Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
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
2.
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
3.
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
4.
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
5.
Front Immunol ; 11: 2122, 2020.
Article in English | MEDLINE | ID: mdl-32983170

ABSTRACT

Moraxella catarrhalis is a human-specific commensal of the respiratory tract and an opportunistic pathogen. It is one of the leading cause of otitis media in children and of acute exacerbations in patients with chronic obstructive pulmonary disease, resulting in significant morbidity and economic burden. Vaccines and new immunotherapeutic strategies to treat this emerging pathogen are needed. Complement is a key component of innate immunity that mediates the detection, response, and subsequent elimination of invading pathogens. Many pathogens including M. catarrhalis have evolved complement evasion mechanisms, which include the binding of human complement inhibitors such as C4b-binding protein (C4BP) and Factor H (FH). Inhibiting C4BP and FH acquisition by M. catarrhalis may provide a novel therapeutic avenue to treat infections. To achieve this, we created two chimeric proteins that combined the Moraxella-binding domains of C4BP and FH fused to human immunoglobulin Fcs: C4BP domains 1 and 2 and FH domains 6 and 7 fused to IgM and IgG Fc, respectively. As expected, FH6-7/IgG displaced FH from the bacterial surface while simultaneously activating complement via Fc-C1q interactions, together increasing pathogen elimination. C4BP1-2/IgM also increased serum killing of the bacteria through enhanced complement deposition, but did not displace C4BP from the surface of M. catarrhalis. These Fc fusion proteins could act as anti-infective immunotherapies. Many microbes bind the complement inhibitors C4BP and FH through the same domains as M. catarrhalis, therefore these Fc fusion proteins may be promising candidates as adjunctive therapy against many different drug-resistant pathogens.


Subject(s)
Complement C4b-Binding Protein/pharmacology , Complement Factor H/pharmacology , Immunoglobulin Fc Fragments/pharmacology , Moraxella catarrhalis/drug effects , Recombinant Fusion Proteins/pharmacology , Animals , Binding, Competitive , Blood Bactericidal Activity , CHO Cells , Complement C3b/analysis , Complement C3d/analysis , Complement C4b-Binding Protein/genetics , Complement C4b-Binding Protein/metabolism , Complement Factor H/genetics , Complement Factor H/metabolism , Cricetinae , Cricetulus , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Immunoglobulin G/pharmacology , Immunoglobulin M/genetics , Immunoglobulin M/metabolism , Immunoglobulin M/pharmacology , Moraxella catarrhalis/metabolism , Protein Binding , Protein Domains , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
6.
Sheng Wu Gong Cheng Xue Bao ; 36(4): 693-699, 2020 Apr 25.
Article in Chinese | 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
7.
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
8.
Front Immunol ; 9: 1945, 2018.
Article in English | MEDLINE | ID: mdl-30210498

ABSTRACT

The complement system is a tightly regulated network of proteins involved in defense against pathogens, inflammatory processes, and coordination of the innate and adaptive immune responses. Dysregulation of the complement cascade is associated with many inflammatory disorders. Thus, inhibition of the complement system has emerged as an option for treatment of a range of different inflammatory diseases. MAP-1 is a pattern recognition molecule (PRM)-associated inhibitor of the lectin pathway of the complement system, whereas C4b-binding protein (C4BP) regulates both the classical and lectin pathways. In this study we generated chimeric proteins consisting of MAP-1 and the first five domains of human C4BP (C4BP1-5) in order to develop a targeted inhibitor acting at different levels of the complement cascade. Two different constructs were designed and expressed in CHO cells where MAP-1 was fused with C4BP1-5 in either the C- or N-terminus. The functionality of the chimeric proteins was assessed using different in vitro complement activation assays. Both chimeric proteins displayed the characteristic Ca2+-dependent dimerization and binding to PRMs of native MAP-1, as well as the co-factor activity of native C4BP. In ELISA-based complement activation assays they could effectively inhibit the lectin and classical pathways. Notably, MAP-1:C4BP1-5 was five times more effective than rMAP-1 and rC4BP1-5 applied at the same time, emphasizing the advantage of a single inhibitor containing both functional domains. The MAP-1/C4BP chimeras exert unique complement inhibitory properties and represent a novel therapeutic approach targeting both upstream and central complement activation.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins , Complement C4b-Binding Protein , Complement Pathway, Mannose-Binding Lectin/immunology , Recombinant Fusion Proteins , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , CHO Cells , Complement C4b-Binding Protein/chemistry , Complement C4b-Binding Protein/genetics , Complement C4b-Binding Protein/immunology , Cricetulus , Enzyme-Linked Immunosorbent Assay , Humans
9.
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
10.
Immunobiology ; 223(1): 125-134, 2018 01.
Article in English | MEDLINE | ID: mdl-29017821

ABSTRACT

The use of C3d, the final degradation product of complement protein C3, as a "natural" adjuvant has been widely examined since the initial documentation of its immunogenicity-enhancing properties as a consequence of binding to complement receptor 2. Subsequently it was demonstrated that these effects are most evident when oligomeric, rather than when monomeric forms of C3d, are linked to various test protein antigens. In this study, we examined the feasibility of enhancing the adjuvant properties of human C3d further by utilizing C4b-binding protein (C4BP) to provide an oligomeric arrayed scaffold fused to the model antigen, tetanus toxin C fragment (TTCF). High molecular weight, C3d-containing oligomeric vaccines were successfully expressed, purified from mammalian cells and used to immunize groups of mice. Surprisingly, anti-TTCF antibody responses measured in these mice were poor. Subsequently we established by in vitro and in vivo analysis that, in the presence of mouse C3, human C3d does not interact with either mouse or even human complement receptor 2. These data confirm the requirement to develop murine versions of C3d based adjuvant compounds to test in mice or that mice would need to be developed that express both human C3 and human CR2 to allow the testing of human C3d based adjuvants in mouse in any capacity.


Subject(s)
B-Lymphocytes/physiology , Complement C3d/immunology , Complement C4b-Binding Protein/genetics , Peptide Fragments/immunology , Tetanus Toxin/immunology , Vaccines, Synthetic/immunology , Adjuvants, Immunologic , Animals , Antibodies/blood , Cell Line , Complement C3d/genetics , Complement C4b-Binding Protein/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Peptide Fragments/genetics , Protein Multimerization/genetics , Receptors, Complement 3d/genetics , Receptors, Complement 3d/metabolism , Tetanus Toxin/genetics , Vaccination , Vaccines, Synthetic/genetics
11.
Mol Med Rep ; 16(2): 1289-1297, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28627632

ABSTRACT

A previous study of the authors using microarray analysis indicated that the expression of complement component 4 binding protein (C4BP)A is upregulated in essential hypertension (EH) patients, but the association between C4BPA variations and EH has not yet been clearly demonstrated. Since the 5' upstream region is known to serve important roles in the gene expression regulation, the present study aimed to identify and analyze the association of single nucleotide polymorphisms (SNPs) in the 5' upstream region between the C4BPA gene with EH in a case­control study among a northeastern Han Chinese population through direct sequencing as well as genotype detection. A total of 822 unrelated participants were included. The higher expression level of C4BPA in the peripheral blood of patients with EH was verified through reverse transcription­quantitative polymerase chain reaction and ELISA. A total of four SNPs, rs73079108, rs74148971, rs77660718 and rs11120211 were identified in the 5' upstream region of C4BPA. Association analysis demonstrated that the genotypic frequencies of rs73079108 were significantly different between EH and the control groups (P=0.011), and A allelic frequency was lower in EH (P<0.001). Logistic regression analysis indicated that the rs73079108 polymorphism was closely associated with EH (AA:GA:GG genetic model: P=0.007, odds ratio (OR)=0.604, 95% confidence interval (CI) [0.418­0.873]; AA+GA:GG genetic model: P=0.005, OR=0.806, 95% CI[0.382­0.841]), and the A allele may be a protective factor. Subgroup analysis by sex and BMI presented concordant conclusions in female and non­obese samples. Further analysis indicated that rs73079108 was associated with systolic blood pressure (P<0.001), diastolic blood pressure (P=0.001) and fast blood glucose (FBG) (P=0.021). In addition, rs73079108 GA and GG carriers reported a significant increase in the level of the protein encoded by C4BPA than those of AA carriers. The rs73079108 polymorphism in the 5' upstream region of C4BPA was associated with EH, and rs73079108­A may be an independent predictor.


Subject(s)
5' Untranslated Regions , Asian People/genetics , Complement C4b-Binding Protein/genetics , Essential Hypertension/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Adult , Aged , Alleles , Case-Control Studies , China , Complement C4b-Binding Protein/metabolism , Essential Hypertension/metabolism , Female , Gene Frequency , Genotype , Humans , Male , Middle Aged , Sequence Analysis, DNA
12.
PLoS One ; 12(1): e0168814, 2017.
Article in English | MEDLINE | ID: mdl-28125581

ABSTRACT

The surface protein SdrE, a microbial surface components recognizing adhesive matrix molecule (MSCRAMM) family protein expressed on the surface of Staphylococcus aureus (S. aureus), can recognize human complement regulator Factor H and C4BP, thus making it a potentially promising vaccine candidate. In this study, SdrE278-591 was found to directly affect S. aureus host cell invasion. Additionally, the crystal structure of SdrE278-591 at a resolution of 1.25 Å was established, with the three-dimensional structure revealing N2-N3 domains which fold in a manner similar to an IgG fold. Furthermore, a putative ligand binding site located at a conserved charged groove formed by the interface between N2 and N3 domains was identified, with ß2 suspected to occupy the ligand recognizing site and undergo a structural rearrangement to allow ligand binding. Overall, these findings have further contributed to the understanding of SdrE as a key factor for S. aureus invasivity and will enable a better understanding of bacterial infection processes.


Subject(s)
Bacterial Proteins/chemistry , Complement C4b-Binding Protein/chemistry , Complement Factor H/chemistry , Mutation , Staphylococcus aureus/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Binding Sites , Cell Line, Tumor , Cloning, Molecular , Complement C4b-Binding Protein/genetics , Complement C4b-Binding Protein/immunology , Complement Factor H/genetics , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , HeLa Cells , Host-Pathogen Interactions , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Ligands , Models, Molecular , Osteoblasts/immunology , Osteoblasts/microbiology , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Folding , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Staphylococcus aureus/pathogenicity
13.
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
14.
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
15.
Immunol Lett ; 169: 82-92, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26658464

ABSTRACT

C4b-binding protein (C4BP) is best known as a potent soluble inhibitor of the classical and lectin pathways of the complement system. This large 500 kDa multimeric plasma glycoprotein is expressed mainly in the liver but also in lung and pancreas. It consists of several identical 75 kDa α-chains and often also one 40 kDa ß-chain, both of which are mainly composed of complement control protein (CCP) domains. Structure-function studies revealed that one crucial binding site responsible for inhibition of complement is located to CCP1-3 of the α-chain. Binding of anticoagulant protein S to the CCP1 of the ß-chain provides C4BP with the ability to strongly bind apoptotic and necrotic cells in order to prevent inflammation arising from activation of complement by these cells. Further, C4BP interacts strongly with various types of amyloid and enhances fibrillation of islet amyloid polypeptide secreted from pancreatic beta cells, which may attenuate pro-inflammatory and cytotoxic effects of this amyloid. Full deficiency of C4BP has not been identified but non-synonymous alterations in its sequence have been found in haemolytic uremic syndrome and recurrent pregnancy loss. Furthermore, C4BP is bound by several bacterial pathogens, notably Streptococcus pyogenes, which due to inhibition of complement and enhancement of bacterial adhesion to endothelial cells provides these bacteria with a survival advantage in the host. Thus, depending on the context, C4BP has a protective or detrimental role in the organism.


Subject(s)
Abortion, Spontaneous/immunology , Atypical Hemolytic Uremic Syndrome/immunology , Complement C4b-Binding Protein/metabolism , Pregnancy Complications, Hematologic/immunology , Abortion, Spontaneous/genetics , Animals , Atypical Hemolytic Uremic Syndrome/genetics , Complement Activation/genetics , Complement C4b-Binding Protein/genetics , Complement C4b-Binding Protein/immunology , Female , Genetic Predisposition to Disease , Homeostasis , Humans , Mutation/genetics , Polymorphism, Genetic , Pregnancy , Pregnancy Complications, Hematologic/genetics
16.
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
17.
Neurosci Lett ; 590: 189-92, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25660618

ABSTRACT

Epidemiological studies have indicated that both maternal bacterial and viral infections during pregnancy increase the risk of schizophrenia among offspring, but to date there is not clear explanation for this increased risk. Previously, the decreased C4b-binding protein (C4BP), a potent circulating soluble inhibitor of the classical and lectin pathways of complement, was reported to be associated with risk of schizophrenia. Here, we analyzed 4 common single nucleotide polymorphisms (SNPs) of C4BPB and 5 SNPs of C4BPA in a group of 556 schizophrenia patients and a matched group of 610 healthy controls to see if the genes C4BPB and C4BPA, which encode C4BP, may confer a susceptibility to schizophrenia. Comparing the genotype and allele frequencies of those SNPs between cases and controls, we found no association between the C4BPB/C4BPA variants and schizophrenia. Our results provided preliminary evidence that C4BPB/C4BPA may not confer susceptibility to schizophrenia among Han Chinese. Further genetic studies from large-scale population are required to obtain more conclusive results.


Subject(s)
Complement C4b-Binding Protein/genetics , Histocompatibility Antigens/genetics , Schizophrenia/genetics , Adult , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Male , Polymorphism, Single Nucleotide
18.
Immunobiology ; 220(4): 467-75, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25468721

ABSTRACT

C4b-binding protein (C4BP) is known as one of the circulating complement regulators that prevents excessive activation of the host-defense complement system. We have reported previously that C4BP is expressed abundantly in the rodent epididymis, one of the male reproductive organs connecting the testis and vas deferens, where immature spermatozoa acquire their motility and fertilizing ability during their transit through the duct. Epididymal C4BP (EpC4BP) is synthesized androgen-dependently by the epithelial cells, secreted into the lumen, and bound to the outer membrane of the passing spermatozoa. In this study, we found that EpC4BP is secreted as a large oligomer, similar to the serum C4BP, but is digested during the epididymal transit and is almost lost from both the luminal fluid and the sperm surface in the vas deferens. Such a processing pattern is not known in serum C4BP, suggesting that EpC4BP and serum C4BP might have different functional mechanisms, and that there is a novel function of EpC4BP in reproduction. In addition, the disappearance of EpC4BP from the sperm surface prior to ejaculation suggests that EpC4BP works only in the epididymis and would not work in the female reproductive tract to protect spermatozoa from complement attack. Next, we generated C4BP-deficient (C4BP-/-) mice to examine the possible role of EpC4BP in reproduction. However, the C4BP-/- mice were fertile and no significant differences were observed between the C4BP-/- and wild-type mouse spermatozoa in terms of morphology, motility, and rate of the spontaneous acrosome reaction. These results suggest that EpC4BP is involved in male reproduction, but not essential for sperm maturation.


Subject(s)
Complement C4b-Binding Protein/metabolism , Epididymis/metabolism , Fertility , Acrosome/metabolism , Animals , Complement C4b-Binding Protein/genetics , Epididymis/ultrastructure , Female , Fertility/genetics , Gene Expression , Gene Order , Gene Targeting , Male , Mice , Mice, Knockout , Models, Animal , Organ Specificity/genetics , Phenotype , Protein Transport , Proteolysis , Sperm Maturation/genetics , Sperm Motility/genetics , Spermatozoa/metabolism , Spermatozoa/ultrastructure
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...