Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
PLoS Pathog ; 17(11): e1010051, 2021 11.
Article in English | MEDLINE | ID: mdl-34752492

ABSTRACT

Complement proteins can form membrane attack complex (MAC) pores that directly kill Gram-negative bacteria. MAC pores assemble by stepwise binding of C5b, C6, C7, C8 and finally C9, which can polymerize into a transmembrane ring of up to 18 C9 monomers. It is still unclear if the assembly of a polymeric-C9 ring is necessary to sufficiently damage the bacterial cell envelope to kill bacteria. In this paper, polymerization of C9 was prevented without affecting binding of C9 to C5b-8, by locking the first transmembrane helix domain of C9. Using this system, we show that polymerization of C9 strongly enhanced damage to both the bacterial outer and inner membrane, resulting in more rapid killing of several Escherichia coli and Klebsiella strains in serum. By comparing binding of wildtype and 'locked' C9 by flow cytometry, we also show that polymerization of C9 is impaired when the amount of available C9 per C5b-8 is limited. This suggests that an excess of C9 is required to efficiently form polymeric-C9. Finally, we show that polymerization of C9 was impaired on complement-resistant E. coli strains that survive killing by MAC pores. This suggests that these bacteria can specifically block polymerization of C9. All tested complement-resistant E. coli expressed LPS O-antigen (O-Ag), compared to only one out of four complement-sensitive E. coli. By restoring O-Ag expression in an O-Ag negative strain, we show that the O-Ag impairs polymerization of C9 and results in complement-resistance. Altogether, these insights are important to understand how MAC pores kill bacteria and how bacterial pathogens can resist MAC-dependent killing.


Subject(s)
Blood Bactericidal Activity , Cell Wall/pathology , Complement C9/chemistry , Complement Membrane Attack Complex/pharmacology , Escherichia coli/growth & development , Klebsiella/growth & development , Polymerization , Cell Wall/drug effects , Escherichia coli/drug effects , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Humans , Klebsiella/drug effects , Klebsiella Infections/drug therapy , Klebsiella Infections/microbiology
2.
Fish Shellfish Immunol ; 104: 101-110, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32464273

ABSTRACT

The ninth complement component (C9) is a terminal complement component (TCC) that is involved in creating the membrane attack complex (MAC) on the target cell surface. In this study, the CsC9 (C9 of Cynoglossus semilaevis) cDNA sequence was cloned and characterized. The full-length CsC9 cDNA measured 2,150 bp, containing an open reading frame (ORF) of 1,803 bp, a 5'-untranslated region (UTR) of 24 bp and a 3'-UTR of 323 bp. A domain search revealed that the CsC9 protein contains five domains, including two TSP1s, an LDLRA, an EGF, and a MACPF. Quantitative real-time PCR analysis showed that CsC9 at the mRNA level was expressed in all the tested tissues, with the highest expression being observed in the liver. CsC9 expression is significantly upregulated in the tested tissues after challenge with Vibrio anguillarum. To further characterize the role of CsC9, peripheral blood mononuclear cells of C. semilaevis were used for transcriptome analysis after incubation with recombinant CsC9 (rCsC9) protein. A total of 3,775 significant differentially expressed genes (DEGs) were identified between the control and the rCsC9-treated group, including 2,063 upregulated genes and 1,712 downregulated genes. KEGG analyses revealed that the DEGs were enriched in cell adhesion molecules, cytokine-cytokine receptor interactions, T cell receptor signaling pathways, B cell receptor signaling pathways and Toll-like receptor signaling pathways. The results of this study indicate that in addition to participating in MAC formation, CsC9 might play multiple roles in the innate and adaptive immunity of C. semilaevis.


Subject(s)
Complement C9/genetics , Complement C9/immunology , Fish Diseases/immunology , Flatfishes/genetics , Flatfishes/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Adaptive Immunity , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Complement C9/chemistry , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/veterinary , Leukocytes/metabolism , Phylogeny , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sequence Alignment/veterinary , Transcriptome , Vibrio , Vibrio Infections
3.
Ecotoxicology ; 29(7): 837-845, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32656652

ABSTRACT

Vibrio alginolyticus is posting an increasing threat to survival of grouper. Classical complement cascade can trigger initiation of immunity, while complement 9 (C9) is a major complement molecule involved in final step of membrane attack complex (MAC) formation. In this study, full-length EcC9 contained an ORF sequence of 1779 bp, encoding a polypeptide of 592 amino acids. A high-level expression of EcC9 mRNA was observed in liver. Following vibrio challenge, increased expression levels of EcC1q, EcBf/C2, EcC4, EcC6, EcC7 and EcC9 mRNA were detected in liver and kidney. These results implied that elevated expression level of classical complement pathway (CCP) and terminal complement components (TCCs) may assess toxicological effect of V. alginolyticus.


Subject(s)
Bass/genetics , Bass/immunology , Complement C9/genetics , Complement C9/immunology , Fish Diseases/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Amino Acid Sequence , Animals , Base Sequence , Complement C9/chemistry , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/veterinary , Phylogeny , Sequence Alignment/veterinary , Vibrio alginolyticus/physiology
4.
Fish Shellfish Immunol ; 86: 449-458, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30508672

ABSTRACT

The complement system plays an important role in host defense against invading microorganisms. Complement component C9 is the last component that is involved in the formation of the membrane attack complex (MAC) on the surface of target cells. In the present study, the full length C9 cDNA sequence of 1984 bp with an open reading frame (ORF) of 1809 bp was cloned from southern catfish (Silurus meridionalis). The deduced amino acid sequence showed similarity with other teleost fish. The mRNA expression of C9 was detected in the liver, spleen, stomach, intestine, and head kidney, with highest levels detected in the liver. The mRNA of C9 was first detected in the yolk syncytial layer at 34 h post fertilization (hpf) with whole mount in situ hybridization, followed by the liver at 36 h post hatching (hph). The mRNA expression of C9 was upregulated significantly in the liver, spleen, and intestine following the injection with Aeromonas hydrophila, suggesting that C9 played an important role in defense against invading pathogens in southern catfish. Therefore, these results provide important information to understand the functions of C9 during fish early development in fish.


Subject(s)
Catfishes/genetics , Catfishes/immunology , Complement C9/genetics , Complement C9/immunology , Fish Diseases/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Aeromonas hydrophila/physiology , Amino Acid Sequence , Animals , Base Sequence , Complement C9/chemistry , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/veterinary , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/veterinary , Sequence Alignment/veterinary
5.
Anal Chem ; 89(6): 3483-3491, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28221766

ABSTRACT

The human complement C9 protein (∼65 kDa) is a member of the complement pathway. It plays an essential role in the membrane attack complex (MAC), which forms a lethal pore on the cellular surface of pathogenic bacteria. Here, we charted in detail the structural microheterogeneity of C9 purified from human blood serum, using an integrative workflow combining high-resolution native mass spectrometry and (glyco)peptide-centric proteomics. The proteoform profile of C9 was acquired by high-resolution native mass spectrometry, which revealed the co-occurrence of ∼50 distinct mass spectrometry (MS) signals. Subsequent peptide-centric analysis, through proteolytic digestion of C9 and liquid chromatography (LC)-tandem mass spectrometry (MS/MS) measurements of the resulting peptide mixtures, provided site-specific quantitative profiles of three different types of C9 glycosylation and validation of the native MS data. Our study provides a detailed specification, validation, and quantification of 15 co-occurring C9 proteoforms and the first direct experimental evidence of O-linked glycans in the N-terminal region. Additionally, next to the two known glycosylation sites, a third novel, albeit low abundant, N-glycosylation site on C9 is identified, which surprisingly does not possess the canonical N-glycosylation sequence N-X-S/T. Our data also reveal a binding of up to two Ca2+ ions to C9. Mapping all detected and validated sites of modifications on a structural model of C9, as present in the MAC, hints at their putative roles in pore formation or receptor interactions. The applied methods herein represent a powerful tool for the unbiased in-depth analysis of plasma proteins and may advance biomarker discovery, as aberrant glycosylation profiles may be indicative of the pathophysiological state of the patients.


Subject(s)
Complement C9/metabolism , Proteomics , Chromatography, Liquid , Complement C9/chemistry , Glycosylation , Humans , Mass Spectrometry , Models, Molecular , Protein Conformation , Protein Interaction Mapping
6.
J Immunol ; 194(5): 2309-18, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25637016

ABSTRACT

Shiga toxin (Stx)-producing Escherichia coli (STEC) cause hemolytic uremic syndrome (HUS). This study investigated whether Stx2 induces hemolysis and whether complement is involved in the hemolytic process. RBCs and/or RBC-derived microvesicles from patients with STEC-HUS (n = 25) were investigated for the presence of C3 and C9 by flow cytometry. Patients exhibited increased C3 deposition on RBCs compared with controls (p < 0.001), as well as high levels of C3- and C9-bearing RBC-derived microvesicles during the acute phase, which decreased after recovery. Stx2 bound to P1 (k) and P2 (k) phenotype RBCs, expressing high levels of the P(k) Ag (globotriaosylceramide), the known Stx receptor. Stx2 induced the release of hemoglobin and lactate dehydrogenase in whole blood, indicating hemolysis. Stx2-induced hemolysis was not demonstrated in the absence of plasma and was inhibited by heat inactivation, as well as by the terminal complement pathway Ab eculizumab, the purinergic P2 receptor antagonist suramin, and EDTA. In the presence of whole blood or plasma/serum, Stx2 induced the release of RBC-derived microvesicles coated with C5b-9, a process that was inhibited by EDTA, in the absence of factor B, and by purinergic P2 receptor antagonists. Thus, complement-coated RBC-derived microvesicles are elevated in HUS patients and induced in vitro by incubation of RBCs with Stx2, which also induced hemolysis. The role of complement in Stx2-mediated hemolysis was demonstrated by its occurrence only in the presence of plasma and its abrogation by heat inactivation, EDTA, and eculizumab. Complement activation on RBCs could play a role in the hemolytic process occurring during STEC-HUS.


Subject(s)
Coated Vesicles/drug effects , Erythrocytes/drug effects , Escherichia coli Infections/blood , Escherichia coli O157/pathogenicity , Hemolytic-Uremic Syndrome/blood , Shiga Toxin/toxicity , Adult , Aged , Antibodies, Monoclonal, Humanized/pharmacology , Child , Child, Preschool , Coated Vesicles/chemistry , Coated Vesicles/immunology , Complement Activation/drug effects , Complement C3/chemistry , Complement C9/chemistry , Complement Membrane Attack Complex/chemistry , Edetic Acid/pharmacology , Erythrocytes/chemistry , Erythrocytes/immunology , Erythrocytes/pathology , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Escherichia coli O157/immunology , Escherichia coli O157/metabolism , Female , Gene Expression , Hemolysis/drug effects , Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/microbiology , Hemolytic-Uremic Syndrome/pathology , Humans , Infant , L-Lactate Dehydrogenase/metabolism , Male , Middle Aged , Purinergic P2 Receptor Antagonists/pharmacology , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2/immunology , Shiga Toxin/chemistry , Shiga Toxin/immunology , Suramin/pharmacology , Trihexosylceramides/immunology
7.
Infect Immun ; 83(3): 888-97, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25534939

ABSTRACT

Upon infection, pathogenic Leptospira species bind several complement regulators in order to overcome host innate immunity. We previously characterized a 20-kDa leptospiral surface protein which interacts with C4b binding protein (C4BP): leptospiral complement regulator-acquiring protein A (LcpA). Here we show that LcpA also interacts with human factor H (FH), which remains functionally active once bound to the protein. Antibodies directed against short consensus repeat 20 (SCR20) inhibited binding of FH to LcpA by approximately 90%, thus confirming that this particular domain is involved in the interaction. We have also shown for the first time that leptospires bind human vitronectin and that the interaction is mediated by LcpA. Coincubation with heparin blocked LcpA-vitronectin interaction in a dose-dependent manner, strongly suggesting that binding may occur through the heparin binding domains of vitronectin. LcpA also bound to the terminal pathway component C9 and inhibited Zn(2+)-induced polymerization and membrane attack complex (MAC) formation. Competitive binding assays indicated that LcpA interacts with C4BP, FH, and vitronectin through distinct sites. Taken together, our findings indicate that LcpA may play a role in leptospiral immune evasion.


Subject(s)
Bacterial Proteins/chemistry , Leptospira interrogans/chemistry , Leptospira/chemistry , Peptide Fragments/chemistry , Vitronectin/chemistry , Antibodies, Monoclonal/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/immunology , Binding Sites , Binding, Competitive , Complement Activation , Complement C4b-Binding Protein/chemistry , Complement C4b-Binding Protein/immunology , Complement C9/chemistry , Complement C9/immunology , Complement Factor H/chemistry , Complement Factor H/immunology , Complement Membrane Attack Complex/chemistry , Heparin/chemistry , Humans , Immune Evasion , Leptospira/immunology , Leptospira/pathogenicity , Leptospira interrogans/immunology , Leptospira interrogans/pathogenicity , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/immunology , Protein Binding , Vitronectin/immunology , Zinc/chemistry
8.
J Biol Chem ; 287(39): 32913-21, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22854970

ABSTRACT

The human lectin complement pathway activation molecules comprise mannose-binding lectin (MBL) and ficolin-1, -2, and -3 in complex with associated serine proteases MASP-1, -2, and -3 and the non-enzymatic small MBL associated protein or sMAP. Recently, a novel plasma protein named MBL/ficolin-associated protein-1 (MAP-1) was identified in humans. This protein is the result of a differential splicing of the MASP1 gene and includes the major part of the heavy chain but lacks the serine protease domain. We investigated the direct interactions of MAP-1 and MASP-3 with ficolin-3 and MBL using surface plasmon resonance and found affinities around 5 nm and 2.5 nm, respectively. We studied structural aspects of MAP-1 and could show by multi-angle laser light scattering that MAP-1 forms a calcium-dependent homodimer in solution. We were able to determine the crystal structure of MAP-1, which also contains a head-to-tail dimer ∼146 Šlong. This structure of MAP-1 also enables modeling and assembly of the MASP-1 molecule in its entirety. Finally we found that MAP-1 competes with all three MASPs for ligand binding and is able to mediate a strong dose-dependent inhibitory effect on the lectin pathway activation, as measured by levels of C3 and C9.


Subject(s)
Complement Pathway, Mannose-Binding Lectin/physiology , Glycoproteins , Lectins , Mannose-Binding Lectin , Mannose-Binding Protein-Associated Serine Proteases , Protein Multimerization/physiology , Alternative Splicing/physiology , Animals , CHO Cells , Complement C3/chemistry , Complement C3/metabolism , Complement C9/chemistry , Complement C9/metabolism , Cricetinae , Cricetulus , Glycoproteins/chemistry , Glycoproteins/metabolism , Humans , Lectins/chemistry , Lectins/metabolism , Mannose-Binding Lectin/chemistry , Mannose-Binding Lectin/metabolism , Mannose-Binding Protein-Associated Serine Proteases/chemistry , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Models, Molecular , Protein Structure, Secondary
9.
J Biol Chem ; 287(11): 8092-100, 2012 Mar 09.
Article in English | MEDLINE | ID: mdl-22267731

ABSTRACT

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


Subject(s)
Arthritis, Rheumatoid/metabolism , Complement Membrane Attack Complex/antagonists & inhibitors , Complement Membrane Attack Complex/metabolism , Extracellular Matrix Proteins/metabolism , Glycoproteins/metabolism , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Basement Membrane/chemistry , Basement Membrane/metabolism , Basement Membrane/pathology , Complement Activation/genetics , Complement C3-C5 Convertases/chemistry , Complement C3-C5 Convertases/genetics , Complement C3-C5 Convertases/metabolism , Complement C4b-Binding Protein/chemistry , Complement C4b-Binding Protein/genetics , Complement C4b-Binding Protein/metabolism , Complement C9/chemistry , Complement C9/genetics , Complement C9/metabolism , Complement Membrane Attack Complex/chemistry , Complement Membrane Attack Complex/genetics , Complement System Proteins/chemistry , Complement System Proteins/genetics , Complement System Proteins/metabolism , Down-Regulation/genetics , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/genetics , Female , Glycoproteins/chemistry , Glycoproteins/genetics , HEK293 Cells , Humans , Male
10.
Fish Shellfish Immunol ; 35(2): 599-606, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23684808

ABSTRACT

Complement system is known as highly sophisticated immune defense mechanism for antigen recognition as well as effector functions. Activation of the terminal pathway of the complement system leads to the assembly of terminal complement complexes (C5b-9), which induces the characteristic complement-mediated cytolysis. The lytic activity of shark complement involves functional analogues of mammalian C8 and C9. In this article, a full-length cDNA of C9 (CpC9) is identified from cartilaginous species, the whitespotted bambooshark, Chiloscyllium plagiosum by RACE. The CpC9 cDNA is 2263 bp in length, encoding a protein of 603 amino acids, which shares 42% and 43% identity with human and Xenopus C9 respectively. Through sequence alignment and comparative analysis, the CpC9 protein was found well conserved, with the typical modular architecture in TCCs and nearly unanimous cysteine composition from fish to mammal. Phylogenetic analysis places it in a clade with C9 orthologs in higher vertebrate and as a sister taxa to the Xenopus. Expression analysis revealed that CpC9 is constitutively highly expressed in shark liver, with much less or even undetectable expression in other tissues; demonstrating liver is the primary tissue for C9synthesis. To sum up, the structural conservation and distinctive phylogenetics might indicate the potentially vital role of CpC9 in shark immune response, though it remains to be confirmed by further study.


Subject(s)
Complement C9/genetics , Fish Proteins/genetics , Sharks/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Complement C9/chemistry , Complement C9/metabolism , DNA, Complementary/genetics , DNA, Complementary/metabolism , Fish Proteins/chemistry , Fish Proteins/metabolism , Gene Expression Profiling , Molecular Sequence Data , Organ Specificity , Phylogeny , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Alignment , Sharks/metabolism
11.
Commun Biol ; 6(1): 42, 2023 01 13.
Article in English | MEDLINE | ID: mdl-36639734

ABSTRACT

The Membrane Attack Complex (MAC) is responsible for forming large ß-barrel channels in the membranes of pathogens, such as gram-negative bacteria. Off-target MAC assembly on endogenous tissue is associated with inflammatory diseases and cancer. Accordingly, a human C5b-9 specific antibody, aE11, has been developed that detects a neoepitope exposed in C9 when it is incorporated into the C5b-9 complex, but not present in the plasma native C9. For nearly four decades aE11 has been routinely used to study complement, MAC-related inflammation, and pathophysiology. However, the identity of C9 neoepitope remains unknown. Here, we determined the cryo-EM structure of aE11 in complex with polyC9 at 3.2 Å resolution. The aE11 binding site is formed by two separate surfaces of the oligomeric C9 periphery and is therefore a discontinuous quaternary epitope. These surfaces are contributed by portions of the adjacent TSP1, LDLRA, and MACPF domains of two neighbouring C9 protomers. By substituting key antibody interacting residues to the murine orthologue, we validated the unusual binding modality of aE11. Furthermore, aE11 can recognise a partial epitope in purified monomeric C9 in vitro, albeit weakly. Taken together, our results reveal the structural basis for MAC recognition by aE11.


Subject(s)
Complement C9 , Complement Membrane Attack Complex , Humans , Animals , Mice , Complement Membrane Attack Complex/metabolism , Complement C5b , Complement C9/chemistry , Complement C9/metabolism , Complement System Proteins/metabolism , Epitopes
12.
J Biol Chem ; 286(20): 17585-92, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21454577

ABSTRACT

C8 is one of five complement proteins that assemble on bacterial membranes to form the lethal pore-like "membrane attack complex" (MAC) of complement. The MAC consists of one C5b, C6, C7, and C8 and 12-18 molecules of C9. C8 is composed of three genetically distinct subunits, C8α, C8ß, and C8γ. The C6, C7, C8α, C8ß, and C9 proteins are homologous and together comprise the MAC family of proteins. All contain N- and C-terminal modules and a central 40-kDa membrane attack complex perforin (MACPF) domain that has a key role in forming the MAC pore. Here, we report the 2.5 Å resolution crystal structure of human C8 purified from blood. This is the first structure of a MAC family member and of a human MACPF-containing protein. The structure shows the modules in C8α and C8ß are located on the periphery of C8 and not likely to interact with the target membrane. The C8γ subunit, a member of the lipocalin family of proteins that bind and transport small lipophilic molecules, shows no occupancy of its putative ligand-binding site. C8α and C8ß are related by a rotation of ∼22° with only a small translational component along the rotation axis. Evolutionary arguments suggest the geometry of binding between these two subunits is similar to the arrangement of C9 molecules within the MAC pore. This leads to a model of the MAC that explains how C8-C9 and C9-C9 interactions could facilitate refolding and insertion of putative MACPF transmembrane ß-hairpins to form a circular pore.


Subject(s)
Complement C8/chemistry , Models, Chemical , Models, Molecular , Complement C8/immunology , Complement C8/metabolism , Complement C9/chemistry , Complement C9/immunology , Complement C9/metabolism , Complement Membrane Attack Complex/chemistry , Complement Membrane Attack Complex/immunology , Complement Membrane Attack Complex/metabolism , Crystallography, X-Ray , Humans , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship
13.
J Biol Chem ; 286(23): 20952-62, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21507937

ABSTRACT

CD59 is a glycosylphosphatidylinositol-anchored protein that inhibits the assembly of the terminal complement membrane attack complex (MAC) pore, whereas Streptococcus intermedius intermedilysin (ILY), a pore forming cholesterol-dependent cytolysin (CDC), specifically binds to human CD59 (hCD59) to initiate the formation of its pore. The identification of the residues of ILY and hCD59 that form their binding interface revealed a remarkably deep correspondence between the hCD59 binding site for ILY and that for the MAC proteins C8α and C9. ILY disengages from hCD59 during the prepore to pore transition, suggesting that loss of this interaction is necessary to accommodate specific structural changes associated with this transition. Consistent with this scenario, mutants of hCD59 or ILY that increased the affinity of this interaction decreased the cytolytic activity by slowing the transition of the prepore to pore but not the assembly of the prepore oligomer. A signature motif was also identified in the hCD59 binding CDCs that revealed a new hCD59-binding member of the CDC family. Although the binding site on hCD59 for ILY, C8α, and C9 exhibits significant homology, no similarity exists in their binding sites for hCD59. Hence, ILY and the MAC proteins interact with common amino acids of hCD59 but lack detectable conservation in their binding sites for hCD59.


Subject(s)
Bacteriocins/metabolism , CD59 Antigens/metabolism , Complement C8/metabolism , Amino Acid Motifs , Animals , Bacteriocins/chemistry , Bacteriocins/genetics , Binding Sites , CD59 Antigens/chemistry , CD59 Antigens/genetics , CHO Cells , Complement C8/chemistry , Complement C8/genetics , Complement C9/chemistry , Complement C9/genetics , Complement C9/metabolism , Cricetinae , Cricetulus , Humans , Mutation , Peptide Mapping/methods , Streptococcus intermedius/chemistry , Streptococcus intermedius/genetics , Streptococcus intermedius/metabolism
14.
Nat Commun ; 12(1): 6086, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34667172

ABSTRACT

Unregulated complement activation causes inflammatory and immunological pathologies with consequences for human disease. To prevent bystander damage during an immune response, extracellular chaperones (clusterin and vitronectin) capture and clear soluble precursors to the membrane attack complex (sMAC). However, how these chaperones block further polymerization of MAC and prevent the complex from binding target membranes remains unclear. Here, we address that question by combining cryo electron microscopy (cryoEM) and cross-linking mass spectrometry (XL-MS) to solve the structure of sMAC. Together our data reveal how clusterin recognizes and inhibits polymerizing complement proteins by binding a negatively charged surface of sMAC. Furthermore, we show that the pore-forming C9 protein is trapped in an intermediate conformation whereby only one of its two transmembrane ß-hairpins has unfurled. This structure provides molecular details for immune pore formation and helps explain a complement control mechanism that has potential implications for how cell clearance pathways mediate immune homeostasis.


Subject(s)
Complement Membrane Attack Complex/chemistry , Complement Membrane Attack Complex/immunology , Complement C8/chemistry , Complement C8/metabolism , Complement C9/chemistry , Complement C9/immunology , Cryoelectron Microscopy , Humans , Protein Binding , Protein Conformation, alpha-Helical , Protein Domains
15.
Nat Commun ; 9(1): 3266, 2018 08 15.
Article in English | MEDLINE | ID: mdl-30111885

ABSTRACT

Complement component 9 (C9) functions as the pore-forming component of the Membrane Attack Complex (MAC). During MAC assembly, multiple copies of C9 are sequentially recruited to membrane associated C5b8 to form a pore. Here we determined the 2.2 Å crystal structure of monomeric murine C9 and the 3.9 Å resolution cryo EM structure of C9 in a polymeric assembly. Comparison with other MAC proteins reveals that the first transmembrane region (TMH1) in monomeric C9 is uniquely positioned and functions to inhibit its self-assembly in the absence of C5b8. We further show that following C9 recruitment to C5b8, a conformational change in TMH1 permits unidirectional and sequential binding of additional C9 monomers to the growing MAC. This mechanism of pore formation contrasts with related proteins, such as perforin and the cholesterol dependent cytolysins, where it is believed that pre-pore assembly occurs prior to the simultaneous release of the transmembrane regions.


Subject(s)
Complement C9/chemistry , Complement Membrane Attack Complex/chemistry , Membrane Proteins/chemistry , Protein Domains , Animals , Complement C9/genetics , Complement C9/metabolism , Complement Membrane Attack Complex/metabolism , Complement Membrane Attack Complex/ultrastructure , Complement System Proteins/chemistry , Complement System Proteins/genetics , Complement System Proteins/metabolism , Cryoelectron Microscopy , Crystallography, X-Ray , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Models, Molecular , Protein Binding
16.
Nat Commun ; 9(1): 5316, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30552328

ABSTRACT

The membrane attack complex (MAC) is one of the immune system's first responders. Complement proteins assemble on target membranes to form pores that lyse pathogens and impact tissue homeostasis of self-cells. How MAC disrupts the membrane barrier remains unclear. Here we use electron cryo-microscopy and flicker spectroscopy to show that MAC interacts with lipid bilayers in two distinct ways. Whereas C6 and C7 associate with the outer leaflet and reduce the energy for membrane bending, C8 and C9 traverse the bilayer increasing membrane rigidity. CryoEM reconstructions reveal plasticity of the MAC pore and demonstrate how C5b6 acts as a platform, directing assembly of a giant ß-barrel whose structure is supported by a glycan scaffold. Our work provides a structural basis for understanding how ß-pore forming proteins breach the membrane and reveals a mechanism for how MAC kills pathogens and regulates cell functions.


Subject(s)
Complement Membrane Attack Complex/chemistry , Complement Membrane Attack Complex/ultrastructure , Cryoelectron Microscopy/methods , Lipid Bilayers/chemistry , Complement C6/chemistry , Complement C6/metabolism , Complement C6/ultrastructure , Complement C7/chemistry , Complement C7/metabolism , Complement C7/ultrastructure , Complement C8/chemistry , Complement C8/metabolism , Complement C8/ultrastructure , Complement C9/chemistry , Complement C9/metabolism , Complement C9/ultrastructure , Complement Membrane Attack Complex/metabolism , Humans , Image Processing, Computer-Assisted , Lipid Bilayers/metabolism , Liposomes , Models, Molecular , Polysaccharides/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Spectrum Analysis/methods
17.
Biochim Biophys Acta ; 1209(1): 117-22, 1994 Nov 16.
Article in English | MEDLINE | ID: mdl-7947973

ABSTRACT

Purified C9 with expected hemolytic and polymerizing activities was found to contain approximately 0.2 mol of sulfhydryl groups/mol of C9. By proteolysis of C9 with labeled SH groups, the SH residues on intact C9 were mapped to Cys-359 and Cys-384 which, presumably, form an intra-domain disulfide bond in the intact molecule. The blocking of these sulfhydryl residues by alkylation, however, had minimal influence on the functions of C9. On the other hand, reduction of C9 by 1 mM dithiothreitol (DTT) (6-fold molar excess over Cys residues) followed by alkylation resulted in a complete block of polymerization activity and a 50% loss of C9 hemolytic activity. In contrast, the ability of C9 to bind EAC1-8 remained largely unaffected. The loss of poly-C9 formation activity correlated with the alkylation of approx. 6 liberated sulfhydryl groups. Hemolytic activity was abolished by treatment with > 5 mM DTT which allowed the liberation of approximately 18 sulfhydryl groups. Most of the DTT-susceptible disulfides were within the C9a fragment (an N-terminal peptide derived by thrombin). Thus, three major functions of C9, EAC1-8 binding, polymerization, and hemolytic activity, are sustained by disulfide bond-dependent conformational motifs with different susceptibility to reducing reagents. The maintenance of the N-terminal C9a region is essential for polymerization, but not EAC1-8 binding activity of C9. Taken together, the results of the present study differentiate in molecular terms several of the functional portions of C9, and stress the significance of intra-chain disulfide linkages in maintaining the structural components necessary for the functions of C9.


Subject(s)
Complement C9/chemistry , Complement C9/physiology , Disulfides/chemistry , Animals , Dithiothreitol , Electrophoresis, Polyacrylamide Gel , Fluorescent Dyes , Humans , In Vitro Techniques , Maleimides , Mice , Oxidation-Reduction , Sheep , Structure-Activity Relationship
18.
Mol Immunol ; 30(12): 1097-106, 1993 Aug.
Article in English | MEDLINE | ID: mdl-8366860

ABSTRACT

Electron microscopy of specimens of C9 tilted through 90 degrees visualized this protein to be a globular ellipsoid with dimensions of 77 x 70 x 52 A. To check the congruence of this observation with physical properties of the molecule, hydrodynamic parameters for C9 were determined. From this work a frictional ratio of 1.32 was calculated. C9 was compared with several other proteins of similar frictional ratios whose tertiary structures are known. All examples found of such proteins whose frictional ratios were between 1.26 and 1.37 are either heart-shaped or globular ellipsoids, but none are prolate ellipsoids. By comparison the size and shape of C9 determined by electron microscopy are congruent with its hydrodynamic parameters. Both electron microscopy and physical measurements suggest that the length (110-120 A) of C9 determined by neutron and X-ray scattering experiments is an overestimate. The source of the discrepancy was identified by the demonstration that the high concns of C9 employed in neutron and X-ray scattering work lead to aggregation of the protein. Thus, investigations involving neutron and X-ray scattering were measuring polydisperse solutions of C9. The deduced value of the radius of gyration from that work (33-35 A) is now recognized as being statistical and significantly higher than the correct value of monomeric C9 (26 A), which was calculated from electron microscopy measurements. Also high-resolution electron microscopy clearly visualized poly(C9) to be a barrel-stave construct. These results suggest that monomeric C9 must undergo a major conformational alteration to extend by 55-70 A in order to self-associate laterally in order to fashion the cylindrical poly(C9).


Subject(s)
Complement C9/chemistry , Humans , Microscopy, Electron , Neutrons , Protein Binding , Protein Conformation , Scattering, Radiation , X-Rays
19.
Mol Immunol ; 33(7-8): 725-33, 1996.
Article in English | MEDLINE | ID: mdl-8760284

ABSTRACT

Lack of hemolytic activity of horse serum is an inherent property of horse C9. To understand the molecular reasons for this deficiency we have cloned C9 cDNA from a horse liver cDNA library and have sequenced the cDNA yielding the complete coding sequence for horse C9. Purification of C9 from horse plasma and microsequencing established the N-terminus of the mature protein and verified that the correct horse C9 cDNA clone had been isolated. The deduced amino acid sequence corresponds to a mature protein of 526 amino acids that is 77% identical to human C9. It has the same domain structure as human C9 and contains 22 cysteines and four invariant tryptophans. The few differences include the N-terminus, which is an unblocked glycine in horse C9 but pyroglutamine in human C9, and three potential N-glycosylation sites compared to two in human C9. The N-terminal difference is unimportant since microsequencing of bovine C9, which is strongly hemolytic, established that it also has an unblocked glycine identical to horse C9. There are no obvious structural differences apparent that could resolve the differences in hemolytic potency between the two molecules. Aside from a few conservative replacements, both C9 sequences are identical between positions 250 and 360. This region includes the membrane interaction domain in C9 and the postulated transmembrane segment that is thought to constitute the wall of a putative transmembrane pore and, therefore, should be required for cytotoxicity. In agreement with this prediction we have observed that, in contrast to the marked decrease in hemolytic activity, horse C9 is very efficient in killing a variety of Gram-negative bacteria. These results demonstrate that horse C9 is a structurally competent molecule with efficient cytotoxic activity. Its inability to lyse erythrocytes may be related to the action of control proteins on target cell membranes.


Subject(s)
Complement C9/isolation & purification , Cytotoxicity, Immunologic , Horses/immunology , Amino Acid Sequence , Animals , Biopolymers/genetics , Biopolymers/isolation & purification , Blood Bactericidal Activity , Cattle , Cloning, Molecular , Complement C9/chemistry , Complement C9/genetics , Epitopes/genetics , Epitopes/isolation & purification , Horses/blood , Horses/genetics , Humans , Molecular Sequence Data
20.
Mol Immunol ; 33(11-12): 925-32, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8960116

ABSTRACT

Complement components corresponding to mammalian C8 and C9 were isolated from carp (Cyprinus carpio) serum. Carp C8 (M(r) 146,000) proved to be a gamma-globulin composed of three polypeptide chains (alpha-chain, M(r) 62,000; beta-chain, M(r) 62,000; gamma-chain, M(r) 22,000). The alpha-chain was disulfide-linked to the gamma-chain and the beta-chain was non-covalently associated with the alpha-gamma chain, in fair agreement with mammalian C8. However, the N-terminal amino acid sequences of the three subunits showed no homology with those of human C8. Carp C9 was an alpha-globulin composed of a single polypeptide (M(r) 91,000) and the N-terminus was blocked. Carp serum depleted of C8 did not hemolyse either carp antibody-sensitized sheep erythrocytes or non-sensitized rabbit erythrocytes, while C9-depleted carp serum did not hemolyse the former, but did hemolyse the latter target cells, as in the case of C9-depleted human serum.


Subject(s)
Carps/immunology , Complement C8/chemistry , Complement C8/isolation & purification , Complement C9/chemistry , Complement C9/isolation & purification , Animals , Complement C8/deficiency , Complement C9/deficiency , Hemolysis/immunology
SELECTION OF CITATIONS
SEARCH DETAIL