyte lysis by both the reactive lysis and classical pathway mechanisms. These results demonstrate that the interaction between the C345C domain of C5 and the FIMs of C7, which mediates reversible binding of C5 to C7 in solution, also plays an essential role in MAC formation and complement lytic activity.


Subject(s)
Complement C5/physiology , Complement C7/antagonists & inhibitors , Complement C7/physiology , Complement Factor I/physiology , Complement Membrane Attack Complex/antagonists & inhibitors , Cysteine , Recombinant Proteins/pharmacology , Amino Acid Motifs , Animals , Binding, Competitive , Complement C5/metabolism , Complement C5b , Complement C7/metabolism , Complement Inactivator Proteins/antagonists & inhibitors , Complement Inactivator Proteins/metabolism , Complement Inactivator Proteins/physiology , Complement Membrane Attack Complex/metabolism , Complement Pathway, Classical/immunology , Hemolysis/immunology , Humans , Kinetics , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sheep , Surface Plasmon Resonance/instrumentation , Surface Plasmon Resonance/methods , Surface Properties
8.
J Immunol ; 173(1): 367-75, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15210795

ABSTRACT

Complement factor I (fI) plays a major role in the regulation of the complement system. It circulates in an active form and has very restricted specificity, cleaving only C3b or C4b in the presence of a cofactor such as factor H (fH), complement receptor type 1, membrane cofactor protein, or C4-binding protein. Using peptide-7-amino-4-methylcoumarin derivatives, we investigated the substrate specificity of fI. There is no previous report of synthetic substrate cleavage by fI, but five substrates were found in this study. A survey of 15 substrates and a range of inhibitors showed that fI has specificity similar to that of thrombin, but with much lower catalytic activity than that of thrombin. fI amidolytic activity has a pH optimum of 8.25, typical of serine proteases and is insensitive to ionic strength. This is in contrast to its proteolytic activity within the fI-C3b-fH reaction, in which the pH optimum for C3b cleavage is <5.5 and the reaction rate is highly dependent on ionic strength. The rate of cleavage of tripeptide 7-amino-4-methylcoumarins by fI is unaffected by the presence of fH or C3(NH(3)). The amidolytic activity is inhibited by the synthetic thrombin inhibitor Z-D-Phe-Pro-methoxypropylboroglycinepinanediol ester, consistent with previous reports, and by benzenesulfonyl fluorides such as Pefabloc SC. Suramin inhibits fI directly at concentration of 1 mM. Within a range of metal ions tested, only Cr(2+) and Fe(3+) were found to inhibit both the proteolytic and amidolytic activity of fI.


Subject(s)
Complement Factor I/physiology , Complement Factor I/antagonists & inhibitors , Hot Temperature , Humans , Hydrogen-Ion Concentration , Magnesium/pharmacology , Osmolar Concentration , Substrate Specificity , Zinc/pharmacology
9.
J Biol Chem ; 274(45): 31811-8, 1999 Nov 05.
Article in English | MEDLINE | ID: mdl-10542204

ABSTRACT

In order to elucidate the function of complement component C6, truncated C6 molecules were expressed recombinantly. These were either deleted of the factor I modules (FIMs) (C6des-748-913) or both complement control protein (CCP) modules and FIMs (C6des-611-913). C6des-748-913 exhibited approximately 60-70% of the hemolytic activity of full-length C6 when assayed for Alternative Pathway activity, but when measured for the Classical Pathway, C6des-748-914 was only 4-6% as effective as C6. The activity difference between C6 and C6des-748-913 for the two complement pathways can be explained by a greater stability of newly formed metastable C5b* when produced by the Alternative Pathway compared with that made by the Classical Pathway. The half-lives of metastable C5b* and the decay of (125)I-C5b measured from cells used to activate the Alternative Pathway were found to be about 5-12-fold longer than those same parameters derived from cells that had activated the Classical Pathway. (125)I-C5 binds reversibly to C6 in an ionic strength-dependent fashion, but (125)I-C5 binds only weakly to C6des-FIMs and not at all to C6des-CCP/FIMs. Therefore, although the FIMs are not required absolutely for C6 activity, these modules promote interaction of C6 with C5 enabling a more efficient bimolecular coupling ultimately leading to the formation of the C5b-6 complex.


Subject(s)
Complement C6/physiology , Complement Factor I/physiology , Animals , CHO Cells , Complement C6/chemistry , Complement C9/chemistry , Complement C9/physiology , Complement Factor I/chemistry , Complement Pathway, Alternative , Complement Pathway, Classical , Cricetinae , Humans , Rabbits , Sheep , Structure-Activity Relationship
10.
J Immunol ; 162(7): 4088-93, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10201932

ABSTRACT

Previous studies based on the use of human serum as a source of C have provided evidence for the C-dependent enhancement of cell infection by HIV-1. The present study was undertaken to distinguish C from other serum factors and to identify the proteins and the mechanisms involved in C-dependent cell infection by HIV-1. The classical C activation pathway was reconstituted from the proteins C1q, C1r, C1s, C4, C2, C3, factor H, and factor I; each were purified to homogeneity. A mixture of these proteins at physiological concentrations was shown to reproduce the ability of normal human serum to enhance the infection of MT2 cells by HIV-1 at low doses of virus. This enhancing effect was abolished when heat-inactivated serum and C2- or C3-depleted serum were used, and was restored upon addition of the corresponding purified proteins. A mixture of two synthetic peptides corresponding to positions 10-15 and 90-97 of human C receptor type 2 (CD21) as well as soluble CD4 both inhibited the C-dependent infection process. These data provide unambiguous evidence that HIV-1 triggers a direct activation of the classical C pathway in vitro and thereby facilitates the infection of MT2 cells at low doses of virus. These findings are consistent with a mechanism involving increased interaction between the virus opsonized by C3b-derived fragment(s) and the CD21 cell receptors and subsequent virus entry through CD4 receptors.


Subject(s)
Complement System Proteins/physiology , HIV-1/immunology , Models, Immunological , Acquired Immunodeficiency Syndrome/immunology , Acquired Immunodeficiency Syndrome/virology , Cell Line , Complement C2/deficiency , Complement C2/physiology , Complement C3/deficiency , Complement C3/physiology , Complement C4/isolation & purification , Complement C4/physiology , Complement Factor H/isolation & purification , Complement Factor H/physiology , Complement Factor I/isolation & purification , Complement Factor I/physiology , Dose-Response Relationship, Immunologic , Drug Synergism , Humans , Receptors, Complement 3d/chemistry , Receptors, Complement 3d/metabolism
11.
Mol Immunol ; 31(3): 227-40, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8114768

ABSTRACT

This study was designed to explore the role of Factor I in the release of immune complexes (IC) from human erythrocytes (E). The interactions between E and IC constructed with murine monoclonal antibodies were examined using, as a complement source, autologous plasma, plasma depleted of Factor I by > 90%, or Factor I-depleted plasma reconstituted with purified Factor I. Striking differences were observed in the interactions between E and different types of IC in Factor I-depleted plasma. The release of IC constructed with IgG1, IgG3, IgM or IgA antibodies was abolished by Factor I depletion whereas IC containing IgG2a or IgG2b antibodies were still released from E in Factor I-depleted plasma. Moreover, when IC containing IgG2a antibodies were incubated briefly in Factor I-depleted plasma, under conditions in which the IC were bound but not released, and then resuspended in the presence or absence of Factor I, as little as 5% of the normal physiologic level of Factor I released the IC from E. Thus, IC containing IgG2a antibodies appear to be exquisitely susceptible to release from E by Factor I. Additional differences in the susceptibility of IC containing IgG1, IgG3, IgM or IgA antibodies to release from E were revealed when Factor I-depleted plasma was reconstituted with Factor I. Under these conditions, the relative susceptibility of IC to release was: IC containing IgG1 or IgA antibodies > IC containing IgM antibodies > IC constructed with IgG3 antibodies. While isotype was critical in determining susceptibility to release, some clonotypic differences between isotype-matched pairs of IC were also evident. Differences in IC release from E by Factor I may reflect antibody matrix-mediated differential susceptibility of IC-bound C3b and/or C4b to cleavage by Factor I and may have implications for immunoregulation, host effector cell mechanisms and the pathophysiology of IC diseases.


Subject(s)
Antigen-Antibody Complex/metabolism , Complement Factor I/physiology , Erythrocytes/ultrastructure , Receptors, Complement 3b/metabolism , Complement Factor I/deficiency , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin G/immunology
12.
Mol Immunol ; 30(9): 841-8, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8321249

ABSTRACT

Three allotypes of mouse factor H, H.1, H.2, and H.3 were purified from the sera of mice with different factor H allotypes, and their functional properties were investigated. The three allotypes all bound to heparin, DNA, Con A, and methylamine-treated mouse C3 (C3(MA)mo) with similar affinities for each protein immobilized, showed identical mobilities on SDS-PAGE, and were reacted well with rabbit polyclonal antibody against H.1 and H.2. Factor I-cofactor activity of these factor H allotypes was measured using highly purified material of mouse, guinea-pig, and human origin. In a homologous system, these allotypes expressed indistinguishable mouse factor I (Imo)-cofactor activity for the cleavage of C3(MA)mo. Imo-cofactor activity was again indistinguishable in these allotypes when methylamine-treated human C3 (C3(MA)hu) or methylamine-treated guinea-pig C3 (C3(MA)gp) was substituted for the C3(MA)mo substrate. The cofactor activity of these factor H allotypes, however, was augmented 4-5 times if C3(MA)hu) was used instead of C3(MA)mo, and was barely detected if C3(MA)gp was employed. In contrast, differences in the potency of the cofactor activity for the three allotypes were revealed if human factor 1 (Ihu) was substituted for Imo: the order of the efficiency for the cleavage of C3(MA)hu was H.2 > H.1 = H.3. These results, taken together with the finding that the homologous combinations of mouse and human factors H and I expressed greater activity for the cleavage of C3(MA)hu than did the heterologous combinations of factor H and factor I, suggest that mouse factor H allotypes discriminate species of protease factor I but not those of substrate (C3(MA), and H.2 possesses the best compatibility for Ihu in C3(MA)hu inactivation.


Subject(s)
Complement Factor H/physiology , Complement Factor I/physiology , Animals , Complement Factor H/chemistry , Complement Factor H/isolation & purification , Humans , Mice , Mice, Inbred BALB C , Structure-Activity Relationship
13.
J Immunol ; 150(9): 4052-60, 1993 May 01.
Article in English | MEDLINE | ID: mdl-8473749

ABSTRACT

B lymphocytes and transformed B lymphoblastoid cell lines express CR2 (CD21, C3d/EBV-receptor) that is specific for C3 fragments generated by cleavage of C3b or spontaneously hydrolyzed native C3 (C3i) by the serum enzyme factor I and its cofactor, factor H. It had been shown previously that the Raji B cell line could be cultivated in serum-free medium supplemented with only transferrin and either OKB7 anti-CR2 mAb, C3d, or C3d-derived peptides containing the CR2 binding site. Because these agents appeared to function through ligation of CR2, it was unclear how native C3 could also serve as a growth factor, because C3 does not bind to CR2. It appeared possible that Raji cells might be able to use endogenous factors H and I to generate a CR2 ligand from C3, because previous studies had shown that Raji cells synthesized factor H and probably also synthesized factor I. PCR analysis was used to demonstrate factor I mRNA in Raji cells. Secretion of Raji cell factor I protein was confirmed by a sensitive mAb ELISA. Several B cell lines were examined for C3-dependent growth. Raji cells required both C3 (or OKB7) and transferrin for growth, whereas Wil-2 cells grew with transferrin alone and C3 enhanced the growth-promoting activity of transferrin. Two other B cell lines (Daudi and U698M), the T cell line 8402, and the U937 monocytoid cell line could not be sustained with transferrin plus C3. The C3-dependent growth of Raji cells was inhibited almost completely by either OX-23 anti-factor H or 052.11.3 anti-factor I mAb that also blocked the activity of serum-derived factor H or I, respectively. By contrast, there was no inhibition of growth by either OX-24 anti-factor H or OX-21 anti-factor I mAb that did not block factors H and I activity. After the spontaneous hydrolysis of native C3 to C3i, it is hypothesized that Raji cells convert C3i to iC3i with endogenous factors H and I, and then this iC3i serves as a growth factor by binding to membrane CR2.


Subject(s)
B-Lymphocytes/physiology , Complement C3/metabolism , Complement Factor H/physiology , Complement Factor I/physiology , Growth Substances/metabolism , Animals , Base Sequence , Cell Division/drug effects , Cell Line , Complement C3/pharmacology , Complement Factor H/biosynthesis , Complement Factor I/biosynthesis , Culture Media, Serum-Free , Humans , Mice , Molecular Sequence Data , Transferrin/pharmacology
14.
J Immunol ; 149(8): 2592-9, 1992 Oct 15.
Article in English | MEDLINE | ID: mdl-1401896

ABSTRACT

The human complement regulatory protein, factor H, was examined by high resolution transmission electron microscopy. Results of electron microscopy confirm hydrodynamic analysis and indicate that factor H is a monomer of M(r) approximately 155,000. Factor H is an extended flexible molecule with a contour length of 495 A and a cross-sectional diameter of 34 A. Most images of factor H indicate that its polypeptide chain typically folds back on itself with the result that the average length of a factor H molecule is about half its contour length. Only one end of factor H associates with C3b. When bound to C3b, factor H still shows considerable conformational flexibility. Factor I is a bilobal protein of 130 A in length, and its two globular parts have maximal diameters of 54 and 49 A. The results establish that factor I is a two domain protein where the smaller subunit is a protease and the larger one is involved with binding C3b. Factor I binds C3b with a one-to-one stoichiometry in an ionic strength-dependent fashion. In the absence of sodium chloride an affinity constant of 5.7 x 10(5) M-1 was determined for factor I interaction with C3b. Whereas the Scatchard plot of factor I binding to C3b in the absence of factor H is linear, in the presence of factor H a curvilinear graph is obtained. The strong binding sites on C3b for factor I have an affinity at least 15-fold higher in the presence of factor H than in its absence. The results of both electron microscopy and binding studies were combined to compose a scheme envisioning how factors H and I cooperate for the processing of C3b.


Subject(s)
Complement Factor H/physiology , Complement Factor H/ultrastructure , Complement Factor I/physiology , Complement Factor I/ultrastructure , Binding Sites , Complement C3b/metabolism , Humans , Microscopy, Electron
SELECTION OF CITATIONS
SEARCH DETAIL
...