Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
2.
Methods Mol Biol ; 1100: 43-60, 2014.
Article in English | MEDLINE | ID: mdl-24218249

ABSTRACT

C1r and C1s are the proteases responsible for the activation and proteolytic activity of the C1 complex of the classical complement pathway, respectively. They are assembled into a Ca(2+)-dependent C1s-C1r-C1r-C1s tetramer which in turn associates with the recognition protein C1q. The C1 complex circulates in serum as a zymogen and is activated upon binding of C1q to appropriate targets, such as antigen-antibody complexes. This property is used for the purification of C1r and C1s from human serum after binding of C1 to insoluble immune complexes. Disruption of the bound C1 complex by EDTA releases C1r and C1s which are further separated by ion-exchange chromatography; both proteins can be reassembled in the presence of calcium ions and the reconstituted tetramer isolated by gel filtration. In this chapter, we describe the purification of the activated and proenzyme forms of C1r and C1s and of the proenzyme C1s-C1r-C1r-C1s tetramer as well as methods for their biochemical and functional characterization. The production of recombinant C1s and of the proenzyme tetramer in a baculovirus-insect cell system, and their purification by affinity chromatography is also presented.


Subject(s)
Complement C1r/immunology , Complement C1s/immunology , Complement Pathway, Classical , Animals , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/isolation & purification , Antigen-Antibody Complex/metabolism , Complement C1r/isolation & purification , Complement C1s/isolation & purification , Gene Expression , Humans , Protein Binding/immunology , Protein Multimerization , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Sf9 Cells , Solubility
3.
PLoS One ; 8(7): e67962, 2013.
Article in English | MEDLINE | ID: mdl-23861840

ABSTRACT

Mannan-binding lectin (MBL), ficolins and collectin-11 are known to associate with three homologous modular proteases, the MBL-Associated Serine Proteases (MASPs). The crystal structures of the catalytic domains of MASP-1 and MASP-2 have been solved, but the structure of the corresponding domain of MASP-3 remains unknown. A link between mutations in the MASP1/3 gene and the rare autosomal recessive 3MC (Mingarelli, Malpuech, Michels and Carnevale,) syndrome, characterized by various developmental disorders, was discovered recently, revealing an unexpected important role of MASP-3 in early developmental processes. To gain a first insight into the enzymatic and structural properties of MASP-3, a recombinant form of its serine protease (SP) domain was produced and characterized. The amidolytic activity of this domain on fluorescent peptidyl-aminomethylcoumarin substrates was shown to be considerably lower than that of other members of the C1r/C1s/MASP family. The E. coli protease inhibitor ecotin bound to the SP domains of MASP-3 and MASP-2, whereas no significant interaction was detected with MASP-1, C1r and C1s. A tetrameric complex comprising an ecotin dimer and two MASP-3 SP domains was isolated and its crystal structure was solved and refined to 3.2 Å. Analysis of the ecotin/MASP-3 interfaces allows a better understanding of the differential reactivity of the C1r/C1s/MASP protease family members towards ecotin, and comparison of the MASP-3 SP domain structure with those of other trypsin-like proteases yields novel hypotheses accounting for its zymogen-like properties in vitro.


Subject(s)
Escherichia coli Proteins/chemistry , Mannose-Binding Protein-Associated Serine Proteases/chemistry , Molecular Docking Simulation , Periplasmic Proteins/chemistry , Binding Sites , Catalytic Domain , Complement C1r/chemistry , Complement C1s/chemistry , Coumarins , Crystallography, X-Ray , Enzyme Assays , Fluorescent Dyes , Humans , Mannose-Binding Protein-Associated Serine Proteases/genetics , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Substrate Specificity , Thrombin/chemistry , Trypsin/chemistry
4.
Proc Natl Acad Sci U S A ; 110(21): 8650-5, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23650384

ABSTRACT

Complement C1q is a hexameric molecule assembled from 18 polypeptide chains of three different types encoded by three genes. This versatile recognition protein senses a wide variety of immune and nonimmune ligands, including pathogens and altered self components, and triggers the classical complement pathway through activation of its associated proteases C1r and C1s. We report a method for expression of recombinant full-length human C1q involving stable transfection of HEK 293-F mammalian cells and fusion of an affinity tag to the C-terminal end of the C chain. The resulting recombinant (r) C1q molecule is similar to serum C1q as judged from biochemical and structural analyses and exhibits the characteristic shape of a bunch of flowers. Analysis of its interaction properties by surface plasmon resonance shows that rC1q retains the ability of serum C1q to associate with the C1s-C1r-C1r-C1s tetramer, to recognize physiological C1q ligands such as IgG and pentraxin 3, and to trigger C1r and C1s activation. Functional analysis of rC1q variants carrying mutations of LysA59, LysB61, and/or LysC58, in the collagen-like stems, demonstrates that LysB61 and LysC58 each play a key role in the interaction with C1s-C1r-C1r-C1s, with LysA59 being involved to a lesser degree. We propose that LysB61 and LysC58 both form salt bridges with outer acidic Ca(2+) ligands of the C1r and C1s CUB (complement C1r/C1s, Uegf, bone morphogenetic protein) domains. The expression method reported here opens the way for deciphering the molecular basis of the unusual binding versatility of C1q by mapping the residues involved in the sensing of its targets and the binding of its receptors.


Subject(s)
Complement Activation/physiology , Complement C1q/metabolism , Complement C1r/metabolism , Complement C1s/metabolism , Amino Acid Substitution , Binding Sites , C-Reactive Protein/chemistry , C-Reactive Protein/metabolism , Calcium/metabolism , Complement C1q/chemistry , Complement C1q/genetics , Complement C1r/chemistry , Complement C1r/genetics , Complement C1s/chemistry , Complement C1s/genetics , Gene Expression , HEK293 Cells , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Mutation, Missense , Protein Binding/physiology , Protein Structure, Quaternary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serum Amyloid P-Component/chemistry , Serum Amyloid P-Component/metabolism , Surface Plasmon Resonance
5.
Biochem J ; 439(2): 185-93, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21954942

ABSTRACT

CUB domains are 110-residue protein motifs exhibiting a ß-sandwich fold and mediating protein-protein interactions in various extracellular proteins. Recent X-ray structural and mutagenesis studies have led to the identification of a particular CUB domain subset, cbCUB (Ca(2+)-binding CUB domain). Unlike other CUB domains, these harbour a homologous Ca(2+)-binding site that underlies a conserved binding site mediating ionic interaction between two of the three conserved acidic Ca(2+) ligands and a basic (lysine or arginine) residue of a protein ligand, similar to the interactions mediated by the low-density lipoprotein receptor family. cbCUB-mediated protein-ligand interactions usually involve multipoint attachment through several cbCUBs, resulting in high-affinity binding through avidity, despite the low affinity of individual interactions. The aim of the present review is to summarize our current knowledge about the structure and functions of cbCUBs, which represent the majority of the known CUB repertoire and are involved in a variety of major biological functions, including immunity and development, as well as in various cancer types. Examples discussed in the present review include a wide range of soluble and membrane-associated human proteins, as well as some archaeal and invertebrate proteins. The fact that these otherwise unrelated proteins share a common Ca(2+)-dependent ligand-binding ability suggests a mechanism inherited from very primitive ancestors. The information provided in the present review should stimulate further investigations on the crucial interactions mediated by cbCUB-containing proteins.


Subject(s)
Amino Acid Motifs , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Amino Acid Sequence , Ligands , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid
6.
J Lipids ; 2011: 376092, 2011.
Article in English | MEDLINE | ID: mdl-21490800

ABSTRACT

Several studies suggest that the complement system is involved in atherogenesis. To further investigate this question, we have studied the ability of native and modified forms of LDL to bind and activate C1, the complex protease that triggers the classical pathway of complement. Unlike native LDL, oxidized (oxLDL) and enzymatically modified (E-LDL) derivatives were both recognized by the C1q subunit of C1, but only E-LDL particles, obtained by sequential treatment with a protease and then with cholesterol esterase, had the ability to trigger C1 activation. Further investigations revealed that C1q recognizes a lipid component of E-LDL. Several approaches, including reconstitution of model lipid vesicles, cosedimentation, and electron microscopy analyses, provided evidence that C1 binding to E-LDL particles is mediated by the C1q globular domain, which senses unesterified fatty acids generated by cholesterol esterase. The potential implications of these findings in atherogenesis are discussed.

7.
J Immunol ; 186(10): 5815-22, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21490156

ABSTRACT

Ficolins and pentraxins are soluble oligomeric pattern-recognition molecules that sense danger signals from pathogens and altered self-cells and might act synergistically in innate immune defense and maintenance of immune tolerance. The interaction of M-ficolin with the long pentraxin pentraxin 3 (PTX3) has been characterized using surface plasmon resonance spectroscopy and electron microscopy. M-ficolin was shown to bind PTX3 with high affinity in the presence of calcium ions. The interaction was abolished in the presence of EDTA and inhibited by N-acetyl-D-glucosamine, indicating involvement of the fibrinogen-like domain of M-ficolin. Removal of sialic acid from the single N-linked carbohydrate of the C-terminal domain of PTX3 abolished the interaction. Likewise, an M-ficolin mutant with impaired sialic acid-binding ability did not interact with PTX3. Interaction was also impaired when using the isolated recognition domain of M-ficolin or the monomeric C-terminal domain of PTX3, indicating requirement for oligomerization of both proteins. Electron microscopy analysis of the M-ficolin-PTX3 complexes revealed that the M-ficolin tetramer bound up to four PTX3 molecules. From a functional point of view, immobilized PTX3 was able to trigger M-ficolin-dependent activation of the lectin complement pathway. These data indicate that interaction of M-ficolin with PTX3 arises from its ability to bind sialylated ligands and thus differs from the binding to the short pentraxin C-reactive protein and from the binding of L-ficolin to PTX3. The M-ficolin-PTX3 interaction described in this study represents a novel case of cross-talk between soluble pattern-recognition molecules, lending further credit to the integrated view of humoral innate immunity that emerged recently.


Subject(s)
C-Reactive Protein/metabolism , Lectins/metabolism , Serum Amyloid P-Component/metabolism , Signal Transduction , Acetylglucosamine/metabolism , C-Reactive Protein/chemistry , Calcium/chemistry , Humans , Immune Tolerance , Immunity, Humoral , Lectins/chemistry , Ligands , Microscopy, Electron , Mutant Proteins/chemistry , Mutant Proteins/metabolism , N-Acetylneuraminic Acid/chemistry , Protein Binding , Protein Structure, Tertiary , Serum Amyloid P-Component/chemistry , Surface Plasmon Resonance , Ficolins
8.
J Mol Biol ; 408(2): 277-90, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21352829

ABSTRACT

Both C1q and calreticulin (CRT) are involved in the recognition of apoptotic cells. CRT was initially characterized as a receptor for the C1q collagen-like fragment (CLF), whereas C1q was shown to bind apoptotic cells through its globular region (GR). Using purified CRT and recombinant CRT domains, we now provide unambiguous experimental evidence that, in addition to its CLF, the C1q GR also binds CRT and that both types of interactions are mediated by the CRT globular domain. Surface plasmon resonance analyses revealed that the C1q CLF and GR domains each bind individually to immobilized CRT and its globular domain with K(D) values of (2.6-8.3) × 10(-7) M. Further evidence that CRT binds to the C1q GR was obtained by electron microscopy. The role of CRT in the recognition of apoptotic HeLa cells by C1q was analyzed. The C1q GR partially colocalized with CRT on the surface of early apoptotic cells, and siRNA (small interfering RNA)-induced CRT deficiency resulted in increased apoptotic cell binding to C1q. The interaction between CRT and phosphatidylserine (PS), a known C1q ligand on apoptotic cells, was also investigated. The polar head of PS was shown to bind to CRT with a 10-fold higher affinity (K(D)=1.5 × 10(-5) M) than that determined for C1q, and, accordingly, the C1q GR-PS interaction was impaired in the presence of CRT. Together, these observations indicate that CRT, C1q, and PS are all closely involved in the uptake of apoptotic cells and strongly suggest a combinatorial role of these three molecules in the recognition step.


Subject(s)
Apoptosis , Calreticulin/metabolism , Cell Communication , Complement C1q/metabolism , Phosphatidylserines/metabolism , Blotting, Western , Calreticulin/antagonists & inhibitors , Calreticulin/genetics , Flow Cytometry , HeLa Cells , Humans , Membrane Proteins/metabolism , Phagocytosis , Protein Binding , RNA, Small Interfering/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Surface Plasmon Resonance
9.
PLoS One ; 6(3): e17886, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21423620

ABSTRACT

In the endoplasmic reticulum, calreticulin acts as a chaperone and a Ca(2+)-signalling protein. At the cell surface, it mediates numerous important biological effects. The crystal structure of the human calreticulin globular domain was solved at 1.55 Å resolution. Interactions of the flexible N-terminal extension with the edge of the lectin site are consistently observed, revealing a hitherto unidentified peptide-binding site. A calreticulin molecular zipper, observed in all crystal lattices, could further extend this site by creating a binding cavity lined by hydrophobic residues. These data thus provide a first structural insight into the lectin-independent binding properties of calreticulin and suggest new working hypotheses, including that of a multi-molecular mechanism.


Subject(s)
Calreticulin/chemistry , Calreticulin/metabolism , Peptides/metabolism , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Humans , Lectins/metabolism , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemistry , Peptides/chemistry , Protein Binding , Protein Structure, Tertiary , Surface Properties
10.
ACS Nano ; 5(2): 730-7, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-21214219

ABSTRACT

The classical pathway of complement is an essential component of the human innate immune system involved in the defense against pathogens as well as in the clearance of altered self-components. Activation of this pathway is triggered by C1, a multimolecular complex comprising a recognition protein C1q associated with a catalytic subunit C1s-C1r-C1r-C1s. We report here the direct observation of organized binding of C1 components C1q and C1s-C1r-C1r-C1s on carbon nanotubes, an ubiquitous component in nanotechnology research. Electron microscopy imaging showed individual multiwalled carbon nanotubes with protein molecules organized along the length of the sidewalls, often over 1 µm long. Less well-organized protein attachment was also observed on double-walled carbon nanotubes. Protein-solubilized nanotubes continued to attract protein molecules after their surface was fully covered. Despite the C1q binding properties, none of the nanotubes activated the C1 complex. We discuss these results on the adsorption mechanisms of macromolecules on carbon nanotubes and the possibility of using carbon nanotubes for structural studies of macromolecules. Importantly, the observations suggest that carbon nanotubes may interfere with the human immune system when entering the bloodstream. Our results raise caution in the applications of carbon nanotubes in biomedicine but may also open possibilities of novel applications concerning the many biochemical processes involving the versatile C1 macromolecule.


Subject(s)
Complement C1/chemistry , Immunity, Innate , Nanotubes, Carbon/chemistry , Complement C1/immunology , Complement C1/metabolism , Crystallization , Humans , Protein Multimerization , Protein Structure, Quaternary , Protein Subunits/chemistry , Protein Subunits/immunology , Protein Subunits/metabolism , Surface Properties
11.
Front Immunol ; 2: 92, 2011.
Article in English | MEDLINE | ID: mdl-22566881

ABSTRACT

C1q, the ligand-binding unit of the C1 complex of complement, is a pattern recognition molecule with the unique ability to sense an amazing variety of targets, including a number of altered structures from self, such as apoptotic cells. The three-dimensional structure of its C-terminal globular domain, responsible for its recognition function, has been solved by X-ray crystallography, revealing a tightly packed heterotrimeric assembly with marked differences in the surface patterns of the subunits, and yielding insights into its versatile binding properties. In conjunction with other approaches, this same technique has been used recently to decipher the mechanisms that allow this domain to interact with various non-immune self ligands, including molecules known to provide eat-me signals on apoptotic cells, such as phosphatidylserine and DNA. These investigations provide evidence for a common binding area for these ligands located in subunit C of the C1q globular domain, and suggest that ligand recognition through this area down-regulates C1 activation, hence contributing to the control of the inflammatory reaction. The purpose of this article is to give an overview of these advances which represent a first step toward understanding the recognition mechanisms of C1q and their biological implications.

12.
Biochemistry ; 49(39): 8608-17, 2010 Oct 05.
Article in English | MEDLINE | ID: mdl-20825197

ABSTRACT

The α and ß subunits of the human mitochondrial trifunctional protein (TFP), the multienzyme complex involved in fatty acid ß-oxidation, were coexpressed in Escherichia coli and purified to homogeneity by nickel affinity chromatography. The resulting α/His-ß construct was analyzed by gel filtration, sedimentation velocity, and electron microscopy, indicating a predominance of α(2)ß(2) and α(4)ß(4) complexes, with higher order oligomers. Electron microscopy indicated that the elementary species α(2)ß(2) had overall structural similarity with its bacterial homologue. As shown by cosedimentation and surface plasmon resonance analyses, recombinant TFP interacted strongly with cardiolipin and phosphatidylcholine, suggesting that the natural complex associates with the inner mitochondrial membrane through direct interactions with phospholipids. Recombinant TFP displayed 2-enoyl-CoA hydratase (ECH), l-3-hydroxyacyl-CoA dehydrogenase (HACD), and 3-ketoacyl-CoA thiolase (KACT) activities, and ECH and HACD each reached equilibrium when the downstream enzymes (HACD and KACT, respectively) were made inactive, indicating feed-back inhibition. The KACT activity was optimal at pH 9.5, sensitive to ionic strength, and inhibited at concentrations of its substrate 3-ketohexadecanoyl-CoA >5 µM. Its kinetic constants (k(cat) = 169 s(-1), K(m) = 4 µM) were consistent with those determined previously on a purified porcine TFP preparation. Using different assays, trimetazidine, an efficient antiaginal agent, had no significant inhibitory effect on any of the three enzymatic activities of the recombinant TFP preparation, in contrast with other reports. This study provides the first detailed structural and functional characterization of a recombinant human TFP preparation and opens the way to in-depth analyses through site-directed mutagenesis.


Subject(s)
Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Acetyl-CoA C-Acyltransferase/metabolism , Enoyl-CoA Hydratase/metabolism , Escherichia coli/genetics , Gene Expression , Humans , Mitochondrial Trifunctional Protein , Multienzyme Complexes/genetics , Multienzyme Complexes/isolation & purification , Phospholipids/metabolism , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/isolation & purification , Protein Subunits/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Deletion
13.
FEBS J ; 277(17): 3526-37, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20716178

ABSTRACT

C1q-mediated removal of immune complexes and apoptotic cells plays an important role in tissue homeostasis and the prevention of autoimmune conditions. It has been suggested that C1q mediates phagocytosis of apoptotic cells through a receptor complex assembled from CD91 (alpha-2- macroglobulin receptor, or low-density lipoprotein receptor-related protein) and calreticulin, with CD91 being the transmembrane part and calreticulin acting as the C1q-binding molecule. In the present study, we observe that C1q binds cells from a CD91 expressing monocytic cell line as well as monocytes from human blood. C1q binding to monocytes was shown to be correlated with CD91 expression and could be inhibited by the CD91 chaperone, receptor-associated protein. We also report data showing a direct interaction between CD91 and C1q. The interaction was investigated using various protein interaction assays. A direct interaction between purified C1q and CD91 was observed both by ELISA and a surface plasmon resonance assay, with either C1q or CD91 immobilized. The interaction showed characteristics of specificity because it was time-dependent, saturable and could be inhibited by known ligands of both CD91 and C1q. The results obtained show for the first time that CD91 recognizes C1q directly. On the basis of these findings, we propose that CD91 is a receptor for C1q and that this multifunctional scavenger receptor uses a subset of its ligand-binding sites for clearance of C1q and C1q bound material.


Subject(s)
Antigens, CD/metabolism , Complement C1q/metabolism , Antigens, CD/immunology , Calreticulin/metabolism , Complement C1q/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Ligands , Low Density Lipoprotein Receptor-Related Protein-1 , Protein Binding , Surface Plasmon Resonance , Time Factors
14.
J Biol Chem ; 285(42): 32251-63, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20592021

ABSTRACT

C1, the complex that triggers the classic pathway of complement, is a 790-kDa assembly resulting from association of a recognition protein C1q with a Ca(2+)-dependent tetramer comprising two copies of the proteases C1r and C1s. Early structural investigations have shown that the extended C1s-C1r-C1r-C1s tetramer folds into a compact conformation in C1. Recent site-directed mutagenesis studies have identified the C1q-binding sites in C1r and C1s and led to a three-dimensional model of the C1 complex (Bally, I., Rossi, V., Lunardi, T., Thielens, N. M., Gaboriaud, C., and Arlaud, G. J. (2009) J. Biol. Chem. 284, 19340-19348). In this study, we have used a mass spectrometry-based strategy involving a label-free semi-quantitative analysis of protein samples to gain new structural insights into C1 assembly. Using a stable chemical modification, we have compared the accessibility of the lysine residues in the isolated tetramer and in C1. The labeling data account for 51 of the 73 lysine residues of C1r and C1s. They strongly support the hypothesis that both C1s CUB(1)-EGF-CUB(2) interaction domains, which are distant in the free tetramer, associate with each other in the C1 complex. This analysis also provides the first experimental evidence that, in the proenzyme form of C1, the C1s serine protease domain is partly positioned inside the C1q cone and yields precise information about its orientation in the complex. These results provide further structural insights into the architecture of the C1 complex, allowing significant improvement of our current C1 model.


Subject(s)
Complement C1/chemistry , Complement C1r/chemistry , Complement C1s/chemistry , Mass Spectrometry/methods , Protein Structure, Quaternary , Amino Acid Sequence , Binding Sites , Complement Activation , Complement C1/genetics , Complement C1/metabolism , Complement C1r/genetics , Complement C1r/metabolism , Complement C1s/genetics , Complement C1s/metabolism , Humans , Lysine/chemistry , Models, Molecular , Molecular Sequence Data , Molecular Structure , Protein Binding , Staining and Labeling/methods , Surface Properties
15.
J Immunol ; 185(2): 808-12, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20548024

ABSTRACT

C1q, the recognition subunit of the C1 complex of complement, is an archetypal pattern recognition molecule with the striking ability to sense a wide variety of targets, including a number of altered self-motifs. The recognition properties of its globular domain were further deciphered by means of x-ray crystallography using deoxy-D-ribose and heparan sulfate as ligands. Highly specific recognition of deoxy-D-ribose, involving interactions with Arg C98, Arg C111, and Asn C113, was observed at 1.2 A resolution. Heparin-derived tetrasaccharide interacted more loosely through Lys C129, Tyr C155, and Trp C190. These data together with previous findings define a unique binding area exhibiting both polyanion and deoxy-D-ribose recognition properties, located on the inner face of C1q. DNA and heparin compete for C1q binding but are poor C1 activators compared with immune complexes. How the location of this binding area in C1q may regulate the level of C1 activation is discussed.


Subject(s)
Complement C1q/chemistry , Deoxyribose/chemistry , Heparitin Sulfate/chemistry , Protein Structure, Tertiary , Binding Sites , Complement C1q/metabolism , Crystallography, X-Ray , DNA/chemistry , DNA/metabolism , Deoxyribose/metabolism , Heparitin Sulfate/metabolism , Humans , Lysine/chemistry , Lysine/metabolism , Models, Molecular , Protein Binding , Tryptophan/chemistry , Tryptophan/metabolism , Tyrosine/chemistry , Tyrosine/metabolism
16.
J Biol Chem ; 285(25): 19267-76, 2010 Jun 18.
Article in English | MEDLINE | ID: mdl-20410306

ABSTRACT

A growing number of studies have investigated the interaction between C1q and PrP, but the oligomeric form of PrP involved in this interaction remains to be determined. Aggregation of recombinant full-length murine PrP in the presence of 100 mm NaCl allowed us to isolate three different types of oligomers by size-exclusion chromatography. In contrast to PrP monomers and fibrils, these oligomers activate the classical complement pathway, the smallest species containing 8-15 PrP protomers being the most efficient. We used Thioflavine T fluorescence to monitor PrP aggregation and showed that, when added to the reaction, C1q has a cooperative effect on PrP aggregation and leads to the formation of C1q-PrP complexes. In these complexes, C1q interacts through its globular domains preferentially with the smallest oligomers, as shown by electron microscopy, and retains the ability to activate the classical complement pathway. Using two cell lines, we also provide evidence that C1q inhibits the cytotoxicity induced by the smallest PrP oligomers. The cooperative interaction between C1q and PrP could represent an early step in the disease, where it prevents elimination of the prion seed, leading to further aggregation.


Subject(s)
Complement C1q/metabolism , Prions/chemistry , Amyloid/chemistry , Animals , Benzothiazoles , Chromatography/methods , Complement C4/chemistry , Complement System Proteins , Humans , Immunity, Innate , Mice , Microscopy, Electron/methods , Neurons/metabolism , Protein Binding , Thiazoles/chemistry
17.
J Innate Immun ; 2(1): 17-23, 2010.
Article in English | MEDLINE | ID: mdl-20375619

ABSTRACT

Innate immunity relies upon the ability of a variety of recognition molecules to sense pathogens through conserved molecular signatures that are often carbohydrates. Ficolins are oligomeric proteins assembled from collagen-like stalks and fibrinogen-like domains that have the ability to sense these molecular patterns on both pathogens and apoptotic cell surfaces. Three ficolins, termed L, H and M, have been identified in humans. They differ in their localization and concentration in extracellular fluids, their mode of secretion and their recognition properties. From a structural point of view, ficolins are assembled from basal trimeric subunits comprising a collagen-like triple helix and a globular domain composed of 3 fibrinogen-like domains. The globular domains are responsible for sensing danger signals whereas the collagen-like stalks provide a link with immune effectors. This review mainly focuses on the structure and recognition properties of the 3 human ficolins, as revealed by recent crystallographic analysis of their recognition domains. The ligand binding sites have been identified in the 3 ficolins and their recognition mechanisms have been characterized at the atomic level. In the case of M-ficolin, a structural transition at acidic pH disables the binding pocket, and thus likely participates in the functional cycle of this protein.


Subject(s)
Lectins/immunology , Carbohydrates/immunology , Complement System Proteins/immunology , Humans , Immunity, Innate , Lectins/chemistry , N-Acetylneuraminic Acid/immunology , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary , Receptors, Pattern Recognition/immunology , Ficolins
18.
J Immunol ; 184(9): 4982-9, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20351192

ABSTRACT

Variants of the human C1 inhibitor serpin domain containing three N-linked carbohydrates at positions 216, 231, and 330 (C1inhDelta97), a single carbohydrate at position 330 (C1inhDelta97DM), or no carbohydrate were produced in a baculovirus/insect cells system. An N-terminally His-tagged C1inhDelta97 variant was also produced. Removal of the oligosaccharide at position 330 dramatically decreased expression, precluding further analysis. All other variants were characterized chemically and shown to inhibit C1s activity and C1 activation in the same way as native C1 inhibitor. Likewise, they formed covalent complexes with C1s as shown by SDS-PAGE analysis. C1 inhibitor and its variants inhibited the ability of C1r-like protease to activate C1s, but did not form covalent complexes with this protease. The interaction of C1 inhibitor and its variants with heparin was investigated by surface plasmon resonance, yielding K(D) values of 16.7 x 10(-8) M (C1 inhibitor), 2.3 x 10(-8) M (C1inhDelta97), and 3.6 x 10(-8) M (C1inhDelta97DM). C1s also bound to heparin, with lower affinity (K(D) = 108 x 10(-8) M). Using the same technique, 50% inhibition of the binding of C1 inhibitor and C1s to heparin was achieved using heparin oligomers containing eight and six saccharide units, respectively. These values roughly correlate with the size of 10 saccharide units yielding half-maximal potentiation of the inhibition of C1s activity by C1 inhibitor, consistent with a "sandwich" mechanism. Using a thermal shift assay, heparin was shown to interact with the C1s serine protease domain and the C1 inhibitor serpin domain, increasing and decreasing their thermal stability, respectively.


Subject(s)
Complement C1 Inhibitor Protein/physiology , Complement C1/antagonists & inhibitors , Complement C1/metabolism , Heparin/metabolism , Serpins/physiology , Animals , Baculoviridae/genetics , Binding, Competitive/genetics , Binding, Competitive/immunology , Carbohydrates/chemistry , Carbohydrates/genetics , Complement C1 Inhibitor Protein/genetics , Complement C1 Inhibitor Protein/metabolism , Complement C1s/antagonists & inhibitors , Complement C1s/metabolism , Heparin/chemistry , Humans , Molecular Weight , Moths/genetics , Mutagenesis, Site-Directed , Protein Binding/genetics , Protein Structure, Tertiary/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Serpins/genetics , Serpins/metabolism , Spodoptera/genetics , Swine
19.
Biochemistry ; 49(10): 2167-76, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20166680

ABSTRACT

We previously reported that enzymatically modified low-density lipoprotein (E-LDL) particles obtained by LDL treatment with trypsin and then cholesterol esterase are recognized by C1q and activate the C1 complex of complement. The objective of this study was to identify the E-LDL component(s) recognized by C1q. In addition to trypsin, plasmin, thrombin, tryptase, and matrix metalloprotease-2 each yielded E-LDL particles with high C1-activating efficiency, and the C1 activation extent was strictly dependent on cholesterol esterase treatment in all cases. When incorporated into vesicles, the lipid fraction of E-LDL, but not of native LDL, triggered C1 activation, and activation correlated with the amount of unesterified cholesterol generated by cholesterol esterase. Whereas treatment of E-LDL particles with human serum albumin reduced their fatty acid content, both cholesterol and unesterified fatty acids were decreased by methyl-beta-cyclodextrin, both treatments resulting in dose-dependent inhibition of the C1-activating ability of the particles. Incorporation of linoleic acid into phosphatidylcholine-containing model vesicles enabled them to interact with the C1q globular domain and to trigger C1 activation, and cholesterol enhanced both processes by facilitating incorporation of the fatty acid into the vesicles. Direct evidence that C1q binds E-LDL through its globular domains was obtained by electron microscopy. This study demonstrates that C1 binding to E-LDL particles involves recognition by the C1q globular domain of the unesterified fatty acids generated by cholesterol esterase. The potential implications of these findings in atherogenesis are discussed.


Subject(s)
Complement C1q/metabolism , Fatty Acids/metabolism , Lipoproteins, LDL/metabolism , Sterol Esterase/metabolism , Animals , Candida/enzymology , Cattle , Complement C1q/chemistry , Esterification , Fatty Acids/chemistry , Humans , Linoleic Acid/metabolism , Microscopy, Electron , Peptide Hydrolases/metabolism , Protein Binding , Protein Structure, Tertiary , Serum Albumin/pharmacology , beta-Cyclodextrins/pharmacology
20.
J Biol Chem ; 285(9): 6612-22, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20032467

ABSTRACT

Ficolins are oligomeric innate immune recognition proteins consisting of a collagen-like region and a fibrinogen-like recognition domain that bind to pathogen- and apoptotic cell-associated molecular patterns. To investigate their carbohydrate binding specificities, serum-derived L-ficolin and recombinant H- and M-ficolins were fluorescently labeled, and their carbohydrate binding ability was analyzed by glycan array screening. L-ficolin preferentially recognized disulfated N-acetyllactosamine and tri- and tetrasaccharides containing terminal galactose or N-acetylglucosamine. Binding was sensitive to the position and orientation of the bond between N-acetyllactosamine and the adjacent carbohydrate. No significant binding of H-ficolin to any of the 377 glycans probed could be detected, providing further evidence for its poor lectin activity. M-ficolin bound preferentially to 9-O-acetylated 2-6-linked sialic acid derivatives and to various glycans containing sialic acid engaged in a 2-3 linkage. To further investigate the structural basis of sialic acid recognition by M-ficolin, point mutants were produced in which three residues of the fibrinogen domain were replaced by their counterparts in L-ficolin. Mutations G221F and A256V inhibited binding to the 9-O-acetylated sialic acid derivatives, whereas Y271F abolished interaction with all sialic acid-containing glycans. The crystal structure of the Y271F mutant fibrinogen domain was solved, showing that the mutation does not alter the structure of the ligand binding pocket. These analyses reveal novel ficolin ligands such as sulfated N-acetyllactosamine (L-ficolin) and gangliosides (M-ficolin) and provide precise insights into the sialic acid binding specificity of M-ficolin, emphasizing the essential role of Tyr(271) in this respect.


Subject(s)
Carbohydrates/chemistry , Lectins/metabolism , N-Acetylneuraminic Acid/metabolism , Polysaccharides/metabolism , Crystallography, X-Ray , Fibrinogen/chemistry , Fibrinogen/genetics , Humans , Lectins/chemistry , Mutagenesis, Site-Directed , Mutation, Missense , N-Acetylneuraminic Acid/chemistry , Polysaccharides/chemistry , Protein Array Analysis , Protein Binding , Tyrosine , Ficolins
SELECTION OF CITATIONS
SEARCH DETAIL
...