Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Drugs ; 81(12): 1423-1430, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34342834

ABSTRACT

Pegcetacoplan (Empaveli™) is a PEGylated pentadecapeptide developed by Apellis Pharmaceuticals for the treatment of complement-mediated diseases. It binds to complement component 3 (C3) and its activation fragment C3b, controlling the cleavage of C3 and the generation of the downstream effectors of complement activation and thus both C3b-mediated extravascular haemolysis and terminal complement-mediated intravascular haemolysis. Pegcetacoplan is the first C3-targeted paroxysmal nocturnal haemoglobinuria (PNH) therapy to be approved (in May 2021) in the USA, where it is indicated for the treatment of adults with PNH, including those switching from C5 inhibitor therapy with eculizumab and ravulizumab. A regulatory assessment of pegcetacoplan for the treatment of PNH is currently underway in the EU and Australia. Pegcetacoplan is also being investigated as a therapeutic option in other complement-mediated diseases, including age-related macular degeneration, C3 glomerulopathy and autoimmune haemolytic anaemia. The recommended dosage regimen of pegcetacoplan is 1080 mg twice weekly, administered as a subcutaneous infusion via an infusion pump with a ≥ 20 mL reservoir. This article summarizes the milestones in the development of pegcetacoplan leading to this first approval for the treatment of adults with PNH.


Subject(s)
Complement Inactivating Agents/therapeutic use , Hemoglobinuria, Paroxysmal/drug therapy , Peptides, Cyclic/therapeutic use , Anemia, Hemolytic, Autoimmune/drug therapy , Complement C3/antagonists & inhibitors , Complement C3b/antagonists & inhibitors , Complement Inactivating Agents/pharmacology , Geographic Atrophy/drug therapy , Glomerulonephritis/drug therapy , Humans , Peptides, Cyclic/pharmacology
2.
Nature ; 588(7838): 459-465, 2020 12.
Article in English | MEDLINE | ID: mdl-32866962

ABSTRACT

Aberrant aggregation of the RNA-binding protein TDP-43 in neurons is a hallmark of frontotemporal lobar degeneration caused by haploinsufficiency in the gene encoding progranulin1,2. However, the mechanism leading to TDP-43 proteinopathy remains unclear. Here we use single-nucleus RNA sequencing to show that progranulin deficiency promotes microglial transition from a homeostatic to a disease-specific state that causes endolysosomal dysfunction and neurodegeneration in mice. These defects persist even when Grn-/- microglia are cultured ex vivo. In addition, single-nucleus RNA sequencing reveals selective loss of excitatory neurons at disease end-stage, which is characterized by prominent nuclear and cytoplasmic TDP-43 granules and nuclear pore defects. Remarkably, conditioned media from Grn-/- microglia are sufficient to promote TDP-43 granule formation, nuclear pore defects and cell death in excitatory neurons via the complement activation pathway. Consistent with these results, deletion of the genes encoding C1qa and C3 mitigates microglial toxicity and rescues TDP-43 proteinopathy and neurodegeneration. These results uncover previously unappreciated contributions of chronic microglial toxicity to TDP-43 proteinopathy during neurodegeneration.


Subject(s)
Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Neurons/pathology , Progranulins/deficiency , TDP-43 Proteinopathies/metabolism , TDP-43 Proteinopathies/pathology , Aging/genetics , Aging/pathology , Animals , Cell Nucleus/genetics , Cell Nucleus/pathology , Complement Activation/drug effects , Complement Activation/immunology , Complement C1q/antagonists & inhibitors , Complement C1q/immunology , Complement C3b/antagonists & inhibitors , Complement C3b/immunology , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , DNA-Binding Proteins/metabolism , Disease Models, Animal , Female , Male , Mice , Nuclear Pore/metabolism , Nuclear Pore/pathology , Progranulins/genetics , RNA-Seq , Single-Cell Analysis , TDP-43 Proteinopathies/drug therapy , TDP-43 Proteinopathies/genetics , Thalamus/metabolism , Thalamus/pathology , Transcriptome
3.
EMBO Mol Med ; 12(8): e12642, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32559343

ABSTRACT

A novel coronavirus, SARS-CoV-2, has recently emerged in China and spread internationally, posing a health emergency to the global community. COVID-19 caused by SARS-CoV-2 is associated with an acute respiratory illness that varies from mild to the life-threatening acute respiratory distress syndrome (ARDS). The complement system is part of the innate immune arsenal against pathogens, in which many viruses can evade or employ to mediate cell entry. The immunopathology and acute lung injury orchestrated through the influx of pro-inflammatory macrophages and neutrophils can be directly activated by complement components to prime an overzealous cytokine storm. The manifestations of severe COVID-19 such as the ARDS, sepsis and multiorgan failure have an established relationship with activation of the complement cascade. We have collected evidence from all the current studies we are aware of on SARS-CoV-2 immunopathogenesis and the preceding literature on SARS-CoV-1 and MERS-CoV infection linking severe COVID-19 disease directly with dysfunction of the complement pathways. This information lends support for a therapeutic anti-inflammatory strategy against complement, where a number of clinically ready potential therapeutic agents are available.


Subject(s)
Betacoronavirus , Complement Activation/drug effects , Complement Inactivating Agents/therapeutic use , Coronavirus Infections/drug therapy , Pandemics , Pneumonia, Viral/drug therapy , Adult , Alveolar Epithelial Cells/immunology , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/virology , Angiotensin-Converting Enzyme 2 , Animals , Betacoronavirus/physiology , COVID-19 , Child , Complement C3b/antagonists & inhibitors , Complement C3b/physiology , Complement Inactivating Agents/pharmacology , Coronavirus Infections/immunology , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/immunology , Glycosylation , Humans , Immunity, Innate , Ligands , Mice , Models, Animal , Models, Molecular , Pattern Recognition, Automated , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/immunology , Protein Conformation , Protein Processing, Post-Translational , Receptors, Virus/metabolism , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/immunology , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , COVID-19 Drug Treatment
4.
FASEB J ; 33(3): 4448-4457, 2019 03.
Article in English | MEDLINE | ID: mdl-30566365

ABSTRACT

Group B Streptococcus (GBS) colonizes the human lower intestinal and genital tracts and constitutes a major threat to neonates from pregnant carrier mothers and to adults with underlying morbidity. The pathogen expresses cell-surface virulence factors that enable cell adhesion and penetration and that counteract innate and adaptive immune responses. Among these, the complement interfering protein (CIP) was recently described for its capacity to interact with the human C4b ligand and to interfere with the classical- and lectin-complement pathways. In the present study, we provide evidence that CIP can also interact with C3, C3b, and C3d. Immunoassay-based competition experiments showed that binding of CIP to C3d interferes with the interaction between C3d and the complement receptor 2/cluster of differentiation 21 (CR2/CD21) receptor on B cells. By B-cell intracellular signaling assays, CIP was confirmed to down-regulate CR2/CD21-dependent B-cell activation. The CIP domain involved in C3d binding was mapped via hydrogen deuterium exchange-mass spectrometry. The data obtained reveal a new role for this GBS polypeptide at the interface between the innate and adaptive immune responses, adding a new member to the growing list of virulence factors secreted by gram-positive pathogens that incorporate multiple immunomodulatory functions.-Giussani, S., Pietrocola, G., Donnarumma, D., Norais, N., Speziale, P., Fabbrini, M., Margarit, I. The Streptococcus agalactiae complement interfering protein combines multiple complement-inhibitory mechanisms by interacting with both C4 and C3 ligands.


Subject(s)
Bacterial Proteins/metabolism , Complement C3d/antagonists & inhibitors , Complement C4/antagonists & inhibitors , Streptococcus agalactiae/pathogenicity , Virulence Factors/metabolism , Amino Acid Sequence , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Bacterial Proteins/pharmacology , Binding Sites , Calcium Signaling , Cell Line, Tumor , Complement C3b/antagonists & inhibitors , Complement C3b/metabolism , Complement C3d/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Lymphocyte Activation/drug effects , Mass Spectrometry , Protein Binding , Protein Interaction Mapping , Receptors, Complement 3d/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Streptococcus agalactiae/immunology , Streptococcus agalactiae/metabolism , Surface Plasmon Resonance , Virulence , Virulence Factors/pharmacology
5.
J Biol Chem ; 293(12): 4468-4477, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29414776

ABSTRACT

Staphylococcus aureus is a versatile pathogen capable of causing a broad range of diseases in many different hosts. S. aureus can adapt to its host through modification of its genome (e.g. by acquisition and exchange of mobile genetic elements that encode host-specific virulence factors). Recently, the prophage φSaeq1 was discovered in S. aureus strains from six different clonal lineages almost exclusively isolated from equids. Within this phage, we discovered a novel variant of staphylococcal complement inhibitor (SCIN), a secreted protein that interferes with activation of the human complement system, an important line of host defense. We here show that this equine variant of SCIN, eqSCIN, is a potent blocker of equine complement system activation and subsequent phagocytosis of bacteria by phagocytes. Mechanistic studies indicate that eqSCIN blocks equine complement activation by specific inhibition of the C3 convertase enzyme (C3bBb). Whereas SCIN-A from human S. aureus isolates exclusively inhibits human complement, eqSCIN represents the first animal-adapted SCIN variant that functions in a broader range of hosts (horses, humans, and pigs). Binding analyses suggest that the human-specific activity of SCIN-A is related to amino acid differences on both sides of the SCIN-C3b interface. These data suggest that modification of this phage-encoded complement inhibitor plays a role in the host adaptation of S. aureus and are important to understand how this pathogen transfers between different hosts.


Subject(s)
Complement C3-C5 Convertases/metabolism , Complement C3b/antagonists & inhibitors , Complement Inactivator Proteins/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/metabolism , Virulence Factors/metabolism , Animals , Complement C3b/metabolism , Complement Inactivator Proteins/chemistry , Hemolysis , Horses , Host Specificity , Humans , Phagocytosis , Protein Binding , Staphylococcal Infections/metabolism , Staphylococcus aureus/isolation & purification , Swine , Virulence Factors/chemistry
6.
J Am Soc Nephrol ; 29(4): 1141-1153, 2018 04.
Article in English | MEDLINE | ID: mdl-29335241

ABSTRACT

The complement system is essential for host defense, but uncontrolled complement system activation leads to severe, mostly renal pathologies, such as atypical hemolytic uremic syndrome or C3 glomerulopathy. Here, we investigated a novel combinational approach to modulate complement activation by targeting C3 and the terminal pathway simultaneously. The synthetic fusion protein MFHR1 links the regulatory domains of complement factor H (FH) with the C5 convertase/C5b-9 inhibitory fragment of the FH-related protein 1. In vitro, MFHR1 showed cofactor and decay acceleration activity and inhibited C5 convertase activation and C5b-9 assembly, which prevented C3b deposition and reduced C3a/C5a and C5b-9 generation. Furthermore, this fusion protein showed the ability to escape deregulation by FH-related proteins and form multimeric complexes with increased inhibitory activity. In addition to substantially inhibiting alternative and classic pathway activation, MFHR1 blocked hemolysis mediated by serum from a patient with aHUS expressing truncated FH. In FH-/- mice, MFHR1 administration augmented serum C3 levels, reduced abnormal glomerular C3 deposition, and ameliorated C3 glomerulopathy. Taking the unique design of MFHR1 into account, we suggest that the combination of proximal and terminal cascade inhibition together with the ability to form multimeric complexes explain the strong inhibitory capacity of MFHR1, which offers a novel basis for complement therapeutics.


Subject(s)
Atypical Hemolytic Uremic Syndrome/blood , Blood Proteins/deficiency , Complement C3b Inactivator Proteins/genetics , Complement Inactivating Agents/pharmacology , Molecular Targeted Therapy , Recombinant Fusion Proteins/pharmacology , Animals , Atypical Hemolytic Uremic Syndrome/genetics , Atypical Hemolytic Uremic Syndrome/immunology , Complement C3/metabolism , Complement C3-C5 Convertases/antagonists & inhibitors , Complement C3-C5 Convertases/metabolism , Complement C3b/antagonists & inhibitors , Complement C3b Inactivator Proteins/deficiency , Complement C5/metabolism , Complement Factor H/genetics , Complement Inactivating Agents/isolation & purification , Complement Inactivating Agents/therapeutic use , Complement Membrane Attack Complex/biosynthesis , Complement Pathway, Alternative , Drug Design , Drug Evaluation, Preclinical , Kidney Glomerulus/chemistry , Kidney Glomerulus/pathology , Mice , Mice, Knockout , Protein Domains , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/therapeutic use
7.
Mol Immunol ; 93: 266-277, 2018 01.
Article in English | MEDLINE | ID: mdl-28860090

ABSTRACT

Candida albicans the most frequently isolated clinical fungal pathogen can cause local as well as systemic and life-threatening infections particularly in immune-compromised individuals. A better and more detailed understanding how C. albicans evades human immune attack is therefore needed for identifying fungal immune-evasive proteins and develop new therapies. Here, we identified Pra1, the pH-regulated C. albicans antigen as a hierarchical complement inhibitor that targets C3, the central human complement component. Pra1 cleaved C3 at a unique site and further inhibited effector function of the activation fragments. The newly formed C3a-like peptide lacked the C-terminal arginine residue needed for C3a-receptor binding and activation. Moreover, Pra1 also blocked C3a-like antifungal activity as shown in survival assays, and the C3b-like molecule formed by Pra1 was degraded by the host protease Factor I. Pra1 also bound to C3a and C3b generated by human convertases and blocked their effector functions, like C3a antifungal activity shown by fungal survival, blocked C3a binding to human C3a receptor-expressing HEK cells, activation of Fura2-AM loaded cells, intracellular Ca2+ signaling, IL-8 release, C3b deposition, as well as opsonophagocytosis and killing by human neutrophils. Thus, upon infection C. albicans uses Pra1 to destroy C3 and to disrupt host complement attack. In conclusion, candida Pra1 represents the first fungal C3-cleaving protease identified and functions as a fungal master regulator of innate immunity and as a central fungal immune-escape protein.


Subject(s)
Candida albicans/enzymology , Complement C3/antagonists & inhibitors , Fungal Proteins/physiology , Amino Acid Sequence , Binding, Competitive , Calcium Signaling/drug effects , Candida albicans/drug effects , Candida albicans/immunology , Cell Line , Complement C3/immunology , Complement C3/metabolism , Complement C3/pharmacology , Complement C3a/antagonists & inhibitors , Complement C3a/pharmacology , Complement C3b/antagonists & inhibitors , Complement C3b/pharmacology , Fungal Proteins/antagonists & inhibitors , Fungal Proteins/pharmacology , HEK293 Cells , Humans , Interleukin-8/metabolism , Neutrophils/drug effects , Neutrophils/physiology , Opsonin Proteins/immunology , Peptide Fragments/metabolism , Phagocytosis/drug effects , Protease Inhibitors/pharmacology , Proteolysis , Receptors, Complement/antagonists & inhibitors , Receptors, Complement/metabolism , Virulence/immunology
8.
mBio ; 6(5): e01339-15, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26419879

ABSTRACT

UNLABELLED: The capsule from Bacteroides, a common gut symbiont, has long been a model system for studying the molecular mechanisms of host-symbiont interactions. The Bacteroides capsule is thought to consist of an array of phase-variable polysaccharides that give rise to subpopulations with distinct cell surface structures. Here, we report the serendipitous discovery of a previously unknown surface structure in Bacteroides thetaiotaomicron: a surface layer composed of a protein of unknown function, BT1927. BT1927, which is expressed in a phase-variable manner by ~1:1,000 cells in a wild-type culture, forms a hexagonally tessellated surface layer. The BT1927-expressing subpopulation is profoundly resistant to complement-mediated killing, due in part to the BT1927-mediated blockade of C3b deposition. Our results show that the Bacteroides surface structure is capable of a far greater degree of structural variation than previously known, and they suggest that structural variation within a Bacteroides species is important for productive gut colonization. IMPORTANCE: Many bacterial species elaborate a capsule, a structure that resides outside the cell wall and mediates microbe-microbe and microbe-host interactions. Species of Bacteroides, the most abundant genus in the human gut, produce a capsule that consists of an array of polysaccharides, some of which are known to mediate interactions with the host immune system. Here, we report the discovery of a previously unknown surface structure in Bacteroides thetaiotaomicron. We show that this protein-based structure is expressed by a subset of cells in a population and protects Bacteroides from killing by complement, a component of the innate immune system. This novel surface layer protein is conserved across many species of the genus Bacteroides, suggesting an important role in colonization and host immune modulation.


Subject(s)
Bacteroides/chemistry , Gastrointestinal Tract/microbiology , Membrane Glycoproteins/analysis , Bacteroides/genetics , Blood Bactericidal Activity , Complement C3b/antagonists & inhibitors , Humans , Immune Evasion , Membrane Glycoproteins/genetics
9.
J Virol ; 89(2): 989-98, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25355897

ABSTRACT

UNLABELLED: Complement is an innate immune system that most animal viruses must face during natural infections. Given that replication and dissemination of the highly pathogenic Nipah virus (NiV) include exposure to environments rich in complement factors, we tested the in vitro sensitivity of NiV to complement-mediated neutralization. Here we show that NiV was completely resistant to in vitro neutralization by normal human serum (NHS). Treatment of purified NiV with NHS activated complement pathways, but there was very little C3 deposition on virus particles. In in vitro reconstitution experiments, NiV particles provided time- and dose-dependent factor I-like protease activity capable of cleaving C3b into inactive C3b (iC3b). NiV-dependent inactivation of C3b only occurred with the cofactors factor H and soluble CR1 but not with CD46. Purified NiV particles did not support C4b cleavage. Electron microscopy of purified NiV particles showed immunogold labeling with anti-factor I antibodies. Our results suggest a novel mechanism by which NiV evades the human complement system through a unique factor I-like activity. IMPORTANCE: Viruses have evolved mechanisms to limit complement-mediated neutralization, some of which involve hijacking cellular proteins involved in control of inappropriate complement activation. Here we report a previously unknown mechanism whereby NiV provides a novel protease activity capable of in vitro cleavage and inactivation of C3b, a key component of the complement cascade. These data help to explain how an enveloped virus such as NiV can infect and disseminate through body fluids that are rich in complement activity. Disruption of the ability of NiV to recruit complement inhibitors could form the basis for the development of effective therapies and safer vaccines to combat these highly pathogenic emerging viruses.


Subject(s)
Complement C3b/antagonists & inhibitors , Complement C3b/metabolism , Fibrinogen/metabolism , Immune Evasion , Nipah Virus/physiology , Viral Structural Proteins/metabolism , Complement Factor H/metabolism , Humans , Hydrolysis , Microscopy, Immunoelectron , Neutralization Tests , Receptors, Complement 3b/metabolism
10.
Clin Vaccine Immunol ; 21(11): 1505-11, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25185576

ABSTRACT

Neisseria meningitidis causes disease only in humans. An important mechanism underlying this host specificity is the ability of the organism to resist complement by recruiting the complement downregulator factor H (FH) to the bacterial surface. In previous studies, binding of FH to one of the major meningococcal FH ligands, factor H binding protein (FHbp), was reported to be specific for human FH. Here we report that sera from 23 of 73 rhesus macaques (32%) tested had high FH binding to FHbp. Similar to human FH, binding of macaque FH to the meningococcal cell surface inhibited the complement alternative pathway by decreasing deposition of C3b. FH contains 20 domains (or short consensus repeats), with domains 6 and 7 being responsible for binding of human FH to FHbp. DNA sequence analyses of FH domains 6 and 7 from macaques with high or low FH binding showed a polymorphism at residue 352 in domain 6, with Tyr being associated with high binding and His with low binding. A recombinant macaque FH 6,7/Fc fragment with Tyr352 showed higher binding to FHbp than the corresponding fragment with His352. In previous studies in human FH transgenic mice, binding of FH to FHbp vaccines decreased protective antibody responses, and mutant FHbp vaccines with decreased FH binding elicited serum antibodies with greater protective activity. Thus, macaques with high FH binding to FHbp represent an attractive nonhuman primate model to investigate further the effects of FH binding on the immunogenicity of FHbp vaccines.


Subject(s)
Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Complement C3b/antagonists & inhibitors , Complement Factor H/metabolism , Meningococcal Vaccines/immunology , Neisseria meningitidis/immunology , Amino Acid Substitution , Animals , Complement Factor H/genetics , Macaca mulatta , Polymorphism, Genetic , Protein Binding , Protein Structure, Tertiary
11.
Clin Vaccine Immunol ; 21(9): 1330-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25030055

ABSTRACT

The vaccinia virus complement control protein (VCP) is a secreted viral protein that binds the C3b and C4b complement components and inhibits the classic and alternative complement pathways. Previously, we reported that an attenuated smallpox vaccine, LC16m8, which was derived from the Lister strain of vaccinia virus (VV-Lister), expressed a glycosylated form of VCP, whereas published sequence data at that time indicated that the VV-Lister VCP has no motif for N-linked glycosylation. We were interested in determining whether the glycosylation of VCP impairs its biological activity, possibly contributing to the attenuation of LC16m8, and the likely origin of the glycosylated VCP. Expression analysis indicated that VV-Lister contains substrains expressing glycosylated VCP and substrains expressing nonglycosylated VCP. Other strains of smallpox vaccine, as well as laboratory strains of vaccinia virus, all expressed nonglycosylated VCP. Individual Lister virus clones expressing either the glycosylated VCP or the nonglycosylated species were isolated, and partially purified VCP from the isolates were found to be functional equivalents in binding human C3b and C4b complement proteins and inhibiting hemolysis and in immunogenicity. Recombinant vaccinia viruses expressing FLAG-tagged glycosylated VCP (FLAG-VCPg) and nonglycosylated VCP (FLAG-VCP) were constructed based on the Western Reserve strain. Purified FLAG-VCP and FLAG-VCPg bind human C3b and C4b and blocked complement-mediated hemolysis. Our data suggest that glycosylation did not affect the biological activity of VCP and thus may not have contributed to the attenuation of LC16m8. In addition, the LC16m8 virus likely originated from a substrain of VV-Lister that expresses glycosylated VCP.


Subject(s)
Complement C3b/antagonists & inhibitors , Complement C4b/antagonists & inhibitors , Vaccinia virus/immunology , Viral Proteins/genetics , Viral Proteins/metabolism , Animals , Glycosylation , Hemolysis , Humans , Male , Mice, Inbred BALB C , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Binding , Vaccinia virus/genetics
12.
Immunol Lett ; 160(2): 163-71, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24491679

ABSTRACT

The alternative pathway of complement is implicated in the pathogenesis of several renal diseases, such as atypical hemolytic uremic syndrome, dense deposit disease and other forms of C3 glomerulopathy. The underlying complement defects include genetic and/or acquired factors, the latter in the form of autoantibodies. Because the autoimmune forms require a specific treatment, in part different from that of the genetic forms, it is important to detect the autoantibodies as soon as possible and understand their characteristics. In this overview, we summarize the types of anti-complement autoantibodies detected in such diseases, i.e. autoantibodies to factor H, factor I, C3b, factor B and those against the C3 convertases (C3 nephritic factor and C4 nephritic factor). We draw attention to newly described autoantibodies and their characteristics, and highlight similarities and differences in the autoimmune forms of these diseases.


Subject(s)
Atypical Hemolytic Uremic Syndrome/immunology , Autoantibodies/biosynthesis , Glomerulonephritis, IGA/immunology , Atypical Hemolytic Uremic Syndrome/genetics , Atypical Hemolytic Uremic Syndrome/pathology , Autoantibodies/genetics , Autoantibodies/immunology , Complement C3-C5 Convertases/antagonists & inhibitors , Complement C3-C5 Convertases/genetics , Complement C3-C5 Convertases/immunology , Complement C3b/antagonists & inhibitors , Complement C3b/genetics , Complement C3b/immunology , Complement Factor B/antagonists & inhibitors , Complement Factor B/genetics , Complement Factor B/immunology , Complement Factor H/antagonists & inhibitors , Complement Factor H/genetics , Complement Factor H/immunology , Complement System Proteins/genetics , Complement System Proteins/immunology , Fibrinogen/antagonists & inhibitors , Fibrinogen/genetics , Fibrinogen/immunology , Gene Expression , Glomerulonephritis, IGA/genetics , Glomerulonephritis, IGA/pathology , Humans
13.
J Immunol ; 191(4): 1775-84, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23863906

ABSTRACT

Staphylococcus aureus is a major human pathogen causing more than a tenth of all septicemia cases and often superficial and deep infections in various tissues. One of the immune evasion strategies of S. aureus is to secrete proteins that bind to the central complement opsonin C3b. One of these, extracellular complement binding protein (Ecb), is known to interfere directly with functions of C3b. Because C3b is also the target of the physiological plasma complement regulator, factor H (FH), we studied the effect of Ecb on the complement regulatory functions of FH. We show that Ecb enhances acquisition of FH from serum onto staphylococcal surfaces. Ecb and FH enhance mutual binding to C3b and also the function of each other in downregulating complement activation. Both Ecb and the C-terminal domains 19-20 of FH bind to the C3d part of C3b. We show that the mutual enhancing effect of Ecb and FH on binding to C3b depends on binding of the FH domain 19 to the C3d part of C3b next to the binding site of Ecb on C3d. Our results show that Ecb, FH, and C3b form a tripartite complex. Upon exposure of serum-sensitive Haemophilus influenzae to human serum, Ecb protected the bacteria, and this effect was enhanced by the addition of the C-terminal domains 19-20 of FH. This finding indicates that the tripartite complex formation could give additional protection to bacteria and that S. aureus is thereby able to use host FH and bacterial Ecb in a concerted action to eliminate C3b at the site of infection.


Subject(s)
Bacterial Proteins/physiology , Complement C3b/metabolism , Complement Factor H/physiology , Complement Inactivator Proteins/physiology , Complement Pathway, Alternative , Immune Evasion/immunology , Staphylococcus aureus/immunology , Virulence Factors/physiology , Bacterial Proteins/chemistry , Binding Sites , Complement C3b/antagonists & inhibitors , Complement C3b/chemistry , Complement Factor H/chemistry , Complement Factor H/genetics , Complement Inactivator Proteins/chemistry , Haemophilus influenzae/immunology , Humans , Immunity, Innate , Models, Molecular , Multiprotein Complexes , Peptide Fragments/metabolism , Point Mutation , Protein Binding , Protein Conformation , Protein Interaction Mapping , Protein Structure, Tertiary , Serum/immunology , Staphylococcal Infections , Virulence Factors/chemistry
14.
J Immunol ; 191(2): 912-21, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23772024

ABSTRACT

Complement is an essential humoral component of innate immunity; however, its inappropriate activation leads to pathology. Polymorphisms, mutations, and autoantibodies affecting factor H (FH), a major regulator of the alternative complement pathway, are associated with various diseases, including age-related macular degeneration, atypical hemolytic uremic syndrome, and C3 glomerulopathies. Restoring FH function could be a treatment option for such pathologies. In this article, we report on an engineered FH construct that directly combines the two major functional regions of FH: the N-terminal complement regulatory domains and the C-terminal surface-recognition domains. This minimal-size FH (mini-FH) binds C3b and has complement regulatory functions similar to those of the full-length protein. In addition, we demonstrate that mini-FH binds to the FH ligands C-reactive protein, pentraxin 3, and malondialdehyde epitopes. Mini-FH was functionally active when bound to the extracellular matrix and endothelial cells in vitro, and it inhibited C3 deposition on the cells. Furthermore, mini-FH efficiently inhibited complement-mediated lysis of host-like cells caused by a disease-associated FH mutation or by anti-FH autoantibodies. Therefore, mini-FH could potentially be used as a complement inhibitor targeting host surfaces, as well as to replace compromised FH in diseases associated with FH dysfunction.


Subject(s)
Complement C3b/immunology , Complement Factor H/genetics , Complement Factor H/immunology , Amino Acid Sequence , Base Sequence , C-Reactive Protein/metabolism , Complement Activation , Complement C3b/antagonists & inhibitors , Complement C3b/metabolism , Complement Pathway, Alternative , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Humans , Malondialdehyde/metabolism , Protein Binding , Protein Engineering , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Sequence Analysis, DNA , Serum Amyloid P-Component/metabolism
15.
J Immunol ; 189(3): 1431-9, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22732591

ABSTRACT

Variola and vaccinia viruses, the two most important members of the family Poxviridae, are known to encode homologs of the human complement regulators named smallpox inhibitor of complement enzymes (SPICE) and vaccinia virus complement control protein (VCP), respectively, to subvert the host complement system. Intriguingly, consistent with the host tropism of these viruses, SPICE has been shown to be more human complement-specific than VCP, and in this study we show that VCP is more bovine complement-specific than SPICE. Based on mutagenesis and mechanistic studies, we suggest that the major determinant for the switch in species selectivity of SPICE and VCP is the presence of oppositely charged residues in the central complement control modules, which help enhance their interaction with factor I and C3b, the proteolytically cleaved form of C3. Thus, our results provide a molecular basis for the species selectivity in poxviral complement regulators.


Subject(s)
Complement C3b/antagonists & inhibitors , Complement Pathway, Alternative/immunology , Vaccinia virus/immunology , Viral Proteins/physiology , Amino Acid Sequence , Amino Acid Substitution/genetics , Amino Acid Substitution/immunology , Animals , Cattle , Complement C3b/genetics , Complement C3b/metabolism , Complement Factor I/antagonists & inhibitors , Complement Factor I/metabolism , Complement Pathway, Alternative/genetics , Humans , Immune Evasion/genetics , Molecular Sequence Data , Species Specificity , Static Electricity , Vaccinia virus/metabolism , Viral Proteins/genetics
16.
J Immunol ; 186(7): 4269-77, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21339361

ABSTRACT

Exposure of nonself surfaces such as those of biomaterials or transplanted cells and organs to host blood frequently triggers innate immune responses, thereby affecting both their functionality and tolerability. Activation of the alternative pathway of complement plays a decisive role in this unfavorable reaction. Whereas previous studies demonstrated that immobilization of physiological regulators of complement activation (RCA) can attenuate this foreign body-induced activation, simple and efficient approaches for coating artificial surfaces with intact RCA are still missing. The conjugation of small molecular entities that capture RCA with high affinity is an intriguing alternative, as this creates a surface with autoregulatory activity upon exposure to blood. We therefore screened two variable cysteine-constrained phage-displayed peptide libraries for factor H-binding peptides. We discovered three peptide classes that differed with respect to their main target binding areas. Peptides binding to the broad middle region of factor H (domains 5-18) were of particular interest, as they do not interfere with either regulatory or binding activities. One peptide in this group (5C6) was further characterized and showed high factor H-capturing activity while retaining its functional integrity. Most importantly, when 5C6 was coated to a model polystyrene surface and exposed to human lepirudin-anticoagulated plasma, the bound peptide captured factor H and substantially inhibited complement activation by the alternative pathway. Our study therefore provides a promising and novel approach to produce therapeutic materials with enhanced biocompatibility.


Subject(s)
Complement Pathway, Alternative/immunology , Peptide Fragments/metabolism , Peptide Fragments/therapeutic use , Biocompatible Materials/metabolism , Cloning, Molecular , Complement C3b/antagonists & inhibitors , Complement C3b/metabolism , Complement Factor H/metabolism , Complement Factor H/therapeutic use , Complement Factor I/antagonists & inhibitors , Complement Factor I/metabolism , Complement Inactivator Proteins/metabolism , Hemolysis , Humans , Peptide Library , Protein Binding/immunology , Surface Properties
17.
J Immunol ; 184(4): 1946-55, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20083651

ABSTRACT

We recently characterized an interaction between the Staphylococcus aureus immune evasion molecule Staphylococcus aureus binder of Ig (Sbi) and complement C3, an interaction mediated primarily through the binding of C3d(g) to Sbi domain IV. Events related to these studies prompted us to investigate via mutagenesis the binding interface of C3d for Sbi domain IV (Sbi-IV), as well as to revisit the controversial issue of the complement receptor 2 (CR2) binding site of C3d. Specifically, we had shown that Sbi domains III and IV fragment binding to C3dg inhibited the latter's binding to CR2. Moreover, a published cocrystal structure of C3d bound to complement inhibitory C-terminal domain of extracellular fibrinogen-binding protein (Efb-C), a structural and functional homolog of Sbi-IV, showed Efb-C binding to a region on the concave face of C3d previously implicated in CR2 binding by our mutagenesis data but not confirmed in the CR2(short consensus repeat [SCR]1-2):C3d cocrystal structure. We have now analyzed by surface plasmon resonance the binding of a series of variant C3dg molecules to biosensor-bound Sbi-IV or CR2(SCR1-2). We found that mutations to the concave face acidic pocket of C3d significantly affected binding to both Sbi-IV and CR2, although there was divergence in which residues were most important in each case. By contrast, no binding defects were seen for mutations made to the sideface of C3d implicated from the cocrystal structure to be involved in binding CR2(SCR1-2). The results with Sbi-IV suggest a mode of binding highly similar to that visualized in the Efb-C:C3d complex. The results with CR2 confirm our earlier mapping studies and cast even further doubt on the physiologic relevance of the complex visualized in the C3d:CR2 cocrystal.


Subject(s)
Bacterial Proteins/chemistry , Carrier Proteins/chemistry , Complement C3d/chemistry , Immune Evasion , Receptors, Complement 3d/chemistry , Staphylococcus aureus/immunology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Complement C3b/antagonists & inhibitors , Complement C3b/genetics , Complement C3b/metabolism , Complement C3d/genetics , Complement C3d/metabolism , Crystallization , Crystallography, X-Ray , DNA Mutational Analysis , Humans , Immune Evasion/genetics , Mice , Mutagenesis, Site-Directed , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/genetics , Peptide Fragments/metabolism , Receptors, Complement 3d/antagonists & inhibitors , Receptors, Complement 3d/genetics , Staphylococcus aureus/genetics
18.
Invest Ophthalmol Vis Sci ; 50(3): 1392-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19029031

ABSTRACT

PURPOSE: Studies implicate activation of complement among the processes involved in the pathogenesis of age-related macular degeneration (AMD). Questions pertain to the trigger(s) responsible for the complement-associated events. The authors previously reported that photooxidation products of A2E can activate complement. Here they have further explored these events. METHODS: In vitro assays using human serum as a source of complement were used, and the C3 split product iC3b was measured by enzyme immunoassay. Serum was placed in contact with ARPE-19 cells and polarized human fetal retinal pigment epithelium that had accumulated A2E and were irradiated (430 nm). Serum was also incubated in wells precoated with bisretinoid pigments of lipofuscin and their oxidized forms. iC3b generation in normal human serum (NHS) was compared with that in factor B-depleted and C1q-depleted human serum. RESULTS: iC3b levels were elevated in NHS placed in contact with A2E-laden retinal pigment epithelium that were irradiated to generate A2E photooxidation products. iC3b was also increased in serum incubated in wells precoated with peroxy-A2E, the lipofuscin pigment all-trans-retinal dimer, and oxidized forms of all-trans-retinal dimer. Substitution of NHS with factor B-depleted sera abrogated these increases in iC3b. Complement activation was also suppressed by the addition of C-reactive protein and by a C3 cleavage inhibitor. CONCLUSIONS: The authors suggest that bisretinoid pigments of retinal pigment epithelial lipofuscin, subsequent to photoactivation and cleavage, serve to activate complement. Complement activation by this mechanism is dependent on the alternative pathway and can be modulated by an inhibitor of C3 cleavage. These events in the setting of complement dysregulation could contribute to the chronic inflammation that underlies AMD pathogenesis.


Subject(s)
Complement Activation/physiology , Complement C3b/biosynthesis , Lipofuscin/metabolism , Pyridinium Compounds/metabolism , Retinal Pigment Epithelium/metabolism , Retinoids/metabolism , Apoptosis/drug effects , C-Reactive Protein/pharmacology , Cells, Cultured , Complement C3b/antagonists & inhibitors , Complement C3b Inactivator Proteins/pharmacology , Complement Pathway, Alternative/physiology , Etoposide/pharmacology , Humans , Immunoenzyme Techniques , Lipofuscin/chemistry , Oxidation-Reduction , Peptides, Cyclic/pharmacology , Pyridinium Compounds/chemistry , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/embryology , Retinoids/chemistry
19.
J Immunol ; 181(12): 8624-32, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19050282

ABSTRACT

Fusobacterium necrophorum subspecies funduliforme is an obligate anaerobic Gram-negative rod causing invasive infections such as the life-threatening Lemierre's syndrome (sore throat, septicemia, jugular vein thrombosis, and disseminated infection). The aim of our study was to understand if and how F. necrophorum avoids C activation. We studied 12 F. necrophorum subsp. funduliforme strains isolated from patients with sepsis. All strains were resistant to serum killing after a 1-h incubation in 20% serum. The bacteria bound, at different levels, the C inhibitor factor H (fH). Binding was ionic and specific in nature and occurred via sites on both the N terminus and the C terminus of fH. Bound fH remained functionally active as a cofactor for factor I in the cleavage of C3b. Interestingly, patients with the most severe symptoms carried strains with the strongest ability to bind fH. An increased C3b deposition and membrane attack complex formation on the surface of a weakly fH-binding strain was observed and its survival in serum at 3.5 h was impaired. This strain had not caused a typical Lemierre's syndrome. These data, and the fact that fH-binding correlated with the severity of disease, suggest that the binding of fH contributes to virulence and survival of F. necrophorum subsp. funduliforme in the human host. Our data show, for the first time, that an anaerobic bacterium is able to bind the C inhibitor fH to evade C attack.


Subject(s)
Bacterial Adhesion/immunology , Blood Bactericidal Activity/immunology , Complement Pathway, Alternative/immunology , Fusobacterium necrophorum/growth & development , Fusobacterium necrophorum/immunology , Anaerobiosis/immunology , Complement C3b/antagonists & inhibitors , Complement C3b/metabolism , Complement C3b/physiology , Complement Factor H/metabolism , Complement Factor H/physiology , Culture Media, Conditioned , Dose-Response Relationship, Immunologic , Fusobacterium necrophorum/pathogenicity , Humans , Protein Binding/immunology , Staphylococcus aureus/growth & development , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity , Streptococcus pneumoniae/growth & development , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/pathogenicity
20.
J Immunol ; 181(9): 6328-36, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18941224

ABSTRACT

Enterococcus faecalis (Ef) accounts for most cases of enterococcal bacteremia, which is one of the principal causes of nosocomial bloodstream infections (BSI). Among several virulence factors associated with the pathogenesis of Ef, an extracellular gelatinase (GelE) has been known to be the most common factor, although its virulence mechanisms, especially in association with human BSI, have yet to be demonstrated. In this study, we describe the complement resistance mechanism of Ef mediated by GelE. Using purified GelE, we determined that it cleaved the C3 occurring in human serum into a C3b-like molecule, which was inactivated rapidly via reaction with water. This C3 convertase-like activity of GelE was shown to result in a consumption of C3 and thus inhibited the activation of the complement system. Also, GelE was confirmed to degrade an iC3b that was deposited on the Ag surfaces without affecting the bound C3b. This proteolytic effect of GelE against the major complement opsonin resulted in a substantial reduction in Ef phagocytosis by human polymorphonuclear leukocytes. In addition, we verified that the action of GelE against C3, which is a central component of the complement cascade, was human specific. Taken together, it was suggested that GelE may represent a promising molecule for targeting human BSI associated with Ef.


Subject(s)
Complement C3/metabolism , Complement C3b/metabolism , Enterococcus faecalis/enzymology , Enterococcus faecalis/immunology , Extracellular Fluid/enzymology , Gelatinases/physiology , Amino Acid Sequence , Animals , Blood Bactericidal Activity/immunology , Chickens , Complement Activation/immunology , Complement C3/antagonists & inhibitors , Complement C3/physiology , Complement C3b/antagonists & inhibitors , Complement Pathway, Alternative/immunology , Dogs , Extracellular Fluid/immunology , Gram-Positive Bacterial Infections/enzymology , Gram-Positive Bacterial Infections/immunology , Gram-Positive Bacterial Infections/microbiology , Guinea Pigs , Humans , Hydrolysis , Mice , Molecular Sequence Data , Neutrophils/immunology , Phagocytosis/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...