Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
J Pept Sci ; 6(8): 398-412, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10969869

ABSTRACT

Additional structure-activity relationship studies on potent cyclic peptide inhibitors of very late antigen-4 (VLA-4) are reported. The new N- to C-terminal cyclic hexa-, hepta- and octapeptide inhibitors like cyclo(MeIle/MePhe-Leu-Asp-Val-X) (X = 2-4 amino acids containing hydrophobic and/or basic side chains) were synthesized using solid phase peptide synthesis methods. The peptides were evaluated in in vitro cell adhesion assays and in in vivo inflammation models. Many of the peptides like cyclo(MePhe-Leu-Asp-Val-D-Arg-D-Arg) (ZD7349) (17), cyclo(MeIle-Leu-Asp-Val-D-Arg-D-Arg-D-Phe) (20), cyclo(MeIle-Leu-Asp-Val-D-Arg-D-Arg-MePhe) (21) and cyclo(MePhe-Leu-Asp-Val-D-Arg-D-Arg-D-Ala-D-Ala) (23) were potent inhibitors of VLA-4-mediated cell adhesion and inhibited ovalbumin-induced delayed type hypersensitivity (DTH) response in mice. The more potent compounds were highly selective and did not affect U937 cell adhesion to fibronectin (VLA-5), phorbolmyristate acetate or PMA-differentiated U937 cell adhesion to intercellular cell adhesion molecule-1 (ICAM-1)-expressing Chinese hamster ovary cells (LFA-1) and adenosine diphosphate (ADP)-induced platelet aggregation (GPIIb/IIIa). In contrast to the inhibitors like Ac-cyclo(D-Lys-D-Ile-Leu-Asp-Val) and cyclo(CH2CO-Ile-Leu-Asp-Val-Pip-CH2CO-Ile-Leu-Asp-Val-Pip) described earlier, the new compounds were much more compatible with the depot formulations based on poly(DL-lactide-co-glycolide) polymers. The hexapeptide cyclo(MePhe-Leu-Asp-Val-D-Arg-D-Arg) (ZD7349) (17) inhibited MOLT-4 cell adhesion to fibronectin and vascular cell adhesion molecule-1 (VCAM-1) with IC50 values of 260 and 330 nM, respectively, and did not show any significant effect against other integrins (IC50 > 300 microM). ZD7349 inhibited ovalbumin-induced DTH response in mice when administered continuously using a mini-pump (ED50 0.01 mg/kg/day) or when given as an s.c. or i.v. bolus injection at a dose of 1-10 mg/kg. ZD7349 was also active in type II collagen-induced arthritis (CIA) and experimental autoimmune encephalomyelitis (EAE) tests at a dose of 3-10 mg/kg. The peptide was released from some formulations over a period of 10-20 days. ZD7349 is currently undergoing pre-clinical investigation.


Subject(s)
Integrins/antagonists & inhibitors , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Receptors, Lymphocyte Homing/antagonists & inhibitors , Animals , Cell Adhesion/drug effects , Cell Line , Cricetinae , Humans , Integrin alpha4beta1 , Mice , Structure-Activity Relationship
2.
J Pept Sci ; 6(7): 321-41, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10946997

ABSTRACT

Potent monomeric and dimeric cyclic peptide very late antigen-4 (VLA-4) inhibitors have been designed based on a tetrapeptide (Ile-Leu-Asp-Val) sequence present in a 25-amino acid peptide (CS-1) reported in the literature. The peptides, synthesized by the SPPS techniques, were evaluated in the in vitro cell adhesion assays and in the in vivo inflammation models. The N- to C-terminal cyclic peptides such as cyclo(Ile-Leu-Asp-Val-NH-(CH2)2-S-(CH2)2-CO) (28) and cyclo(MeIle-Leu-Asp-Val-D-Ala-D-Ala) (31), monomeric and dimeric peptides containing piperazine (Pip) or homopiperazine (hPip) residues as linking groups, e.g. cyclo(MeIle-Leu-Asp-Val-Pip-CH2CO-NH-(CH2)2-S-CH2-CO) (49) and cyclo(MeIle-Leu-Asp-Val hPip-CH2CO-MeIle-Leu-Asp-Val-hPip-CH2CO) (58) and cyclic peptides containing an amide bond between the side chain amino group of an amino acid such as Lys and the C-terminal Val carboxyl group, e.g. Ac-cyclo(D-Lys-D-Ile-Leu-Asp-Val) (62) and beta-Ala-cyclo(D-Lys-D-Leu-Leu-Asp-Val) (68) were more potent than CS-1 in inhibiting the adhesion of the VLA-4-expressing MOLT-4 cells to fibronectin. The more potent compounds were highly selective and did not affect U937 cell adhesion to fibronectin (VLA-5), PMA-differentiated U937 cell adhesion to intercellular cell adhesion molecule- 1-expressing Chinese hamster ovary cells (LFA-1) and ADP-induced platelet aggregation (GPIIb/IIIa). A number of the more potent compounds inhibited ovalbumin-induced delayed type hypersensitivity in mice and some were 100-300 times more potent (ED50 = 0.003-0.009 mg/kg/day, s.c.) than CS-1. Two peptides, Ac-cyclo(D-Lys D-Ile-Leu-Asp-Val) (62) and cyclo(CH2CO-Ile-Leu-Asp-Val-Pip-CH2CO-Ile-Leu-Asp-Val-Pip) (55), were formulated in poly(DL-lactide-co-glycolide) depots and the release profile was investigated in vitro over a 30-day period.


Subject(s)
Cell Adhesion/drug effects , Integrins/antagonists & inhibitors , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/pharmacology , Receptors, Lymphocyte Homing/antagonists & inhibitors , Amino Acid Sequence , Animals , CHO Cells , Cell Line , Cricetinae , Delayed-Action Preparations , Dimerization , Drug Stability , Humans , Hypersensitivity, Delayed/prevention & control , Integrin alpha4beta1 , Lactic Acid , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Peptides, Cyclic/chemistry , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Polymers , U937 Cells
3.
Ultrasound Med Biol ; 25(6): 905-14, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10461718

ABSTRACT

We have investigated the ability to quantitate atherosclerosis in the aortic arch of the Watanabe rabbit using noninvasive 3-D ultrasound. Our methodology utilizes postprocessing of videotaped freehand 2-D interrogations to form a compound 3-D data block. Structures may then be segmented on the attributed grey-scale level and volumes measured. Analysis of 3-D reconstructions revealed a low echo structure in the aortic arch of atherosclerotic rabbits, absent in nonatherosclerotic rabbits, at recognized sites of plaque predilection. This structure volume correlated closely with fatty streak volume determined from histology (r = 0.890). During a 30-week study, this structure volume increased in untreated animals, but was blocked by treatment with the antiatherosclerotic agent probucol. Thus, a new 3-D ultrasound methodology has been used noninvasively to detect and quantitate a low echo structure corresponding to fatty streaks in the Watanabe rabbit aortic arch. This new methodology could potentially aid plaque burden quantification in human peripheral arteries.


Subject(s)
Aortic Diseases/diagnostic imaging , Arteriosclerosis/diagnostic imaging , Algorithms , Animals , Anticholesteremic Agents/therapeutic use , Aorta, Thoracic/diagnostic imaging , Aorta, Thoracic/pathology , Aortic Diseases/drug therapy , Aortic Diseases/pathology , Arteriosclerosis/drug therapy , Arteriosclerosis/pathology , Female , Humans , Hyperlipidemias/pathology , Image Processing, Computer-Assisted , Probucol/therapeutic use , Rabbits , Time Factors , Ultrasonography/methods
4.
Br J Pharmacol ; 126(8): 1751-60, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10372817

ABSTRACT

1. Small, N- to C-terminal cyclized peptides containing the leucyl-aspartyl-valine (LDV) motif from fibronectin connecting segment-1 (CS-1) have been investigated for their effects on the adhesion of human T-lymphoblastic leukaemia cells (MOLT-4) to human plasma fibronectin in vitro mediated by the integrin Very Late Antigen (VLA)-4 (alpha4beta1, CD49d/CD29). 2. Cyclo(-isoleucyl-leucyl-aspartyl-valyl-aminohexanoyl-) (c(ILDV-NH(CH2)5CO)) was approximately 5 fold more potent (IC50 3.6+/-0.44 microM) than the 25-amino acid linear CS-1 peptide. Cyclic peptides containing two more or one less methylene groups had similar potency to c(ILDV-NH(CH2)5CO) while a compound containing three less methylene groups, c(ILDV-NH(CH2)2CO), was inactive at 100 microM. 3. c(ILDV-NH(CH2)5CO) had little effect on cell adhesion mediated by two other integrins, VLA-5 (alpha5,beta1, CD49e/CD29) (K562 cell adhesion to fibronectin) or Leukocyte Function Associated molecule-1 (LFA-1, alphabeta2, CD11a/CD18) (U937 cell adhesion to Chinese hamster ovary cells transfected with intercellular adhesion molecule-1) at concentrations up to 300 microM. 4. c(ILDV-NH(CH2)5CO) inhibited ovalbumin delayed-type hypersensitivity or oxazolone contact hypersensitivity in Balb/c mice when dosed continuously from subcutaneous osmotic mini-pumps (0.1-10 mg kg(-1) day(-1)). Maximum inhibition (approximately 40%) was similar to that caused by the monoclonal antibody PS/2 (7.5 mg kg(-1) i.v.) directed against the alpha4 integrin subunit. 5. c(ILDV-NH(CH2)5CO) also inhibited oxazolone contact hypersensitivity when dosed intravenously 20 h after oxazolone challenge (1-10 mg kg(-1)). Ear swelling was reduced at 3 h and 4 h but not at 1 h and 2 h post-dose (10 mg kg(-1)). 6. Small molecule VLA-4 inhibitors derived from c(ILDV-NH(CH2)5CO) may be useful as anti-inflammatory agents.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Integrins/antagonists & inhibitors , Peptides/pharmacology , Receptors, Lymphocyte Homing/antagonists & inhibitors , Amino Acid Sequence , Animals , CHO Cells/cytology , CHO Cells/metabolism , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cricetinae , Dermatitis, Contact/drug therapy , Dermatitis, Contact/immunology , Female , Fibronectins/metabolism , Humans , Hypersensitivity, Delayed/drug therapy , Hypersensitivity, Delayed/immunology , Inflammation/drug therapy , Inflammation/immunology , Integrin alpha4beta1 , Integrins/physiology , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Intercellular Adhesion Molecule-1/physiology , Intercellular Signaling Peptides and Proteins , Leukemia, Erythroblastic, Acute/pathology , Leukemia, T-Cell/pathology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Ovalbumin/immunology , Oxazolone/immunology , Rats , Receptors, Lymphocyte Homing/physiology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Transfection
5.
Arterioscler Thromb Vasc Biol ; 18(5): 723-31, 1998 May.
Article in English | MEDLINE | ID: mdl-9598830

ABSTRACT

Hypertriglyceridemia may contribute to the development of atherosclerosis by increasing expression of cell adhesion molecules (CAMs). Although the cellular expression of CAMs is difficult to assess clinically, soluble forms of CAMs (sCAMs) are present in the circulation and may serve as markers for CAMs. In this study, we examined the association between sCAMs and other risk factors occurring with hypertriglyceridemia, the effect of triglyceride reduction on sCAM levels, and the role of soluble vascular cell adhesion molecule-1 (sVCAM-1) in monocyte adhesion in vitro. Compared with normal control subjects (n=20), patients with hypertriglyceridemia and low HDL (n=39) had significantly increased levels of soluble intercellular adhesion molecule-1 (sICAM-1) (316+/-28.8 versus 225+/-16.6 ng/mL), sVCAM-1 (743+/-52.2 versus 522+/-43.6 ng/mL), and soluble E-selectin (83+/-5.9 versus 49+/-3.6 ng/mL). ANCOVA showed that the higher sCAM levels in patients occurred independently of diabetes mellitus and other risk factors. In 27 patients who received purified n-3 fatty acid (Omacor) 4 g/d for > or =7 months, triglyceride level was reduced by 47+/-4.6%, sICAM-1 level was reduced by 9+/-3.4% (P=.02), and soluble E-selectin level was reduced by 16+/-3.2% (P<.0001), with the greatest reduction in diabetic patients. These results support previous in vitro data showing that disorders in triglyceride and HDL metabolism influence CAM expression and treatment with fish oils may alter vascular cell activation. In a parallel-plate flow chamber, recombinant sVCAM-1 at the concentration seen in patients significantly inhibited adhesion of monocytes to interleukin-1-stimulated cultured endothelial cells under conditions of flow by 27.5+/-7.2%. Thus, elevated sCAMs may negatively regulate monocyte adhesion.


Subject(s)
Cell Adhesion , E-Selectin/metabolism , Hypertriglyceridemia/metabolism , Intercellular Adhesion Molecule-1/metabolism , Monocytes/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Adolescent , Adult , Aged , Cell Adhesion/drug effects , Cells, Cultured , Endothelium, Vascular , Fatty Acids/therapeutic use , Female , Fish Oils/therapeutic use , Humans , Interleukin-1/pharmacology , Male , Middle Aged , Monocytes/drug effects , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Solubility , Time Factors , Tumor Cells, Cultured , Vascular Cell Adhesion Molecule-1/pharmacology
7.
J Cell Physiol ; 167(1): 106-12, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8698827

ABSTRACT

A variety of evidence suggests that vascular smooth muscle cells (SMC) exhibit a more immature phenotype when stimulated by injury to replicate in the adult. One growth characteristic common to immature (embryonic, fetal, and neonatal) SMC is a markedly reduced responsiveness to platelet-derived growth factor (PDGF) and other mitogenic stimuli. Here we demonstrate that SMC isolated from the 14-day neointima of experimentally injured carotid arteries exhibit a similar growth phenotype. The proliferative responses of neointimal cells to the BB homodimer of PDGF, which interacts with both forms of the PDGF receptor, were up to twenty-fold less (as assessed by BrdU immunocytochemistry) than that of adult control tunica media cells over a wide range of PDGF concentrations. Paradoxically, these cells expressed abundant mRNA for the alpha- and beta-subunits of the PDGF receptor (by RT-PCR) and expressed abundant PDGF receptor protein (by Western blotting). Addition of PDGF-BB to neointimal SMC induced significant autophosphorylation of the PDGF receptor, suggesting that the PDGF receptors were fully functional. The chemotactic responses of neointimal SMC to PDGF, in in vitro migration assays, were identical to that of control medial cells. The data further establish the existence of vascular SMC phenotypes characterized by a refractoriness to growth stimulation by specific mitogens, and provide further evidence for the reiteration of developmentally regulated programs following vascular injury in vivo.


Subject(s)
Muscle, Smooth, Vascular/pathology , Platelet-Derived Growth Factor/pharmacology , Tunica Intima/pathology , Animals , Base Sequence , Cell Differentiation , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Molecular Sequence Data , Muscle, Smooth, Vascular/metabolism , Rats , Receptors, Platelet-Derived Growth Factor/metabolism , Tunica Intima/metabolism
8.
Circulation ; 93(5): 1009-19, 1996 Mar 01.
Article in English | MEDLINE | ID: mdl-8598064

ABSTRACT

BACKGROUND: Numerous studies have demonstrated the ability of angiotensin II (Ang II) receptor antagonists and angiotensin-converting enzyme (ACE) inhibitors to inhibit intimal hyperplasia after balloon dilation of noncoronary arteries in small-animal models, suggesting an important role for Ang II in the response to injury. Although ACE inhibitors have not been similarly effective in nonhuman coronary models or in human restenosis trials, questions remain regarding the efficacy ACE inhibitors against tissue ACE and the contributions of ACE-independent pathways of Ang II generation. Unlike ACE inhibitors, Ang II receptor antagonists have the potential to inhibit responses to Ang II independent of its biosynthetic origin. METHODS AND RESULTS: In separate studies, three Ang II receptor antagonists, including AT1 selective (L-158,809), balanced AT1/AT2 (L-163,082), and AT2 selective (L-164,282) agents, were evaluated for their ability to inhibit vascular intimal thickening in a porcine coronary artery model of vascular injury. Preliminary studies in a rat carotid artery model revealed that constant infusion of L-158,809 (0.3 or 1.0 mg X kg-1 X d-1) reduced the neointimal cross-sectional area by up to 37% measured 14 days after balloon dilatation. In the porcine studies, animals were treated with vehicle or test compound beginning 2 days before and extending 28 days after experimental angioplasty. Left anterior descending, left circumflex, and/or right coronary arteries were injured by inflation of commercially available angioplasty balloons with placement of coiled metallic stents. Infusion of L-158,809 (1 mg X kg-1 X d-1), L-163,082 (1 mg X kg-1 X d-1), or L-164,282 (1.5 mg X kg-1 X d-1) in the study animals yielded plasma drug levels sufficient either to chronically block or, for L-164,282, to spare pressor responses to exogenous Ang II. Neither L-158,809, L-163,082, nor L-164,282 had statistically significant effects (P=.12, P=.75, and P=.48, respectively, compared with vehicle-treated controls) on neointimal thickness (normalized for degree of injury) measured by morphometric analysis at day 28 after angioplasty. CONCLUSIONS: These findings indicate that chronic blockade of Ang II receptors by either site-selective or balanced AT1/AT2 antagonists is insufficient to inhibit intimal hyperplasia after experimental coronary vascular injury in the pig. The results further suggest that, unlike in the rat carotid artery, Ang II is not a major mediator of intimal thickening in the pig coronary artery.


Subject(s)
Angiotensin II/physiology , Angiotensin Receptor Antagonists , Coronary Disease/pathology , Coronary Vessels/drug effects , Imidazoles/pharmacology , Sulfonamides/pharmacology , Tetrazoles/pharmacology , Angiotensin II/metabolism , Animals , Coronary Vessels/pathology , Disease Models, Animal , Imidazoles/blood , Rats , Recurrence , Swine , Tetrazoles/blood
9.
Circulation ; 90(6): 3091-7, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7994858

ABSTRACT

BACKGROUND: The brisk fibrinolytic response of canines has impaired efforts to develop a canine model of chronic thromboembolic pulmonary hypertension. Difficulties in retaining chronic embolic residuals were partially overcome by administration of tranexamic acid (TXA) (Circulation. 1991;83:1272-1279.). In this study, we used type 1 plasminogen activator inhibitor (PAI-1), a major inhibitor of the endogenous fibrinolytic system, to determine its efficacy in the suppression of thrombolysis in canines. METHODS AND RESULTS: Thrombus was induced in the inferior vena cava of anesthetized mongrel dogs with thrombin and a special double-balloon catheter; 2 hours later, the thrombus was embolized. In one group of dogs, activated type 1 plasminogen activator inhibitor (PAI-1) (130 micrograms) was delivered directly into the forming thrombus; in another, TXA (110 mg/kg) was given intravenously before thrombus formation; in controls, thrombus was induced without inhibitors. Cross-linked fibrin degradation product (D-dimer) appeared in the blood of control animals within 1 hour of thrombus induction (176 +/- 62.5 versus 1.02 +/- 0.39 ng/mL baseline; mean +/- SEM), was maximal by 4 hours (413 +/- 110 ng/mL) and remained elevated at 24 hours (90.8 +/- 19.5 ng/mL). Compared with controls, PAI-1 and TXA suppressed D-dimer release by 80% and 85%, respectively, over the first 24 hours. One week later, animals were killed, and residual emboli were harvested. Perfusion scan defects persisted in all animals at this time, but there were no scan defect differences among groups. However, emboli recovered from animals receiving PAI-1 still harbored immunoreactive PAI-1 and were, on average, more than twofold greater in mass (393 +/- 56 mg) than emboli recovered from either controls (183 +/- 76 mg) or animals receiving TXA (180 +/- 80 mg). CONCLUSIONS: Intravenous TXA and intrathrombus PAI-1 effectively suppress thrombolysis for 24 hours in canines. Thromboemboli enriched with PAI-1 appear to resist lysis for longer periods of time (up to 1 week). These findings are consistent with the hypothesis that PAI-1 remains associated with the embolus, where it continues to inhibit lysis, whereas TXA eventually diffuses out of the embolus, allowing lysis to ensue.


Subject(s)
Antifibrinolytic Agents/pharmacology , Pulmonary Embolism/blood , Animals , Antigens/analysis , Dogs , Fibrin Fibrinogen Degradation Products/metabolism , Immunohistochemistry , Plasminogen Activator Inhibitor 1/blood , Plasminogen Activator Inhibitor 1/immunology , Plasminogen Activator Inhibitor 1/pharmacology , Pulmonary Embolism/pathology , Tranexamic Acid/pharmacology
10.
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
11.
Biochem Biophys Res Commun ; 190(3): 1001-8, 1993 Feb 15.
Article in English | MEDLINE | ID: mdl-8382482

ABSTRACT

The role of the thrombin receptor tethered ligand hypothesis in mediating the mitogenic responses of cells to thrombin was explored. We have found that small (5-14 amino acid) peptides corresponding to the proposed amino terminus of thrombin activated human and hamster thrombin receptors are mitogenic for the Chinese hamster fibroblast cell line, CCL39. Hirudin and hirugen block the mitogenic effects of thrombin but not the activity of the agonist peptides. Pertussis toxin treated cells do not respond to either alpha-thrombin or the agonist peptides. The data support the idea that the thrombin receptor on CCL39 cells, which is homologous to the thrombin receptor on human platelets, is capable of transmitting mitogenic signals by a mechanism consistent with the tethered ligand hypothesis.


Subject(s)
Mitogens , Receptors, Cell Surface/drug effects , Thrombin/pharmacology , Amino Acid Sequence , Animals , Cells, Cultured , Cricetinae , Gene Expression , Hirudins/analogs & derivatives , Hirudins/pharmacology , In Vitro Techniques , Molecular Sequence Data , Peptide Fragments/pharmacology , Peptides/chemistry , Peptides/pharmacology , RNA, Messenger/genetics , Receptors, Cell Surface/genetics , Receptors, Thrombin , Thrombin/chemistry
12.
J Biol Chem ; 267(24): 17128-35, 1992 Aug 25.
Article in English | MEDLINE | ID: mdl-1512250

ABSTRACT

Plasminogen activator inhibitor-1 (PAI-1) accumulates within thrombi and forming whole blood clots. To explore this phenomenon at the molecular level, PAI-1 binding to fibrin was examined. The experiments were performed by adding 125I-PAI-1, which retains its complete tissue-type plasminogen (t-PA) inhibitory activity, to fibrin matrices formed in 2-cm2 tissue culture wells. Guanidine HCl-activated PAI-1 binding was reversible and was inhibited in the presence of excess, unlabeled PAI-1. Activated 125I-PAI-1 recognized 2 sites on fibrin: a very small number of high affinity sites (Kd less than 1 nM) and principally a large number of low affinity sites with an approximate Kd of 3.8 microM. Latent PAI-1 bound to fibrin at a site indistinguishable from the lower affinity site recognized by activated PAI-1. Fibrin, pretreated with activated PAI-1, was protected from t-PA-mediated plasmin degradation in a PAI-1 dose-responsive manner (IC50 = 12.3 nM). Clot protection correlated with partial occupancy of the low affinity PAI-1 binding site on fibrin and was due to the formation of sodium dodecyl sulfate-stable, PAI-1.t-PA complexes. Latent PAI-1 (27 nM) did not protect the fibrin from dissolution. The localization of PAI-1 to a thrombus by virtue of its fibrin binding potential could result in significant protection of the thrombus from the degradative effects of the fibrinolytic system.


Subject(s)
Fibrin/metabolism , Fibrinogen/metabolism , Fibrinolysis/drug effects , Plasminogen Inactivators/metabolism , Plasminogen Inactivators/pharmacology , Tissue Plasminogen Activator/antagonists & inhibitors , Binding, Competitive , Cell Line , Dexamethasone/pharmacology , Electrophoresis, Polyacrylamide Gel , Fibrinogen/isolation & purification , Fibrosarcoma , Humans , Kinetics , Light , Molecular Weight , Plasminogen Inactivators/isolation & purification , Protein Binding , Scattering, Radiation
13.
Arterioscler Thromb ; 11(5): 1276-86, 1991.
Article in English | MEDLINE | ID: mdl-1911713

ABSTRACT

The effects of both clot-bound and circulating plasminogen activator inhibitor-1 (PAI-1) on endogenous fibrinolysis were investigated in a rat model of pulmonary embolism. Iodine-125 fibrin(ogen)-labeled blood-clot homogenates were delivered through the left jugular vein to the lung microvasculature, and the subsequent extent of the clot lysis was monitored by measuring the release of 125I-fibrin degradation products (FDPs) into the blood. Clots that had incorporated activated PAI-1 ex vivo were subsequently protected from dissolution in vivo in a dose-responsive manner (half-maximal concentration [IC50] = 4.3 micrograms/ml). PAI-1-containing clots also resisted lysis, as measured by the release of the specific FDP D-dimer. Plasma levels of plasminogen activator (PA) and PAI activity were unaltered by administration of PAI-1-containing clots, and the clot-protective effects of clot-bound PAI-1 were reversed by exogenous tissue-type plasminogen activator administration. Clot lysis was also inhibited in a dose-responsive manner (IC50 = 58 micrograms/kg) by intravenous bolus delivery of activated PAI-1 to the circulation. The clot-protective effects of circulating PAI-1 were correlated with dose-dependent increases in plasma PAI-1 antigen and activity levels and decreases in plasma PA levels (IC50 = 37 micrograms/ml). There was no evidence of any accumulation of circulating PAI-1 in the lungs. Latent PAI-1, whether delivered with or delivered after the clot homogenates, did not affect the clot-lytic process. Activated and latent PAI-1 was cleared from the circulation in a monophasic manner, with a half-life of approximately 32 and 7 minutes, respectively. The results indicate that both clot-bound and circulating PAI-1 are potent inhibitors of fibrinolysis in vivo. Clot-bound PAI-1 may inhibit PAs in the immediate vicinity of the clots, whereas circulating PAI-1 may act systemically by controlling overall levels of PAs present in the blood.


Subject(s)
Fibrinolysis/drug effects , Plasminogen Inactivators/pharmacology , Pulmonary Embolism/physiopathology , Animals , Blood Coagulation/physiology , Dose-Response Relationship, Drug , Fibrinolysis/physiology , Male , Plasminogen Inactivators/pharmacokinetics , Rats , Rats, Inbred Strains , Tissue Plasminogen Activator/pharmacology
14.
Circulation ; 84(1): 287-92, 1991 Jul.
Article in English | MEDLINE | ID: mdl-1905594

ABSTRACT

BACKGROUND: Plasminogen activator inhibitor-1 (PAI-1), the specific, fast-acting inhibitor of tissue-type plasminogen activator (t-PA), binds to fibrin and has been found in high concentrations within arterial thrombi. These findings suggest that the localization of PAI-1 to a thrombus protects that same thrombus from fibrinolysis. In this study, clot-bound PAI-1 was assessed for its ability to suppress clot lysis in vivo. METHODS AND RESULTS: Autologous, canine whole blood clots were formed in the presence of increasing amounts of activated PAI-1 (0-30 micrograms/ml). Approximately 6-8% of the PAI-1 bound to the clots under the experimental conditions. Control and PAI-1-enriched clots containing iodine-125-labeled fibrin (ogen) were homogenized, washed to remove nonbound elements, and delivered to the lungs of anesthetized dogs where the homogenates subsequently underwent lysis by the endogeneous fibrinolytic system. 125I-labeled fibrin degradation products appeared in the blood of control animals within 10 minutes and were maximal by 90 minutes. PAI-1 reduced fibrin degradation product release in a dose-responsive manner at all times between 30 minutes and 5 hours (greater than or equal to 76% inhibition at 30 minutes, PAI-1 greater than or equal to 6 micrograms/ml). PAI-1 also suppressed D-dimer release from clots containing small amounts of human fibrin (ogen). t-PA administration attenuated the effects of PAI-1, whereas latent PAI-1 (20 micrograms/ml) had no effect on clot lysis. Blood levels of PA and PAI activity remained unaltered during these experiments. CONCLUSIONS: The results indicate that PAI-1 markedly inhibits endogenous fibrinolysis in vivo and, moreover, suggest that the localization of PAI-1 to a forming thrombus is an important physiological mechanism for subsequent thrombus stabilization.


Subject(s)
Fibrinolysis/drug effects , Plasminogen Inactivators/pharmacology , Pulmonary Embolism/therapy , Thrombolytic Therapy , Animals , Antifibrinolytic Agents/metabolism , Dogs , Female , Fibrin Fibrinogen Degradation Products/metabolism , Iodine Radioisotopes/metabolism , Male , Microcirculation , Plasminogen Inactivators/metabolism , Pulmonary Embolism/metabolism , Pulmonary Embolism/physiopathology , Tissue Plasminogen Activator/pharmacology
15.
J Biol Chem ; 266(15): 9419-27, 1991 May 25.
Article in English | MEDLINE | ID: mdl-2033043

ABSTRACT

Bovine vascular smooth muscle cells (SMC) were examined for production of plasminogen activator inhibitor-1 (PAI-1) which may play a key role in regulating the fibrinolytic system. Growth-arrested SMC released active PAI (101 arbitrary units (AU)/10(6) cells/24 h) and a latent form of PAI (880 AU/10(6) cells/24 h) into the conditioned medium (CM). The levels of PAI were significant since 880 AU of PAI could inhibit approximately 1 microgram of tissue plasminogen activator. The extracellular matrix of SMC also contained PAI activity; however, the level was 17-fold less than that observed in the CM. SMC-PAI was a rapid inhibitor of tissue plasminogen activator (kass greater than 10(7) M-1 S-1) and was identified as a 45-kDa protein immunologically related to endothelial cell PAI-1. PAI-1 comprised 20 and 30%, respectively, of the newly synthesized protein detected in the CM and extracellular matrix of SMC. The SMC growth modulators, platelet-derived growth factor and transforming growth factor-beta, induced PAI-1 activity and protein synthesis by 2- and 3-fold, respectively, in a dose- and time-dependent manner. The increases in PAI-1 activity and protein synthesis were ascribed to elevated levels of PAI-1 mRNA as judged by Northern blot analysis of total RNA prepared from control and platelet-derived growth factor- and transforming growth factor-beta-treated cells. Increases in PAI-1 mRNA levels were evident 1 h after growth factor treatment and were maximal after 4 h. PAI-1 mRNA levels were unaffected by cycloheximide treatment. The results indicate that SMC synthesize and release PAI-1 which could regulate the normal fibrinolytic environment of the arterial wall. During atherosclerosis or after vascular injury increases in platelet-derived or locally produced mitogens may stimulate further PAI-1 synthesis and generate a prothrombotic state.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Plasminogen Inactivators/metabolism , Platelet-Derived Growth Factor/pharmacology , Transforming Growth Factor beta/pharmacology , Animals , Blotting, Northern , Cattle , Cycloheximide/pharmacology , Electrophoresis, Polyacrylamide Gel , Fibrinolysis , Kinetics , Muscle, Smooth, Vascular/cytology , Platelet Aggregation/drug effects , RNA, Messenger/metabolism
16.
J Biol Chem ; 266(12): 7488-93, 1991 Apr 25.
Article in English | MEDLINE | ID: mdl-1708387

ABSTRACT

Plasminogen activator inhibitor-1 (PAI-1) regulates fibrinolysis by inhibiting tissue type plasminogen activator (t-PA). Fibrinogen, heparin, and vitronectin enhance the rate of inhibition of t-PA by PAI-1. Kinetic studies indicate that both fibrinogen and heparin increase the second-order inhibition constant by a maximum of approximately 4-fold, whereas vitronectin increases the rate constant by a maximum of approximately 6-fold. The dissociation constants of fibrinogen, heparin, and vitronectin for the inhibition reaction were 200 nM, 20 nM, and 600 pM, respectively. In addition, PAI-1 inhibition of t-PA may be regulated by the presence of lipoprotein(a) (Lp(a)). Previous studies demonstrated that Lp(a) competes with plasminogen for the active site of fibrinogen- and heparin-bound t-PA. Kinetic studies described here demonstrate that Lp(a) prevents the inhibition of t-PA by PAI-1 in the presence of fibrinogen and heparin, but has no effect on the reaction in the presence of vitronectin or in the absence of either fibrinogen or heparin. The data suggest that fibrinogen and heparin may enhance the rate of inhibition through an interaction with t-PA, and that vitronectin may enhance the inhibition through an interaction with PAI-1. In addition, these experiments indicate that Lp(a) may regulate fibrinolysis by competing with PAI-1 and plasminogen for fibrinogen- and heparin-bound t-PA. These data suggest that PAI-1 inhibition of t-PA in vivo is primarily mediated via interaction with fibrinogen, heparin, vitronectin, and Lp(a), and therefore, the functional levels of PAI-1 activity in the vasculature may be regulated by the presence of these components.


Subject(s)
Fibrinogen/pharmacology , Glycoproteins/pharmacology , Heparin/pharmacology , Lipoproteins/pharmacology , Plasminogen Inactivators/pharmacology , Tissue Plasminogen Activator/antagonists & inhibitors , Lipoprotein(a) , Vitronectin
17.
Blood ; 76(8): 1514-20, 1990 Oct 15.
Article in English | MEDLINE | ID: mdl-2207327

ABSTRACT

The pharmacokinetics of the activated and latent forms of plasminogen activator inhibitor-1 (PAI-1) isolated from HT1080 fibrosarcoma cells (HT1080 PAI-1) and a nonglycosylated form of human PAI-1 isolated from a yeast expression system (rPAI-1) were followed in the rabbit. As assessed by an immunologic assay specific for human PAI-1, guanidine HCI activated HT1080 PAI-1 and rPAI-1 entered the total plasma volume following intravenous bolus administration and exhibited a biphasic clearance pattern. The t1/2s of HT1080 PAI-1 for the initial and beta phases equalled 6.0 and 24.8 minutes, respectively. The t1/2s of rPAI-1 for the initial and beta phases equalled 8.8 and 34.0 minutes, respectively. Similar results were obtained by measuring PAI-1 activity in plasma and with trace amounts of 125I-rPAI-1, suggesting that the above pharmacokinetic behavior could also apply to endogenous PAI-1. The liver was the main site of rPAI-1 clearance. Unactivated, latent PAI-1 exhibited a very different pharmacokinetic profile. Over 80% of latent rPAI-1 cleared from the circulation within 10 minutes (t1/2 = 1.7 minutes). The difference in clearance behavior between activated and latent PAI-1 may be related to the ability of activated PAI-1, but not latent PAI-1, to rapidly form high-molecular-weight complexes with plasma binding factors which were observed in vitro and in vivo. Because PAI-1 could potentially tilt the fibrinolytic balance toward a prothrombotic state, its rapid clearance may represent an important control mechanism governing the circulating levels of this key component of the fibrinolytic pathway.


Subject(s)
Plasminogen Inactivators/pharmacokinetics , Animals , Fibrosarcoma/metabolism , Glycosylation , Guanidine , Guanidines/pharmacology , Half-Life , Humans , Metabolic Clearance Rate , Plasminogen Inactivators/blood , Rabbits , Tissue Distribution , Tumor Cells, Cultured
18.
J Cell Physiol ; 142(2): 342-51, 1990 Feb.
Article in English | MEDLINE | ID: mdl-2154504

ABSTRACT

Rat aortic smooth muscle cells (SMC) have been established by retroviral delivery of the complementary DNA (cDNA) for the simian virus 40 large T antigen (SV40LT) and examined for SMC phenotypic markers and growth characteristics, including responsiveness to the antiproliferative effects of heparin. The transfected cells (SV40LT-SMC) maintain defined SMC characteristics for more than 215 population doublings (PD) as judged by muscle-specific actin expression and growth inhibition by heparin. SV40LT-SMC greater than 129 PD become transformed while SV40LT-SMC less than 77 PD resemble nontransfected SMC morphologically and are nontumorigenic. SV40LT-SMC apparently release a growth factor which acts in an autocrine fashion, since (1) suramin inhibits SV40LT-SMC proliferation, (2) SV40LT-SMC-conditioned medium (CM) contains mitogenic activity, and (3) SV40LT-SMC CM suppresses the binding of platelet-derived growth factor to SMC. Heparin (10-100 micrograms/ml) is a potent inhibitor of both early (less than 80 PD) and late-passage (greater than 80 PD) SV40LT-SMC proliferation. The antiproliferative effects of heparin are similar to those previously observed for SMC by several criteria; the dose-response inhibition curves are indistinguishable from those obtained with nontransfected cells, other glycosaminoglycans have little effect on SV40LT-SMC growth, the antiproliferative effects of heparin are reversed in the presence of epidermal growth factor, and heparin displays high-affinity saturable binding to SV40LT-SMC. In conclusion, SV40LT-SMC are a continuous line of SMC-like cells that are sensitive to the growth inhibitor, heparin. SV40LT-SMC should facilitate studies of heparin inhibition and may be applicable for the study of other SMC characteristics as well.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Heparin/pharmacology , Muscle, Smooth, Vascular/cytology , Simian virus 40/immunology , Transfection , Actins/metabolism , Animals , Aorta, Abdominal , Blood , Cell Division/drug effects , Cell Line, Transformed , DNA/genetics , Fluorescent Antibody Technique , Gene Expression , Genetic Vectors , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Phenotype , Rats , Rats, Inbred Strains , Rats, Nude , Retroviridae/genetics
19.
Biochem Biophys Res Commun ; 160(3): 1047-54, 1989 May 15.
Article in English | MEDLINE | ID: mdl-2471517

ABSTRACT

Several biological effects of recombinant PDGF-BB and PDGF obtained from human platelets were examined with vascular smooth muscle cells. Although PDGF and PDGF-BB were equally potent mitogens for these cells, 5 fold higher levels of PDGF were required to displace 125I-PDGF-BB binding than PDGF-BB itself. Higher concentrations of PDGF relative to PDGF-BB were also required to stimulate the phosphorylation of a 163K protein in membrane preparations. PDGF-BB, but not PDGF, treatment of intact cells resulted in the phosphorylation on tyrosine residues of 168, 53, 48, and 45K proteins. The data suggest that PDGF and PDGF-BB stimulate smooth muscle cell mitogenesis by different mechanisms.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Platelet-Derived Growth Factor/pharmacology , Recombinant Proteins/pharmacology , Animals , Aorta, Thoracic , Binding, Competitive , Blood Platelets/analysis , Cattle , DNA/biosynthesis , Humans , Immunosorbent Techniques , Macromolecular Substances , Membrane Proteins/metabolism , Molecular Weight , Phosphorylation , Phosphotyrosine , Platelet-Derived Growth Factor/metabolism , Radioligand Assay , Tyrosine/analogs & derivatives , Tyrosine/metabolism
20.
J Biol Chem ; 264(12): 6990-5, 1989 Apr 25.
Article in English | MEDLINE | ID: mdl-2651437

ABSTRACT

To gain insight into the mechanism of the antiproliferative effects of heparin on vascular smooth muscle cells (SMC), the influence of this glycosaminoglycan on cell cycle progression and the expression of the c-fos, c-myc, and c-myb proto-oncogenes and two other growth-regulated genes was examined. SMC, synchronized by a serum-deprivation protocol, enter S phase 12-16 h after serum stimulation. Pretreatment with heparin for 48 h blocked the induction of histone H3 RNA, an S phase-expressed product, and prevented cell replication. Thus, heparin prevents entry of cells into S phase. Conversely, heparin had essentially no effect on changes in expression of the c-fos and c-myc proto-oncogenes during the G0 to G1 transition. Normal increases in c-fos and c-myc RNA were observed 30 min and 2 h following serum addition, respectively. However, the increase in expression of the mRNA of the c-myb proto-oncogene and the mitochondrial ATP/ADP carrier protein, 2F1, which begins to occur 8 h following serum addition to SMC, was completely inhibited by heparin. Two-dimensional polyacrylamide gel electrophoresis of the products of a rabbit reticulocyte cell-free translation of RNA isolated at various times confirmed this temporal assessment of the effects of heparin. These results suggest that heparin does not inhibit cell proliferation by blocking the G0 to G1 transition. Rather, heparin may affect a critical event in the mid-G1 phase of the cell cycle which is necessary for subsequent DNA synthesis.


Subject(s)
Cell Cycle/drug effects , Gene Expression Regulation/drug effects , Heparin/pharmacology , Muscle, Smooth/cytology , Animals , Cattle , Culture Media , Electrophoresis, Gel, Two-Dimensional , In Vitro Techniques , Interphase/drug effects , Mitochondrial ADP, ATP Translocases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-myb , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...