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










Publication year range
1.
Colorectal Dis ; 18(9): 910-8, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26456021

ABSTRACT

AIM: This study was performed to determine the impact of a surgical site infection (SSI) reduction strategy on SSI rates following colorectal resection. METHOD: American College of Surgeons National Surgical Quality Improvement Program (NSQIP) data from 2006-14 were utilized and supplemented by institutional review board-approved chart review. The primary end-point was superficial and deep incisional SSI. The inclusion criterion was colorectal resection. The SSI reduction strategy consisted of preoperative (blood glucose, bowel preparation, shower, hair removal), intra-operative (prophylactic antibiotics, antimicrobial incisional drape, wound protector, wound closure technique) and postoperative (wound dressing technique) components. The SSI reduction strategy was prospectively implemented and compared with historical controls (pre-SSI strategy arm). Statistical analysis included Pearson's chi-square test, and Student's t-test performed with spss software. RESULTS: Of 1018 patients, 379 were in the pre-SSI strategy arm, 311 in the SSI strategy arm and 328 were included to test durability. The study arms were comparable for all measured parameters. Preoperative wound class, operation time, resection type and stoma creation did not differ significantly. The SSI strategy arm demonstrated a significant decrease in overall SSI rates (32.19% vs 18.97%) and superficial SSI rates (23.48% vs 8.04%). Deep SSI and organ space rates did not differ. A review of patients testing durability demonstrated continued improvement in overall SSI rates (8.23%). CONCLUSION: The implementation of an SSI reduction strategy resulted in a 41% decrease in SSI rates following colorectal resection over its initial 3 years, and its durability as demonstrated by continuing improvement was seen over an additional 2 years.


Subject(s)
Abdominal Wound Closure Techniques , Colectomy/methods , Colonic Diseases/surgery , Rectum/surgery , Surgical Wound Infection/prevention & control , Aged , Antibiotic Prophylaxis/methods , Bandages , Case-Control Studies , Chlorhexidine/therapeutic use , Disinfectants/therapeutic use , Enema , Female , Hair Removal/methods , Historically Controlled Study , Humans , Hygiene , Hyperglycemia/drug therapy , Male , Middle Aged , Prospective Studies
3.
Tech Coloproctol ; 19(3): 125-6, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25708683
4.
J Thromb Haemost ; 8(6): 1333-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20345708

ABSTRACT

BACKGROUND: The principal inhibitor of fibrinolysis in vivo is plasminogen activator inhibitor-1 (PAI-1). PAI-749 is a small molecule inhibitor of PAI-1 with proven antithrombotic efficacy in several preclinical models. OBJECTIVE: To assess the effect of PAI-749, by using an established ex vivo clinical model of thrombosis and a range of complementary in vitro human plasma-based and whole blood-based models of fibrinolysis. METHODS: In a double-blind, randomized, crossover study, ex vivo thrombus formation was assessed using the Badimon chamber in 12 healthy volunteers during extracorporeal administration of tissue-type plasminogen activator (t-PA) in the presence of PAI-749 or control. t-PA-mediated lysis of plasma clots and of whole blood model thrombi were assessed in vitro. The role of vitronectin was examined by assessing lysis of fibrin clots generated from purified plasma proteins. RESULTS: There was a dose-dependent reduction in ex vivo thrombus formation by t-PA (P < 0.0001). PAI-749 had no effect on in vitro or ex vivo thrombus formation or fibrinolysis in the presence or absence of t-PA. Inhibition of PAI-1 with a blocking antibody enhanced fibrinolysis in vitro (P < 0.05). CONCLUSIONS: Despite its efficacy in a purified human system and in preclinical models of thrombosis, the current study suggests that PAI-749 does not affect thrombus formation or fibrinolysis in a range of established human plasma and whole blood-based systems.


Subject(s)
Fibrinolysis/drug effects , Indoles/pharmacology , Plasminogen Activator Inhibitor 1 , Tetrazoles/pharmacology , Adult , Cross-Over Studies , Double-Blind Method , Humans , Models, Biological
5.
J Thromb Haemost ; 7(11): 1915-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19691482

ABSTRACT

BACKGROUND: Interaction of P-selectin with its glycoprotein ligand (P-selectin glycoprotein ligand type 1) mediates inflammatory processes that may also include vascular thrombosis. Platelet P-selectin expression is increased in patients with coronary heart disease, and its antagonism represents a potential future therapeutic target for the prevention and treatment of atherothrombosis. AIM: To investigate the effects of the novel small molecule P-selectin antagonist PSI-697 on thrombus formation in humans. METHODS AND RESULTS: In a double-blind randomized crossover study, thrombus formation was measured in 12 healthy volunteers, using the Badimon ex vivo perfusion chamber under conditions of low and high shear stress. Saline placebo, low-dose (2 m) and high-dose (20 m) PSI-697 and the glycoprotein IIb-IIIa receptor antagonist tirofiban (50 ng mL(-1)) were administered into the extracorporeal circuit prior to the perfusion chamber. As compared with saline placebo, blockade of platelet glycoprotein IIb-IIIa receptor with tirofiban produced 28% and 56% reductions in thrombus formation in the low-shear and high-shear chambers, respectively. PSI-697 caused a dose-dependent, but more modest, reduction in thrombus formation. Low-dose PSI-796 (2 m) reduced total thrombus area by 14% (P = 0.04) and 30% (P = 0.0002) in the low-shear and high-shear chambers, respectively. At the high dose (20 m), PSI-697 reduced total thrombus area by 18% (P = 0.0094) and 41% (P = 0.0008) in the low-shear and high-shear chambers, respectively. CONCLUSIONS: P-selectin antagonism with PSI-697 reduces ex vivo thrombus formation in humans. These findings provide further evidence that P-selectin antagonism may be a potential target for the prevention and treatment of cardiovascular disease.


Subject(s)
P-Selectin/antagonists & inhibitors , Thrombosis/prevention & control , Adolescent , Adult , Blood/drug effects , Cells, Cultured , Cross-Over Studies , Double-Blind Method , Humans , Hydroxyquinolines/pharmacology , Perfusion , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Thrombosis/drug therapy , Tirofiban , Tyrosine/analogs & derivatives , Tyrosine/pharmacology , Young Adult
6.
J Mol Cell Cardiol ; 36(1): 111-9, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14734053

ABSTRACT

In the search for a readily available source of native cardiac cells, we investigated the molecular and pharmacological properties of the immortalized cardiac atrial myocyte cell line, HL-1 cells. This work focused on the expression pattern of voltage-gated Ca2+ channels (VGCC). Reverse transcription-polymerase chain reaction analysis revealed that HL-1 cells have mRNA for several types of Ca2+ channels including the L-types, alpha1C and alpha1D, as well as T-types, alpha1H and alpha1G, but are lacking N-type, alpha1B and the T-type, alpha1I. Western blot analysis demonstrated significant alpha1C protein subunit expression, with less alpha1D subunit apparent, while alpha1A, alpha1B and alpha1E subunit expression was undetectable. Immunocytochemical staining showed that the alpha1C protein subunit is expressed predominantly on the cell surface, whereas the alpha1D protein is expressed mostly intracellularly. Whole-cell patch-clamp measurements demonstrated the presence of low (ICa,T) and high (ICa,L) voltage-activated Ca2+ currents, with preferential sensitivity to mibefradil and nimodipine, respectively. Addition of increasing external Ca2+ concentrations, [Ca2+]o, resulted in Ca2+ influx measured by fluorometric imaging with an EC50 of 0.8 mM [Ca2+]o. At a fixed [Ca2+]o of 0.125 mM, Ca2+ influx was also triggered by increasing the extracellular K+ concentration, [K+]o, with an EC50 of 3.7 mM [K+]o. As increasing [K+]o depolarizes the cell, this latter result is consistent with Ca2+ influx through a voltage-dependent mechanism. L-type (nimodipine and verapamil) and T-type (mibefradil and pimozide) Ca2+ channel blockers inhibited Ca2+ influx with IC50s of 1, 2, 0.4 and 0.2 microM, respectively. Antagonists of N-type (omega-conotoxins GVIA) and P/Q-type (MVIIC or omega-agatoxin IVA) did not inhibit Ca2+ influx, consistent with the lack of expression of N-, P-, or Q-type channels observed in the molecular studies. Taken together, these findings indicate that HL-1 cells express L- and T-subtypes of VGCC and are a unique in vitro model system for the study of native, mammalian cardiac Ca2+ channels.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Channels, T-Type/metabolism , Calcium/metabolism , Myocytes, Cardiac/metabolism , Animals , Calcium/pharmacology , Calcium Channels, L-Type/genetics , Calcium Channels, T-Type/genetics , Cell Line , Electric Conductivity , Heart Atria/cytology , Ion Transport/drug effects , Mibefradil/pharmacology , Mice , Myocytes, Cardiac/drug effects , Nimodipine/pharmacology , Patch-Clamp Techniques , Pimozide/pharmacology , Potassium/pharmacology , Verapamil/pharmacology
7.
Article in English | MEDLINE | ID: mdl-15317290

ABSTRACT

The thrombin receptor on human platelets is the first member identified of a new family of G-protein coupled receptors referred to as protease activated receptors (PARs). These receptors are activated by a unique mechanism involving proteolytic cleavage of a portion of the extracellular domain to generate a new N-terminus which then acts as a tethered or intramolecular ligand (agonist) for the receptor. The hexapeptide SFLLRN-NH2 comprising the new N-terminus is referred to as the Thrombin Receptor Activating Peptide, or "TRAP" Thrombin is the most potent agonist for platelet aggregation and selective blockade of the intramolecular activation step without effecting the proteolytic activity of thrombin should result in moderation of platelet activation and aggregation without interfering with the other coagulation cascade effects of thrombin. Screening of combinatorial libraries identified a novel, non-peptide PAR-1 thrombin receptor antagonist. Examination of structure-activity relationships revealed that portions of the molecule could be replaced resulting in simpler molecules of lower molecular weight that were at the same time more potent. Molecules in this series were effective antagonists of TRAP-stimulated platelet activation, but had limited activity when thrombin was the agonist. Additional directed screening and subsequent lead refinement resulted in a second series of isoxazole based compounds. Some of the resultant molecules were potent PAR-1 antagonists that were effective against both TRAP- and thrombin-stimulated receptor activation. These compounds do not inhibit the proteolytic effects of thrombin but rather interfere with the intramolecular binding of the tethered ligand (SFLLRN) to the transmembrane portion of the thrombin receptor. They represent promising leads for future explorations of antithrombotic activity of thrombin receptor antagonists.


Subject(s)
Isoxazoles/pharmacology , Receptor, PAR-1/antagonists & inhibitors , Urea/analogs & derivatives , Urea/pharmacology , Blood Platelets/drug effects , Humans , Inhibitory Concentration 50 , Isoxazoles/chemistry , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/pharmacology , Structure-Activity Relationship , Urea/chemistry
8.
Bioorg Med Chem Lett ; 11(20): 2691-6, 2001 Oct 22.
Article in English | MEDLINE | ID: mdl-11591503

ABSTRACT

Thrombin is the most potent agonist of platelet activation, and its effects are predominantly mediated by platelet thrombin receptors. Therefore, antagonists of the thrombin receptor have potential utility for the treatment of thrombotic disorders. Screening of combinatorial libraries revealed 2 to be a potent antagonist of the thrombin receptor. Modifications of this structure produced 11k, which inhibits thrombin receptor stimulated secretion and aggregation of platelets.


Subject(s)
Receptors, Thrombin/antagonists & inhibitors , Urea/pharmacology , Platelet Activation/drug effects , Receptor, PAR-1 , Structure-Activity Relationship , Urea/chemistry
9.
Acta Crystallogr D Biol Crystallogr ; 57(Pt 8): 1071-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11468390

ABSTRACT

Leech anti-platelet protein (LAPP) from the leech Haementeria officinalis is a collagen-binding protein that inhibits the collagen-mediated adhesion of blood platelets. The crystal structure of recombinant LAPP has been determined using single isomorphous replacement with anomalous scattering combined with solvent flattening and threefold molecular averaging. The model of LAPP has been refined to 2.2 A resolution (R factor 21.5%; free R factor 24.0%). LAPP contains an 89-residue C-terminal domain consisting of a central six-stranded antiparallel beta-sheet flanked on one side by an alpha-helix and on the other side by two extended loops with little secondary structure. A 36-residue N-terminal region is not visible in the electron-density map. This region is rich in glycine and lacks hydrophobic residues. It probably does not have a compact globular fold, but instead has an extended conformation and is flexible. The crystal packing suggests that LAPP may form tightly interacting dimers. The fold of the C-terminal domain of LAPP closely resembles that of the N-domain of hepatocyte growth factor (HGF), which classifies LAPP as a PAN domain. However, no significant sequence homology exists between LAPP and other PAN domains. Common structural features between LAPP and the HGF N-domain include two disulfide bonds that link the alpha-helix to the central region of the protein and five residues with a conserved hydrophobic nature that are located in the core of the domain. These conserved structural features may be an important determinant of the PAN-domain type of fold.


Subject(s)
Hemostasis/drug effects , Leeches/chemistry , Salivary Proteins and Peptides/chemistry , Amino Acid Sequence , Animals , Binding Sites , Collagen/metabolism , Crystallization , Crystallography, X-Ray , Hepatocyte Growth Factor/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Structure, Tertiary , Salivary Proteins and Peptides/metabolism , Salivary Proteins and Peptides/pharmacology , Sequence Homology, Amino Acid
10.
Biochem Biophys Res Commun ; 241(3): 744-8, 1997 Dec 29.
Article in English | MEDLINE | ID: mdl-9434779

ABSTRACT

Disagregin, a 6 kDa protein isolated from salivary glands of the tick Ornithodoros Moubata, is a potent and selective inhibitor of fibrinogen dependent platelet aggregation and of the adhesion of platelets to fibrinogen (Karczewski et al. (1994) J. Biol. Chem. 269, 6702-6708). In the current study the interaction of disagregin with purified glycoprotein IIb-IIIa (GPIIb-IIIa) was examined. Biotin-labeled disagregin (b-disagregin) bound to GPIIb-IIIa immobilized on the surface of the ELISA plate. This binding was specific, dependent on divalent cations, and was blocked by the peptides fibrinogen gamma-chain fg gamma (400-411), GPIIb(296-306) and by the RGD-containing peptide, GRGDSP. Disagregin also bound to soluble GPIIb-IIIa as demonstrated in studies using the chemical crosslinker, BS3. This binding was inhibited by the peptides fg gamma (400-411) and GPIIb(296-306). In contrast to the results in the solid phase, peptide GRGDSP had no effect on the binding of b-disagregin to soluble GPIIb-IIIa. These data demonstrate that disagregin binds to GPIIb-IIIa through a mechanism distinct from that used by RGD-containing disintegrins. Further analysis of the region(s) of disagregin which bind to GPIIb-IIIa should provide useful information for molecular modeling of the fibrinogen binding site on GPIIb-IIIa and for the design of a new class of potent fibrinogen receptor antagonists.


Subject(s)
Platelet Aggregation Inhibitors/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Salivary Proteins and Peptides/metabolism , Animals , Binding, Competitive , Cross-Linking Reagents , Humans , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Rats , Salivary Proteins and Peptides/blood , Solubility , Ticks
11.
Arterioscler Thromb Vasc Biol ; 15(9): 1424-31, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7670958

ABSTRACT

Salivary glands of the leech Haementeria officinalis contain a protein, leech antiplatelet protein (LAPP). This protein was cloned and expressed in yeast and blocks collagen-mediated platelet aggregation and the adhesion of platelets to collagen-coated plates under static conditions. In the current study we investigated the effect of rLAPP on platelet deposition to collagen and collagen-rich surfaces under flow conditions. rLAPP completely inhibited platelet adhesion on collagen types I, III, and IV with IC50 values of 70, 600, and 90 nmol/L, respectively (shear rate = 1600 s-1). Approximately 10-fold more rLAPP was required to obtain a similar inhibition at a low shear rate of 375 s-1. rLAPP caused a concentration-dependent inhibition of binding of 125I-von Willebrand factor (vWF) to collagen type III and was able to displace prebound vWF even after 24 hours. Since platelet adhesion at low shear rate is less dependent on vWF than at high shear rate, this property of rLAPP may explain why less rLAPP is needed at high shear rate than at low shear rate to produce the same effect. Platelet adhesion to collagen type VI was only partially inhibited by rLAPP (maximal 44% with 3 mumol/L rLAPP). rLAPP also caused a pronounced inhibition of platelet deposition to cross sections of human atherosclerotic coronary arteries but had no effect on matrices of cultured human umbilical vein endothelial cells. rLAPP is a potent platelet adhesion inhibitor at high shear rate, which binds to collagen and works by inhibiting binding of vWF to collagen.


Subject(s)
Blood Platelets/physiology , Collagen/metabolism , Leeches , Platelet Adhesiveness/drug effects , Platelet Aggregation Inhibitors/pharmacology , Salivary Proteins and Peptides/pharmacology , Animals , Biomechanical Phenomena , Coronary Artery Disease/pathology , Coronary Vessels/pathology , Endothelium, Vascular/pathology , Extracellular Matrix/pathology , Female , Fibronectins/metabolism , Humans , Recombinant Proteins/pharmacology , Rheology , von Willebrand Factor/metabolism
12.
J Med Chem ; 38(20): 4125-30, 1995 Sep 29.
Article in English | MEDLINE | ID: mdl-7562949

ABSTRACT

The N-terminal thrombin receptor peptide H-Ser-Phe-Leu-Leu-Arg-Asn-Pro-Asn-Asp-Lys-Tyr-Glu-Pro-Phe-OH (1) fully activates the thrombin receptor with an EC50 of 10 microM. Structural features in the tetradecapeptide which are responsible for receptor activation have been elucidated. Agonist potency has been enhanced 1000-fold with the design of the shortened peptide H-Ala-Phe(p-F)-Arg-Cha-HArg-Tyr-NH2 (56). This analog exhibits an EC50 of 0.01 microM and is the most potent agonist for receptor activation reported to date. The monoiodinated derivative H-Ala-Phe(p-F)-Arg-Cha-HArg-Tyr(3-I)-NH2 (59) exhibits an EC50 of 0.03 microM, a level sufficient for development of a radioligand.


Subject(s)
Oligopeptides/pharmacology , Receptors, Thrombin/agonists , Amino Acid Sequence , Humans , Ligands , Molecular Sequence Data , Structure-Activity Relationship
13.
Circulation ; 91(12): 2961-71, 1995 Jun 15.
Article in English | MEDLINE | ID: mdl-7796507

ABSTRACT

BACKGROUND: Thrombin inhibitors have been shown to be efficacious in animal models of thrombosis and in initial human clinical trials. It is unknown if their efficacy is due to their prevention of thrombin-mediated fibrin formation or to an inhibitory effect on thrombin-stimulated platelet activation. Appropriate tools to address this question have not been available. Therefore, to evaluate the role of the platelet thrombin receptor in intravascular thrombus formation, a polyclonal antibody was raised against a peptide derived from the thrombin-binding exosite region of the cloned human thrombin receptor. This antibody serves as a selective inhibitor of the thrombin receptor for in vivo evaluation. METHODS AND RESULTS: The immune IgG (IgG 9600) inhibited thrombin-stimulated aggregation and secretion of human platelets. In contrast, it had no effect on platelet activation induced by other agonists including ADP, collagen, or the thrombin receptor-derived peptide SFLLR-NH2. IgG 9600 also inhibited thrombin-induced aggregation of African Green monkey (AGM) platelets. By Western blot analysis, the IgG identified a protein of approximately 64 kD in homogenates of both human and AGM platelets. The effect of thrombin receptor blockade by this antibody on arterial thrombosis was evaluated in an in vivo model of platelet-dependent cyclic flow reductions (CFRs) in the carotid artery of the AGM. The intravenous administration of IgG 9600 (10 mg/kg) abolished CFRs in three monkeys and reduced CFR frequency by 50% in a fourth monkey. Ex vivo platelet aggregation in response to up to 100 nmol/L thrombin was completely inhibited during the 120-minute postbolus observation period in all four animals. There was a twofold increase in bleeding time, which was not statistically different from baseline, and ex vivo clotting time (APTT) was not changed. The glycoprotein IIb/IIIa receptor antagonist MK-0852 and the thrombin inhibitor recombinant hirudin also demonstrated inhibitory effects on CFRs at doses that did not significantly prolong template bleeding time. Control IgG had no effect on CFRs, ex vivo platelet aggregation, bleeding time, or APTT. CONCLUSIONS: These results demonstrate that blockade of the platelet thrombin receptor can prevent arterial thrombosis in this animal model without significantly altering hemostatic parameters and suggest that the thrombin receptor is an attractive antithrombotic target.


Subject(s)
Antibodies/therapeutic use , Receptors, Thrombin/immunology , Thrombosis/drug therapy , Animals , Antibody Formation , Chlorocebus aethiops , Platelet Aggregation/drug effects , Receptors, Thrombin/chemistry , Receptors, Thrombin/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology
14.
Biochem Biophys Res Commun ; 208(2): 532-41, 1995 Mar 17.
Article in English | MEDLINE | ID: mdl-7695604

ABSTRACT

An inhibitor of the adhesion of human platelets to collagen was identified in soluble extracts of the soft tick Ornithodoros Moubata and purified by four chromatographic steps. The isolated inhibitor, TAI (Tick Adhesion Inhibitor), is a approximately 15-kDa protein that completely blocks the adhesion of platelets to collagen-coated microtiter plates with an IC50 of 8 nM. In the same concentration range it does not inhibit collagen-induced platelet aggregation or platelet adhesion to fibrinogen and has a partial inhibitory effect on platelet adhesion to fibronectin. TAI also blocks the adhesion of human endothelial cells to collagen, thus its inhibitory effect is not limited to platelets. TAI competes for the binding to platelets of a radiolabeled monoclonal antibody against the platelet glycoprotein Ia-IIa integrin complex. Based on its selective activity and small size, TAI is a promising new molecule for exploring cell-collagen interactions.


Subject(s)
Cell Adhesion/drug effects , Collagen/metabolism , Platelet Adhesiveness/drug effects , Salivary Proteins and Peptides/pharmacology , Ticks/chemistry , Animals , Binding, Competitive , Endothelium, Vascular/cytology , Humans , Integrins/antagonists & inhibitors , Integrins/metabolism , Molecular Weight , Receptors, Collagen , Salivary Glands/chemistry , Salivary Proteins and Peptides/chemistry
15.
Thromb Res ; 77(5): 453-63, 1995 Mar 01.
Article in English | MEDLINE | ID: mdl-7778060

ABSTRACT

Optimal conditions for a method to simultaneously measure aggregation in 96 samples using a microplate reader were developed. The temperature of the assay was set at 25 degrees C, the optimal platelet concentration range was determined to be from 1-3 x 10(8) per mL, the assay volume was determined to be best at 100 microL and an agitation rate of setting #5 on the vortex was found to yield the most reliable aggregation response. After these initial assay parameters were established, EC50 values for standard platelet agonists including ADP, thrombin, collagen and thrombin receptor activating peptides were determined using the plate assay and compared to those obtained by measuring light transmittance in an aggregometer. The results were quantitatively similar, and qualitatively the shapes of the aggregations as monitored by both methods were characteristic of those expected for each agonist. The use of this assay was then extended to quantitate the inhibition of aggregation by antagonists of the fibrinogen receptor as well as by an inactive thrombin receptor peptide and by antibodies against the thrombin receptor. This method provided useful data for characterization of both platelet agonists and antagonists and should be useful for future platelet aggregation studies.


Subject(s)
Hematology/methods , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/physiology , Adenosine Diphosphate/pharmacology , Blood Platelets/cytology , Blood Platelets/drug effects , Blood Platelets/physiology , Collagen/pharmacology , Humans , In Vitro Techniques , Peptide Fragments/pharmacology , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/drug effects , Receptors, Thrombin/drug effects , Thrombin/pharmacology
16.
J Pharmacol Toxicol Methods ; 33(1): 53-61, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7727808

ABSTRACT

Acute thrombotic reocclusion and restenosis after successful coronary angioplasty are limitations of the procedure. Although the restenotic process is not completely understood, acute platelet deposition and thrombosis are considered important initiating mechanisms. The effort to identify pharmacologic agents capable of modifying acute platelet action following mechanical injury requires an animal model mimicking the clinical pathophysiology as closely as possible. We developed a model of angioplasty-induced injury in atherosclerotic rabbit femoral arteries. Acute 111indium-labelled platelet deposition and thrombosis were assessed four hours after balloon-injury in arteries subjected to prior endothelial damage (air desiccation) and cholesterol supplementation (one month). The effects of recombinant tick anticoagulant peptide (rTAP), a blood coagulation factor Xa (fXa) inhibitor and of recombinant leech antiplatelet protein (rLAPP), a platelet adhesion inhibitor, were compared to heparin (HEP) and aspirin (ASA). Recombinant TAP and HEP, but not rLAPP or ASA, successfully prevented thrombus formation and reduced platelet deposition in balloon-injured vessel segments to levels not significantly different from those observed in the contralateral atherosclerotic non-balloon-injured vessels. Therefore, this model, incorporating balloon catheter dilation of arteries exhibiting neointimal growth and atherosclerotic plaque formation, may be useful for evaluation of possible adjunctive therapies during angioplasty.


Subject(s)
Angioplasty, Balloon, Coronary/adverse effects , Arteriosclerosis/pathology , Aspirin/pharmacology , Blood Platelets/drug effects , Factor Xa Inhibitors , Heparin/pharmacology , Animals , Arthropod Proteins , Diet, Atherogenic , Disease Models, Animal , Intercellular Signaling Peptides and Proteins , Male , Peptides/pharmacology , Platelet Adhesiveness/drug effects , Rabbits , Recombinant Proteins/pharmacology , Salivary Proteins and Peptides/pharmacology , Thrombosis/pathology
17.
Thromb Haemost ; 72(4): 627-33, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7878643

ABSTRACT

The aggregation of platelets from a variety of animal species in response to thrombin receptor-derived activating peptides was evaluated. A series of 14-(SFLLRNPNDKYEPF), 7-(SFLLRNP-NH2), 6-(SFLLRN-HN2) or 5-(SFLLR-NH2) residue peptides, the structures of which were based on the deduced amino acid sequence of the human thrombin receptor, promoted full aggregation of platelets in plasma from humans, African Green and Rhesus monkeys, baboons and guinea pigs at 4-50 microM depending on the peptide used. Platelets in plasma from rabbit, dog, pig, and hamster underwent a shape change but failed to aggregate in response to these peptides over 3 log units of peptide up to 800 microM, despite being fully responsive to human thrombin. However, because the receptor peptides induced shape change in the platelets from these non-aggregating species, they apparently can activate some of the intracellular signaling system(s) usually initiated by thrombin in these platelets. In contrast, platelets from rats did not undergo shape change or aggregate in response to the peptides. A 7-residue receptor-derived peptide based on the deduced amino acid sequence of the clone of the hamster thrombin receptor (SFFLRNP-N2) was nearly as efficacious as the corresponding human receptor-derived 7-residue peptide to promote aggregation of human platelets. However, the hamster peptide could not promote aggregation of hamster platelets in plasma at up to 800 microM peptide, while a shape change response was elicited. Platelets from rats, rabbits and pigs also did not aggregate in response to this peptide derived from the hamster thrombin receptor, but all species except the rat underwent a shape change.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Mammals/blood , Peptide Fragments/pharmacology , Platelet Aggregation/drug effects , Receptors, Thrombin/physiology , Amino Acid Sequence , Animals , Blood Platelets/ultrastructure , Cell Line , Cricetinae , DNA Replication/drug effects , Dogs/blood , Fibroblasts/drug effects , Guinea Pigs , Humans , Male , Molecular Sequence Data , Muscle, Smooth, Vascular/drug effects , Peptide Fragments/chemical synthesis , Primates/blood , Rats , Receptors, Thrombin/chemistry , Rodentia/blood , Species Specificity , Swine/blood
18.
J Biol Chem ; 269(9): 6702-8, 1994 Mar 04.
Article in English | MEDLINE | ID: mdl-8120028

ABSTRACT

A platelet aggregation inhibitor was identified in the salivary gland of the tick Ornithodoros moubata and isolated by gel filtration and reverse phase high pressure liquid chromatography. The purified inhibitor is a approximately 6-kDa protein, which we have named disagregin. It inhibits ADP-stimulated platelet aggregation in plasma with an IC50 = 104 +/- 17 nM. Disagregin also inhibits platelet aggregation induced by other agonists, including collagen, epinephrine, platelet-activating factor, thrombin, and the thrombin receptor peptide SFLLRNPNDKYEPF. It does not, however, affect platelet shape change induced by these agonists or thrombin-induced dense granule release. Disagregin inhibits platelet aggregation by binding to the platelet fibrinogen receptor. 125I-Disagregin forms a specific complex with both subunits of the fibrinogen receptor, glycoproteins IIb and IIIa, in the presence of a chemical cross-linker. It binds to unstimulated platelets with a Kd = 42.5 +/- 7.5 nM (23,800 +/- 1600 sites/platelet) and to ADP-stimulated platelets with Kd = 39.4 +/- 6.6 nM (24,050 +/- 1500 sites/platelet). Unlabeled disagregin and the snake venom disintegrin echistatin both compete for this binding. Disagregin also completely blocks platelet adhesion to fibrinogen while partially inhibiting platelet adhesion to fibronectin and having little effect on platelet adhesion to collagen. Disagregin had no effect on the adhesion of human umbilical cord vein endothelial cells to fibrinogen or vitronectin. These cells lack the glycoprotein IIb-IIIa complex; therefore, this result is consistent with the ability of disagregin to bind selectively to platelet glycoproteins IIb and IIIa. Sequence analysis of disagregin revealed 60 residues composing a unique protein. Unlike other fibrinogen antagonists it does not contain the Arg-Gly-Asp cell recognition sequence or a conservative substitution, and it has no structural homology with the Arg-Gly-Asp-containing snake venom disintegrins. Thus, disagregin is unique both in structure and function and may serve as a useful tool for the design of therapeutically useful antithrombotic agents.


Subject(s)
Blood Platelets/physiology , Endothelium, Vascular/physiology , Platelet Aggregation Inhibitors/pharmacology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Salivary Proteins and Peptides/pharmacology , Ticks/metabolism , Adenosine Diphosphate/pharmacology , Amino Acid Sequence , Animals , Blood Platelets/drug effects , Cell Adhesion/drug effects , Chromatography, Gel , Chromatography, High Pressure Liquid , Collagen/pharmacology , Endothelium, Vascular/drug effects , Epinephrine/pharmacology , Humans , Kinetics , Molecular Sequence Data , Platelet Activating Factor/pharmacology , Platelet Aggregation Inhibitors/isolation & purification , Platelet Aggregation Inhibitors/metabolism , Salivary Glands/chemistry , Salivary Glands/metabolism , Salivary Proteins and Peptides/isolation & purification , Salivary Proteins and Peptides/metabolism , Thrombin/pharmacology , Umbilical Cord
19.
J Clin Invest ; 93(2): 615-32, 1994 Feb.
Article in English | MEDLINE | ID: mdl-8113399

ABSTRACT

A ruptured atherosclerotic plaque leads to exposure of deeper layers of the plaque to flowing blood and subsequently to thrombus formation. In contrast to the wealth of data on the occurrence of thrombi, little is known about the reasons why an atherosclerotic plaque is thrombogenic. One of the reasons is the relative inaccessibility of the atherosclerotic plaque. We have circumvented this problem by using 6-microns cryostat cross sections of human coronary arteries. These sections were mounted on coverslips that were exposed to flowing blood in a rectangular perfusion chamber. In normal-appearing arteries, platelet deposition was seen on the luminal side of the intima and on the adventitia. In atherosclerotic arteries, strongly increased platelet deposition was seen on the connective tissue of specific parts of the atherosclerotic plaque. The central lipid core of an advanced plaque was not reactive towards platelets. The results indicate that the atherosclerotic plaque by itself is more thrombogenic than the normal vessel wall. To study the cause of the increased thrombus formation on the atherosclerotic plaque, perfusion studies were combined with immunohistochemical studies. Immunohistochemical studies of adhesive proteins showed enrichment of collagen types I, III, V, and VI, vitronectin, fibronectin, fibrinogen/fibrin, and thrombospondin in the atherosclerotic plaque. Laminin and collagen type IV were not enriched. von Willebrand Factor (vWF) was not present in the plaque. The pattern of increased platelet deposition in serial cross sections corresponded best with areas in which collagen types I and III were enriched, but there were also areas in the plaque where both collagens were enriched but no increased reactivity was seen. Inhibition of platelet adhesion with a large range of antibodies or specific inhibitors showed that vWF from plasma and collagen types I and/or III in the plaque were involved. Fibronectin from plasma and fibronectin, fibrinogen, laminin, and thrombospondin in the vessel wall had no effect on platelet adhesion. We conclude that the increased thrombogenicity of atherosclerotic lesions is due to changes in quantity and nature of collagen types I and/or III.


Subject(s)
Arteriosclerosis/pathology , Blood Platelets/physiology , Coronary Vessels/pathology , Adult , Aged , Arteriosclerosis/physiopathology , Blood Platelets/ultrastructure , Cadaver , Coronary Vessels/physiopathology , Coronary Vessels/ultrastructure , Extracellular Matrix/physiology , Female , Humans , In Vitro Techniques , Male , Microscopy, Electron, Scanning , Middle Aged , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Platelet Aggregation , Reference Values
20.
Arterioscler Thromb ; 13(11): 1593-601, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8218100

ABSTRACT

Leech antiplatelet protein (LAPP) is a specific inhibitor of collagen-induced human platelet aggregation and adhesion to collagen under static conditions. Recombinant LAPP (rLAPP) and L-366,763 (acetylated-Cys-Asn-Pro-Arg-Gly-Asp-Cys-NH2), a peptidyl fibrinogen receptor antagonist, were evaluated in an anesthetized baboon thrombosis model using a collagen-coated graft segment of an arteriovenous shunt to elicit thrombus formation. Animals were randomized to receive systemic intravenous administration of rLAPP (100 micrograms.kg-1 x min-1; n = 5), L-366,763 (8.5 micrograms.kg-1 x min-1; n = 3), or saline (n = 3). Despite complete and selective inhibition of type I collagen-induced ex vivo aggregation of platelets, rLAPP had no significant effect on the rate or the extent of 111-In-labeled platelet deposition onto the collagen graft and no effect on template bleeding time. In contrast, L-366,763 completely prevented platelet deposition, maintained blood flow, and significantly prolonged bleeding time at the dosage that inhibited ex vivo aggregation in response to all agonists studied. In this study, the absence of an antithrombotic benefit of rLAPP contrasted sharply with the efficacy of the fibrinogen receptor antagonist. These results demonstrate that specific inhibition of collagen-mediated platelet aggregation alone is not sufficient to prevent platelet-dependent thrombosis in this baboon model.


Subject(s)
Collagen/pharmacology , Platelet Aggregation Inhibitors/therapeutic use , Platelet Aggregation/drug effects , Salivary Proteins and Peptides/therapeutic use , Thrombosis/prevention & control , Amino Acid Sequence , Animals , Arteriovenous Shunt, Surgical , Bleeding Time , Fibrinopeptide A/metabolism , Kinetics , Male , Molecular Sequence Data , Oligopeptides/pharmacology , Papio , Partial Thromboplastin Time , Platelet Aggregation Inhibitors/pharmacology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Salivary Proteins and Peptides/administration & dosage , Salivary Proteins and Peptides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...