Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Front Immunol ; 10: 2209, 2019.
Article in English | MEDLINE | ID: mdl-31572401

ABSTRACT

Complement split products (CSPs), such as the fragments C4d and C3d, which are generated as a consequence of complement regulatory processes, are established markers for disease activity in autoimmunity or antibody-mediated graft rejection. Since immunoglobulin-like transcript 4 (ILT4) was previously shown to interact with soluble CSPs, but not with CSPs covalently-bound to target surfaces following classical complement activation, the present study aimed to identify novel cellular receptors interacting with covalently-deposited CSPs. By applying an unbiased screening approach using a cDNA mammalian expression library generated from human monocyte-derived dendritic cells and probed with recombinant human C4d, we identified neuropilin-1 (NRP1) as a novel receptor for C4d, C3d, and iC3b. NRP1, a highly conserved type 1 transmembrane protein, plays important roles in the development of the nervous and cardiovascular system as well as in tumorigenesis through interaction with its established binding partners, such as vascular endothelial growth factor (VEGF) and semaphorin 3A (Sema3A). NRP1 is also expressed on immune cells and serves as a marker for murine Tregs. Although NRP1 contains domains homologous to ones found in some complement proteins, it has not been linked to the complement system. We demonstrate that binding of C4d to NRP1 expressing cells was dose-dependent and saturable, and had a KD value of 0.71 µM. Importantly, and in contrast to ILT4, NRP1 interacted with CSPs that were covalently bound to target surfaces in the course of complement activation, therefore representing a classical complement receptor. The binding site of CSPs was mapped to the b1 domain of the coagulation factor V/VIII homology domain of NRP1. Taken together, our results demonstrate a novel role for NRP1 as a receptor for CSPs deposited on surfaces during complement activation. Further work is required to elucidate the functional consequences of the NRP1-CSP interactions in immunity.


Subject(s)
Complement System Proteins/metabolism , Neuropilin-1/metabolism , Receptors, Complement/metabolism , Semaphorin-3A/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Line, Tumor , Complement Activation , Complement C3b/metabolism , Complement C3d/metabolism , Complement C4b/metabolism , Humans , Jurkat Cells , Mice , Peptide Fragments/metabolism , Protein Binding
2.
Cell Host Microbe ; 23(5): 644-652.e5, 2018 05 09.
Article in English | MEDLINE | ID: mdl-29746835

ABSTRACT

In physiological settings, the complement protein C3 is deposited on all bacteria, including invasive pathogens. However, because experimental host-bacteria systems typically use decomplemented serum to avoid the lytic action of complement, the impact of C3 coating on epithelial cell responses to invasive bacteria remains unexplored. Here, we demonstrate that following invasion, intracellular C3-positive Listeria monocytogenes is targeted by autophagy through a direct C3/ATG16L1 interaction, resulting in autophagy-dependent bacterial growth restriction. In contrast, Shigella flexneri and Salmonella Typhimurium escape autophagy-mediated growth restriction in part through the action of bacterial outer membrane proteases that cleave bound C3. Upon oral infection with Listeria, C3-deficient mice displayed defective clearance at the intestinal mucosa. Together, these results demonstrate an intracellular role of complement in triggering antibacterial autophagy and immunity against intracellular pathogens. Since C3 indiscriminately associates with foreign surfaces, the C3-ATG16L1 interaction may provide a universal mechanism of xenophagy initiation.


Subject(s)
Autophagy/drug effects , Autophagy/immunology , Bacteria/immunology , Carrier Proteins/immunology , Complement C3/immunology , Complement C3/pharmacology , Host-Pathogen Interactions/immunology , Animals , Autophagy-Related Proteins , Bacteria/pathogenicity , Bacterial Outer Membrane Proteins/immunology , Dysentery, Bacillary/immunology , Dysentery, Bacillary/microbiology , Epithelial Cells , Female , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Listeria monocytogenes/immunology , Listeria monocytogenes/pathogenicity , Listeriosis/immunology , Listeriosis/microbiology , Male , Mice , Mice, Inbred C57BL , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Shigella flexneri/immunology , Shigella flexneri/pathogenicity , THP-1 Cells
3.
Sci Rep ; 7: 45623, 2017 03 30.
Article in English | MEDLINE | ID: mdl-28358123

ABSTRACT

Degenerative disc disease (DDD) is associated with spinal pain often leading to long-term disability. However, the non-chondrodystrophic canine intervertebral disc is protected from the development of DDD, ostensibly due to its retention of notochordal cells (NC) in the nucleus pulposus (NP). In this study, we hypothesized that secretome analysis of the NC-rich NP will lead to the identification of key proteins that delay the onset of DDD. Using mass-spectrometry, we identified 303 proteins including components of TGFß- and Wnt-signaling, anti-angiogeneic factors and proteins that inhibit axonal ingrowth in the bioactive fractions of serum free, notochordal cell derived conditioned medium (NCCM). Ingenuity Pathway Analysis revealed TGFß1 and CTGF as major hubs in protein interaction networks. In vitro treatment with TGFß1 and CTGF promoted the synthesis of healthy extra-cellular matrix proteins, increased cell proliferation and reduced cell death in human degenerative disc NP cells. A single intra-discal injection of recombinant TGFß1 and CTGF proteins in a pre-clinical rat-tail disc injury model restored the NC and stem cell rich NP. In conclusion, we demonstrate the potential of TGFß1 and CTGF to mitigate the progression of disc degeneration and the potential use of these molecules in a molecular therapy to treat the degenerative disc.


Subject(s)
Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Degeneration/therapy , Notochord/metabolism , Nucleus Pulposus/metabolism , Animals , CCN Intercellular Signaling Proteins/metabolism , Cell Survival , Cells, Cultured , Connective Tissue Growth Factor/metabolism , Culture Media, Conditioned , Disease Models, Animal , Female , Humans , Intervertebral Disc Degeneration/pathology , Mass Spectrometry , Protein Interaction Maps , Rats, Wistar , Repressor Proteins/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism
4.
FASEB J ; 30(4): 1492-503, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26678451

ABSTRACT

Complement regulation leads to the generation of complement split products (CSPs) such as complement component (C)4d, a marker for disease activity in autoimmune syndromes or antibody-mediated allograft rejection. However, the physiologic role of C4d has been unknown. By screening murine thymoma BW5147 cells expressing a cDNA library generated from human monocyte-derived dendritic cells with recombinant human C4d, we identified Ig-like transcript (ILT)4 and ILT5v2 as cellular receptors for C4d. Both receptors, expressed on monocytes, macrophages, and dendritic cells, also interacted with the CSPs C3d, C4b, C3b, and iC3b. However, C4d did not bind to classic complement receptors (CRs). Interaction between cell surface-resident ILT4 and soluble monomeric C4d resulted in endocytosis of C4d. Surprisingly, binding of soluble ILT4 to C4d covalently immobilized to a cellular surface following classic complement activation could not be detected. Remarkably, C4d immobilized to a solid phaseviaits intrinsic thioester conferred a dose-dependent inhibition of TNF-α and IL-6 secretion in monocytes activatedviaFc-cross-linking of up to 50% as compared to baseline. Similarly, C4d conferred an attenuation of intracellular Ca(2+)flux in monocytes activatedviaFc-cross-linking. In conclusion, ILT4 represents a scavenger-type endocytotic CR for soluble monomeric C4d, whereas attenuation of monocyte activation by physiologically oriented C4d on a surface appears to be dependent on a yet to be identified C4d receptor.-Hofer, J., Forster, F., Isenman, D. E., Wahrmann, M., Leitner, J., Hölzl, M. A., Kovarik, J. K., Stockinger, H., Böhmig, G. A., Steinberger, P., Zlabinger, G. J. Ig-like transcript 4 as a cellular receptor for soluble complement fragment C4d.


Subject(s)
Complement C4b/metabolism , Membrane Glycoproteins/metabolism , Peptide Fragments/metabolism , Receptors, Complement/metabolism , Receptors, Immunologic/metabolism , Animals , Calcium/metabolism , Cell Line, Tumor , Complement C3b/metabolism , Complement C3d/metabolism , Dendritic Cells/metabolism , Endocytosis , Flow Cytometry , Humans , Immunoblotting , Interleukin-6/metabolism , Macrophages/metabolism , Mice , Monocytes/metabolism , Protein Binding , Tumor Necrosis Factor-alpha/metabolism
5.
Immunity ; 37(2): 199-207, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22921118

ABSTRACT

The complement system of innate immunity is important in regulating humoral immunity largely through the complement receptor CR2, which forms a coreceptor on B cells during antigen-induced activation. However, CR2 also retains antigens on follicular dendritic cells (FDCs). Display of antigen on FDCs is critical for clonal selection and affinity maturation of activated B cells. This review will discuss the role of complement in adaptive immunity in general with a focus on the interplay between CR2-associated antigen on B cells with CR2 expressed on FDCs. This latter interaction provides an opportunity for memory B cells to sample antigen over prolonged periods. The cocrystal structure of CR2 with its ligand C3d provides insight into how the complement system regulates access of antigen by B cells with implications for therapeutic manipulations to modulate aberrant B cell responses in the case of autoimmunity.


Subject(s)
B-Lymphocytes/immunology , Complement C3d/immunology , Dendritic Cells, Follicular/immunology , Immunity, Humoral/immunology , Receptors, Complement 3d/immunology , Animals , Antigen Presentation/immunology , Antigens/immunology , Antigens, CD19/immunology , Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , Complement Activation/immunology , Complement C3d/chemistry , Complement C3d/metabolism , Dendritic Cells, Follicular/metabolism , Humans , Immunity, Innate , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Receptors, Complement 3d/chemistry , Receptors, Complement 3d/metabolism , Tetraspanin 28/immunology
6.
Science ; 332(6029): 608-11, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21527715

ABSTRACT

The interaction of complement receptor 2 (CR2)--which is present on B cells and follicular dendritic cells--with its antigen-bound ligand C3d results in an enhanced antibody response, thus providing an important link between the innate and adaptive immune systems. Although a cocrystal structure of a complex between C3d and the ligand-binding domains of CR2 has been published, several aspects of this structure, including the position in C3d of the binding interface, remained controversial because of disagreement with biochemical data. We now report a cocrystal structure of a CR2(SCR1-2):C3d complex at 3.2 angstrom resolution in which the interaction interfaces differ markedly from the previously published structure and are consistent with the biochemical data. It is likely that, in the previous structure, the interaction was influenced by the presence of zinc acetate additive in the crystallization buffer, leading to a nonphysiological complex. Detailed knowledge of the binding interface now at hand gives the potential to exploit the interaction in vaccine design or in therapeutics directed against autoreactive B cells.


Subject(s)
Complement C3d/chemistry , Receptors, Complement 3d/chemistry , Binding Sites , Complement C3d/metabolism , Crystallization , Crystallography, X-Ray , Humans , Hydrogen Bonding , Ligands , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Receptors, Complement 3d/genetics , Receptors, Complement 3d/metabolism , Zinc Acetate
7.
Proc Natl Acad Sci U S A ; 108(7): 2897-902, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21285368

ABSTRACT

The alternative pathway of complement is important in innate immunity, attacking not only microbes but all unprotected biological surfaces through powerful amplification. It is unresolved how host and nonhost surfaces are distinguished at the molecular level, but key components are domains 19-20 of the complement regulator factor H (FH), which interact with host (i.e., nonactivator surface glycosaminoglycans or sialic acids) and the C3d part of C3b. Our structure of the FH19-20:C3d complex at 2.3-Å resolution shows that FH19-20 has two distinct binding sites, FH19 and FH20, for C3b. We show simultaneous binding of FH19 to C3b and FH20 to nonactivator surface glycosaminoglycans, and we show that both of these interactions are necessary for full binding of FH to C3b on nonactivator surfaces (i.e., for target discrimination). We also show that C3d could replace glycosaminoglycan binding to FH20, thus providing a feedback control for preventing excess C3b deposition and complement amplification. This explains the molecular basis of atypical hemolytic uremic syndrome, where mutations on the binding interfaces between FH19-20 and C3d or between FH20 and glycosaminoglycans lead to complement attack against host surfaces.


Subject(s)
Complement C3d/metabolism , Complement Factor H/metabolism , Complement Pathway, Alternative/immunology , Glycosaminoglycans/metabolism , Immunity, Innate/immunology , Analysis of Variance , Atypical Hemolytic Uremic Syndrome , Binding Sites , Chromatography, Affinity , Complement C3d/genetics , Complement C3d/immunology , Complement Factor H/genetics , Complement Factor H/immunology , Crystallization , Crystallography, X-Ray , DNA Primers/genetics , Escherichia coli , Glycosaminoglycans/genetics , Glycosaminoglycans/immunology , Hemolytic-Uremic Syndrome/immunology , Humans , Mutagenesis, Site-Directed , Pichia , Reverse Transcriptase Polymerase Chain Reaction , Surface Plasmon Resonance
8.
J Immunol ; 186(5): 3120-9, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21270401

ABSTRACT

The complement system plays an important role in eliminating invading pathogens. Activation of complement results in C3b deposition (opsonization), phagocytosis, anaphylatoxin (C3a, C5a) release, and consequently cell lysis. Moraxella catarrhalis is a human respiratory pathogen commonly found in children with otitis media and in adults with chronic obstructive pulmonary disease. The species has evolved multiple complement evasion strategies, which among others involves the ubiquitous surface protein (Usp) family consisting of UspA1, A2, and A2 hybrid. In the present study, we found that the ability of M. catarrhalis to bind C3 correlated with UspA expression and that C3 binding contributed to serum resistance in a large number of clinical isolates. Recombinantly expressed UspA1 and A2 inhibit both the alternative and classical pathways, C3b deposition, and C3a generation when bound to the C3 molecule. We also revealed that the M. catarrhalis UspA-binding domain on C3b was located to C3d and that the major bacterial C3d-binding domains were within UspA1(299-452) and UspA2(165-318). The interaction with C3 was not species specific since UspA-expressing M. catarrhalis also bound mouse C3 that resulted in inhibition of the alternative pathway of mouse complement. Taken together, the binding of C3 to UspAs is an efficient strategy of Moraxella to block the activation of complement and to inhibit C3a-mediated inflammation.


Subject(s)
Antigens, Bacterial/physiology , Antigens, Surface/physiology , Bacterial Outer Membrane Proteins/physiology , Complement C3d/metabolism , Immune Evasion/immunology , Moraxella catarrhalis/immunology , Adult , Animals , Antigens, Bacterial/metabolism , Antigens, Surface/metabolism , Bacterial Outer Membrane Proteins/biosynthesis , Bacterial Outer Membrane Proteins/metabolism , Child , Complement Activation/immunology , Complement C3a/antagonists & inhibitors , Complement C3a/physiology , Complement Inactivator Proteins/physiology , Complement Pathway, Alternative/immunology , Humans , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Mice , Moraxella catarrhalis/isolation & purification , Moraxella catarrhalis/pathogenicity , Moraxellaceae Infections/immunology , Moraxellaceae Infections/microbiology , Moraxellaceae Infections/pathology , Protein Binding/immunology , Rabbits , Sheep
9.
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
10.
J Biol Chem ; 284(23): 15650-8, 2009 Jun 05.
Article in English | MEDLINE | ID: mdl-19351878

ABSTRACT

Atypical hemolytic uremic syndrome (aHUS) is a thrombotic microangiopathy associated with mutations in complement proteins, most frequently in the main plasma alternative pathway regulator factor H (FH). The hotspot for the FH mutations is in domains 19-20 (FH19-20) that are indispensable for FH activity on C3b bound covalently to host cells. In aHUS, down-regulation of cell-bound C3b by FH is impaired, but it is not clear whether this is due to an altered FH binding to surface-bound C3b or to cell surface structures. To explore the molecular pathogenesis of aHUS we tested binding of 14 FH19-20 point mutants to C3b and its C3d fragment, mouse glomerular endothelial cells (mGEnC-1), and heparin. The cell binding correlated well, but not fully, with heparin binding and the cell binding site was overlapping but distinct from the C3b/C3d binding site that was shown to extend to domain 19. Our results show that aHUS-associated FH19-20 mutants have different combinations of three primary defects: impaired binding to C3b/C3d, impaired binding to the mGEnC-1 cells/heparin, and, as a novel observation, an enhanced mGEnC-1 cell or heparin binding. We propose a model of the molecular pathogenesis of aHUS where all three mechanisms lead eventually to impaired control of C3b on the endothelial cell surfaces. Based on the results with the aHUS patient mutants and the overlap in FH19-20 binding sites for mGEnC-1/heparin and C3b/C3d we conclude that binding of FH19-20 to C3b/C3d is essential for target discrimination by the alternative pathway.


Subject(s)
Complement Factor H/genetics , Complement Factor H/metabolism , Hemolytic-Uremic Syndrome/genetics , Amino Acid Substitution , Animals , Complement C3b/chemistry , Complement C3b/genetics , Complement C3b/metabolism , Complement C3d/chemistry , Complement C3d/genetics , Complement C3d/metabolism , Complement Factor H/chemistry , DNA Primers , Endothelial Cells/physiology , Humans , Inflammation/physiopathology , Kidney Glomerulus/physiology , Kinetics , Mice , Mutagenesis , Mutation , Point Mutation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
11.
Eur J Immunol ; 38(11): 3114-26, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18991288

ABSTRACT

Fibrinogen-like protein 2 (FGL2) is a multifunctional protein, which has been implicated in the pathogenesis of allograft and xenograft rejection. Previously, FGL2 was shown to inhibit maturation of BM-derived DC and T-cell proliferation. The mechanism of the immunosuppressive activity of FGL2 remains poorly elucidated. Here, we focus on identification of FGL2-specific receptor(s) and their ability to modulate APC activity and allograft survival. Using flow cytometry and surface plasmon resonance analysis, we show that FGL2 binds specifically to Fc gamma receptor (FcgammaR)IIB and FcgammaRIII receptors, which are expressed on the surface of APC, including B lymphocytes, macrophages and DC. Antibody to FcgammaRIIB and FcgammaRIII, or deficiency of these receptors, abrogated FGL2 binding. FGL2 inhibited the maturation of BMDC from FcgammaRIIB+/+ mice but not from FcgammaRIIB(-/-) mice and induced apoptosis in the FcgammaRIIB+ mouse B-cell line (A20) but not the A20IIA1.6 cell line that does not express FcgammaRIIB. Recombinant FGL2 infused into FcgammaRIIB+/+ (C57BL/6J, H-2b) mice but not FcgammaRIIB(-/-) mice inhibited rejection of fully mismatched BALB/cJ (H-2d) skin allografts. The identification of specific receptor binding has important implications for the pathogenesis of immune-mediated disease and suggests a potential for targeted FGL2 therapy.


Subject(s)
Fibrinogen/physiology , Immune Tolerance , Receptors, IgG/physiology , Animals , Antigen-Presenting Cells/immunology , Dendritic Cells/physiology , Female , Graft Survival , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Skin Transplantation/immunology , Transplantation, Homologous
12.
J Mol Biol ; 382(4): 910-9, 2008 Oct 17.
Article in English | MEDLINE | ID: mdl-18692506

ABSTRACT

The monoclonal antibody 2F5 neutralizes a broad range of human immunodeficiency virus-1 isolates via a conserved epitope on the viral glycoprotein gp41. The conformation of the principal epitope is a type I beta-turn centered on gp41 residues (664)DKW(666); in addition, binding studies indicate that residues N- and C-terminal to this core as well as structurally more distant parts of gp41 also contribute to the interaction. Ab2/3H6 is an anti-idiotypic antibody that inhibits the interaction between 2F5 and gp41 and as such, Ab2/3H6 may, in principle, possess a paratope that mimics the gp41 epitope. To establish the potential of Ab2/3H6 to serve as a guide for the design of vaccine components against human immunodeficiency virus, we investigated the crystal structure of the heterodimeric complex of Ab2/3H6 F(ab) and 2F5 F(ab)'. Ab2/3H6 F(ab) binds to 2F5 F(ab)' via a helix-like protrusion formed by residues (58(H))RYSPSLNTRL(67(H)) of the 2F5 F(ab)' variable domain and proximal to but not overlapping with the gp41 (664)DKW(666) epitope-binding pocket. This defines Ab2/3H6 as an anti-idiotypic antibody of the Ab2gamma class, i.e., an antigen-inhibitable idiotype that does not carry the internal image of the linear primary gp41 (662)ELDKWAS(668) epitope.


Subject(s)
Antibodies, Anti-Idiotypic/chemistry , Antibodies, Monoclonal/chemistry , HIV Antibodies/chemistry , HIV-1/immunology , Immunoglobulin Fab Fragments/chemistry , Amino Acid Sequence , Animals , Antibodies, Anti-Idiotypic/genetics , Antibodies, Anti-Idiotypic/immunology , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Binding Sites , Crystallography, X-Ray , Epitopes/chemistry , Epitopes/genetics , HIV Antibodies/genetics , HIV Antibodies/immunology , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/genetics , HIV Envelope Protein gp41/immunology , Humans , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Mice , Models, Molecular , Molecular Sequence Data , Neutralization Tests
13.
J Biol Chem ; 283(32): 22113-20, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18550524

ABSTRACT

Among the recently discovered Staphylococcus aureus immune evasion proteins, Sbi is unique in its ability to interact with components of both the adaptive and innate immune systems of the host. Sbi domains I and II (Sbi-I and Sbi-II) bind IgG. Sbi domain IV (residues 198-266) binds the central complement protein C3. When linked to Sbi-III, Sbi-IV induces a futile consumption of complement via alternative pathway activation, whereas isolated Sbi-IV specifically inhibits the alternative pathway without complement consumption. Here we have determined the three-dimensional structure of Sbi-IV by NMR spectroscopy, showing that Sbi-IV adopts a three-helix bundle fold similar to those of the S. aureus complement inhibitors Efb-C, Ehp, and SCIN. The (1)H-(15)N HSQC spectrum of Sbi-III indicates that this domain, essential for futile complement consumption, is natively unfolded, at least when isolated from the rest of Sbi. Sbi-IV and Sbi-III-IV both bind C3dg with 1:1 stoichiometry and submicromolar affinity. Despite low overall sequence identity, Sbi possesses the same residues as Efb at two positions essential for Efb-C binding to C3d. Mutation to alanine of either of these residues, Arg-231 and Asn-238, abolishes both Sbi-IV binding to C3dg and Sbi-IV alternative pathway inhibition. The almost complete conservation of Sbi-III and Sbi-IV amino acid sequences across more than 30 strains isolated from human and animal hosts indicates that the unique mechanism of Sbi in complement system subversion is a feature of infections of both humans and economically important animals.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Models, Molecular , Staphylococcus aureus/chemistry , Staphylococcus aureus/metabolism , Amino Acid Sequence , Bacterial Proteins/immunology , Binding Sites , Carrier Proteins/immunology , Complement Inactivator Proteins/chemistry , Complement Inactivator Proteins/metabolism , Complement Pathway, Alternative , Humans , Molecular Sequence Data , Mutation , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Folding , Protein Structure, Tertiary , Sequence Alignment , Staphylococcus aureus/immunology , Structure-Activity Relationship
14.
J Biol Chem ; 283(25): 17579-93, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18434316

ABSTRACT

Staphylococcal immunoglobulin-binding protein, Sbi, is a 436-residue protein produced by many strains of Staphylococcus aureus. It was previously characterized as being cell surface-associated and having binding capacity for human IgG and beta(2)-glycoprotein I. Here we show using small angle x-ray scattering that the proposed extracellular region of Sbi (Sbi-E) is an elongated molecule consisting of four globular domains, two immunoglobulin-binding domains (I and II) and two novel domains (III and IV). We further show that together domains III and IV (Sbi-III-IV), as well as domain IV on its own (Sbi-IV), bind complement component C3 via contacts involving both the C3dg fragment and the C3a anaphylatoxin domain. Preincubation of human serum with either Sbi-E or Sbi-III-IV is inhibitory to all complement pathways, whereas domain IV specifically inhibits the alternative pathway. Monitoring C3 activation in serum incubated with Sbi fragments reveals that Sbi-E and Sbi-III-IV both activate the alternative pathway, leading to consumption of C3. By contrast, inhibition of this pathway by Sbi-IV does not involve C3 consumption. The observation that Sbi-E activates the alternative pathway is counterintuitive to intact Sbi being cell wall-associated, as recruiting complement to the surface of S. aureus would be deleterious to the bacterium. Upon re-examination of this issue, we found that Sbi was not associated with the cell wall fraction, but rather was found in the growth medium, consistent with it being an excreted protein. As such, our data suggest that Sbi helps mediate bacterial evasion of complement via a novel mechanism, namely futile fluid-phase consumption.


Subject(s)
Bacterial Proteins/chemistry , Carrier Proteins/chemistry , Complement C3/chemistry , Gene Expression Regulation, Bacterial , Staphylococcus aureus/metabolism , Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Cell Wall/metabolism , Cloning, Molecular , Humans , Immunoglobulin G/chemistry , Models, Biological , Models, Molecular , Models, Statistical , Protein Binding , Protein Structure, Tertiary , Receptors, Complement 3d/chemistry , Surface Plasmon Resonance
15.
Biochemistry ; 45(27): 8378-92, 2006 Jul 11.
Article in English | MEDLINE | ID: mdl-16819837

ABSTRACT

C4b-binding protein (C4BP) is a multimeric serum protein that is a potent regulator of the classical and lectin complement pathways. The binding site for C4b has been localized to complement control protein (CCP) domains 1-3 of the C4BP alpha-chain and, in particular, to a cluster of positively charged amino acids predicted to be at the interface between CCP 1 and CCP 2. To determine the regions of C4b contributing to C4BP binding, we have examined via surface plasmon resonance technology the binding of the C4c and C4dg subfragments of C4b to C4BP. At half-physiologic ionic strength, specific and saturable binding was observed for both C4c and C4dg. C4c exhibited much greater ionic strength sensitivity in its binding than did C4dg. Analysis of the effect on binding of the subfragments to various C4b-binding-defective C4BP mutants, together with cross-competition experiments, suggests that the subsites in C4BP for C4c and C4dg are adjacent, but distinct. Additionally, we observed synergy in subsite filling such that the presence of C4dg enhanced the extent of C4c binding over its basal level, and vice versa. The enhanced binding of C4c in the presence of C4dg was not due to an increase in affinity but rather reflected a 2-3-fold increase in the number of sites capable of binding C4c. This suggests the existence of a conformational equilibrium between high- and low-affinity states in the C4c binding subsite within each C4BP subunit, an equilibrium which is shifted in favor of the high-affinity state by the filling of the C4dg subsite.


Subject(s)
Complement C4/chemistry , Complement C4b-Binding Protein/chemistry , Amino Acid Sequence , Binding Sites , Complement C4b-Binding Protein/genetics , Heparin/chemistry , Humans , Ligands , Mutation , Osmolar Concentration , Peptide Fragments/chemistry , Surface Plasmon Resonance
16.
J Biol Chem ; 281(18): 12767-75, 2006 May 05.
Article in English | MEDLINE | ID: mdl-16524885

ABSTRACT

MARCO is a class A scavenger receptor capable of binding both gram-negative and -positive bacteria. Using the surface plasmon resonance technique, we show here that a recombinant, soluble form of MARCO, sMARCO, binds the major gram-negative and -positive bacterial surface components, lipopolysaccharide and lipoteichoic acid. Yet, the interaction of these two polyanions with sMARCO is of much lower affinity than that of polyinosinic acid, a polyanionic inhibitor of bacterial binding to MARCO. To further elucidate the ligand-binding functions of MARCO, we performed a phage display screen with sMARCO. The screening resulted in the enrichment of only a handful of phage clones. Contrary to expectations, no polyanionic peptides, but only those with a predominantly hydrophobic nature, were enriched. One peptide, VRWGSFAAWL, was displayed on two-thirds of the phages recovered after four rounds of screening. The VRWGSFAAWL phage-sMARCO interaction had significantly slower dissociation kinetics than that between sMARCO and lipopolysaccharide or lipoteichoic acid. Further work with this phage, and the second most enriched phage, displaying the peptide RLNWAWWLSY, demonstrated that both peptides bind to the SRCR domain of MARCO, and that they probably bind to the same site. Data base searches suggested that the VRWGSFAAWL peptide represents complement component C4, but we could not convincingly confirm this suggestion. A study with chimeric scavenger receptors indicated that even minor sequence changes in the MARCO scavenger receptor cysteine-rich (SRCR) domain can have profound effects on the binding of the prototypic scavenger receptor ligand, acetylated low density lipoprotein. As shown by differential binding of glutathione S-transferase-VR-WGSFAAWL, these differences were very likely due to conformational changes. These findings led to experiments that demonstrated a crucial role of the SRCR domain for acetylated low density lipoprotein binding in MARCO. Thus, our results strengthen the notion that the SRCR domain is the major ligand-binding domain in MARCO. Furthermore, they suggest that the domain may contain multiple ligand-binding sites.


Subject(s)
Lipoproteins, LDL/chemistry , Receptors, Immunologic/physiology , Receptors, Scavenger/chemistry , Acetylation , Animals , Binding Sites , CHO Cells , Cricetinae , Ligands , Mice , Peptide Library , Protein Binding , Protein Structure, Tertiary , Rats , Receptors, Immunologic/metabolism
17.
J Immunol ; 172(3): 1670-80, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14734749

ABSTRACT

Several previous reports concluded that the C4b fragment of human C4A (C4Ab) binds with higher affinity to CR1 than does C4Bb. Because the isotypic residues, (1101)PCPVLD and (1101)LSPVIH in C4A and C4B, respectively, are located within the C4d region, one may have expected a direct binding contribution of C4d to the interaction with CR1. However, using surface plasmon resonance as our analytical tool, with soluble rCR1 immobilized on the biosensor chip, we failed to detect significant binding of C4d of either isotype. By contrast, binding of C4c was readily detectable. C4A and C4B, purified from plasma lacking one of the isotypes, were Cs converted to C4Ab and C4Bb. Spontaneously formed disulfide-linked dimers were separated from monomers and higher oligomers by sequential chromatographic steps. The binding sensorgrams of C4Ab and C4Bb monomers as analytes reached steady state plateaus, and these equilibrium data yielded essentially superimposable saturation curves that were well fit by a one-site binding model. Although a two-site model was required to fit the equilibrium-binding data for the dimeric forms of C4b, once again there was little difference in the K(D) values obtained for each isotype. Independent verification of our surface plasmon resonance studies came from ELISA-based inhibition experiments in which monomers of C4Ab and C4Bb were equipotent in inhibiting the binding of soluble CR1 to plate-bound C4b. Although divergent from previous reports, our results are consistent with recent C4Ad structural data that raised serious doubts about there being a conformational basis for the previously reported isotypic differences in the C4b-CR1 interaction.


Subject(s)
Complement C4a/metabolism , Complement C4b/metabolism , Receptors, Complement 3b/metabolism , Binding, Competitive/immunology , Complement C4/metabolism , Complement C4a/antagonists & inhibitors , Complement C4b/antagonists & inhibitors , Complement Inactivator Proteins/metabolism , Dimerization , Enzyme-Linked Immunosorbent Assay , Humans , Peptide Fragments/metabolism , Protein Binding/immunology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Receptors, Complement 3b/antagonists & inhibitors , Recombinant Proteins/metabolism , Solubility , Surface Plasmon Resonance
18.
J Mol Biol ; 322(5): 1103-15, 2002 Oct 04.
Article in English | MEDLINE | ID: mdl-12367531

ABSTRACT

C4 fulfills a vital role in the propagation of the classical and lectin pathways of the complement system. Although there are no reports to date of a C4 functional activity that is mediated solely by the C4d region, evidence clearly points to it having a vital role in a number of the properties of native C4 and its major activation fragment, C4b. Contained within the C4d region are the thioester-forming residues, the four isotype-specific residues controlling the C4A/C4B transacylation preferences, a binding site for nascent C3b important in assembling the classical pathway C5 convertase and determinants for the Chido/Rodgers (Ch/Rg) blood group antigens. In view of its functional importance, we undertook to determine the three-dimensional structure of C4d by X-ray crystallography. Here we report the 2.3A resolution structure of C4Ad, the C4d fragment derived from the human C4A isotype. Although the approximately 30% sequence identity between C4Ad and the corresponding fragment of C3 might be expected to establish a general fold similarity between the two molecules, C4Ad in fact displays a fold that is essentially superimposable on the structure of C3d. By contrast, the electrostatic characteristics of the various faces of the C4Ad molecule show marked differences from the corresponding faces of C3d, likely reflecting the differences in function between C3 and C4. Residues previously predicted to form the major Ch/Rg epitopes were proximately located and accessible on the concave surface of C4Ad. In addition to providing further insights on the current models for the covalent binding reaction, the C4Ad structure allows one to rationalize why C4d is not a ligand for complement receptor 2. Finally the structure allows for the visualization of the face of the molecule containing the binding site for C3b utilized in the assembly of classical pathway C5 convertase.


Subject(s)
Complement C4/chemistry , Peptide Fragments/chemistry , Protein Structure, Tertiary , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Structure, Secondary , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...