Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Mol Biol ; 431(5): 885-903, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30653992

ABSTRACT

Proprotein convertase subtilisin/kexin 9 (PCSK9) is a key regulator of lipid metabolism by degrading liver LDL receptors. Structural studies have provided molecular details of PCSK9 function. However, the N-terminal acidic stretch of the PCSK9 prodomain (Q31-T60) has eluded structural investigation, since it is in a disordered state. The interest in this region is intensified by the presence of human missense mutations associated with low and high LDL-c levels (E32K, D35Y, and R46L, respectively), as well as two posttranslationally modified sites, sulfated Y38 and phosphorylated S47. Herein we show that a segment within this region undergoes disorder-to-order transition. Experiments with acidic stretch-derived peptides demonstrated that the folding is centered at the segment Y38-L45, which adopts an α-helix as determined by NMR analysis of free peptides and by X-ray crystallography of peptides in complex with antibody 6E2 (Ab6E2). In the Fab6E2-peptide complexes, the structured region features a central 2 1/4-turn α-helix and encompasses up to 2/3 of the length of the acidic stretch, including the missense mutations and posttranslationally modified sites. Experiments with helix-breaking proline substitutions in peptides and in PCSK9 protein indicated that Ab6E2 specifically recognizes the helical conformation of the acidic stretch. Therefore, the observed quantitative binding of Ab6E2 to native PCSK9 from various cell lines suggests that the disorder-to-order transition is a true feature of PCSK9 and not limited to peptides. Because the helix provides a constrained spatial orientation of the missense mutations and the posttranslationally modified residues, it is probable that their biological functions take place in the context of an ordered conformational state.


Subject(s)
Proprotein Convertase 9/genetics , Amino Acid Sequence , Cell Line, Tumor , Crystallography, X-Ray/methods , Hep G2 Cells , Humans , Mutation, Missense/genetics , Peptides/genetics , Protein Processing, Post-Translational/genetics , Receptors, LDL/genetics , Serine Endopeptidases/genetics
2.
Protein Eng Des Sel ; 30(9): 619-625, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28475752

ABSTRACT

The well-studied B-domain from Staphylococcal protein A is a 59 amino acid three-helix bundle that binds the Fc portion of IgG with a dissociation constant of ~35 nM. The B-domain variant bearing a Gly to Ala mutation (=Z-domain) has been the subject of efforts to minimize a domain's size while retaining its function. We report X-ray crystallographic characterization of three steps in such a process using complexes with Fc: the full three-helix Z-domain, a 34 amino acid two-helix version called Z34C and a 13 amino acid single helix stabilized with an exo-helix tether, called LH1.


Subject(s)
Alanine/chemistry , Glycine/chemistry , Immunoglobulin Fc Fragments/chemistry , Staphylococcal Protein A/chemistry , Alanine/metabolism , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Glycine/metabolism , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Kinetics , Models, Molecular , Mutation , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Staphylococcal Protein A/genetics , Staphylococcal Protein A/metabolism , Staphylococcus aureus/chemistry , Thermodynamics
3.
Biochemistry ; 40(32): 9522-31, 2001 Aug 14.
Article in English | MEDLINE | ID: mdl-11583151

ABSTRACT

A new inhibitory peptide binding exosite on the protease domain of coagulation Factor VIIa (FVIIa) has been identified. A novel series of peptide inhibitors of FVIIa, termed the "A-series" peptides, identified from peptide phage libraries and exemplified by peptide A-183 [Dennis, M. S., Roberge, M., Quan, C., and Lazarus, R. A. (2001) Biochemistry 40, 9513-9521], specifically bind at a site that is distinct from both the active site and the exosite of another recently described peptide inhibitor of FVIIa, E-76 [Dennis, M. S., Eigenbrot, C., Skelton, N. J., Ultsch, M. H., Santell, L., Dwyer, M. A., O'Connell, M. P., and Lazarus, R. A. (2000) Nature 404, 465-4701. Peptide A-183 prolonged TF-dependent clotting in human, but not rabbit plasma. Thus, a panel of human FVIIa mutants, containing 70 of the 76 rabbit sequence differences in the protease domain, localized the binding site to residues in the 60s loop and the C-terminus. The location of the exosite was refined by a series of FVIIa alanine mutants, which showed that proximal residues Trp 61 and Leu 251 were critical for binding. Kinetic and equilibrium binding constants for zymogen FVII, FVIIa and TF x FVIIa were determined using immobilized N-terminal biotinylated A-183 by surface plasmon resonance. No peptide binding to nine other human serine proteases was observed. Key residues on the peptide were determined from binding to FVIIa and inhibition of FX activation using a series of alanine mutants of A-183 fused to the Z domain of protein A. Analysis of the mutagenesis data is presented in the context of a crystal structure of A-183 in complex with a version of zymogen FVII [Eigenbrot, C., Kirchhofer, D., Dennis, M. S., Santell, L., Lazarus, R. A., Stamos, J., and Ultsch, M. H. (2001) Structure 9, 627-636]. The shape and proximity of this exosite to the active site may lend itself towards the design of new anticoagulants that inhibit FVIIa.


Subject(s)
Blood Coagulation/drug effects , Factor VIIa/antagonists & inhibitors , Peptides/pharmacology , Protease Inhibitors/pharmacology , Amino Acid Sequence , Animals , Binding Sites , Dose-Response Relationship, Drug , Factor VIIa/chemistry , Factor VIIa/metabolism , Humans , Kinetics , Models, Molecular , Molecular Sequence Data , Peptide Library , Peptides/chemistry , Peptides/metabolism , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Rabbits , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Structure-Activity Relationship
4.
Structure ; 9(7): 627-36, 2001 Jul 03.
Article in English | MEDLINE | ID: mdl-11470437

ABSTRACT

BACKGROUND: Coagulation factor VIIa (FVIIa) contains a Trypsin-like serine protease domain and initiates the cascade of proteolytic events leading to Thrombin activation and blood clot formation. Vascular injury allows formation of the complex between circulating FVIIa and its cell surface bound obligate cofactor, Tissue Factor (TF). Circulating FVIIa is nominally activated but retains zymogen-like character and requires TF in order to complete the zymogen-to-enzyme transition. The manner in which TF exerts this effect is unclear. The structure of TF/FVIIa is known. Knowledge of the zymogen structure is helpful for understanding the activation transition in this system. RESULTS: The 2 A resolution crystal structure of a zymogen form of FVII comprising the EGF2 and protease domains is revealed in a complex with the exosite binding inhibitory peptide A-183 and a vacant active site. The activation domain, which includes the N terminus, differs in ways beyond those that are expected for zymogens in the Trypsin family. There are large differences in the TF binding region. An unprecedented 3 residue shift in registration between beta strands B2 and A2 in the C-terminal beta barrel and hydrogen bonds involving Glu154 provide new insight into conformational changes accompanying zymogen activation, TF binding, and enzymatic competence. CONCLUSIONS: TF-mediated allosteric control of the activity of FVIIa can be rationalized. The reregistering beta strand connects the TF binding region and the N-terminal region. The zymogen registration allows H bonds that prevent the N terminus from attaining a key salt bridge with the active site. TF binding may influence an equilibrium by selecting the enzymatically competent registration.


Subject(s)
Factor VII/chemistry , Factor VII/metabolism , Aspartic Acid/chemistry , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Enzyme Precursors/chemistry , Factor VII/genetics , Glutamic Acid/chemistry , Isoleucine/chemistry , Models, Molecular , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thromboplastin/metabolism
5.
Biochemistry ; 40(3): 675-82, 2001 Jan 23.
Article in English | MEDLINE | ID: mdl-11170384

ABSTRACT

Tissue factor is the cell membrane-anchored cofactor for factor VIIa and triggers the coagulation reactions. The initial step is the conversion of factor VII to factor VIIa which, in vitro, is efficiently catalyzed by low concentrations of factor Xa. To identify the tissue factor region that interacts with the activator factor Xa during this process, we evaluated a panel of soluble tissue factor (1-219) mutants for their ability to support factor Xa-mediated activation of factor VII. The tissue factor residues identified as most important for this interaction (Tyr157, Lys159, Ser163, Gly164, Lys165, Lys166, and Tyr185) were identical to those found to be important for the interaction of substrate factor X with the tissue factor.factor VIIa complex. The residues form a continuous surface-exposed patch with an area of about 500 A(2), which appears to be located outside the tissue factor-factor VII contact zone. In agreement, the two monoclonal antibodies 5G6 and D3H44-F(ab')(2), whose epitopes overlap with this identified region, inhibited the rates of factor VII activation by 86% and 95%, respectively. These antibodies also strongly inhibited the conversion of (125)I-labeled factor VII when cell membrane-expressed, full-length tissue factor (1-263) was employed. Together the results suggest the usage of a common surface region of tissue factor in its dual role-as a cofactor for factor Xa-mediated factor VII activation and as a cofactor for factor VIIa-mediated factor X activation. The finding that factor Xa and factor X may engage in similar, if not identical, molecular interactions with tissue factor further indicates that factor Xa and factor X are similarly oriented toward their respective interaction partners in the ternary catalytic complexes.


Subject(s)
Factor VII/metabolism , Factor Xa/metabolism , Thromboplastin/metabolism , 1-Carboxyglutamic Acid/metabolism , Cell Line , Cell Membrane/enzymology , Cell Membrane/genetics , Cell Membrane/metabolism , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Enzyme Precursors/metabolism , Factor VIIa/metabolism , Humans , Iodine Radioisotopes , Mutagenesis, Site-Directed , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Mapping , Protein Binding/genetics , Protein Structure, Tertiary/genetics , Solubility , Surface Plasmon Resonance , Thromboplastin/genetics
6.
Cytokine ; 13(1): 1-7, 2001 Jan 07.
Article in English | MEDLINE | ID: mdl-11145836

ABSTRACT

IL-18, or IGIF (interferon-gamma inducing factor), is an IL-1-related, pro-inflammatory cytokine, which plays a pivotal role in systemic and local inflammation. We have identified and characterized IL-1H, a novel IL-1-related molecule. IL-1H appears to be expressed in most tissues with relatively high levels in testis, thymus and uterus. The IL-1H transcripts were stimulated by phorbol ester (PMA) in human cell lines (A431, THP-1 and KG-1) and peripheral blood mononuclear cells (HPBMC) and dendritic cells (NHDC). The protein sequence of IL-1H is mostly related to IL-1ra with a similarity of 36%. A short form of IL-1H was identified, and lacks a 40-amino acid segment in the amino-terminal region of the protein. When expressed in mammalian cells, two secreted polypeptides of IL-1H were identified: an uncleaved and a cleaved form starting with amino acid Val-46. Furthermore, IL-1H binds the IL-18 receptor, but not the IL-1 receptor. These findings suggest that IL-1H may be another ligand for the IL-18 receptor and a new player in the inflammatory and immune responses mediated by the IL-18/IL-18R axis.


Subject(s)
Proteins/metabolism , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , Cell Line , Cloning, Molecular , Cricetinae , DNA Primers/genetics , Female , Gene Expression Regulation/drug effects , Humans , In Vitro Techniques , Interleukin-18 Receptor alpha Subunit , Male , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interleukin/chemistry , Receptors, Interleukin-18 , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Tetradecanoylphorbol Acetate/pharmacology , Tissue Distribution , Transfection
7.
Biochemistry ; 39(25): 7380-7, 2000 Jun 27.
Article in English | MEDLINE | ID: mdl-10858285

ABSTRACT

The enzymatic activity of coagulation factor VIIa is controlled by its cellular cofactor tissue factor (TF). TF binds factor VIIa with high affinity and, in addition, participates in substrate interaction through its C-terminal fibronectin type III domain. We analyzed surface-exposed residues in the C-terminal TF domain to more fully determine the area on TF important for substrate activation. Soluble TF (sTF) mutants were expressed in E. coli, and their ability to support factor VIIa-dependent substrate activation was measured in the presence of phospholipid vesicles or SW-13 cell membranes. The results showed that factor IX and factor X interacted with the same TF region located proximal to the putative phospholipid surface. According to the degree of activity loss of the sTF mutants, this TF region can be divided into a main region (residues Tyr157, Lys159, Ser163, Gly164, Lys165, Lys166, Tyr185) forming a solvent-exposed patch of 488 A(2) and an extended region which comprises an additional 7-8 residues, including the distally positioned Asn199, Arg200, and Asp204. Some of the identified TF residues, such as Trp158 and those within the loop Lys159-Lys165, are near the factor VIIa gamma-carboxyglutamic acid (Gla) domain, suggesting that the factor VIIa Gla-domain may also participate in substrate interaction. Moreover, the surface identified as important for substrate interaction carries a net positive charge, suggesting that charge interactions may significantly contribute to TF-substrate binding. The calculated surface-exposed area of this substrate interaction region is about 1100 A(2), which is approximately half the size of the TF area that is in contact with factor VIIa. Therefore, a substantial portion of the TF surface (3000 A(2)) is engaged in protein-protein interactions during substrate catalysis.


Subject(s)
Factor IX/metabolism , Factor X/metabolism , Thromboplastin/metabolism , Amides/metabolism , Antibodies/immunology , Enzyme Activation , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Phospholipids/metabolism , Recombinant Proteins/metabolism , Substrate Specificity , Thromboplastin/chemistry , Thromboplastin/genetics , Thromboplastin/immunology
8.
Nature ; 404(6777): 465-70, 2000 Mar 30.
Article in English | MEDLINE | ID: mdl-10761907

ABSTRACT

Potent anticoagulants have been derived by targeting the tissue factor-factor VIIa complex with naive peptide libraries displayed on M13 phage. The peptides specifically block the activation of factor X with a median inhibitory concentration of 1 nM and selectively inhibit tissue-factor-dependent clotting. The peptides do not bind to the active site of factor VIIa; rather, they work by binding to an exosite on the factor VIIa protease domain, and non-competitively inhibit activation of factor X and amidolytic activity. One such peptide (E-76) has a well defined structure in solution determined by NMR spectroscopy that is similar to the X-ray crystal structure when complexed with factor VIIa. These structural and functional studies indicate an allosteric 'switch' mechanism of inhibition involving an activation loop of factor VIIa and represent a new framework for developing inhibitors of serine proteases.


Subject(s)
Anticoagulants/pharmacology , Factor VIIa/antagonists & inhibitors , Oligopeptides/pharmacology , Peptides/pharmacology , Serine Proteinase Inhibitors/pharmacology , Amino Acid Sequence , Animals , Binding Sites , Consensus Sequence , Crystallography, X-Ray , Enzyme Precursors/metabolism , Factor VIIa/chemistry , Factor X/antagonists & inhibitors , Factor X/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Oligopeptides/isolation & purification , Peptide Library , Peptides/isolation & purification , Protein Conformation , Rabbits , Serine Proteinase Inhibitors/analysis , Thromboplastin/metabolism
9.
Proteins ; 38(4): 361-7, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10707023

ABSTRACT

The "ELR" (Glu-Leu-Arg) tripeptide sequence near the N-terminus of interleukin-8 (IL-8) contributes a large part of the receptor binding free energy. Prior X-ray and nuclear magnetic resonance (NMR) structures of IL-8 have shown this region of the molecule to be highly mobile. We reasoned that a hydrophobic interaction between the leucine and the neighboring beta-turn might exist in the receptor binding conformation of the N-terminus. To test this hypothesis, we mutated two residues to cysteine and connected the N-terminus to the beta-turn. The mutant retains receptor binding affinity reasonably close to wild type and allows the characterization of a high-affinity conformation that may be useful in the design of small IL-8 mimics. The L5C/H33C mutant is refined to R-values of R = 20.6% and Rfree = 27.7% at 2.35 A resolution. Other receptor binding determinants reside in the "N-loop" found after "ELR" and preceding the first beta-strand. All available structures of IL-8 have been found with one of two distinct N-loop conformations. One of these is relevant for receptor binding, based on NMR results with receptor peptides. The other conformation obscures the receptor-peptide binding surface and may have an undetermined but necessarily different function.


Subject(s)
Interleukin-8/chemistry , Amino Acid Motifs , Amino Acid Substitution , Animals , Antigens, CD/chemistry , Crystallography, X-Ray , Humans , Interleukin-8/genetics , Magnetic Resonance Spectroscopy , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding , Protein Conformation , Receptors, Cytokine/chemistry , Receptors, Interleukin/chemistry , Receptors, Interleukin-8A
10.
Thromb Haemost ; 84(6): 1072-81, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11154116

ABSTRACT

Tissue factor (TF), the cellular cofactor for the serine protease factor VIIa (F.VIIa), triggers blood coagulation and is involved in the pathogenesis of various thrombosis-related disorders. Therefore, agents which specifically target tissue factor, such as monoclonal antibodies, may provide promising new antithrombotic therapy. We mapped the epitopes of several anti-TF antibodies using a panel of soluble TF mutants. They bound to three distinct TF regions. The epitope of the 7G11 antibody included Phe50 and overlapped with a TF-F.VIIa light chain contact area. The common epitope of the antibodies 6B4 and HTF1 included residues Tyr94 and Phe76 both of which make critical contacts to the catalytic domain of F.VIIa. The antibodies D3 and 5G6 had a common epitope outside the TF-F.VIIa contact region. It included residues Lys 165, Lys 166, Asn199, Arg200 and Lys201 and thus overlapped with the substrate interaction region of tissue factor. The antibodies 5G6 and D3 were potent anticoagulants when infused to flowing human blood in an ex-vivo thrombosis model. Plasma fibrinopeptide A levels and fibrin deposition were completely inhibited. In contrast, 6B4 was a weak inhibitor in this ex-vivo thrombosis model, and HTF1 displayed no inhibition at all. These disparate activities were also reflected in TF-dependent F.X activation assays performed with human plasma. The potency differences could neither be explained by the determined binding affinities nor by the on-rates of antibodies. Therefore, the results suggest that antibody binding epitope and hence the particular mechanism of inhibition, is the main determinative factor of anticoagulant potency of anti-TF antibodies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Epitopes/analysis , Thromboplastin/immunology , Antibody Affinity , Anticoagulants/pharmacology , Binding Sites , Blood Coagulation/drug effects , Blood Flow Velocity , Crystallography, X-Ray , Epitopes/pharmacology , Factor VIIa/metabolism , Humans , Mutation , Thromboplastin/genetics , Thromboplastin/metabolism
11.
Cell ; 91(5): 695-704, 1997 Nov 28.
Article in English | MEDLINE | ID: mdl-9393862

ABSTRACT

Vascular endothelial growth factor (VEGF) is a homodimeric hormone that induces proliferation of endothelial cells through binding to the kinase domain receptor and the Fms-like tyrosine kinase receptor (Flt-1), the extracellular portions of which consist of seven immunoglobulin domains. We show that the second and third domains of Flt-1 are necessary and sufficient for binding VEGF with near-native affinity, and that domain 2 alone binds only 60-fold less tightly than wild-type. The crystal structure of the complex between VEGF and the second domain of Flt-1 shows domain 2 in a predominantly hydrophobic interaction with the "poles" of the VEGF dimer. Based on this structure and on mutational data, we present a model of VEGF bound to the first four domains of Flt-1.


Subject(s)
Endothelial Growth Factors/chemistry , Endothelial Growth Factors/genetics , Lymphokines/chemistry , Lymphokines/genetics , Proto-Oncogene Proteins/chemistry , Receptor Protein-Tyrosine Kinases/chemistry , Cells, Cultured , Crystallography , Endothelial Growth Factors/metabolism , Lymphokines/metabolism , Molecular Sequence Data , Mutagenesis , Peptide Fragments/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Platelet-Derived Growth Factor/genetics , Sequence Homology, Amino Acid , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
13.
Proteins ; 27(4): 556-66, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9141135

ABSTRACT

The characteristic CXC chemokine disulfide core of interleukin-8 (IL-8) has been rearranged in a variant replacing the 9-50 disulfide with a 9-38 disulfide. The new variant has been characterized by its binding affinity to IL-8 receptors A and B and the erythrocyte receptor DARC. This variant binds the three receptors with affinities between 500- and 2,500-fold lower than wild-type IL-8. Binding affinity results are also reported for the variant with alanine substituted for both cysteines 9 and 50. The Glu38-->Cys/Cys50-->Ala IL-8 crystallizes in space group P2(1)2(1)2(1) with cell parameters a = 46.4, b = 49.2, and c = 69.5 A, and has been refined to an R-value of 19.4% for data from 10 to 2 A resolution. Analysis of the structure confirms the new disulfide arrangement and suggests that changes at Ile10 may be the principal cause of the lowered affinities.


Subject(s)
Antigens, CD/metabolism , Cysteine/genetics , Disulfides/chemistry , Interleukin-8/chemistry , Interleukin-8/metabolism , Receptors, Interleukin/metabolism , Binding Sites/genetics , Binding, Competitive , Computer Simulation , Crystallography, X-Ray , Erythrocytes/metabolism , Interleukin-8/genetics , Models, Molecular , Mutation , Neutrophils/metabolism , Protein Conformation , Receptors, Interleukin-8A , Water/chemistry
14.
Proteins ; 18(1): 49-62, 1994 Jan.
Article in English | MEDLINE | ID: mdl-7908437

ABSTRACT

X-ray crystal structures of fragments from two different humanized anti-CD18 antibodies are reported. The Fv fragment of the nonbinding version has been refined in space group C2 with a = 64.2 A, b = 61.3 A, c = 51.8 A, and beta = 99 degrees to an R-value of 18.0% at 1.9 A, and the Fab fragment of the tight-binding version has been refined in space group P3 with a = 101. A and c = 45.5 A to an R-value of 17.8% at 3.0 A resolution. The very large difference in their binding affinity (> 1000-fold) is attributed to large and local structural differences in the C-terminal part of CDR-H2, and from this we conclude there is direct contact between this region and antigen when they combine. X-ray structures of antibody-antigen complexes available in the literature have yet to show this part of CDR-H2 in contact with antigen, despite its hypervariable sequence. Implications of this result for antibody humanization are discussed.


Subject(s)
Antibodies, Monoclonal/chemistry , Antigens, CD/immunology , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fragments/chemistry , Immunoglobulin Variable Region/chemistry , Amino Acid Sequence , Binding, Competitive , CD18 Antigens , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Recombinant Proteins/chemistry , T-Lymphocytes/immunology
15.
J Mol Biol ; 229(4): 969-95, 1993 Feb 20.
Article in English | MEDLINE | ID: mdl-8095303

ABSTRACT

The X-ray structures of 1 Fv and 2 Fab humanized anti-p185HER2 antibody fragments (IgG1-kappa) have been determined at a resolution between 2.7 A and 2.2 A. The antibodies are three different versions of a human antibody framework onto which the antigen recognition loops from a murine antibody (4D5) have been grafted. The sequences of the three versions differ in the framework region at positions L55, H78 and H102. The version 8 Fv fragment crystallizes in space group P2(1) with cell parameters a = 37.6 A, b = 63.4 A, c = 90.2 A, beta = 98.2 degrees, with two molecules per asymmetric unit, and has been refined against data 10.0 A-2.2 A to an R-factor of 18.3%. Versions 4 and 7 Fabs crystallize in space group P1 with cell parameters a = 39.2 A, b = 80.2 A, c = 86.1 A, alpha = 113.1 degrees, beta = 92.7 A, gamma = 102.6 A and two molecules per asymmetric unit. Version 4 has been refined against data 10.0 A-2.5 A resolution to an R-factor of 17.9%. Version 7 has been refined against data 10 A-2.7 A to an R-factor of 17.1%. The X-ray structures have been used to assess the accuracy of structural predictions made via molecular modeling, and they confirm the structural role of certain framework residues and the conformations of five of six complementarity determining regions (CDRS). The average deviation of the model from the X-ray structures is within the range observed among the X-ray structures for 81% of the C alpha atoms. Of the hydrogen bonds common to the X-ray structures, 94% of the main-chain-main-chain and 79% of the main-chain-side-chain ones were predicted by the model. The side-chain conformation was predicted correctly for 79% of the buried residues. The third CDR in the heavy chain is variable, differing by up to 8 A between molecules within an asymmetric unit. The structural relationship between variable domains of light and heavy chains is not significantly altered by the absence of constant domains in the Fv molecule. The antigen-binding potential of an unusual light chain sequence has been confirmed. The arginine at position 66 interacts with the first light chain CDR, but in a fashion somewhat different than predicted. A substitution of a leucine for an alanine side-chain directed between the beta-sheets has only relatively small and local effects.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Antibodies/immunology , Binding Sites, Antibody , Oncogene Proteins, Viral/immunology , Amino Acid Sequence , Animals , Antibodies/chemistry , Computer Simulation , Humans , Hydrogen Bonding , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/immunology , Mice , Models, Molecular , Molecular Sequence Data , Oncogene Proteins, Viral/chemistry , Protein Conformation , Receptor, ErbB-2 , Sequence Homology, Amino Acid , X-Ray Diffraction
16.
Proteins ; 14(1): 65-74, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1384033

ABSTRACT

The structures of five basic pancreatic trypsin inhibitor (BPTI) molecules are compared to establish the extent and nature of the conformational variability resulting from crystal packing effects. BPTI is an ideal system to evaluate such factors because of the availability of high resolution X-ray models of five different BPTI structures, each in a different crystal packing environment. Differences observed among the structures are found to be distributed throughout the molecule, although the regions that display most variability are associated with the loop structures (residues 14-17 and 24-29). The regions of structure that show the largest rms deviations from the mean of the five packing motifs correlate well with the presence of intermolecular contacts in the crystal lattice. For most of the molecules there is also a correspondence between a larger number of intermolecular contacts and systematically higher B-factors, although it is not apparent whether this is induced by the crystal contact or results from the fact that the contacts are made predominantly through surface loops. The conformational differences seen among the X-ray models constitute more than local shifts at the lattice contact surfaces, and in fact involve in some cases the making and breaking of intramolecular H-bonds. The magnitudes of the differences among packing models are significantly larger than those usually associated with changes induced by mutagenesis; for instance; the structural differences at the site of mutation observed on removing an internal disulfide from the molecule are significantly less than those associated with lattice contact effects. The crystal packing conformations are compared to representative structures of BPTI generated during a 96-psec molecular dynamics (MD) simulation. This comparison shows a high level of correspondence between the protein flexibility indicated by the X-ray and MD analyses, and specifically between those regions that are most variable. This suggests that the regions that show most variability among the crystal packing models are not artifacts of crystallization, but rather represent true low-energy conformers that have been preferentially selected by crystallization factors.


Subject(s)
Aprotinin/chemistry , Amino Acid Sequence , Computer Simulation , Crystallization , Hydrogen Bonding , Molecular Sequence Data , Protein Conformation , Temperature , Thermodynamics , X-Ray Diffraction
17.
Proteins ; 14(1): 75-87, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1384034

ABSTRACT

The X-ray structure of the C30V/C51A disulfide mutant of basic pancreatic trypsin inhibitor (BPTI) has been analyzed at 1.6 A resolution. The mutant crystallizes in a cell having two molecules in the asymmetric unit. The packing environments of these two molecules are quite different, allowing for an assessment of which among the observed structural changes result from the mutation and which are produced by lattice packing considerations. The removal of the 30-51 disulfide bridge has little apparent affect on the B-factors of segments of adjacent polypeptide chain, although there are distinct differences in the structure compared to wild-type BPTI crystal structures. Both of the two C30V/C51A molecules show differences at the mutation site when compared to another 30-51 disulfide mutant, C30A/C51A, presumably due to the larger steric bulk of a valine versus an alanine at residue 30. A comparison of the two independent C30V/C51A molecules indicates that there are significant differences between them even at the site of mutation. The description of the specific structural differences of each molecule differs in detail and suggests different conclusions about the nature of structural perturbation near 30-51. In addition, when these two molecules are compared to two different wild-type structures, which had been determined from different space groups, a somewhat different pattern of changes is observed. These findings indicate that crystal packing can influence the observed perturbations in mutant structures.


Subject(s)
Aprotinin/genetics , Alanine , Animals , Aprotinin/chemistry , Cattle , Crystallization , Models, Molecular , Molecular Structure , Mutagenesis , Protein Conformation , Valine , X-Ray Diffraction
18.
Curr Opin Biotechnol ; 3(4): 333-7, 1992 Aug.
Article in English | MEDLINE | ID: mdl-1368433

ABSTRACT

The way a protein responds to mutation provides key insights into its architecture and energetics. Mutations are improving the understanding both of protein folding and stability, and of the adaptability of the hydrophobic core. The importance of intermolecular effects in crystal structures is being emphasized and new insights into the correspondence between crystal and solution structures are being developed.


Subject(s)
Mutation , Proteins/genetics , Protein Conformation , Proteins/chemistry , X-Ray Diffraction
19.
Biochemistry ; 31(24): 5434-41, 1992 Jun 23.
Article in English | MEDLINE | ID: mdl-1351741

ABSTRACT

The murine monoclonal antibody 4D5 (anti-p185HER2) inhibits the proliferation of human tumor cells overexpressing p185HER2 in vitro and has been "humanized" [Carter, P., Presta, L., Gorman, C. M., Ridgway, J. B. B., Henner, D., Wong, W.-L. T., Rowland, A. M., Kotts, C., Carver, M. E., & Shepard, H. M. (1992) Proc. Natl. Acad. Sci. U.S.A. (in press)] for use in human cancer therapy. We have determined the antigen binding thermodynamics and the antiproliferative activities of chimeric 4D5 Fab (ch4D5 Fab) fragment and a series of eight humanized Fab (hu4D5 Fab) fragments differing by amino acid substitutions in the framework regions of the variable domains. Fab fragments were expressed by secretion from Escherichia coli and purified from fermentation supernatants by using affinity chromatography on immobilized streptococcal protein G or staphylococcal protein A for ch4D5 and hu4D5, respectively. Circular dichroism spectroscopy indicates correct folding of the E. coli produced Fab, and scanning calorimetry shows a greater stability for hu4D5 (Tm = 82 degrees C) as compared with ch4D5 Fab (Tm = 72 degrees C). KD values for binding to the extracellular domain (ECD) of p185HER2 were determined by using a radioimmunoassay; the delta H and delta Cp for binding were determined by using isothermal titration calorimetry. ch4D5 Fab and one of the humanized variants (hu4D5-8 Fab) bind p185HER2-ECD with comparable affinity (delta G degrees = -13.6 kcal mol-1).(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antibodies, Monoclonal/metabolism , Proto-Oncogene Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Calorimetry, Differential Scanning , Circular Dichroism , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/metabolism , Mice , Motion , Protein Conformation , Protein Denaturation , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Radioimmunoassay , Receptor, ErbB-2 , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Solubility , Structure-Activity Relationship , Thermodynamics , X-Ray Diffraction
20.
J Med Chem ; 35(11): 2040-8, 1992 May 29.
Article in English | MEDLINE | ID: mdl-1597855

ABSTRACT

Stimulation of platelets activates GPIIbIIIa, the heterodimeric integrin receptor, to bind fibrinogen (Fg), which results in platelet aggregation. GPIIbIIIa/Fg binding inhibitors are potentially suitable for acute use during and after thrombolytic therapy as antithrombotic agents. Incorporation of the tripeptide sequence Arg-Gly-Asp (RGD), a common structural element of many integrin ligands, into cyclic peptides produced a series of peptides of the general structure BrAc-(AA1)-RGD-Cys-OH, which were prepared by solid-phase peptide synthesis. Cyclization was accomplished by reaction of the N-terminal bromoacetyl group with the cysteine sulfhydryl at pH 8 at high dilution, resulting in thioether-bridged cyclic peptides [cyclo-S-Ac-(AA1)-RGD-Cys-OH]. Use of alpha-substituted bromoacetyl groups gave rise to an analogous series of acetyl-substituted thioether-bridged cyclic peptides. Oxidation of the thioethers produced separable diastereomeric sulfoxide-bridged cyclic peptides. After thorough evaluation in a GPIIbIIIa ELISA assay and a platelet aggregation assay, G-4120 (70A; AA1 = D-Tyr; sulfoxide bridge) was selected for further investigation as an antithrombotic agent. G-4120 was equipotent in the platelet aggregation assay to kistrin, a highly potent inhibitor of fibrinogen-mediated platelet aggregation isolated from snake venom (IC50 = 0.15 microM).


Subject(s)
Fibrinolytic Agents/chemical synthesis , Peptides, Cyclic/chemical synthesis , Peptides/chemical synthesis , Platelet Aggregation Inhibitors/chemical synthesis , Receptors, Immunologic/chemistry , Receptors, Peptide , Sulfoxides/chemical synthesis , Amino Acid Sequence , Cyclization , Enzyme-Linked Immunosorbent Assay , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/pharmacology , Humans , Molecular Conformation , Molecular Sequence Data , Molecular Structure , Peptides/chemistry , Peptides/pharmacology , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/pharmacology , Platelet Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Structure-Activity Relationship , Sulfoxides/chemistry , Sulfoxides/pharmacology , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL
...