Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 275(30): 22756-63, 2000 Jul 28.
Article in English | MEDLINE | ID: mdl-10801853

ABSTRACT

Factor XII deficiency has been postulated to be a risk factor for thrombosis suggesting that factor XII is an antithrombotic protein. The biochemical mechanism leading to this clinical observation is unknown. We have previously reported high molecular weight kininogen (HK) inhibition of thrombin-induced platelet aggregation by binding to the platelet glycoprotein (GP) Ib-IX-V complex. Although factor XII will bind to the intact platelet through GP Ibalpha (glycocalicin) without activation, we now report that factor XIIa (0. 37 microm), but not factor XII zymogen, is required for the inhibition of thrombin-induced platelet aggregation. Factor XIIa had no significant effect on SFLLRN-induced platelet aggregation. Moreover, an antibody to the thrombin site on protease-activated receptor-1 failed to block factor XII binding to platelets. Inhibition of thrombin-induced platelet aggregation was demonstrated with factor XIIa but not with factor XII zymogen or factor XIIf, indicating that the conformational exposure of the heavy chain following proteolytic activation is required for inhibition. However, inactivation of the catalytic activity of factor XIIa did not affect the inhibition of thrombin-induced platelet aggregation. Factor XII showed displacement of biotin-labeled HK (30 nm) binding to gel-filtered platelets and, at concentrations of 50 nm, was able to block 50% of the HK binding, suggesting involvement of the GP Ib complex. Antibodies to GP Ib and GP IX, which inhibited HK binding to platelets, did not block factor XII binding. However, using a biosensor, which monitors protein-protein interactions, both HK and factor XII bind to GP Ibalpha. Factor XII may serve to regulate thrombin binding to the GP Ib receptor by co-localizing with HK, to control the extent of platelet aggregation in vivo.


Subject(s)
Factor XII/metabolism , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thrombin/antagonists & inhibitors , Amino Acid Sequence , Antibodies/immunology , Binding, Competitive , Blotting, Western , Humans , Platelet Glycoprotein GPIb-IX Complex/immunology , Protein Binding , Receptor, PAR-1 , Receptors, Thrombin/metabolism , Thrombin/pharmacology
2.
Blood ; 90(4): 1508-15, 1997 Aug 15.
Article in English | MEDLINE | ID: mdl-9269768

ABSTRACT

We and others have shown that both high and low molecular mass kininogens are able to inhibit the thrombin-induced aggregation of gel-filtered platelets, indicating that the locus for inhibition resides in the heavy chain. The inhibitory site is present in domain 3, confined to the C-terminal portion of the region encoded by exon 7 (K270-G292), and the minimal effective sequence is a heptapeptide (L271-A277; Kunapuli et al, J Biol Chem 271:11228, 1996). Kininogens inhibit thrombin binding to platelets and thus inhibit thrombin-induced aggregation. The molecular mechanism by which kininogens inhibit thrombin-induced aggregation of platelets is unknown. Thrombin has previously been shown to bind to two receptors on the platelet surface, glycoprotein (GP) Ib-IX-V complex and the hepta-spanning transmembrane receptor coupled to G protein(s). We now show that, unlike its effect on normal platelets, kininogen (2 micromol/L) did not inhibit the thrombin-induced aggregation of Bernard-Soulier platelets, which lack the GP Ib-IX-V complex, suggesting that kininogen interacts either directly or indirectly with that complex and restricts access by thrombin to this receptor. We further show that both recombinant K270-G292 polypeptide and the synthetic peptide L271-A277 derived from high molecular mass kininogen lower thrombin binding to platelets in a manner similar to monoclonal antibodies to or ligands (von Willebrand factor and echicetin) of GP Ib-IX. The anti-GP Ib-IX-V complex antibodies, TM-60 and SZ 2, can inhibit 125I-high molecular mass kininogen binding to platelets. Conversely, kininogen could block the binding of biotinylated TM-60 or of 125I-SZ 2. Kininogen inhibited the binding of biotinylated thrombin bound to a mouse fibroblast cell line transfected with the GP Ib-IX-V complex. These results indicated that kininogen binds to the GP Ib-IX-V complex modulating thrombin binding to platelets and the consequent platelet aggregation. Kininogen can thus serve as an important regulator of the early stages of platelet stimulation by thrombin.


Subject(s)
Blood Platelets/metabolism , Kininogens/physiology , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thrombin/metabolism , Animals , Antibodies, Monoclonal/metabolism , Bernard-Soulier Syndrome/blood , Blood Platelets/drug effects , Cell Line , Enzyme-Linked Immunosorbent Assay , Humans , Kininogens/pharmacology , Mice , Peptide Fragments/pharmacology , Peptides/metabolism , Platelet Aggregation/drug effects , Transfection
3.
J Protein Chem ; 15(6): 519-25, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8895098

ABSTRACT

Domain 3 (D3) of human kininogens, the major cysteine proteinase inhibitors in plasma, has been shown to be the tightest binding inhibitory domain for cathepsins B and H. D3 was expressed in three fragments as its exon products as follows: exon 7 (Gly235-Gln292), exon 8 (Gln292-Gly328), and exon 9 (Gln329-Met357). Exon products 7, 8, and 9 alone as well as exon product 7 + 9 each exhibited an 1C50 value 5- to 30-fold higher (5-30 microM) than exon products 7 + 8 and 8 + 9 (0.9-1.3 microM) for cathepsins B and H, respectively. However, in turn, the exon products 7 + 8 and 8 + 9 seemed to be less potent inhibitors than the intact D3 (10, 200 nM) or HK (200, 500 nM) molecule. These results clearly indicate that an intact molecule of HK or its domain 3 as a whole is required for optimal inhibition of cathepsins B and H.


Subject(s)
Cathepsin B/antagonists & inhibitors , Cathepsin B/chemistry , Cathepsins/antagonists & inhibitors , Cathepsins/chemistry , Cysteine Endopeptidases , Enzyme Inhibitors/pharmacology , Kininogens/pharmacology , Amino Acid Sequence , Cathepsin H , Humans , Kinetics , Molecular Sequence Data
4.
J Biol Chem ; 271(19): 11228-35, 1996 May 10.
Article in English | MEDLINE | ID: mdl-8626672

ABSTRACT

The ability of kininogens to modulate thrombin-induced aggregation of human platelets has been assigned to domain 3 (D3) in the common heavy chain coded for by exons 7, 8, and 9 of kininogen gene. We expressed each of the exons 7, 8, and 9, and various combinations as glutathione S-transferase fusion proteins in Escherichia coli. Each of the exon products 7 (Lys236-Gln292), 9 (Val293-Gly328), and 8 (Gln329-Met357), and their combinations were evaluated for the ability to inhibit thrombin induced platelet aggregation. Only products containing exon 7 inhibited platelet aggregation induced by thrombin with an IC50 of > 20 microM. A deletion mutant of exon 7 product, polypeptide 7A product (Lys236-Lys270) did not block thrombin-induced platelet aggregation, while 7B product (Thr255-Gln292) and 7C product (Leu271-Gln292) inhibited aggregation. These findings indicated that the inhibitory activity is localized to residues Leu271-Gln292. Peptides Phe279-Ile283 and Phe281-Gln292 did not block thrombin, and Asn275-Phe279 had only minimal inhibitory activity. A heptapeptide Leu271-Ala277 inhibited thrombin-induced aggregation of platelets with an IC50 of 65 microM. The effect is specific for the activation of platelets by thrombin but not ADP or collagen. No evidence for a thrombin-kininogen complex was found, and neither HK nor its derivatives directly inhibited thrombin activity. Knowledge of the critical sequence of kininogen should allow design of compounds that can modulate thrombin activation of platelets.


Subject(s)
Kininogens/pharmacology , Peptide Fragments/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Thrombin/pharmacology , Adenosine Diphosphate/pharmacology , Amino Acid Sequence , Base Sequence , Collagen/pharmacology , Exons , Fibrinogen/metabolism , Humans , In Vitro Techniques , Kinetics , Kininogens/biosynthesis , Kininogens/chemistry , Macromolecular Substances , Models, Molecular , Molecular Sequence Data , Oligodeoxyribonucleotides , Peptide Fragments/biosynthesis , Peptide Fragments/chemistry , Plasmids , Protein Conformation , Recombinant Fusion Proteins/biosynthesis , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Sequence Deletion , Structure-Activity Relationship , Thrombin/antagonists & inhibitors
6.
Chem Biol Interact ; 98(3): 211-22, 1995 Dec 22.
Article in English | MEDLINE | ID: mdl-8548860

ABSTRACT

Chronic exposure of humans to benzene causes severe bone marrow cell depression leading to aplastic anemia. Marrow stromal macrophage dysfunction and deficient interleukin-1 production has been reported for patients with severe aplastic anemia. The stromal macrophage, a target of benzene toxicity, is involved in hematopoietic regulation through the synthesis of several cytokines including interleukin-1, which is required for production by stromal fibroblasts of a number of cytokines required for the survival of hematopoietic progenitor cells. We have previously demonstrated that hydroquinone, a major toxic metabolite of benzene in marrow, prevents the proteolytic conversion of 31 kDa pre-interleukin-1 alpha to the 17 kDa cytokine by calpain in purified murine stromal macrophages. Furthermore, stromal macrophages from benzene-treated mice produce the 31 kDa pre-interleukin-1 alpha when stimulated in culture with endotoxin, but cannot convert the precursor to interleukin-1 alpha. In this report, we show that 1,4-benzoquinone, the oxidation product of hydroquinone in the cell, causes a concentration-dependent inhibition of highly purified human platelet calpain with an IC50 of 3 microM. Hydroquinone also inhibits the processing of pre-interleukin-1 beta by interleukin-1 beta convertase. The addition of 2 microM hydroquinone to B1 cells that undergo autocrine stimulation by interleukin-1 beta resulted in the cessation of autocrine cell growth and interleukin-1 beta secretion into the culture medium, as determined by Western immunoblots of the culture supernatants. Purified converting enzyme treated with 3 microM benzoquinone was incapable of converting 31 kDa recombinant pre-interleukin-1 beta to the 17 kDa mature cytokine as analyzed by polyacrylamide gel electrophoresis and Western immunoblotting. These findings support our observations in a mouse model that benzene-induced bone marrow cell depression results from a lack of interleukin-1 alpha subsequent to an inhibition by benzoquinone of calpain, the protease required for converting pre-interleukin-1 alpha to active cytokine. The results may provide a basis for studying benzene-induced aplastic anemia in a mouse model.


Subject(s)
Benzoquinones/toxicity , Calpain/antagonists & inhibitors , Cysteine Endopeptidases/metabolism , Interleukin-1/metabolism , Protein Precursors/metabolism , Benzene/metabolism , Benzene/toxicity , Blotting, Western , Caspase 1 , Cell Division/drug effects , Cysteine Proteinase Inhibitors/toxicity , Humans , Hydroquinones/toxicity , Indomethacin/toxicity , Macrophages/drug effects , Macrophages/metabolism , Recombinant Proteins/metabolism , Tumor Cells, Cultured
7.
Pept Res ; 7(1): 32-5, 1994.
Article in English | MEDLINE | ID: mdl-8019060

ABSTRACT

Thrombin-induced platelet aggregation has been suggested to play an important role in reocclusion following thrombolytic therapy or angioplasty for treatment of myocardial infarction. We previously demonstrated that thrombin-induced platelet aggregation is indirectly mediated by intracellularly activated calpain expressed on the platelet surface through the cleavage of aggregin, a putative ADP-receptor, and that high molecular weight kininogen (HK), a naturally occurring thiol protease inhibitor, modulates thrombin-induced platelet aggregation. Considering the substrate specificity of calpain and the conserved sequence in HK, we studied selective inhibitors of thrombin-induced platelet aggregation by the affinity labeling approach with an S-3-nitro-2-pyridinesulfenyl (Npys) group. H-Phe-Gln-Val-Val-Cys (Npys)-Gly-NH2, which combines chemical and structural features of calpain substrate specificity and the conserved sequence in HK, selectively inhibited thrombin-induced platelet aggregation. It did not inhibit the aggregatory effects of other platelet agonists, and did not inhibit amidolytic activity of thrombin and thrombin-induced platelet shape change. The design and synthesis of such inhibitors could lead to the development of a new class of inhibitors that selectively block thrombin-induced platelet aggregation.


Subject(s)
Kininogens/chemical synthesis , Kininogens/pharmacology , Peptides/chemical synthesis , Peptides/pharmacology , Platelet Aggregation Inhibitors/chemical synthesis , Platelet Aggregation Inhibitors/pharmacology , Thrombin/pharmacology , Amino Acid Sequence , Calpain/antagonists & inhibitors , Molecular Sequence Data , Pyridines/chemistry , Structure-Activity Relationship
8.
J Biol Chem ; 268(35): 26546-51, 1993 Dec 15.
Article in English | MEDLINE | ID: mdl-8253784

ABSTRACT

Both high molecular weight kininogen (HK) and low molecular weight kininogens (LK) are potent tight binding inhibitors of platelet calpain (Ki = 2 nM), but the molecular basis for the inhibitory function is not well delineated. The amino acid sequences of the calpain inhibitory domain 2 from human and rat HK were compared for homology with the noninhibitory domains from human and rat domain 3 and from domain 2 of rat T-kininogen, and two areas of nonconserved differences were detected. Computer three-dimensional models were constructed on a template built using the x-ray crystallographic data for cystatin, an evolutionary precursor of HK. Two nonconserved regions in the calpain inhibitory domains flank the highly conserved motif QVVAG to form a continuous surface for interaction with cysteine proteases. Three peptide sequences, components of the modeled surface, were chosen for synthesis from HK D-2: VHPISTQSPDLE (peptide 146-156, NH2-terminal), CTDNAYIDIQLRIASFSQNC (peptide 229-248, COOH-terminal), and CQRQVVAGLNFRIC (185-189, central) containing QVVAG. This last peptide differs from the natural sequence by substitutions of A185C and T195C. Peptides 185-198 and 229-248 were folded by air oxidation of their cysteine residues and then tested for their ability to inhibit calpain and papain. The folded peptide 229-248 inhibited calpain with an IC50 35 microM and unfolding reduced this effect. The folded peptide 185-198 did not inhibit calpain, but when preincubated with calpain, could block the inhibition by HK indicating a probable enzyme binding site. Peptide 146-157 did not inhibit calpain but could inhibit papain with an IC50 of 20 microM. We have thus defined separate binding and inhibitory sequences on HK which form a contiguous surface for thiol protease interactions.


Subject(s)
Blood Platelets/metabolism , Calpain/antagonists & inhibitors , Kininogens/metabolism , Amino Acid Sequence , Animals , Binding Sites , Computer Graphics , Humans , Kininogens/chemistry , Molecular Sequence Data , Protein Binding , Protein Conformation , Rats , Rats, Wistar , Sequence Homology, Amino Acid
9.
Eur J Biochem ; 214(1): 233-41, 1993 May 15.
Article in English | MEDLINE | ID: mdl-8389701

ABSTRACT

Thrombin-induced platelet aggregation has been suggested to play an important role in reocclusion following thrombolytic therapy of angioplasty for treatment of myocardial infarction. We previously demonstrated that aggregation of washed platelets by thrombin is accompanied by cleavage of aggregin, a putative ADP receptor, and that these events are indirectly mediated by calpain, expressed on the surface of the external membrane. High-molecular-mass kininogen (HK) contains, in its heavy chain, domain 2, which is responsible for its action as a potent inhibitor of platelet calpain. Domain 3 of the heavy chain of HK directly inhibits binding of thrombin to platelets, confounding mechanistic studies using the entire molecule. Moreover, HK, a protease of 120 kDa, is unsuitable as a potential pharmacological agent. The highly conserved sequence Gln-Val-Val-Ala-Gly, present in HK and its evolutionary precursors, the cystatins, is thought to be involved in the binding of cysteine proteases but is, itself, not inhibitory. An affinity analog, Phe-Gln-Val-Val-Cys(Npys)-Gly-NH2(Npys, 3-nitro-2-sulfenylpyridine), P1, corresponding to the thiol-protease-binding sequence in HK and containing a ligand, Npys, that can react with the free sulfhydryl group in the active site of calpain, was synthesized. P1 was an irreversible inhibitor of platelet calpain. P1 selectively inhibited thrombin-induced aggregation of washed platelets and platelets in plasma, but did not inhibit the aggregatory effects of other platelet agonists. P1 did not inhibit the amidolytic activity and coagulant activity of thrombin. Unlike HK, P1 did not inhibit binding of thrombin to washed platelets. P1 did not inhibit thrombin-induced platelet-shape change. P1 neither raised intracellular levels of cAMP nor did it interfere with the ability of thrombin to antagonize the rise in intracellular levels of cAMP induced by iloprost, an analog of prostaglandin I2. The design and synthesis of P1 could leave to the development of a new class of inhibitors that selectively block thrombin-induced platelet aggregation while sparing other functions of this pathophysiological protease and without inhibiting the action of other platelet agonists.


Subject(s)
Calpain/antagonists & inhibitors , Oligopeptides/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Thrombin/pharmacology , Amino Acid Sequence , Blood Platelets/metabolism , Calpain/isolation & purification , Cyclic AMP/blood , Cysteine Endopeptidases/metabolism , Electrophoresis, Polyacrylamide Gel , Fibrinogen/metabolism , Humans , Kininogens/chemistry , Kininogens/metabolism , Molecular Sequence Data , Oligopeptides/chemical synthesis , Platelet Aggregation Inhibitors/chemical synthesis , Platelet Membrane Glycoproteins/metabolism , Sequence Alignment , Thrombin/metabolism
10.
Biochem J ; 270(1): 83-90, 1990 Aug 15.
Article in English | MEDLINE | ID: mdl-2396995

ABSTRACT

The plasma kininogens, high-molecular-mass and low-molecular-mass kininogens, are the most potent plasma inhibitors of platelet calpain. We explored the kinetic mechanisms for kininogen inhibition of calpain by comparing calpain inactivation by human high-molecular-mass kininogen (HK) and human low-molecular-mass kininogen (LK). With a [14C]methylated alpha-casein substrate, the inhibition of calpain by HK did not follow classic Michaelis-Menten kinetics. With the use of a fluorogenic assay with the dipeptide substrate for calpain, 3-carboxypropionyl-leucyltyrosine 7-(4-methyl)coumarylamide, the inhibition by HK and LK fitted a kinetic model of tight-binding inhibition. LK was found to be a non-competitive inhibitor of platelet calpain with a Ki of 2.7 nM. HK showed mixed non-competitive inhibition of calpain with a Ki of 2.3 nM in the absence of substrate and Ki of 0.71 nM in the presence of saturating substrate, almost 4-fold tighter than LK. Proteolysis of HK by plasma and tissue kallikreins did not influence its ability to inhibit calpain. Digestion of the HK light chain by Factor XIa also did not alter its calpain-inhibitory function. These studies indicate that the kininogens are tight-binding non-competitive inhibitors of platelet calpain, the inhibitory domain in each case being mainly on the heavy chain. The light chain of HK appears to influence its kinetic behaviour.


Subject(s)
Blood Platelets/enzymology , Calpain/antagonists & inhibitors , Kininogens/pharmacology , Caseins/metabolism , Humans , In Vitro Techniques , Kinetics , Molecular Weight , Peptide Fragments/pharmacology , Structure-Activity Relationship
11.
Blood ; 75(6): 1273-81, 1990 Mar 15.
Article in English | MEDLINE | ID: mdl-2310827

ABSTRACT

Platelet calpain has many platelet substrates, including external membrane proteins. We thus investigated whether platelet calpain II was associated with platelet membranes in unstimulated and thrombin-activated platelets. A monospecific, goat polyclonal antibody was reared to purified platelet calpain II. Sixteen whole platelet lysates were found to contain 4.5 +/- 0.7 micrograms calpain antigen II per 10(8) platelets (mean +/- SEM) as determined by a competitive enzyme-linked immunosorbent assay. Using the dipeptide fluorogenic substrate, Suc-Leu-Tyr-MCA, 17 human platelet lysates contained 3.6 +/- 0.4 micrograms calpain activity per 10(8) platelets. Platelet calpain II was associated with the Triton X-100 insoluble platelet cytoskeletons from both unstimulated and thrombin-activated platelets. When compared with the total cell content of platelet calpain II, calpain antigen (10% to 13%) and calpain activity (24% to 28%) was associated with platelet cytoskeletons in unstimulated and thrombin-activated platelets, respectively. On immunoblot, the heavy chain (80 Kd) of calpain II was detected in platelet cytoskeletons. Subcellular fractionation studies on both unstimulated and thrombin-activated platelets, revealed that half of the total platelet calpain II antigen was associated with cytosol, and the other half was associated with the membrane fraction. Platelet calpain II was not seen on the surface of unstimulated, paraformaldehyde fixed platelets by immunofluorescence. However, on thrombin-activated platelets, rim immunofluorescence was seen, indicating that activated platelets externalize their calpain. This observation was confirmed by the finding that about 2,000 molecules per platelet of an 125I-anti-calpain II Fab' specifically bound to thrombin-activated but not unstimulated platelets. Both dibucaine (1 mmol/L) and platelet activating factor (1.86 mumol/L) in the absence of external Ca++, but not collagen (5 micrograms/mL) or ionophore A23187 (2.5 mumol/L) in the absence of external Ca++, were also able to externalize platelet calpain II antigen, as indicated by a similar level of specific 125I-anti-calpain II Fab'-platelet binding. These combined studies indicate that platelet calpain II is a major protein, comprising 2% of total platelet protein, a substantial portion of which is membrane-associated. When platelets are activated by thrombin and platelet activating factor, calpain II antigen also becomes present on the external platelet surface.


Subject(s)
Blood Platelets/metabolism , Calpain/metabolism , Cell Membrane/metabolism , Antibodies/analysis , Antibodies/immunology , Blood Platelets/analysis , Blood Platelets/ultrastructure , Calpain/analysis , Calpain/immunology , Cell Fractionation , Cell Membrane/ultrastructure , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Humans , Immunohistochemistry , Platelet Activation/drug effects , Platelet Activation/physiology , Thrombin/pharmacology
12.
Thromb Res ; 54(2): 115-23, 1989 Apr 15.
Article in English | MEDLINE | ID: mdl-2473540

ABSTRACT

High molecular weight kininogen has been shown to be the principal plasma inhibitor of cellular thiol proteases including cathepsins B, H and L and calpains 1 and 2. Since these same enzymes have been reported to be elevated in animals with muscular dystrophy, we studied plasmas from hamsters with muscular dystrophy and compared these to normal hamster plasma. The ability of plasma to inhibit purified platelet calpain was assayed and found to be 62% of normal. Since low molecular weight kininogen can also inhibit calpain, the coagulant activity of kininogen, an activity unique for high molecular weight kininogen, was determined in dystrophic hamster plasma and found to be 69% of normal in close agreement with the calpain inhibitory activity. The contribution of the other plasma calpain inhibitor alpha 2-macroglobulin appeared small since inactivation with methylamine did not alter the ability to inhibit calpain in either normal or dystrophic plasma. We conclude that there is a selective deficiency of plasma high molecular weight kininogen in dystrophic hamsters, an abnormality which could play a role in the pathogenesis of this disorder.


Subject(s)
Kininogens/deficiency , Muscular Dystrophy, Animal/metabolism , Protease Inhibitors/metabolism , Age Factors , Animals , Calpain/blood , Cricetinae , Whole Blood Coagulation Time , alpha-Macroglobulins/analysis
13.
Trans Assoc Am Physicians ; 102: 13-9, 1989.
Article in English | MEDLINE | ID: mdl-2561638

ABSTRACT

1. A synthetic peptide disulfide, Gln-Val-Val-Cys(NpyS)-Gly-NH2 (P1) inhibited thrombin and plasmin-induced platelet aggregation and cleavage of aggregin. P1 did not inhibit platelet aggregation induced by other agonists nor did it inhibit shape change. 2. P1 also inhibited purified platelet calpain II. 3. The correspondence between the molecular structure of P1 and inhibitory sequence of the peptide in domain 2 of high molecular weight kininogen has shed light on the molecular nature of the cellular mechanism underlying thrombin- and plasmin-induced platelet aggregation and the inhibition by P1. 4. P1 may prove to be useful in designing and improving future protocols of thrombolytic therapy to prevent reocclusion. P1 may also have a role in inhibiting thrombin formed during angioplasty and thus preventing restenosis.


Subject(s)
Oligopeptides/pharmacology , Platelet Aggregation/drug effects , Amino Acid Sequence , Calpain/antagonists & inhibitors , Fibrinolysin/pharmacology , Humans , In Vitro Techniques , Kininogens/pharmacology , Molecular Sequence Data , Platelet Aggregation Inhibitors , Thrombin/pharmacology
14.
Blood ; 71(2): 388-94, 1988 Feb.
Article in English | MEDLINE | ID: mdl-2827812

ABSTRACT

Platelets are known to process human factor V during secretion and/or membrane binding. We studied the functional and structural changes produced in human factor V by purified human platelet calpain (calcium-activated thiol protease) and compared the alterations with those induced by thrombin. A maximum increase in coagulant activity of 2.5-fold was observed when factor V (1 U/mL, 33 nmol/L) was incubated with calpain (0.03 U/mL, 2.7 nmol/L) in comparison with a 8.8-fold increment for alpha-thrombin (0.7 U/mL, 8 nmol/L) at 25 degrees C. Thrombin additions to reactions initiated by calpain resulted in further activation comparable to that of thrombin alone, whereas the subsequent addition of calpain had no effect on the extent or pattern of the activation of factor V by thrombin. The cleavage pattern of factor V produced by these two enzymes are distinctly different. Although thrombin activation eventually results in four final components designated C1 (150 kd), D (105 kd), E (71 kd), and F1F2 (71 to 74 kd), calpain yields initial components of 200 kd and 160 kd within one minute. Further digestion of the 200 kd species by calpain gives rise first to a polypeptide of 160 kd that is converted to a 140 kd and a 120 kd species by two minutes with an increase in coagulant activity. Immunoblotting of these fragments with the monoclonal antibody (MoAb) B10 directed to factor V and the thrombin-generated C1 fragment yields results demonstrating a common epitope in these calpain-generated components of 200, 160, 140 and 120 kd. The degradation of the initial 160 kd polypeptide gives rise to polypeptides of 100 and 65 kd, both undetectable on immunoblotting with MoAb B10. The 130, 87, 58, and 48 kd components are of less certain origin. Thus, platelet calpain generates a complex but reproducible cleavage pattern different from thrombin that may explain the partial activation observed. Nevertheless, calpain processing may play a role in early hemostatic reactions involving platelets before the appearance of the first thrombin molecule.


Subject(s)
Blood Platelets/enzymology , Calpain/metabolism , Factor V/metabolism , Thrombin/metabolism , Antibodies, Monoclonal , Blood Coagulation , Enzyme Activation , Humans , Kinetics , Molecular Weight , Peptide Fragments/analysis
15.
J Biol Chem ; 262(3): 1405-11, 1987 Jan 25.
Article in English | MEDLINE | ID: mdl-2433279

ABSTRACT

In normal human plasma two forms of kininogen exist, low molecular weight kininogen (LMWK) and high molecular weight kininogen (HMWK). When these proteins are cleaved they are found to have a common heavy chain and bradykinin, but each has a unique light chain. Monoclonal antibodies to the heavy and light chains of HMWK have been developed, and the effects of each on the function of this protein are defined. Initial studies showed that an antibody, C11C1, completely neutralized the coagulant activity of plasma HMWK whereas another antibody, 2B5, did not. On a competitive enzyme-linked immunosorbent assay (CELISA) the C11C1 antibody was consumed by kininogen antigen in normal plasma but not by kininogen antigen in HMWK-deficient plasma. On immunoblot, the C11C1 antibody recognized one kininogen protein in normal plasma and did not recognize any kininogen antigen in HMWK-deficient plasma. These combined studies indicated that the C11C1 antibody was directed to an epitope on the unique 46-kDa light chain of HMWK. In contrast, the 2B5 antibody on a CELISA was consumed by kininogen antigen in both normal plasma and HMWK-deficient plasma but not by total kininogen-deficient plasma. On immunoblot, the 2B5 antibody recognized both kininogens in normal plasma but only LMWK in HMWK-deficient plasma. These combined studies indicated that the 2B5 antibody was directed to the common 64-kDa heavy chain of the plasma kininogens. Utilizing direct binding studies or competition kinetic experiments, the 2B5 and C11C1 antibodies bound with high affinity (1.71 and 0.77 nM, respectively) to their antigenic determinants on the HMWK molecule. The 2B5 antibody did neutralize the ability of HMWK to inhibit platelet calpain. These studies with monoclonal antibodies directed to each of the HMWK chains indicate that HMWK is a bifunctional molecule that can serve as a cofactor for serine zymogen activation and an inhibitor of cysteine proteases.


Subject(s)
Antibodies, Monoclonal/immunology , Kininogens/blood , Animals , Antibody Affinity , Binding, Competitive , Blood Coagulation , Blood Platelets/enzymology , Calpain/antagonists & inhibitors , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Female , Humans , Immunosorbent Techniques , Kininogens/immunology , Kininogens/pharmacology , Male , Mice , Mice, Inbred BALB C , Peptide Fragments/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...