Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Biotechnol Bioeng ; 118(1): 423-432, 2021 01.
Article in English | MEDLINE | ID: mdl-32970320

ABSTRACT

Vascular leak is a key driver of organ injury in diseases, and strategies that reduce enhanced permeability and vascular inflammation are promising therapeutic targets. Activation of the angiopoietin-1 (ANG1)-Tie2 tyrosine kinase signaling pathway is an important regulator of vascular quiescence. Here we describe the design and construction of a new soluble ANG1 mimetic that is a potent activator of endothelial Tie2 in vitro and in vivo. Using a chimeric fusion strategy, we replaced the extracellular matrix (ECM) binding and oligomerization domain of ANG1 with a heptameric scaffold derived from the C-terminus of serum complement protein C4-binding protein α. We refer to this new fusion protein biologic as Hepta-ANG1, which forms a stable heptamer and induces Tie2 phosphorylation in cultured cells, and in the lung following intravenous injection of mice. Injection of Hepta-ANG1 ameliorates vascular endothelial growth factor- and lipopolysaccharide-induced vascular leakage, in keeping with the known functions of Angpt1-Tie2 in maintaining quiescent vascular stability. The new Hepta-ANG1 fusion is easy to produce and displays remarkable stability with high multimericity that can potently activate Tie2. It could be a new candidate ANG1 mimetic therapy for treatments of inflammatory vascular leak, such as acute respiratory distress syndrome and sepsis.


Subject(s)
Angiopoietin-1 , Complement C4b-Binding Protein , Human Umbilical Vein Endothelial Cells/metabolism , Recombinant Fusion Proteins , Vascular Diseases/drug therapy , Angiopoietin-1/biosynthesis , Angiopoietin-1/genetics , Angiopoietin-1/pharmacology , Animals , Complement C4b-Binding Protein/biosynthesis , Complement C4b-Binding Protein/genetics , Complement C4b-Binding Protein/pharmacology , Female , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Protein Domains , Rabbits , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology , Sepsis/drug therapy , Sepsis/metabolism , Sepsis/pathology , Vascular Diseases/metabolism , Vascular Diseases/pathology
2.
Anesth Analg ; 132(1): e1-e5, 2021 01 01.
Article in English | MEDLINE | ID: mdl-30169415

ABSTRACT

Opioids may influence inflammation. We compared genes associated with pain and inflammation in patients who consumed opioids (3-120 mg of oral morphine equivalents per day) with those who did not for differential expression. White blood cells were assayed in 20 patients presenting for total lower extremity joint replacement. We focused on messenger ribonucleic acid expression of complement proteins. We report that the expression of a complement inhibitor, complement 4 binding protein A, was reduced, and the expression of a complement activator, complement factor D, was increased in opioid-consuming patients. We conclude that opioid consumption may influence expression of complement activators and inhibitors.


Subject(s)
Analgesics, Opioid/administration & dosage , Complement C4b-Binding Protein/biosynthesis , Elective Surgical Procedures/trends , Complement C4b-Binding Protein/antagonists & inhibitors , Complement C4b-Binding Protein/genetics , Complement System Proteins , Female , Gene Expression , Humans , Male , Pain, Postoperative/blood , Pain, Postoperative/genetics , Pain, Postoperative/prevention & control
3.
J Hum Genet ; 65(8): 657-665, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32277175

ABSTRACT

Age-related macular degeneration (AMD) is the leading cause of irreversible blindness among the elderly population. To accelerate the understanding of the genetics of AMD, we conducted a meta-analysis of genome-wide association studies (GWAS) combining data from the International AMD Genomics Consortium AMD-2016 GWAS (16,144 advanced AMD cases and 17,832 controls), AMD-2013 GWAS (17,181 cases and 60,074 controls), and new data on 4017 AMD cases and 14,984 controls from Genetic Epidemiology Research on Aging study. We identified 12 novel AMD loci near or within C4BPA-CD55, ZNF385B, ZBTB38, NFKB1, LINC00461, ADAM19, CPN1, ACSL5, CSK, RLBP1, CLUL1, and LBP. We then replicated the associations of the novel loci in independent cohorts, UK Biobank (5860 cases and 126,726 controls) and FinnGen (1266 cases and 47,560 control). In general, the concordance in effect sizes was very high (correlation in effect size estimates 0.89), 11 of 12 novel loci were in the expected direction, 5 were associated with AMD at a nominal significance level, and rs3825991 (near gene RLBP1) after Bonferroni correction. We identified an additional 21 novel genes using a gene-based test. Most of the novel genes are expressed in retinal tissue and could be involved in the pathogenesis of AMD (i.e., complement, inflammation, and lipid pathways). These findings enhance our understanding of the genetic architecture of AMD and shed light on the biological process underlying AMD pathogenesis.


Subject(s)
Macular Degeneration/genetics , ADAM Proteins/genetics , Acute-Phase Proteins/genetics , CD55 Antigens/genetics , Carrier Proteins/genetics , Coenzyme A Ligases/genetics , Complement C4b-Binding Protein/genetics , Databases, Genetic , Eye Proteins/genetics , Genetic Loci , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Membrane Glycoproteins/genetics , NF-kappa B p50 Subunit/genetics , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Repressor Proteins/genetics , Retina/metabolism , Retina/pathology
4.
Hum Genet ; 137(4): 305-314, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29675612

ABSTRACT

Single nucleotide variants (SNVs) within and surrounding the complement receptor 1 (CR1) gene show some of the strongest genome-wide association signals with late-onset Alzheimer's disease. Some studies have suggested that this association signal is due to a duplication allele (CR1-B) of a low copy repeat (LCR) within the CR1 gene, which increases the number of complement C3b/C4b-binding sites in the mature receptor. In this study, we develop a triplex paralogue ratio test assay for CR1 LCR copy number allowing large numbers of samples to be typed with a limited amount of DNA. We also develop a CR1-B allele-specific PCR based on the junction generated by an historical non-allelic homologous recombination event between CR1 LCRs. We use these methods to genotype CR1 and measure CR1-B allele frequency in both late-onset and early-onset cases and unaffected controls from the United Kingdom. Our data support an association of late-onset Alzheimer's disease with the CR1-B allele, and confirm that this allele occurs most frequently on the risk haplotype defined by SNV alleles. Furthermore, regression models incorporating CR1-B genotype provide a better fit to our data compared to incorporating the SNV-defined risk haplotype, supporting the CR1-B allele as the variant underlying the increased risk of late-onset Alzheimer's disease.


Subject(s)
Alzheimer Disease/genetics , DNA Copy Number Variations/genetics , Genetic Predisposition to Disease , Receptors, Complement 3b/genetics , Adult , Aged , Aged, 80 and over , Alleles , Alzheimer Disease/physiopathology , Binding Sites , Complement C3b/genetics , Complement C4b-Binding Protein/genetics , Female , Gene Duplication/genetics , Genome-Wide Association Study , Genotype , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Risk Factors , United Kingdom
5.
Methods ; 111: 12-20, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27592383

ABSTRACT

Differential expression (DE) analysis is commonly used to identify biomarker candidates that have significant changes in their expression levels between distinct biological groups. One drawback of DE analysis is that it only considers the changes on single biomolecule level. Recently, differential network (DN) analysis has become popular due to its capability to measure the changes on biomolecular pair level. In DN analysis, network is typically built based on correlation and biomarker candidates are selected by investigating the network topology. However, correlation tends to generate over-complicated networks and the selection of biomarker candidates purely based on network topology ignores the changes on single biomolecule level. In this paper, we propose a novel approach, INDEED, that builds sparse differential network based on partial correlation and integrates DE and DN analyses for biomarker discovery. We applied this approach on real proteomic and glycomic data generated by liquid chromatography coupled with mass spectrometry for hepatocellular carcinoma (HCC) biomarker discovery study. For each omic data, we used one dataset to select biomarker candidates, built a disease classifier and evaluated the performance of the classifier on an independent dataset. The biomarker candidates, selected by INDEED, were more reproducible across independent datasets, and led to a higher classification accuracy in predicting HCC cases and cirrhotic controls compared with those selected by separate DE and DN analyses. INDEED also identified some candidates previously reported to be relevant to HCC, such as intercellular adhesion molecule 2 (ICAM2) and c4b-binding protein alpha chain (C4BPA), which were missed by both DE and DN analyses. In addition, we applied INDEED for survival time prediction based on transcriptomic data acquired by analysis of samples from breast cancer patients. We selected biomarker candidates and built a regression model for survival time prediction based on a gene expression dataset and patients' survival records. We evaluated the performance of the regression model on an independent dataset. Compared with the biomarker candidates selected by DE and DN analyses, those selected through INDEED led to more accurate survival time prediction.


Subject(s)
Antigens, CD/genetics , Biomarkers, Tumor/genetics , Cell Adhesion Molecules/genetics , Complement C4b-Binding Protein/genetics , Proteomics/methods , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Chromatography, Liquid , Gene Expression Regulation, Neoplastic , Glycomics/methods , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mass Spectrometry , Transcriptome/genetics
6.
J Biol Chem ; 290(30): 18333-42, 2015 Jul 24.
Article in English | MEDLINE | ID: mdl-26067271

ABSTRACT

The complement, coagulation, and fibrinolytic systems are crucial for the maintenance of tissue homeostasis. To date numerous interactions and cross-talks have been identified between these cascades. In line with this, here we propose a novel, hitherto unknown interaction between the complement inhibitor C4b-binding protein (C4BP) and plasminogen of the fibrinolytic pathway. Binding of C4BP to Streptococcus pneumoniae is a known virulence mechanism of this pathogen and it was increased in the presence of plasminogen. Interestingly, the acute phase variant of C4BP lacking the ß-chain and protein S binds plasminogen much stronger than the main isoform containing the ß-chain and protein S. Indeed, the complement control protein (CCP) 8 domain of C4BP, which would otherwise be sterically hindered by the ß-chain, primarily mediates this interaction. Moreover, the lysine-binding sites in plasminogen kringle domains facilitate the C4BP-plasminogen interaction. Furthermore, C4BP readily forms complexes with plasminogen in fluid phase and such complexes are present in human serum and plasma. Importantly, whereas the presence of plasminogen did not affect the factor I cofactor activity of C4BP, the activation of plasminogen by urokinase-type plasminogen activator to active plasmin was significantly augmented in the presence of C4BP. Taken together, our data demonstrate a novel interaction between two proteins of the complement and fibrinolytic system. Most complexes might be formed during the acute phase of inflammation and have an effect on the homeostasis at the site of injury or acute inflammation.


Subject(s)
Complement C4b-Binding Protein/metabolism , Inflammation/metabolism , Plasminogen/metabolism , Streptococcus pneumoniae/metabolism , Complement C4b-Binding Protein/genetics , Fibrinogen/metabolism , Fibrinolysin/metabolism , Fibrinolysis/genetics , Humans , Inflammation/pathology , Lysine/metabolism , Plasminogen/genetics , Plasminogen Activators/metabolism , Protein Binding , Protein Interaction Maps , Protein S/metabolism , Streptococcus pneumoniae/pathogenicity , Surface Plasmon Resonance , Urokinase-Type Plasminogen Activator/metabolism
7.
Mol Ecol ; 25(6): 1324-39, 2016 03.
Article in English | MEDLINE | ID: mdl-26836579

ABSTRACT

The complement system is an innate immunity effector mechanism; its action is antagonized by a wide array of pathogens and complement evasion determines the virulence of several infections. We investigated the evolutionary history of the complement system and of bacterial-encoded complement-interacting proteins. Complement components targeted by several pathogens evolved under strong selective pressure in primates, with selection acting on residues at the contact interface with microbial/viral proteins. Positively selected sites in CFH and C4BPA account for the human specificity of gonococcal infection. Bacterial interactors, evolved adaptively as well, with selected sites located at interaction surfaces with primate complement proteins. These results epitomize the expectation under a genetic conflict scenario whereby the host's and the pathogen's genes evolve within binding avoidance-binding seeking dynamics. In silico mutagenesis and protein-protein docking analyses supported this by showing that positively selected sites, both in the host's and in the pathogen's interacting partner, modulate binding.


Subject(s)
Biological Evolution , Complement System Proteins/genetics , Host-Pathogen Interactions/genetics , Primates/genetics , Animals , Bacteria/pathogenicity , Complement Activation , Complement C4b-Binding Protein/genetics , Complement Factor H/genetics , Genetics, Population , Humans , Immunity, Innate , Molecular Docking Simulation , Phylogeny , Protein Interaction Mapping , Selection, Genetic , Sequence Analysis, DNA
8.
J Immunol ; 190(6): 2857-72, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23390292

ABSTRACT

The classical pathway complement regulator C4b-binding protein (C4BP) is composed of two polypeptides (α- and ß-chains), which form three plasma oligomers with different subunit compositions (α7ß1, α7ß0, and α6ß1). We show in this article that the C4BP α7ß0 isoform (hereafter called C4BP[ß(-)] [C4BP lacking the ß-chain]), overexpressed under acute-phase conditions, induces a semimature, tolerogenic state on human monocyte-derived dendritic cells (DCs) activated by a proinflammatory stimulus. C4BP isoforms containing ß-chain (α7ß1 and α6ß1; C4BP[ß(+)]) neither interfered with the normal maturation of DCs nor competed with C4BP(ß(-)) activity on these cells. Immature DCs (iDCs) treated with C4BP(ß(-)) retained high endocytic activity, but, upon LPS treatment, they did not upregulate surface expression of CD83, CD80, and CD86. Transcriptional profiling of these semimature DCs revealed that treatment with C4BP(ß(-)) prevented the induction of IDO and BIC-1, whereas TGF-ß1 expression was maintained to the level of iDCs. C4BP(ß(-))-treated DCs were also unable to release proinflammatory Th1 cytokines (IL-12, TNF-α, IFN-γ, IL-6, IL-8) and, conversely, increased IL-10 secretion. They prevented surface CCR7 overexpression and, accordingly, displayed reduced chemotaxis, being morphologically indistinguishable from iDCs. Moreover, C4BP(ß(-))-treated DCs failed to enhance allogeneic T cell proliferation, impairing IFN-γ production in these cells and, conversely, promoting CD4(+)CD127(low/neg)CD25(high)Foxp3(+) T cells. Deletion mutant analysis revealed that the complement control protein-6 domain of the α-chain is necessary for the tolerogenic activity of C4BP(ß(-)). Our data demonstrate a novel anti-inflammatory and immunomodulatory function of the complement regulator C4BP, suggesting a relevant role of the acute-phase C4BP(ß(-)) isoform in a number of pathophysiological conditions and potential applications in autoimmunity and transplantation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cell Differentiation/immunology , Complement C4b-Binding Protein/physiology , Dendritic Cells/chemistry , Dendritic Cells/immunology , Histocompatibility Antigens/physiology , Cell Differentiation/genetics , Complement C4b-Binding Protein/chemistry , Complement C4b-Binding Protein/genetics , Dendritic Cells/pathology , HEK293 Cells , Histocompatibility Antigens/chemistry , Histocompatibility Antigens/genetics , Humans , Immune Tolerance/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/prevention & control , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/physiology
9.
J Biol Chem ; 288(38): 27494-27504, 2013 Sep 20.
Article in English | MEDLINE | ID: mdl-23928307

ABSTRACT

Group A Streptococcus pyogenes (GAS) strain AP53 is a primary isolate from a patient with necrotizing fasciitis. These AP53 cells contain an inactivating mutation in the sensor component of the cluster of virulence (cov) responder (R)/sensor (S) two-component gene regulatory system (covRS), which enhances the virulence of the primary strain, AP53/covR(+)S(-). However, specific mechanisms by which the covRS system regulates the survival of GAS in humans are incomplete. Here, we show a key role for covRS in the regulation of opsonophagocytosis of AP53 by human neutrophils. AP53/covR(+)S(-) cells displayed potent binding of host complement inhibitors of C3 convertase, viz. Factor H (FH) and C4-binding protein (C4BP), which concomitantly led to minimal C3b deposition on AP53 cells, further showing that these plasma protein inhibitors are active on GAS cells. This resulted in weak killing of the bacteria by human neutrophils and a corresponding high death rate of mice after injection of these cells. After targeted allelic alteration of covS(-) to wild-type covS (covS(+)), a dramatic loss of FH and C4BP binding to the AP53/covR(+)S(+) cells was observed. This resulted in elevated C3b deposition on AP53/covR(+)S(+) cells, a high level of opsonophagocytosis by human neutrophils, and a very low death rate of mice infected with AP53/covR(+)S(+). We show that covRS is a critical transcriptional regulator of genes directing AP53 killing by neutrophils and regulates the levels of the receptors for FH and C4BP, which we identify as the products of the fba and enn genes, respectively.


Subject(s)
Bacterial Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neutrophils/metabolism , Phagocytosis , Repressor Proteins/metabolism , Streptococcus pyogenes/pathogenicity , Virulence Factors/metabolism , Animals , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Complement C3b/genetics , Complement C3b/metabolism , Complement C4b-Binding Protein/genetics , Complement C4b-Binding Protein/metabolism , Complement Factor H/genetics , Complement Factor H/metabolism , Female , Fructose-Bisphosphate Aldolase , Histidine Kinase , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Neutrophils/microbiology , Neutrophils/pathology , Protein Binding , Repressor Proteins/genetics , Streptococcal Infections/genetics , Streptococcal Infections/metabolism , Streptococcal Infections/pathology , Streptococcus pyogenes/genetics , Streptococcus pyogenes/metabolism , Virulence Factors/genetics
10.
Eur J Immunol ; 43(6): 1617-29, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23508668

ABSTRACT

Since a tightly regulated complement system is needed for a successful pregnancy, we hypothesized that alterations in complement inhibitors may be associated with idiopathic, recurrent miscarriage. We sequenced all exons coding for three complement inhibitors: C4b-binding protein (C4BP), CD46, and CD55 in 384 childless women with at least two miscarriages that could not be explained by known risk factors. Several alterations were found in C4BPA, of which the R120H, I126T, and the G423T mutations affected the expression level and/or the ability of recombinant C4BP to serve as cofactor for factor I. The only variant in C4BPB was located in the C-terminal part, and did not impair the polymerization of the molecule. Our results identify for the first time alterations in C4BP in women experiencing recurrent miscarriages. We also found four CD46 alterations in individual patients that were not found in healthy controls. One of the rare variants, P324L, showed decreased expression, whereas N213I resulted in deficient protein processing as well as an impaired cofactor activity in the degradation of both C4b and C3b. The identified alterations may result in in vivo consequences and contribute to the disorder but the degree of association must be evaluated in larger cohorts.


Subject(s)
Abortion, Habitual/genetics , Abortion, Habitual/immunology , CD55 Antigens/genetics , Complement C4b-Binding Protein/genetics , Membrane Cofactor Protein/genetics , Adult , DNA Mutational Analysis , Female , Fibrinogen/metabolism , Humans , Mutation/genetics , Polymorphism, Genetic , Pregnancy
11.
J Biol Chem ; 287(11): 8092-100, 2012 Mar 09.
Article in English | MEDLINE | ID: mdl-22267731

ABSTRACT

PRELP is a 58-kDa proteoglycan found in a variety of extracellular matrices, including cartilage and at several basement membranes. In rheumatoid arthritis (RA), the cartilage tissue is destroyed and fragmented molecules, including PRELP, are released into the synovial fluid where they may interact with components of the complement system. In a previous study, PRELP was found to interact with the complement inhibitor C4b-binding protein, which was suggested to locally down-regulate complement activation in joints during RA. Here we show that PRELP directly inhibits all pathways of complement by binding C9 and thereby prevents the formation of the membrane attack complex (MAC). PRELP does not interfere with the interaction between C9 and already formed C5b-8, but inhibits C9 polymerization thereby preventing formation of the lytic pore. The alternative pathway is moreover inhibited already at the level of C3-convertase formation due to an interaction between PRELP and C3. This suggests that PRELP may down-regulate complement attack at basement membranes and on damaged cartilage and therefore limit pathological complement activation in inflammatory disease such as RA. The net outcome of PRELP-mediated complement inhibition will highly depend on the local concentration of other complement modulating molecules as well as on the local concentration of available complement proteins.


Subject(s)
Arthritis, Rheumatoid/metabolism , Complement Membrane Attack Complex/antagonists & inhibitors , Complement Membrane Attack Complex/metabolism , Extracellular Matrix Proteins/metabolism , Glycoproteins/metabolism , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Basement Membrane/chemistry , Basement Membrane/metabolism , Basement Membrane/pathology , Complement Activation/genetics , Complement C3-C5 Convertases/chemistry , Complement C3-C5 Convertases/genetics , Complement C3-C5 Convertases/metabolism , Complement C4b-Binding Protein/chemistry , Complement C4b-Binding Protein/genetics , Complement C4b-Binding Protein/metabolism , Complement C9/chemistry , Complement C9/genetics , Complement C9/metabolism , Complement Membrane Attack Complex/chemistry , Complement Membrane Attack Complex/genetics , Complement System Proteins/chemistry , Complement System Proteins/genetics , Complement System Proteins/metabolism , Down-Regulation/genetics , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/genetics , Female , Glycoproteins/chemistry , Glycoproteins/genetics , HEK293 Cells , Humans , Male
12.
J Biol Chem ; 285(42): 32038-46, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20693287

ABSTRACT

The anticoagulant vitamin K-dependent protein S (PS) circulates in plasma in two forms, 30% free and 70% being bound to the complement regulatory protein C4b-binding protein (C4BP). The major C4BP isoform consists of 7 α-chains and 1 ß-chain (C4BPß(+)), the chains being linked by disulfide bridges. PS binds to the ß-chain with high affinity. In plasma, PS is in molar excess over C4BPß(+) and due to the high affinity, all C4BPß(+) molecules contain a bound PS. Taken together with the observation that PS-deficient patients have decreased levels of C4BPß(+), this raises the question of whether PS is important for secretion of the ß-chain from the cell. To test this hypothesis, HEK293 cells were stably and transiently transfected with ß-chain cDNA in combinations with cDNAs for PS and/or the α-chain. The concentration of ß-chains in the medium increased after co-transfection with PS cDNA, but not by α-chain cDNA, suggesting secretion of the ß-chains from the cells to be dependent on concomitant synthesis of PS, but not of the α-chains. Thus, ß-chains that were not disulfide-linked to the α-chains were secreted in complex with PS, either as monomers or dimers. Pulse-chase demonstrated that the complexes between PS and ß-chain were formed intracellularly, in the endoplasmic reticulum. In conclusion, our results demonstrate that successful secretion of ß-chains depends on intracellular complex formation with PS, but not on the α-chains. This provides an explanation for the decreased ß-chain levels observed in PS-deficient patients.


Subject(s)
Complement C4b-Binding Protein/chemistry , Complement C4b-Binding Protein/metabolism , Eukaryotic Cells/metabolism , Protein S/metabolism , Protein Subunits/chemistry , Protein Subunits/metabolism , Animals , Barium/metabolism , Cell Line , Complement C4b-Binding Protein/genetics , Humans , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein S/genetics , Protein Subunits/genetics
13.
J Immunol ; 182(3): 1518-25, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19155499

ABSTRACT

Components derived from cartilage have been suggested to maintain the inflammation in joints in arthritis. Small leucine-rich repeat proteins (SLRPs) are structural components of cartilage important in organizing the meshwork of extracellular matrix components. It has recently been shown that the SLRP fibromodulin interacts with complement initiator C1q, leading to complement activation. The complement response is limited since fibromodulin also interacts with the complement inhibitor factor H. We have now found that osteoadherin, chondroadherin, fibromodulin, and proline arginine-rich end leucine-rich repeat protein bind to the complement inhibitor C4b-binding protein (C4BP). Using direct binding assays with C4BP fragments and C4BP mutants lacking individual domains in combination with electron microscopy, we have demonstrated that mainly the central core of C4BP mediated binding to SLRPs. Binding of SLRPs to C4BP did not affect its ability to inhibit complement. Osteoadherin, fibromodulin, and chondroadherin, which bind C1q and activate complement, were found to cause significantly higher C9 deposition in C4BP-depleted serum compared with Igs, indicating that the level of complement activation initiated by SLRPs is regulated by simultaneous binding to C4BP. A similar dual binding of C1q and complement inhibitors was observed previously for other endogenous ligands (amyloid, prions, C-reactive protein, and apoptotic cells) but not for exogenous activators (bacteria-bound Igs). These interactions can be significant during inflammatory joint diseases, such as rheumatoid arthritis, where cartilage is degraded, and cartilage components are released into synovial fluid, where they can interact with factors of the complement system.


Subject(s)
Complement C4b-Binding Protein/metabolism , Complement Inactivating Agents/metabolism , Extracellular Matrix Proteins/metabolism , Glycoproteins/metabolism , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Collagen Type XI/metabolism , Complement Activation/genetics , Complement Activation/immunology , Complement C4b-Binding Protein/genetics , Fibromodulin , Glycoproteins/genetics , Humans , Protein Binding/immunology , Protein S/metabolism , Proteoglycans/metabolism , Repetitive Sequences, Amino Acid , Sequence Deletion/immunology , Synovial Fluid/metabolism
14.
J Immunol ; 182(12): 7865-77, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19494311

ABSTRACT

The complement system constitutes an important component of the innate immune system. To colonize their host and/or to cause disease, many pathogens have evolved strategies to avoid complement-mediated bacterial lysis and opsonophagocytosis. In this study, using a collection of 55 clinical isolates of Streptococcus pneumoniae, we demonstrate for the first time that pneumococci bind the complement inhibitor C4b-binding protein (C4BP). C4BP binding seems to be restricted to certain serotypes such as serotype 4, 6B, 7F, and 14, of which the strains of serotype 14 are the strongest binders. We show that bacteria-bound C4BP retains its functional activity and down-regulates the activation of the classical pathway. Thus, this major respiratory pathogen may escape immune recognition and eradication by the complement system. Furthermore, we show that C4BP binding varies between strains but is dependent on the expression of pneumococcal surface protein C, PspC of group 4. The study of the distribution of group 4 pspC locus shows that most of high-binder serotype 14 isolates harbor an allelic variant of group 4 pspC. Using PspC-negative mutant strains, we identified a new allelic variant of PspC (PspC4.4) as a major ligand for C4BP, revealing a new function for this important pneumococcal virulence factor. Thus pneumococci exploit host C4BP for complement evasion in a PspC allele-dependent manner.


Subject(s)
Alleles , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Complement C4b-Binding Protein/immunology , Complement C4b-Binding Protein/metabolism , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/metabolism , Bacterial Proteins/genetics , Cell Line , Complement C4b-Binding Protein/genetics , Humans , Ligands , Mutation/genetics , Phylogeny , Protein Binding , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/isolation & purification
15.
Mol Ther ; 18(5): 987-92, 2010 May.
Article in English | MEDLINE | ID: mdl-20179675

ABSTRACT

One of the major obstacles in the use of baculovirus vectors for in vivo gene transfer is the virus inactivation by serum complement. In this study, we investigated the effect of decay-accelerating factor (DAF), factor H (FH)-like protein-1 (FHL-1), C4b-binding protein (C4BP), and membrane cofactor protein (MCP) on protection of baculovirus vectors from the complement-mediated inactivation. Complement regulatory proteins were displayed on baculovirus surface as fusions to membrane anchor of the vesicular stomatitis virus-G (VSV-G) protein. This strategy resulted in abundant expression of recombinant proteins on the viral envelope while viral titers comparable to control virus were reached. The surface-modified vectors exhibited complement resistance in vitro, DAF showing the highest level of protection. Intraportal delivery of DAF-displaying baculovirus resulted in increased survival and enhanced gene expression in immunocompetent mice. Mice receiving DAF-displaying baculovirus also exhibited lower level of liver inflammation as evidenced by aspartate aminotransferase (AST). In line with this, macrophages treated with DAF baculovirus produced lower levels of inflammatory cytokines IL-1beta, IL-6, and IL-12p40 compared to control virus. These results suggest that DAF-display can protect the vector against complement inactivation but also reduce complement-mediated inflammation injury. In conclusion, complement shielded baculovirus vectors represent attractive tools for effective in vivo gene delivery.


Subject(s)
Baculoviridae/metabolism , Genetic Therapy/methods , Genetic Vectors/metabolism , Animals , Baculoviridae/genetics , CD55 Antigens/genetics , CD55 Antigens/metabolism , Cell Line , Complement C4b-Binding Protein/genetics , Complement C4b-Binding Protein/metabolism , Complement Factor H/genetics , Complement Factor H/metabolism , Electrophoresis, Polyacrylamide Gel , Genetic Vectors/genetics , Hep G2 Cells , Humans , Immunoblotting , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/metabolism , Mice , Mice, Inbred C57BL
16.
Front Immunol ; 12: 726801, 2021.
Article in English | MEDLINE | ID: mdl-34539665

ABSTRACT

Neisseria gonorrhoeae is the etiological agent of gonorrhea, the second most common bacterial sexually transmitted infection worldwide. Reproductive sequelae of gonorrhea include infertility, ectopic pregnancy and chronic pelvic pain. Most antibiotics currently in clinical use have been rendered ineffective due to the rapid spread of antimicrobial resistance among gonococci. The developmental pipeline of new antibiotics is sparse and novel therapeutic approaches are urgently needed. Previously, we utilized the ability of N. gonorrhoeae to bind the complement inhibitor C4b-binding protein (C4BP) to evade killing by human complement to design a chimeric protein that linked the two N-terminal gonococcal binding domains of C4BP with the Fc domain of IgM. The resulting molecule, C4BP-IgM, enhanced complement-mediated killing of gonococci. Here we show that C4BP-IgM induced membrane perturbation through complement deposition and membrane attack complex pore insertion facilitates the access of antibiotics to their intracellular targets. Consequently, bacteria become more susceptible to killing by antibiotics. Remarkably, C4BP-IgM restored susceptibility to azithromycin of two azithromycin-resistant clinical gonococcal strains because of overexpression of the MtrC-MtrD-MtrE efflux pump. Our data show that complement activation can potentiate activity of antibiotics and suggest a role for C4BP-IgM as an adjuvant for antibiotic treatment of drug-resistant gonorrhea.


Subject(s)
Anti-Bacterial Agents/pharmacology , Complement Activation , Complement C4b-Binding Protein/administration & dosage , Drug Resistance, Bacterial/drug effects , Immunoglobulin M/administration & dosage , Neisseria gonorrhoeae/drug effects , Azithromycin/pharmacology , Ciprofloxacin/pharmacology , Complement C4b-Binding Protein/genetics , Humans , Immunoglobulin M/genetics , Neisseria gonorrhoeae/growth & development , Recombinant Fusion Proteins/administration & dosage , Spectinomycin/pharmacology
17.
Iran J Immunol ; 18(2): 95-102, 2021 06.
Article in English | MEDLINE | ID: mdl-34190690

ABSTRACT

BACKGROUND: The immune evasion of dysplastic cells plays an important role in suppressing the immune response and progression of malignancy. The role of the complement inhibitors in the development of oral epithelial dysplastic lesions and squamous cell carcinoma (SCC) is still unclear. OBJECTIVE: This study aimed to assess the expression of C4 binding protein (C4BP) as a complement inhibitor in oral squamous cell carcinoma and leukoplakia. METHODS: In this study, 94 samples were classified into four groups: leukoplakia with mild to moderate dysplasia, leukoplakia with severe dysplasia or carcinoma in situ, early invasive SCC, and invasive SCC. The expression of C4BP marker was evaluated by immunohistochemistry (IHC) and real-time PCR. The results were analyzed by the Kruskal-Wallis, Bonferroni adjusted Dunn's multiple comparison, and one-way ANOVA tests. RESULTS: The results of IHC revealed the expression patterns of C4BP in oral dysplasia and SCC, and indicated that the C4BP expression was not significantly different between different histopathological grades in epithelial cells and vessels (P=0.157 and P=0.123, respectively) but, it was significantly different in fibroblasts and lymphocytes (P=0.017 and P=0.043, respectively). The real-time PCR showed a significant correlation between the dysplasia grade and expression of C4BP (P<0.05). CONCLUSION: According to the results, C4BP is expressed in the cancerous tissue by the tumor cells and their surrounding stroma. In addition, upregulation of the C4BP gene as an inhibitor of the complement system is a possible strategy adopted by the tumor cells to evade the immune system.


Subject(s)
Complement C4b-Binding Protein/physiology , Leukoplakia, Oral/immunology , Mouth Neoplasms/immunology , Squamous Cell Carcinoma of Head and Neck/immunology , Adult , Aged , Aged, 80 and over , Complement C4b-Binding Protein/analysis , Complement C4b-Binding Protein/genetics , Female , Humans , Immunohistochemistry , Leukoplakia, Oral/pathology , Male , Middle Aged , Mouth Neoplasms/pathology , Squamous Cell Carcinoma of Head and Neck/pathology
18.
PLoS Pathog ; 4(8): e1000140, 2008 Aug 29.
Article in English | MEDLINE | ID: mdl-18769718

ABSTRACT

Many pathogens are equipped with factors providing resistance against the bactericidal action of complement. Yersinia enterocolitica, a Gram-negative enteric pathogen with invasive properties, efficiently resists the deleterious action of human complement. The major Y. enterocolitica serum resistance determinants include outer membrane proteins YadA and Ail. Lipopolysaccharide (LPS) O-antigen (O-ag) and outer core (OC) do not contribute directly to complement resistance. The aim of this study was to analyze a possible mechanism whereby Y. enterocolitica could inhibit the antibody-mediated classical pathway of complement activation. We show that Y. enterocolitica serotypes O:3, O:8, and O:9 bind C4b-binding protein (C4bp), an inhibitor of both the classical and lectin pathways of complement. To identify the C4bp receptors on Y. enterocolitica serotype O:3 surface, a set of mutants expressing YadA, Ail, O-ag, and OC in different combinations was tested for the ability to bind C4bp. The studies showed that both YadA and Ail acted as C4bp receptors. Ail-mediated C4bp binding, however, was blocked by the O-ag and OC, and could be observed only with mutants lacking these LPS structures. C4bp bound to Y. enterocolitica was functionally active and participated in the factor I-mediated degradation of C4b. These findings show that Y. enterocolitica uses two proteins, YadA and Ail, to bind C4bp. Binding of C4bp could help Y. enterocolitica to evade complement-mediated clearance in the human host.


Subject(s)
Adhesins, Bacterial/metabolism , Antibodies, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Complement Activation , Complement C4b-Binding Protein/metabolism , Yersinia enterocolitica/metabolism , Adhesins, Bacterial/genetics , Bacterial Outer Membrane Proteins/genetics , Complement C4b/genetics , Complement C4b/metabolism , Complement C4b-Binding Protein/genetics , Fibrinogen/genetics , Fibrinogen/metabolism , Gene Expression Regulation, Bacterial/genetics , Humans , Mutation , O Antigens/genetics , O Antigens/metabolism , Protein Binding/genetics , Yersinia Infections/genetics , Yersinia Infections/metabolism , Yersinia enterocolitica/genetics
19.
J Immunol ; 181(8): 5537-44, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18832711

ABSTRACT

The periodontal pathogen Porphyromonas gingivalis is highly resistant to the bactericidal activity of human complement, which is present in the gingival crevicular fluid at 70% of serum concentration. All thirteen clinical and laboratory P. gingivalis strains tested were able to capture the human complement inhibitor C4b-binding protein (C4BP), which may contribute to their serum resistance. Accordingly, in serum deficient of C4BP, it was found that significantly more terminal complement component C9 was deposited on P. gingivalis. Moreover, using purified proteins and various isogenic mutants, we found that the cysteine protease high molecular weight arginine-gingipain A (HRgpA) is a crucial C4BP ligand on the bacterial surface. Binding of C4BP to P. gingivalis appears to be localized to two binding sites: on the complement control protein 1 domain and complement control protein 6 and 7 domains of the alpha-chains. Furthermore, the bacterial binding of C4BP was found to increase with time of culture and a particularly strong binding was observed for large aggregates of bacteria that formed during culture on solid blood agar medium. Taken together, gingipains appear to be a very significant virulence factor not only destroying complement due to proteolytic degradation as we have shown previously, but was also inhibiting complement activation due to their ability to bind the complement inhibitor C4BP.


Subject(s)
Adhesins, Bacterial/immunology , Bacteroidaceae Infections/immunology , Blood Bactericidal Activity/immunology , Complement C4b-Binding Protein/immunology , Cysteine Endopeptidases/immunology , Porphyromonas gingivalis/immunology , Virulence Factors/immunology , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Bacteroidaceae Infections/enzymology , Bacteroidaceae Infections/genetics , Blood Bactericidal Activity/genetics , Complement Activation/genetics , Complement Activation/immunology , Complement C4b-Binding Protein/genetics , Complement C4b-Binding Protein/metabolism , Complement C9/genetics , Complement C9/immunology , Complement C9/metabolism , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Gingipain Cysteine Endopeptidases , Humans , Ligands , Mutation , Porphyromonas gingivalis/enzymology , Porphyromonas gingivalis/pathogenicity , Protein Binding/genetics , Protein Binding/immunology , Protein Structure, Secondary/genetics , Protein Structure, Tertiary/genetics , Virulence Factors/genetics , Virulence Factors/metabolism
20.
Sheng Wu Gong Cheng Xue Bao ; 36(4): 693-699, 2020 Apr 25.
Article in Zh | MEDLINE | ID: mdl-32347063

ABSTRACT

To study the interaction between C4b-binding protein (C4BP) and Riemerella anatipestifer (RA), we cloned duck C4BPα, conducted prokaryotic expression and prepared the polyclonal antibody by immunizing mice. Then indirect immunofluorescence assay and dot blotting hybridization assay were used to verify the interaction between C4BP and RA. The full length of duck C4BPα nucleotide sequence was 1 230 bp, with the highest similarity to chicken C4BPα (82.1%). Phylogenetic tree analysis showed that duck C4BPα and chicken C4BPα were on the same phylogenetic tree branch and the genetic evolution relationship between them was the closest. C4BPα was efficiently expressed in Escherichia coli BL21 (DE3). The recombinant proteins existed in intracellular soluble form. The titer of polyclonal antibody was more than 1:10 000 and polyclonal antibodies could specifically recognize the recombinant proteins. The results of indirect immunofluorescence assay and dot blot hybridization assay showed that RA could interact with duck C4BP. The results provide a basis to further reveal the pathogenesis of RA.


Subject(s)
Complement C4b-Binding Protein , Ducks , Gene Expression Regulation , Riemerella , Animals , Cloning, Molecular , Complement C4b-Binding Protein/chemistry , Complement C4b-Binding Protein/genetics , Complement C4b-Binding Protein/metabolism , Ducks/classification , Ducks/genetics , Ducks/microbiology , Mice , Phylogeny , Riemerella/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL