Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 221
Filter
1.
J Biol Chem ; 300(4): 105784, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38401844

ABSTRACT

The introduction of a therapeutic anti-C5 antibody into clinical practice in 2007 inspired a surge into the development of complement-targeted therapies. This has led to the recent approval of a C3 inhibitory peptide, an antibody directed against C1s and a full pipeline of several complement inhibitors in preclinical and clinical development. However, no inhibitor is available that efficiently inhibits all three complement initiation pathways and targets host cell surface markers as well as complement opsonins. To overcome this, we engineered a novel fusion protein combining selected domains of the three natural complement regulatory proteins decay accelerating factor, factor H and complement receptor 1. Such a triple fusion complement inhibitor (TriFu) was recombinantly expressed and purified alongside multiple variants and its building blocks. We analyzed these proteins for ligand binding affinity and decay acceleration activity by surface plasmon resonance. Additionally, we tested complement inhibition in several in vitro/ex vivo assays using standard classical and alternative pathway restricted hemolysis assays next to hemolysis assays with paroxysmal nocturnal hemoglobinuria erythrocytes. A novel in vitro model of the alternative pathway disease C3 glomerulopathy was established to evaluate the potential of the inhibitors to stop C3 deposition on endothelial cells. Next to the novel engineered triple fusion variants which inactivate complement convertases in an enzyme-like fashion, stoichiometric complement inhibitors targeting C3, C5, factor B, and factor D were tested as comparators. The triple fusion approach yielded a potent complement inhibitor that efficiently inhibits all three complement initiation pathways while targeting to surface markers.


Subject(s)
Complement Factor H , Receptors, Complement 3b , Recombinant Fusion Proteins , Humans , Complement Factor H/metabolism , Complement Factor H/genetics , Complement Factor H/chemistry , Complement Factor H/immunology , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Complement Activation/drug effects , CD55 Antigens/genetics , CD55 Antigens/metabolism , Hemolysis/drug effects , Complement Pathway, Alternative/drug effects , Complement Inactivating Agents/pharmacology , Erythrocytes/metabolism
2.
Acta Biomater ; 155: 123-138, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36328123

ABSTRACT

The use of biomaterials in modern medicine has enabled advanced drug delivery strategies and led to reduced morbidity and mortality in a variety of interventions such as transplantation or hemodialysis. However, immune-mediated reactions still present a serious complication of these applications. One of the drivers of such reactions is the complement system, a central part of humoral innate immunity that acts as a first-in-line defense system in its own right but also coordinates other host defense responses. A major regulator of the complement system is the abundant plasma protein factor H (FH), which impairs the amplification of complement responses. Previously, we could show that it is possible to recruit FH to biomedical surfaces using the phage display-derived cyclic peptide 5C6 and, consequently, reduce deposition of C3b, an activation product of the complement system. However, the optimal orientation of 5C6 on surfaces, structural determinants within the peptide for the binding, and the exact binding region on FH remained unknown. Here, we show that the cyclic core and C-terminal region of 5C6 are essential for its interaction with FH and that coating through its N-terminus strongly increases FH recruitment and reduces C3-mediated opsonization in a microparticle-based assay. Furthermore, we could demonstrate that 5C6 selectively binds to FH but not to related proteins. The observation that 5C6 also binds murine FH raises the potential for translational evaluation in animal models. This work provides important insight for the future development of 5C6 as a probe or therapeutic entity to reduce complement activation on biomaterials. STATEMENT OF SIGNIFICANCE: Biomaterials have evolved into core technologies critical to biomedical and drug delivery applications alike, yet their safe and efficient use may be adversely impacted by immune responses to the foreign materials. Taking inspiration from microbial immune evasion strategies, our group developed a peptide-based surface coating that recruits factor H (FH), a host regulator of the complement system, from plasma to the material surface and prevents unwanted activation of this innate immunity pathway. In this study, we identified the molecular determinants that define the interaction between FH and the coated peptide, developed tethering strategies with largely enhanced binding capacity and provided important insight into the target selectivity and species specificity of the FH-binding peptide, thereby paving the way for preclinical development steps.


Subject(s)
Complement C3b , Complement Factor H , Animals , Mice , Complement Factor H/chemistry , Complement Factor H/metabolism , Complement C3b/chemistry , Complement C3b/metabolism , Biocompatible Materials/pharmacology , Protein Binding , Peptides/pharmacology , Peptides/metabolism
3.
J Biol Chem ; 298(6): 101962, 2022 06.
Article in English | MEDLINE | ID: mdl-35452676

ABSTRACT

Atypical hemolytic uremic syndrome (aHUS) is a disease associated with dysregulation of the immune complement system, especially of the alternative pathway (AP). Complement factor H (CFH), consisting of 20 domains called complement control protein (CCP1-20), downregulates the AP as a cofactor for mediating C3 inactivation by complement factor I. However, anomalies related to CFH are known to cause excessive complement activation and cytotoxicity. In aHUS, mutations and the presence of anti-CFH autoantibodies (AAbs) have been reported as plausible causes of CFH dysfunction, and it is known that CFH-related aHUS carries a high probability of end-stage renal disease. Elucidating the detailed functions of CFH at the molecular level will help to understand aHUS pathogenesis. Herein, we used biophysical data to reveal that a heavy-chain antibody fragment, termed VHH4, recognized CFH with high affinity. Hemolytic assays also indicated that VHH4 disrupted the protective function of CFH on sheep erythrocytes. Furthermore, X-ray crystallography revealed that VHH4 recognized the Leu1181-Leu1189CCP20 loop, a known anti-CFH AAbs epitope. We next analyzed the dynamics of the C-terminal region of CFH and showed that the epitopes recognized by anti-CFH AAbs and VHH4 were the most flexible regions in CCP18-20. Finally, we conducted mutation analyses to elucidate the mechanism of VHH4 recognition of CFH and revealed that VHH4 inserts the Trp1183CCP20 residue of CFH into the pocket formed by the complementary determining region 3 loop. These results suggested that anti-CFH AAbs may adopt a similar molecular mechanism to recognize the flexible loop of Leu1181-Leu1189CCP20, leading to aHUS pathogenesis.


Subject(s)
Antibodies, Monoclonal/chemistry , Atypical Hemolytic Uremic Syndrome , Complement Factor H/chemistry , Atypical Hemolytic Uremic Syndrome/metabolism , Autoantibodies/immunology , Complement Activation , Epitopes , Humans , Mutation
4.
Viruses ; 13(7)2021 06 28.
Article in English | MEDLINE | ID: mdl-34203168

ABSTRACT

Robert (Bob) Sim had a profound effect on almost every aspect of my approach to scientific research, acting as a mentor and moral compass through the many different stages of my career [...].


Subject(s)
Immunochemistry/history , Complement Factor H/chemistry , Complement Factor H/history , History, 20th Century , History, 21st Century , United Kingdom
5.
Blood ; 138(22): 2185-2201, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34189567

ABSTRACT

Atypical hemolytic uremic syndrome (aHUS) is a life-threatening thrombotic microangiopathy that can progress, when untreated, to end-stage renal disease. Most frequently, aHUS is caused by complement dysregulation due to pathogenic variants in genes that encode complement components and regulators. Among these genes, the factor H (FH) gene, CFH, presents with the highest frequency (15% to 20%) of variants and is associated with the poorest prognosis. Correct classification of CFH variants as pathogenic or benign is essential to clinical care but remains challenging owing to the dearth of functional studies. As a result, significant numbers of variants are reported as variants of uncertain significance. To address this knowledge gap, we expressed and functionally characterized 105 aHUS-associated FH variants. All FH variants were categorized as pathogenic or benign and, for each, we fully documented the nature of the pathogenicity. Twenty-six previously characterized FH variants were used as controls to validate and confirm the robustness of the functional assays used. Of the remaining 79 uncharacterized variants, only 29 (36.7%) alter FH expression or function in vitro and, therefore, are proposed to be pathogenic. We show that rarity in control databases is not informative for variant classification, and we identify important limitations in applying prediction algorithms to FH variants. Based on structural and functional data, we suggest ways to circumvent these difficulties and, thereby, improve variant classification. Our work highlights the need for functional assays to interpret FH variants accurately if clinical care of patients with aHUS is to be individualized and optimized.


Subject(s)
Atypical Hemolytic Uremic Syndrome/genetics , Complement Factor H/genetics , Atypical Hemolytic Uremic Syndrome/metabolism , Atypical Hemolytic Uremic Syndrome/pathology , Complement Factor H/chemistry , Complement Factor H/metabolism , Gene Expression , Genetic Predisposition to Disease , Genetic Variation , Humans , Models, Molecular , Point Mutation , Polymorphism, Single Nucleotide , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
6.
J Biochem ; 169(5): 543-553, 2021 Jul 03.
Article in English | MEDLINE | ID: mdl-33326036

ABSTRACT

In the complement system, the opsonin C3b binds to the bacterial cell surface and mediates the opsonophagocytosis. However, the cell-wall protein SdrE of Staphylococcus aureus inhibits the C3b activity by recruiting the complement regulatory protein factor H (fH). SdrE binds to fH via its N-terminal N2N3 domain, which are also found in six other staphylococcal cell-wall proteins. In this study, we report that not only the N2N3 domain of SdrE but also those of ClfA, FnbpA and FnbpB can bind to fH. When immobilized on a microplate, the N2N3 domains recruited fH and enhanced the factor I (fI)-mediated cleavage of C3b. When mixed with fH and S. aureus cells, the N2N3 domains inhibited the fH binding to S. aureus cells and reduced the fI-mediated C3b cleavage on the bacterial cell surface. The F(ab)'2 fragments of the rabbit N2N3 antibodies also inhibited the fH binding to the S. aureus cell surface. When added to human blood, the N2N3 antibodies or the N2N3 domain proteins significantly increased the bactericidal activity. Based on these results, we conclude that, in S. aureus, not only SdrE but also ClfA, FnbpA and FnbpB can contribute to the inhibition of C3b-mediated opsonophagocytosis.


Subject(s)
Coagulase/chemistry , Staphylococcus aureus/chemistry , Coagulase/genetics , Coagulase/metabolism , Complement Factor H/chemistry , Complement Factor H/genetics , Complement Factor H/metabolism , Humans , Protein Binding , Protein Domains , Staphylococcus aureus/metabolism
7.
Front Immunol ; 11: 611830, 2020.
Article in English | MEDLINE | ID: mdl-33363547

ABSTRACT

Complement-mediated inflammation or dysregulation in lipid metabolism are associated with the pathogenesis of several diseases. These include age-related macular degeneration (AMD), C3 glomerulonephritis (C3GN), dense deposit disease (DDD), atherosclerosis, and Alzheimer's disease (AD). In all these diseases, formation of characteristic lipid-rich deposits is evident. Here, we will discuss molecular mechanisms whereby dysfunction of complement, and especially of its key regulator factor H, could be involved in lipid accumulation and related inflammation. The genetic associations to factor H polymorphisms, the role of factor H in the resolution of inflammation in lipid-rich deposits, modification of macrophage functions, and complement-mediated clearance of apoptotic and damaged cells indicate that the function of factor H is crucial in limiting inflammation in these diseases.


Subject(s)
Complement Pathway, Alternative , Inflammation/metabolism , Lipid Metabolism , Alzheimer Disease/genetics , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Animals , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Complement Factor H/chemistry , Complement Factor H/genetics , Complement Factor H/metabolism , Glomerulonephritis/genetics , Glomerulonephritis/immunology , Glomerulonephritis/metabolism , Humans , Inflammation/genetics , Inflammation/immunology , Macular Degeneration/genetics , Macular Degeneration/immunology , Macular Degeneration/metabolism , Polymorphism, Genetic , Protein Conformation , Signal Transduction , Structure-Activity Relationship
8.
Mol Vis ; 26: 299-310, 2020.
Article in English | MEDLINE | ID: mdl-32476814

ABSTRACT

Purpose: North Carolina macular dystrophy (NCMD) is an autosomal dominant maculopathy that is considered a non-progressive developmental disorder with variable expressivity. Our study aimed to clinically and genetically characterize macular dystrophy in a family (MOL1154) consisting of six affected subjects with a highly variable maculopathy phenotype in which no correlation between age and severity exists. Methods: Clinical characterization included visual acuity testing and electroretinography. Genetic analysis included Sanger sequencing and whole exome sequencing (WES). Results: WES analysis performed on DNA samples from two individuals revealed a heterozygous deletion of six nucleotides [c.2247_2252del; p.(Leu750_Lys751del)] in the CFH gene. Co-segregation analysis revealed that five of the six NCMD affected subjects carried this deletion, while one individual who had a relatively mild phenotype compatible with dry age-related macular degeneration (AMD) did not carry it. We subsequently analyzed the upstream region of PRDM13 that has previously been reported to be associated with NCMD and identified a unique heterozygous transversion (chr6:100040974A>C) located within the previously described suspected control region in all six affected individuals. This transversion is likely to cause NCMD. Conclusions: NCMD has a wide spectrum of clinical phenotypes that can overlap with AMD, making it challenging to correctly diagnose affected individuals and family members. The DNA sequence variant we found in the CFH gene of some of the affected family members may suggest some role as a modifier gene. However, this variant still does not explain the huge phenotypic variability of NCMD and needs to be studied in other and larger populations.


Subject(s)
Corneal Dystrophies, Hereditary , Histone-Lysine N-Methyltransferase , Transcription Factors , Adult , Aged, 80 and over , Child , Female , Humans , Middle Aged , Color Vision Defects/genetics , Complement Factor H/chemistry , Complement Factor H/genetics , Corneal Dystrophies, Hereditary/blood , Corneal Dystrophies, Hereditary/diagnostic imaging , Corneal Dystrophies, Hereditary/genetics , Corneal Dystrophies, Hereditary/physiopathology , Electrooculography , Electroretinography , Exome Sequencing , Fluorescein Angiography , Genetic Testing , Genotype , Heterozygote , Histone-Lysine N-Methyltransferase/blood , Histone-Lysine N-Methyltransferase/genetics , Jews , Pedigree , Phenotype , Phylogeny , Retinal Dystrophies/genetics , Sequence Deletion , Tomography, Optical Coherence , Transcription Factors/blood , Transcription Factors/genetics
9.
Nat Commun ; 11(1): 1326, 2020 03 12.
Article in English | MEDLINE | ID: mdl-32165615

ABSTRACT

Persistent pathogens have evolved to avoid elimination by the mammalian immune system including mechanisms to evade complement. Infections with African trypanosomes can persist for years and cause human and animal disease throughout sub-Saharan Africa. It is not known how trypanosomes limit the action of the alternative complement pathway. Here we identify an African trypanosome receptor for mammalian factor H, a negative regulator of the alternative pathway. Structural studies show how the receptor binds ligand, leaving inhibitory domains of factor H free to inactivate complement C3b deposited on the trypanosome surface. Receptor expression is highest in developmental stages transmitted to the tsetse fly vector and those exposed to blood meals in the tsetse gut. Receptor gene deletion reduced tsetse infection, identifying this receptor as a virulence factor for transmission. This demonstrates how a pathogen evolved a molecular mechanism to increase transmission to an insect vector by exploitation of a mammalian complement regulator.


Subject(s)
Complement Factor H/metabolism , Trypanosoma/physiology , Tsetse Flies/parasitology , Animals , Antibodies, Monoclonal/metabolism , CHO Cells , Cattle , Cell Membrane/metabolism , Complement C3b/metabolism , Complement Factor H/chemistry , Cricetinae , Cricetulus , Mice, Inbred BALB C , Parasitemia/blood , Protein Binding , Protein Domains , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Receptors, Cell Surface/metabolism , Up-Regulation
10.
J Am Soc Nephrol ; 31(2): 241-256, 2020 02.
Article in English | MEDLINE | ID: mdl-31980588

ABSTRACT

Sequence and copy number variations in the human CFHR-Factor H gene cluster comprising the complement genes CFHR1, CFHR2, CFHR3, CFHR4, CFHR5, and Factor H are linked to the human kidney diseases atypical hemolytic uremic syndrome (aHUS) and C3 glomerulopathy. Distinct genetic and chromosomal alterations, deletions, or duplications generate hybrid or mutant CFHR genes, as well as hybrid CFHR-Factor H genes, and alter the FHR and Factor H plasma repertoire. A clear association between the genetic modifications and the pathologic outcome is emerging: CFHR1, CFHR3, and Factor H gene alterations combined with intact CFHR2, CFHR4, and CFHR5 genes are reported in atypical hemolytic uremic syndrome. But alterations in each of the five CFHR genes in the context of an intact Factor H gene are described in C3 glomerulopathy. These genetic modifications influence complement function and the interplay of the five FHR proteins with each other and with Factor H. Understanding how mutant or hybrid FHR proteins, Factor H::FHR hybrid proteins, and altered Factor H, FHR plasma profiles cause pathology is of high interest for diagnosis and therapy.


Subject(s)
Atypical Hemolytic Uremic Syndrome/genetics , Complement C3/analysis , Glomerulonephritis, Membranoproliferative/genetics , Atypical Hemolytic Uremic Syndrome/etiology , Complement Factor H/chemistry , Complement Factor H/genetics , Complement Factor H/physiology , DNA Copy Number Variations , Genetic Predisposition to Disease , Genetic Variation , Glomerulonephritis, Membranoproliferative/etiology , Humans , Kidney/pathology , Multigene Family
11.
Front Immunol ; 11: 602284, 2020.
Article in English | MEDLINE | ID: mdl-33519811

ABSTRACT

Membranoproliferative glomerulonephritis (MPGN), C3 glomerulopathy (C3G), atypical haemolytic uraemic syndrome (aHUS) and age-related macular degeneration (AMD) have all been strongly linked with dysfunction of the alternative pathway (AP) of complement. A significant proportion of individuals with MPGN, C3G, aHUS and AMD carry rare genetic variants in the CFH gene that cause functional or quantitative deficiencies in the factor H (FH) protein, an important regulator of the AP. In silico analysis of the deleteriousness of rare genetic variants in CFH is not reliable and careful biochemical assessment remains the gold standard. Six N-terminal variants of uncertain significance in CFH were identified in patients with these diseases of the AP and selected for analysis. The variants were produced in Pichia Pastoris in the setting of FH CCPs 1-4, purified by nickel affinity chromatography and size exclusion and characterized by surface plasmon resonance and haemolytic assays as well as by cofactor assays in the fluid phase. A single variant, Q81P demonstrated a profound loss of binding to C3b with consequent loss of cofactor and decay accelerating activity. A further 2 variants, G69E and D130N, demonstrated only subtle defects which could conceivably over time lead to disease progression of more chronic AP diseases such as C3G and AMD. In the variants S159N, A161S, and M162V any functional defect was below the capacity of the experimental assays to reliably detect. This study further underlines the importance of careful biochemical assessment when assigning functional consequences to rare genetic variants that may alter clinical decisions for patients.


Subject(s)
Atypical Hemolytic Uremic Syndrome/genetics , Genetic Variation , Glomerulonephritis, Membranoproliferative/genetics , Macular Degeneration/genetics , Complement Factor H/chemistry , Complement Factor H/genetics , Humans
12.
Front Immunol ; 11: 601895, 2020.
Article in English | MEDLINE | ID: mdl-33552059

ABSTRACT

Complement Factor H (CFH), with 20 short complement regulator (SCR) domains, regulates the alternative pathway of complement in part through the interaction of its C-terminal SCR-19 and SCR-20 domains with host cell-bound C3b and anionic oligosaccharides. In solution, CFH forms small amounts of oligomers, with one of its self-association sites being in the SCR-16/20 domains. In order to correlate CFH function with dimer formation and the occurrence of rare disease-associated variants in SCR-16/20, we identified the dimerization site in SCR-16/20. For this, we expressed, in Pichia pastoris, the five domains in SCR-16/20 and six fragments of this with one-three domains (SCR-19/20, SCR-18/20, SCR-17/18, SCR-16/18, SCR-17 and SCR-18). Size-exclusion chromatography suggested that SCR dimer formation occurred in several fragments. Dimer formation was clarified using analytical ultracentrifugation, where quantitative c(s) size distribution analyses showed that SCR-19/20 was monomeric, SCR-18/20 was slightly dimeric, SCR-16/20, SCR-16/18 and SCR-18 showed more dimer formation, and SCR-17 and SCR-17/18 were primarily dimeric with dissociation constants of ~5 µM. The combination of these results located the SCR-16/20 dimerization site at SCR-17 and SCR-18. X-ray solution scattering experiments and molecular modelling fits confirmed the dimer site to be at SCR-17/18, this dimer being a side-by-side association of the two domains. We propose that the self-association of CFH at SCR-17/18 enables higher concentrations of CFH to be achieved when SCR-19/20 are bound to host cell surfaces in order to protect these better during inflammation. Dimer formation at SCR-17/18 clarified the association of genetic variants throughout SCR-16/20 with renal disease.


Subject(s)
Protein Multimerization , Complement Factor H/chemistry , Complement Factor H/genetics , Humans , Protein Domains
13.
Semin Immunol ; 45: 101341, 2019 10.
Article in English | MEDLINE | ID: mdl-31757608

ABSTRACT

The complement system, while being an essential and very efficient effector component of innate immunity, may cause damage to the host and result in various inflammatory, autoimmune and infectious diseases or cancer, when it is improperly activated or regulated. Factor H is a serum glycoprotein and the main regulator of the activity of the alternative complement pathway. Factor H, together with its splice variant factor H-like protein 1 (FHL-1), inhibits complement activation at the level of the central complement component C3 and beyond. In humans, there are also five factor H-related (FHR) proteins, whose function is poorly characterized. While data indicate complement inhibiting activity for some of the FHRs, there is increasing evidence that FHRs have an opposite role compared with factor H and FHL-1, namely, they enhance complement activation directly and also by competing with the regulators FH and FHL-1. This review summarizes the current stand and recent data on the roles of factor H family proteins in health and disease, with focus on the function of FHR proteins.


Subject(s)
Blood Proteins/metabolism , Carrier Proteins/metabolism , Complement Activation/immunology , Complement Factor H/immunology , Complement Factor H/metabolism , Animals , Blood Proteins/chemistry , Blood Proteins/genetics , Carrier Proteins/chemistry , Carrier Proteins/genetics , Complement Factor H/chemistry , Complement System Proteins/immunology , Complement System Proteins/metabolism , Disease Susceptibility , Humans , Immunomodulation , Ligands , Multigene Family , Protein Binding , Structure-Activity Relationship
14.
J Biol Chem ; 294(52): 20148-20163, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31719147

ABSTRACT

Activation and suppression of the complement system compete on every serum-exposed surface, host or foreign. Potentially harmful outcomes of this competition depend on surface molecules through mechanisms that remain incompletely understood. Combining surface plasmon resonance (SPR) with atomic force microscopy (AFM), here we studied two complement system proteins at the single-molecule level: C3b, the proteolytically activated form of C3, and factor H (FH), the surface-sensing C3b-binding complement regulator. We used SPR to monitor complement initiation occurring through a positive-feedback loop wherein surface-deposited C3b participates in convertases that cleave C3, thereby depositing more C3b. Over multiple cycles of flowing factor B, factor D, and C3 over the SPR chip, we amplified C3b from ∼20 to ∼220 molecules·µm-2 AFM revealed C3b clusters of up to 20 molecules and solitary C3b molecules deposited up to 200 nm away from the clusters. A force of 0.17 ± 0.02 nanonewtons was needed to pull a single FH molecule, anchored to the AFM probe, from its complex with surface-attached C3b. The extent to which FH molecules stretched before detachment varied widely among complexes. Performing force-distance measurements with FH(D1119G), a variant lacking one of the C3b-binding sites and causing atypical hemolytic uremic syndrome, we found that it detached more uniformly and easily. In further SPR experiments, KD values between FH and C3b on a custom-made chip surface were 5-fold tighter than on commercial chips and similar to those on erythrocytes. These results suggest that the chemistry at the surface on which FH acts drives conformational adjustments that are functionally critical.


Subject(s)
Complement C3b/metabolism , Complement Factor H/metabolism , Microscopy, Atomic Force , Surface Plasmon Resonance , Complement Activation , Complement C3b/chemistry , Complement C3d/chemistry , Complement C3d/metabolism , Complement Factor H/chemistry , Humans , Immobilized Proteins/chemistry , Immobilized Proteins/metabolism , Kinetics , Protein Binding
15.
Front Immunol ; 10: 1230, 2019.
Article in English | MEDLINE | ID: mdl-31214187

ABSTRACT

Streptococcus pyogenes infects over 700 million people worldwide annually. Immune evasion strategies employed by the bacteria include binding of the complement inhibitors, C4b-binding protein (C4BP) and Factor H in a human-specific manner. We recently showed that human IgG increased C4BP binding to the bacterial surface, which promoted streptococcal immune evasion and increased mortality in mice. We sought to identify how IgG promotes C4BP binding to Protein H, a member of the M protein family. Dimerization of Protein H is pivotal for enhanced binding to human C4BP. First, we illustrated that Protein H, IgG, and C4BP formed a tripartite complex. Second, surface plasmon resonance revealed that Protein H binds IgG solely through Fc, but not Fab domains, and with high affinity (IgG-Protein H: KD = 0.4 nM; IgG-Fc-Protein H: KD ≤ 1.6 nM). Each IgG binds two Protein H molecules, while up to six molecules of Protein H bind one C4BP molecule. Third, interrupting Protein H dimerization either by raising temperature to 41°C or with a synthetic peptide prevented IgG-Protein H interactions. IgG-Fc fragments or monoclonal human IgG permitted maximal C4BP binding when used at concentrations from 0.1 to 10 mg/ml. In contrast, pooled human IgG enhanced C4BP binding at concentrations up to 1 mg/ml; decreased C4BP binding at 10 mg/ml occurred probably because of Fab-streptococcal interactions at these high IgG concentrations. Taken together, our data show how S. pyogenes exploits human IgG to evade complement and enhance its virulence. Elucidation of this mechanism could aid design of new therapeutics against S. pyogenes.


Subject(s)
Complement C4b-Binding Protein/immunology , Host-Pathogen Interactions/immunology , Immunoglobulin G/immunology , Immunomodulation , Streptococcal Infections/immunology , Streptococcus pyogenes/immunology , Complement C4b-Binding Protein/metabolism , Complement Factor H/chemistry , Complement Factor H/immunology , Complement Factor H/metabolism , Disease Susceptibility/immunology , Disease Susceptibility/metabolism , Humans , Kinetics , Ligands , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Protein Binding , Protein Multimerization , Streptococcal Infections/metabolism , Streptococcal Infections/microbiology
16.
Int J Mol Sci ; 20(10)2019 May 25.
Article in English | MEDLINE | ID: mdl-31130605

ABSTRACT

An understanding of the interaction between the antibody and its targeted antigen and knowing of the epitopes are critical for the development of monoclonal antibody drugs. Complement factor H (CFH) is implied to play a role in tumor growth and metastasis. An autoantibody to CHF is associated with anti-tumor cell activity. The interaction of a human monoclonal antibody Ab42 that was isolated from a cancer patient with CFH polypeptide (pCFH) antigen was analyzed by molecular docking, molecular dynamics (MD) simulation, free energy calculation, and computational alanine scanning (CAS). Experimental alanine scanning (EAS) was then carried out to verify the results of the theoretical calculation. Our results demonstrated that the Ab42 antibody interacts with pCFH by hydrogen bonds through the Tyr315, Ser100, Gly33, and Tyr53 residues on the complementarity-determining regions (CDRs), respectively, with the amino acid residues of Pro441, Ile442, Asp443, Asn444, Ile447, and Thr448 on the pCFH antigen. In conclusion, this study has explored the mechanism of interaction between Ab42 antibody and its targeted antigen by both theoretical and experimental analysis. Our results have important theoretical significance for the design and development of relevant antibody drugs.


Subject(s)
Antibodies, Monoclonal/immunology , Peptides/immunology , Antibodies, Monoclonal/chemistry , Antigen-Antibody Reactions , Autoantibodies/chemistry , Autoantibodies/immunology , Complement Factor H/chemistry , Complement Factor H/immunology , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/immunology , Epitopes/chemistry , Epitopes/immunology , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Neoplasms/immunology , Peptides/chemistry , Protein Conformation
17.
J Thromb Thrombolysis ; 48(1): 95-102, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30915671

ABSTRACT

Coagulation factor XIII (FXIII) covalently crosslinks pre-formed fibrin clots preventing their premature fibrinolysis. In plasma, FXIII circulates as a zymogenic heterotetramer composed of catalytic FXIII-A subunits, and carrier/regulatory FXIII-B subunits. FXIII-A is a well characterized component of this complex, and has been associated with several pleiotropic roles outside coagulation as well. In comparison only protective/regulatory roles towards the FXIII-A subunit have been identified for FXIII-B. Strong homology between FXIII-B and complement regulator Complement factor H suggests a putative role of FXIII-B in complement activation. In the current study we have analyzed the similarities and yet functional divergence of these two proteins using in silico sequence alignment and structural analysis. We have evaluated complement activation post reconstitution of FXIII components into FXIII deficient and CFH deficient plasma. We have also transiently expressed FXIII-B in SH-SY5Y cell lines and evaluated its effect on the endogenous complement activation. Our investigations show no effect of FXIII-B subunit on the rate of complement activation. Therefore we conclude that at a physiological level, FXIII-B subunit plays no role in the complement system, although a vestigial function in altered pathological states might still exist.


Subject(s)
Complement Activation , Factor XIII/chemistry , Blood Specimen Collection , Cell Line , Complement Factor H/chemistry , Complement Factor H/physiology , Computer Simulation , Factor XIII/physiology , Humans , Molecular Structure , Protein Domains , Sequence Alignment , Sequence Homology, Amino Acid , Structural Homology, Protein , Transfection
18.
Biophys J ; 116(2): 215-226, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30616835

ABSTRACT

A single nucleotide polymorphism, tyrosine at position 402 to histidine (Y402H), within the gene encoding complement factor H (FH) predisposes individuals to acquiring age-related macular degeneration (AMD) after aging. This polymorphism occurs in short consensus repeat (SCR) 7 of FH and results in decreased binding affinity of SCR6-8 for heparin. As FH is responsible for regulating the complement system, decreased affinity for heparin results in decreased regulation on surfaces of self. To understand the involvement of the Y402H polymorphism in AMD, we leverage methods from bioinformatics and computational biophysics to quantify structural and dynamical differences between SCR7 isoforms that contribute to decreased pattern recognition in SCR7H402. Our data from molecular and Brownian dynamics simulations suggest a revised mechanism for decreased heparin binding. In this model, transient contacts not observed in structures for SCR7 are predicted to occur in molecular dynamics simulations between coevolved residues Y402 and I412, stabilizing SCR7Y402 in a conformation that promotes association with heparin. H402 in the risk isoform is less likely to form a contact with I412 and samples a larger conformational space than Y402. We observe energy minima for sidechains of Y402 and R404 from SCR7Y402 that are predicted to associate with heparin at a rate constant faster than energy minima for sidechains of H402 and R404 from SCR7H402. As both carbohydrate density and degree of sulfation decrease with age in Bruch's membrane of the macula, the decreased heparin recognition of SCR7H402 may contribute to the pathogenesis of AMD.


Subject(s)
Complement Factor H/chemistry , Macular Degeneration/genetics , Molecular Docking Simulation , Molecular Dynamics Simulation , Mutation, Missense , Binding Sites , Complement Factor H/genetics , Complement Factor H/metabolism , Heparin/chemistry , Humans , Protein Binding
19.
J Phys Chem B ; 122(47): 10653-10658, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30351116

ABSTRACT

Activation of proteins often involves conformational transitions, and these switches are often difficult to characterize in multidomain proteins. Full-length factor H (FH), consisting of 20 small consensus repeat domains (150 kD), is a complement control protein that regulates the activity of the alternative complement pathway. Different preparations of FH can also reduce the disulfide bonds linking large Von Willebrand factor (VWF) multimers into smaller, less adhesive forms. In contrast, commercially available purified FH (pFH) has little or no VWF reductase activity unless the pFH is chemically modified by either ethylenediaminetetraacetic acid (EDTA) or urea. We used atomic force microscopy single molecule force measurements to investigate different forms of FH, including recombinant FH and pFH, in the presence or absence of EDTA and urea, and to correlate the conformational changes to its activities. We found that the FH conformation depends on the method used for sample preparation, which affects the VWF reductase activity of FH.


Subject(s)
Oxidoreductases/chemistry , von Willebrand Factor/chemistry , Catalysis , Complement Factor H/chemistry , Detergents/chemistry , HEK293 Cells , Humans , Microscopy, Atomic Force/methods , Octoxynol/chemistry , Organophosphates/chemistry , Oxidation-Reduction , Protein Conformation , Protein Domains , Recombinant Proteins/chemistry , Thermodynamics
20.
J Biol Chem ; 293(44): 17166-17187, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30217822

ABSTRACT

Factor H (FH) is the major regulator of C3b in the alternative pathway of the complement system in immunity. FH comprises 20 short complement regulator (SCR) domains, including eight glycans, and its Y402H polymorphism predisposes those who carry it to age-related macular degeneration. To better understand FH complement binding and self-association, we have studied the solution structures of both the His-402 and Tyr-402 FH allotypes. Analytical ultracentrifugation revealed that up to 12% of both FH allotypes self-associate, and this was confirmed by small-angle X-ray scattering (SAXS), MS, and surface plasmon resonance analyses. SAXS showed that monomeric FH has a radius of gyration (Rg ) of 7.2-7.8 nm and a length of 25 nm. Starting from known structures for the SCR domains and glycans, the SAXS data were fitted using Monte Carlo methods to determine atomistic structures of monomeric FH. The analysis of 29,715 physically realistic but randomized FH conformations resulted in 100 similar best-fit FH structures for each allotype. Two distinct molecular structures resulted that showed either an extended N-terminal domain arrangement with a folded-back C terminus or an extended C terminus and a folded-back N terminus. These two structures are the most accurate to date for glycosylated full-length FH. To clarify FH functional roles in host protection, crystal structures for the FH complexes with C3b and C3dg revealed that the extended N-terminal conformation accounted for C3b fluid-phase regulation, the extended C-terminal conformation accounted for C3d binding, and both conformations accounted for bivalent FH binding to glycosaminoglycans on the target cell surface.


Subject(s)
Complement C3b , Complement Factor H , Peptide Fragments , Complement C3b/chemistry , Complement C3b/genetics , Complement C3b/metabolism , Complement Factor H/chemistry , Complement Factor H/genetics , Complement Factor H/metabolism , Crystallography, X-Ray , Humans , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding , Protein Conformation , Protein Domains , Surface Plasmon Resonance , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL
...