Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Biochem J ; 422(2): 295-303, 2009 Aug 13.
Article in English | MEDLINE | ID: mdl-19522701

ABSTRACT

The complement system plays crucial roles in the immune system, but incorrect regulation causes inflammation and targeting of self-tissue, leading to diseases such as systemic lupus erythematosus, rheumatoid arthritis and age-related macular degeneration. In vivo, the initiating complexes of the classical complement and lectin pathways are controlled by SERPING1 [(C1 inhibitor) serpin peptidase inhibitor, clade G, member 1], which inactivates the components C1s and MASP-2 (mannan-binding lectin serine peptidase 2). GAGs (glycosaminoglycan) and DXS (dextran sulfate) are able to significantly accelerate SERPING1-mediated inactivation of C1s, the key effector enzyme of the classical C1 complex, although the mechanism is poorly understood. In the present study we have shown that C1s can bind to DXS and heparin and that these polyanions enhanced C1s proteolytic activity at low concentrations and inhibited it at higher concentrations. The recent determination of the crystal structure of SERPING1 has given rise to the hypothesis that both the serpin (serine protease inhibitor)-polyanion and protease-polyanion interactions might be required to accelerate the association rate of SERPING1 and C1s. To determine what proportion of the acceleration was due to protease-polyanion interactions, a chimaeric mutant of alpha1-antitrypsin containing the P4-P1 residues from the SERPING1 RCL (reactive-centre loop) was produced. Like SERPING1, this molecule is able to effectively inhibit C1s, but is unable to bind polyanions. DXS exerted a biphasic effect on the association rate of C1s which correlated strongly with the effect of DXS on C1s proteolytic activity. Thus, whereas polyanions are able to bind C1s and modulate its activity, polyanion interactions with SERPING1 must also play a vital role in the mechanism by which these cofactors accelerate the C1s-SERPING1 reaction.


Subject(s)
Complement C1 Inactivator Proteins/metabolism , Complement C1s/metabolism , Peptide Hydrolases/metabolism , Polymers/metabolism , Complement C1 Inhibitor Protein , Enzyme Activation/physiology , Humans , Hydrolysis , Polyelectrolytes , Protein Binding/physiology
2.
Biol Chem ; 390(5-6): 503-7, 2009.
Article in English | MEDLINE | ID: mdl-19361285

ABSTRACT

Complement is a key component of the immune system, but can contribute to inflammatory diseases. The substrate specificity of C1s protease has been successfully investigated using a combinatorial approach, while a positional scanning method failed. The lack of success of the latter approach is possibly due to cooperativity in the active site, which could confound such analyses. With a panel of peptides devised using factorial design, we show pronounced cooperativity between the S4 and S1' subsites in the active site of the enzyme, and weaker cooperativity between the S1' and S3' subsites. The use of factorial design has promise as a methodology for determining cooperativity in protease active sites.


Subject(s)
Complement C1s/chemistry , Complement C1s/metabolism , Binding Sites , Catalytic Domain , Factor Analysis, Statistical , Peptide Library , Peptides/chemistry , Peptides/metabolism , Research Design , Substrate Specificity
3.
Mol Immunol ; 45(3): 670-7, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17709141

ABSTRACT

Complement is a central component of host defence, but unregulated activation can contribute to disease. The system can be initiated by three pathways: classical, alternative and lectin. The classical and lectin pathways are initiated by the C1 and mannose-binding lectin (MBL) or ficolin complexes, respectively, with C1s the executioner protease of the C1 complex and MASP-2 its counterpart in the lectin complexes. These proteases in turn cleave the C4 and C2 components of the system. Here we have elucidated the cleavage specificity of MASP-2 using a randomised substrate phage display library. Apart from the crucial P1 position, the MASP-2 S2 and S3 subsites (in that order) play the greatest role in determining specificity, with Gly residues preferred at P2 and Leu or hydrophobic residues at P3. Cleavage of peptide substrates representing the known physiological cleavage sequences in C2, C4 or the serpin C1-inhibitor (a likely regulator of MASP-2) revealed that MASP-2 is up to 1000 times more catalytically active than C1s. C1-inhibitor inhibited MASP-2 50-fold faster than C1s and much faster than any other protease tested to date, implying that MASP-2 is a major physiological target of C1-inhibitor.


Subject(s)
Complement C1 Inhibitor Protein/chemistry , Complement Pathway, Mannose-Binding Lectin/physiology , Mannose-Binding Protein-Associated Serine Proteases/chemistry , Complement C1/chemistry , Complement C1/genetics , Complement C1/immunology , Complement C1 Inhibitor Protein/genetics , Complement C1 Inhibitor Protein/immunology , Complement C2/chemistry , Complement C2/genetics , Complement C2/immunology , Complement C4/chemistry , Complement C4/genetics , Complement C4/immunology , Humans , Mannose-Binding Lectin/chemistry , Mannose-Binding Lectin/genetics , Mannose-Binding Lectin/immunology , Mannose-Binding Protein-Associated Serine Proteases/genetics , Mannose-Binding Protein-Associated Serine Proteases/immunology , Peptide Library , Substrate Specificity/physiology
4.
J Biol Chem ; 280(47): 39510-4, 2005 Nov 25.
Article in English | MEDLINE | ID: mdl-16169853

ABSTRACT

The complement system is a central component of host defense but can also contribute to the inflammation seen in pathological conditions. The C1s protease of the first complement component, the C1 complex, initiates the pathway. In this study we have elucidated the full specificity of the enzyme for the first time using a randomized phage display library. It was found that, aside from the crucial P(1) position, the S(3) and S(2) subsites (in that order) played the greatest role in determining specificity. C1s prefers Leu or Val at P(3) and Gly or Ala residues at P(2). Apart from the S(2)' position, which showed specificity for Leu, prime subsites did not greatly affect specificity. It was evident, however, that together they significantly contributed to the efficiency of cleavage of a peptide. A peptide substrate based on the top sequence obtained in the phage display validated these results and produced the best kinetics of any C1s substrate to date. The results allow an understanding of the active site specificity of the C1s protease for the first time and provide a basis for the development of specific inhibitors aimed at controlling inflammation associated with complement activation in adverse pathological situations.


Subject(s)
Complement C1s/metabolism , Complement Pathway, Classical/physiology , Amino Acid Sequence , Base Sequence , Binding Sites/genetics , Catalytic Domain/genetics , Complement C1s/chemistry , DNA/genetics , Humans , In Vitro Techniques , Models, Molecular , Oligopeptides/chemistry , Oligopeptides/genetics , Oligopeptides/metabolism , Peptide Library , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...