Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Cardiovasc Pharmacol ; 70(1): 34-41, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28679129

RESUMEN

Exenatide and cyclosporine A have been shown to moderately protect against myocardial reperfusion injury leading to reduction of infarct size in patients. Our objective was to investigate whether the combined treatment with exenatide (glucagon-like peptide 1 receptor agonist) and cyclosporine A or parstatin 1-26 (inhibitors of mitochondrial permeability transition pore and/or inflammation) is more beneficial than either agent alone. Rabbits underwent 40 minutes of ischemia and 120 minutes of reperfusion. Intravenous bolus administration of exenatide or cyclosporine A, 10 minutes before reperfusion, reduced infarct size by 38% (P < 0.05) and 40% (P < 0.05), and cardiac troponin I (cTnI) plasma levels by 48% (P < 0.05) and 36% (P < 0.05), respectively, compared with control. The combined administration of both agents resulted in an additive decrease of infarct size by 55% (P < 0.05) and cTnI release by 61% (P < 0.05). Also, combined treatment of exenatide and parstatin 1-26 enhanced infarct size reduction (62%, P < 0.05), compared with monotherapies (41% for parstatin 1-26, P < 0.05; 43% for exenatide, P < 0.05). In contrast, the combined administration of parstatin 1-26 and cyclosporine A canceled out the cardioprotective effects observed by monotherapies. These results suggest that, for the therapy of myocardial reperfusion injury the combined administration of exenatide and cyclosporine A or parstatin 1-26 is more effective than monotherapies and may provide advantageous clinical outcome.


Asunto(s)
Ciclosporina/administración & dosificación , Modelos Animales de Enfermedad , Infarto del Miocardio/tratamiento farmacológico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Fragmentos de Péptidos/administración & dosificación , Péptidos/administración & dosificación , Receptor PAR-1/administración & dosificación , Ponzoñas/administración & dosificación , Animales , Quimioterapia Combinada , Exenatida , Masculino , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/patología , Conejos
2.
J Cardiovasc Pharmacol ; 62(3): 270-7, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23644989

RESUMEN

Nerve growth factor (NGF) has been reported to play an important role in physiological and pathological angiogenesis. Based on these observations, we hypothesized that NGF may induce the formation of functional blood vessels in a hindlimb ischemic rabbit model. Hindlimb ischemia was induced in 34 rabbits bilaterally by endovascular embolization of femoral arteries. On the 7th, 14th, and 20th postembolization days, NGF was injected intramuscularly, in 1 ischemic limb, and vehicle was injected in the contralateral control limb. On the 40th day, newly developed collateral vessels (diameter >500 µm) were quantified by transauricular intraarterial subtraction angiography. Perfusion analysis of an in vivo dynamic computed tomography study was performed to the limbs to investigate the hemodynamic recovery of the distal ischemic tissues. Functional estimation of limb perfusion showed a statistically significant increase of blood flow and blood volume for NGF. However, the increase of the collateral vessels was not detectable angiographically, providing evidence for the existence of a NGF-stimulated capillary angiogenic network but not increase of arteriogenesis. The combination of NGF with either tropomyosin-related kinase type A or vascular endothelial growth factor receptor 2 antagonists abolished the NGF-induced hemodynamic recovery. These findings provide new insights into understanding the involvement of NGF in vascular formation and its applications in therapeutic angiogenesis.


Asunto(s)
Inductores de la Angiogénesis/uso terapéutico , Modelos Animales de Enfermedad , Isquemia/tratamiento farmacológico , Músculo Esquelético/efectos de los fármacos , Factor de Crecimiento Nervioso/uso terapéutico , Receptor trkA/agonistas , Receptor 2 de Factores de Crecimiento Endotelial Vascular/agonistas , Inductores de la Angiogénesis/administración & dosificación , Inductores de la Angiogénesis/antagonistas & inhibidores , Inductores de la Angiogénesis/aislamiento & purificación , Animales , Capilares/diagnóstico por imagen , Capilares/efectos de los fármacos , Capilares/patología , Hemodinámica/efectos de los fármacos , Miembro Posterior , Inyecciones Intramusculares , Isquemia/inducido químicamente , Isquemia/diagnóstico por imagen , Isquemia/patología , Masculino , Ratones , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/diagnóstico por imagen , Músculo Esquelético/patología , Neovascularización Fisiológica/efectos de los fármacos , Factor de Crecimiento Nervioso/administración & dosificación , Factor de Crecimiento Nervioso/antagonistas & inhibidores , Factor de Crecimiento Nervioso/aislamiento & purificación , Inhibidores de Proteínas Quinasas/efectos adversos , Conejos , Radiografía , Distribución Aleatoria , Receptor trkA/antagonistas & inhibidores , Receptor trkA/metabolismo , Flujo Sanguíneo Regional/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
3.
Am J Nephrol ; 36(3): 278-86, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22965158

RESUMEN

BACKGROUND/AIMS: Parstatin is a 41-mer peptide formed by proteolytic cleavage on activation of the protease-activated receptor 1. Parstatin was recently found to be cardioprotective against myocardial ischemia/reperfusion (IR) injury. In the present study, it was hypothesized that parstatin would protect the kidneys in acute renal failure. METHODS: We investigated the effects of parstatin on the renal dysfunction and injury caused either by renal IR injury or contrast-induced nephropathy (CIN) in two animal models. Renal IR injury was induced in rats by bilateral occlusion of renal arteries and veins for 45 min followed by 4 h of reperfusion, while CIN was induced in rabbits by intravenous injection of the radiocontrast medium Iopromide. RESULTS: Treatment with parstatin 15 min before or immediately after renal ischemia attenuated the resulting renal dysfunction as demonstrated by the improved biochemical indicators (serum creatinine and fractional excretion of Na(+)) and scintigraphic analysis. The effect was dose depended and provided evidence for a more prominent protection of tubular than glomerulal function. Histopathological examination of the kidneys revealed severe renal damage, which was significantly suppressed by the parstatin. Similarly, administration of a single dose of parstatin before the induction of CIN significantly protected against the resulting renal dysfunction and histologically evidenced renal tubular injury. CONCLUSION: These results suggest that parstatin is able to act as nephroprotective agent and may be useful in enhancing the tolerance of the kidney against renal injury associated with clinical conditions of acute renal failure. Further investigation on the mechanism underlying the nephroprotective properties of parstatin is deemed necessary.


Asunto(s)
Medios de Contraste/efectos adversos , Enfermedades Renales/prevención & control , Fragmentos de Péptidos/fisiología , Receptor PAR-1/fisiología , Daño por Reperfusión/prevención & control , Secuencia de Aminoácidos , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Riñón/efectos de los fármacos , Riñón/lesiones , Riñón/patología , Enfermedades Renales/inducido químicamente , Túbulos Renales/patología , Masculino , Datos de Secuencia Molecular , Fragmentos de Péptidos/metabolismo , Péptidos/química , Conejos , Cintigrafía/métodos , Ratas , Ratas Wistar , Receptor PAR-1/metabolismo , Factores de Tiempo
4.
J Pharmacol Exp Ther ; 332(3): 898-905, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20008957

RESUMEN

Parstatin, the N-terminal 41-amino-acid peptide cleaved by thrombin from the protease-activated receptor 1, protects against rat myocardial ischemia and reperfusion injury. In this study, we determined that the parstatin fragment 1-26, the putative signal peptide of protease-activated receptor 1, contains the functional domain of parstatin. We assessed a synthesized parstatin(1-26) peptide in an in vivo rat model of myocardial regional ischemia-reperfusion injury (n = 6/group). Infarct size in control rat hearts was 58 +/- 1% area at risk. Parstatin(1-26) was able to reduce infarct size to 13 +/- 1% (P < 0.001) and 22 +/- 1% area at risk (P < 0.01) when given before or after reperfusion. The infarct-sparing effects of parstatin(1-26) were abolished by inhibition of G(i) proteins (pertussis toxin), phosphoinositide 3-kinase/Akt (wortmannin), nitric-oxide synthase (NOS; N(G)-monomethyl-l-arginine), soluble guanylyl cyclase [1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ)], and sarcolemmal and mitochondrial K(ATP) channels [glibenclamide, 5-hydroxydecanoic acid, and sodium (5-(2-(5-chloro-2-methoxybenzamido)ethyl)-2-methoxyphenylsulfonyl) (methylcarbamothioyl)amide (HMR 1098)]. Parstatin(1-26) cardioprotection was also abolished by atractyloside, a mitochondrial permeability transition pore (mPTP) opener. The inhibitors and opener alone had no effect on infarct size. Furthermore, preischemic treatment with parstatin(1-26) increased Akt and endothelial NOS phosphorylation at the time of reperfusion. After a 120-min reperfusion, parstatin(1-26) increased nitric oxide levels (12 +/- 0.4 to 17 +/- 0.9 mmol/g tissue) and cyclic GMP levels (87 +/- 21 to 395 +/- 36 pmol/g tissue). Parstatin(1-26) treatment either before or after ischemia results in an extremely efficacious protection against ischemia-reperfusion injury that depends on a G(i) protein-mediated pathway involving mPTP, the end effector of the preconditioning pathway. This suggests that parstatin(1-26) has a potential therapeutic role in the treatment of ischemia and reperfusion injury.


Asunto(s)
Daño por Reperfusión Miocárdica/prevención & control , Fragmentos de Péptidos/farmacología , Señales de Clasificación de Proteína , Receptor PAR-1/química , Animales , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/fisiología , Masculino , Infarto del Miocardio/patología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/patología , Fragmentos de Péptidos/uso terapéutico , Ratas , Ratas Sprague-Dawley , Transducción de Señal
5.
J Pharmacol Exp Ther ; 328(2): 378-89, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18988770

RESUMEN

The proteolytic activation by thrombin of the proteinase-activated receptor 1 unveils the tethered peptide ligand and cleaves a 41-amino acid peptide. In this report, we show that this peptide, which we have designated as "parstatin," is a potent inhibitor of angiogenesis. Synthesized parstatin suppressed both the basic angiogenesis and that stimulated by basic fibroblast growth factor and vascular endothelial growth factor in the chick embryo model in vivo and in the rat aortic ring assay. Parstatin also abrogated endothelial cell migration and capillary-like network formation on the Matrigel and fibrin angiogenesis models in vitro. Treatment of endothelial cells with parstatin resulted in inhibition of cell growth by inhibiting the phosphorylation of extracellular signal-regulated kinases in a specific and reversible fashion and by promoting cell cycle arrest and apoptosis through a mechanism involving activation of caspases. We have shown that parstatin acts as a cell-penetrating peptide, exerting its biological effects intracellularly. The uptake into cells and the inhibitory activity were dependent on parstatin hydrophobic region. These results support the notion that parstatin may represent an important negative regulator of angiogenesis with possible therapeutic applications.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Movimiento Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Receptor PAR-1/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Capilares/efectos de los fármacos , Capilares/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Embrión de Pollo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Humanos , Péptidos/farmacología , Ratas , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
J Vasc Surg ; 49(4): 1000-12, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19217750

RESUMEN

BACKGROUND: Compared with angiogenesis, arteriogenesis is a distinct process based on the remodeling and maturation of pre-existing arterioles into large conductance arteries. Therapeutic angiogenesis has been proposed as a potential treatment for ischemic atherosclerotic diseases. Since a variety of angiogenic factors have been tested with inconsistent so far clinical results, the challenge remains in identifying the factor(s) that will stimulate functional neovascularization. Thrombin has been reported to play a pivotal role in the initiation of angiogenesis by regulating and organizing a network of angiogenic mediators. Also, it was recently demonstrated that thrombin is a potent anti-apoptotic factor for endothelial cells, providing evidence on a potential role of thrombin in vascular protection and maintenance of vessel integrity. Based on these observations, we hypothesized that thrombin may promote the development of mature functional blood vessels. METHODS: Seventy-four (n=74) rabbits underwent bilateral femoral artery surgical excision. On the 20th postsurgical day increasing doses of VEGF or bFGF or thrombin were injected in one ischemic limb per rabbit and an equal volume of normal saline to the contralateral control limbs. Quantification of newly developed collateral vessels (diameter >500 mum) was performed by transauricular intra-arterial subtraction angiography. Computerized quantitative analysis of collateral vessels in angiography images was based on the concept of multiscale structural tensor. Perfusion analysis of an in vivo dynamic computed tomography study was performed to investigate hemodynamic recovery of the distal ischemic limbs. Tissue perfusion analysis was performed with the semiquantitative slope methodology, which focuses on the first-pass arterial phase. RESULTS: A single administration of thrombin exhibited a dose-dependent increase of arteriogenic outcome. Thrombin at 5000 IU induced a 30.2 +/- 7.4% (P < 0.05) increase of total collateral area and length. Both VEGF and bFGF were without any significant effect at the concentrations used. Functional estimation of limb perfusion showed a statistically significant increase of blood flow recovery only for thrombin. The semiquantitative slope method perfusion score differed significantly in the 5000 IU thrombin treated limbs (5.7 +/- 0.3 vs 5.0 +/- 0.3 in control ischemic limbs; P < .05), and was not significantly inferior from the score of normal nonoperated limbs (6.5 +/- 0.3) suggesting a trend towards hemodynamic recovery of distal limb perfusion. CONCLUSIONS: In a rabbit hindlimb ischemia model, thrombin promoted the formation of large collateral vessels and improved the perfusion of distal ischemic tissue. These results provide new insights in understanding the involvement of thrombin in vascular formation and point to a novel role of thrombin in arteriogenesis.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Hemodinámica , Isquemia/metabolismo , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Trombina/metabolismo , Inductores de la Angiogénesis/administración & dosificación , Angiografía de Substracción Digital , Animales , Arterias/crecimiento & desarrollo , Circulación Colateral , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Factor 2 de Crecimiento de Fibroblastos/administración & dosificación , Hemodinámica/efectos de los fármacos , Miembro Posterior , Inyecciones Intramusculares , Isquemia/diagnóstico por imagen , Isquemia/tratamiento farmacológico , Isquemia/fisiopatología , Masculino , Neovascularización Fisiológica/efectos de los fármacos , Imagen de Perfusión/métodos , Conejos , Recuperación de la Función , Flujo Sanguíneo Regional , Trombina/administración & dosificación , Tomografía Computarizada por Rayos X , Factor A de Crecimiento Endotelial Vascular/administración & dosificación
7.
Thromb Haemost ; 92(4): 846-57, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15467917

RESUMEN

In a previous report we have presented evidence that thrombin interacts with alpha(v)beta(3) integrin in endothelial cells at the molecular and cellular level. This interaction was shown to be of functional significance in vitro and in vivo and contributed to activation of angiogenesis by thrombin. In the present study, we have used a synthetic thrombin peptide, TP508, which represents residues 183 to 200 of human thrombin. This peptide lacks the catalytic site of thrombin but contains the thrombin RGD sequence. Immobilized (surface-coated) TP508 peptide, like thrombin, supported alpha(v)beta(3) integrin-dependent endothelial cell attachment and haptotactic migration. These effects were specific (a scrambled TP508 peptide was without effect), and dosedependent. The RGD sequence was essential since a modified TP508 peptide, which contained RAD sequence instead of RGD, was inactive. Immobilized TP508 peptide stimulated phosphorylation of mitogen-activated protein kinases and focal adhesion kinase, the signal transduction pathways characteristic for integrin activation. On the other hand, TP508 peptide, when in solution, did not mimic other thrombin-promoted angiogenic effects, such as that of activation gelatinase A, upregulation of expression of vascular endothelial growth factor receptor mRNA or prostacyclin PGI(2) release in endothelial cells. On the contrary, soluble TP508 acted as an antagonist for the aforementioned effects of thrombin. TP508 peptide inhibited these thrombin-induced effects through a RGD and alpha(v)beta(3)-related mechanism. The antagonism with thrombin or thrombin receptor activating peptide was specific and involved at least in part mitogen-activated protein kinases activation. These results point to the importance of RGD sequence of thrombin in mediating effects on endothelial cells and angiogenesis.


Asunto(s)
Endotelio Vascular/citología , Integrinas/fisiología , Neovascularización Fisiológica/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Receptores de Vitronectina/fisiología , Trombina/farmacología , Adhesión Celular/efectos de los fármacos , Movimiento Celular , Endotelio Vascular/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Oligopéptidos/fisiología , Venas Umbilicales/citología
8.
Eur J Med Chem ; 70: 199-224, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24158013

RESUMEN

A series of pseudodipeptide-based chiral 1,3,4,5-tetrasubstituted-2-oxopiperazines has been designed and synthesized as potential PAR1 antagonists. These highly functionalized piperazines were synthesized from aromatic and basic amino acid derived Ψ[CH(CN)NH]pseudodipeptides through a four step pathway that involves reduction of the cyano group to build the 2-oxopiperazine ring, followed by selective functionalization at the N4-, N1-positions, and at the exocyclic moiety at position C5. This regioselective functionalization required the fine tuning of reaction conditions. All new compounds were screened as inhibitors of human platelet aggregation induced by the PAR1 agonist SFLLRN and as cytotoxic agents in human cancer cell lines. Some of the compounds displayed moderate PAR1 antagonist activity, while, others were cytotoxic at µM concentration. No correlation was observed between both types of activities.


Asunto(s)
Antineoplásicos/farmacología , Peptidomiméticos/farmacología , Piperazinas/farmacología , Receptor PAR-1/antagonistas & inhibidores , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HT29 , Humanos , Conformación Molecular , Peptidomiméticos/síntesis química , Peptidomiméticos/química , Piperazinas/síntesis química , Piperazinas/química , Agregación Plaquetaria/efectos de los fármacos , Relación Estructura-Actividad
9.
Eur J Med Chem ; 58: 98-111, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23123726

RESUMEN

By applying a diversity oriented synthesis strategy for the search of new antagonists of the thrombin receptor PAR1, a series of peptide-based ureas and thioureas, including analogues of the PAR1 reference antagonist RWJ-58259, has been designed and synthesized. The general synthetic scheme involves reduction of basic amino acid-derived amino nitriles by hydrogen transfer from hydrazine monohydrate in the presence of Raney Ni, followed by reaction with diverse isocyanates and isothiocyanates, and protecting group removal. All new compounds have been evaluated as inhibitors of human platelet aggregation induced by the PAR1 agonist SFLLRN. Some protected peptide-based ureas displayed significant antagonist activity.


Asunto(s)
Diseño de Fármacos , Péptidos/química , Receptor PAR-1/antagonistas & inhibidores , Urea/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Agregación Plaquetaria/efectos de los fármacos , Valores de Referencia , Relación Estructura-Actividad , Urea/análogos & derivados , Urea/síntesis química
10.
FEBS Lett ; 586(16): 2351-9, 2012 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-22659187

RESUMEN

The protease-activated receptor 1 (PAR1) is activated by thrombin cleavage releasing the physiologically-relevant parstatin peptide (residues 1-41). However, the actual length of parstatin was unclear since the receptor may also possess a cleavable signal peptide (residues 1-21) according to prediction programs. Here, we show that this putative signal peptide is indeed functional and removed from the PAR1 resolving the question of parstatin length. Moreover, we show that the sequence encoding the signal peptide may surprisingly play a role in stabilization of the PAR1 mRNA, a function which would be novel for a G protein-coupled receptor.


Asunto(s)
Regulación de la Expresión Génica , Señales de Clasificación de Proteína , Receptor PAR-1/fisiología , ADN Complementario/metabolismo , Células HEK293 , Humanos , Microscopía Confocal/métodos , Microscopía Fluorescente/métodos , Conformación de Ácido Nucleico , Fragmentos de Péptidos/química , Plásmidos/metabolismo , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Receptor PAR-1/química , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes de Fusión/química , Trombina/química
11.
Invest Ophthalmol Vis Sci ; 51(11): 5825-32, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20538980

RESUMEN

PURPOSE: Parstatin is a 41-mer peptide formed by proteolytic cleavage on activation of the PAR1 receptor. The authors recently showed that parstatin is a potent inhibitor of angiogenesis. The purpose of the present study was to evaluate the therapeutic effect of parstatin on ocular neovascularization. METHODS: Choroidal neovascularization was generated in mice using laser-induced rupture of Bruch's membrane and was assessed after 14 days after perfusion of FITC-dextran. Oxygen-induced retinal neovascularization was established in neonatal mice by exposing them to 75% O(2) at postnatal day (P)7 for 5 days and then placing them in room air for 5 days. Evaluation was performed on P17 after staining with anti-mouse PECAM-1. The effect of parstatin was tested after intravitreal administration. The effects of subconjunctival-injected parstatin on corneal neovascularization and inflammation in rats were assessed 7 days after chemical burn-induced corneal neovascularization. Retinal leukostasis in mice was assessed after perfusion with FITC-conjugated concanavalin A. RESULTS: Parstatin potently inhibited choroidal neovascularization with an IC(50) of approximately 3 µg and a maximum inhibition of 59% at 10 µg. Parstatin suppressed retinal neovascularization with maximum inhibition of 60% at 3 µg. Ten-microgram and 30-µg doses appeared to be toxic to the neonatal retina. Subconjunctival parstatin inhibited corneal neovascularization, with 200 µg the most effective dose (59% inhibition). In addition, parstatin significantly inhibited corneal inflammation and VEGF-induced retinal leukostasis. In all models tested, scrambled parstatin was without any significant effect. CONCLUSIONS: Parstatin is a potent antiangiogenic agent of ocular neovascularization and may have clinical potential in the treatment of angiogenesis-related ocular disorders.


Asunto(s)
Neovascularización Coroidal/prevención & control , Neovascularización de la Córnea/prevención & control , Modelos Animales de Enfermedad , Queratitis/prevención & control , Fragmentos de Péptidos/uso terapéutico , Receptor PAR-1/uso terapéutico , Animales , Neovascularización Coroidal/patología , Conjuntiva/efectos de los fármacos , Neovascularización de la Córnea/patología , Inyecciones Intravítreas , Queratitis/patología , Leucostasis/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Enfermedades de la Retina/prevención & control , Neovascularización Retiniana/patología , Neovascularización Retiniana/prevención & control
12.
Eur Cytokine Netw ; 20(4): 171-9, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20167556

RESUMEN

A plethora of endogenous modulators of angiogenesis have been identified and their roles in the molecular and cellular events that mediate and regulate angiogenesis have been proposed. In this review, we summarize the recent findings on the role of thrombin/thrombosis on angiogenesis and other related pathophysiological processes. The mechanisms by which thrombin itself and its receptor PAR1 orchestrate many cellular events through interaction with a variety of other factors and cell types are discussed. These new data point to the complexity of the regulatory processes involved in the angiogenic cascade, which may be tissue specific, and dependent upon the pathology involved. The understanding of these events may provide targets for therapeutic intervention in disease states where angiogenesis is disturbed.


Asunto(s)
Neovascularización Patológica/metabolismo , Trombina/metabolismo , Animales , Apoptosis , Coagulación Sanguínea/fisiología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Neovascularización Patológica/fisiopatología , Neovascularización Patológica/terapia , Receptor PAR-1/metabolismo
13.
Cardiovasc Res ; 83(2): 325-34, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19380418

RESUMEN

AIMS: Thrombin activates protease-activated receptor 1 by proteolytic cleavage of the N-terminus. Although much research has focused on the activated receptor, little is known about the 41-amino acid N-terminal peptide (parstatin). We hypothesized that parstatin would protect the heart against ischaemia-reperfusion injury. METHODS AND RESULTS: We assessed the protective role of parstatin in an in vivo and in vitro rat model of myocardial ischaemia-reperfusion injury. Parstatin treatment before, during, and after ischaemia decreased infarct size by 26%, 23%, and 18%, respectively, in an in vivo model of ischaemia-reperfusion injury. Parstatin treatment immediately before ischaemia decreased infarct size by 65% and increased recovery in ventricular function by 23% in an in vitro model. We then assessed whether parstatin induced cardioprotection by activation of a Gi-protein-dependent pathway. Gi-protein inactivation by pertussis toxin completely abolished the cardioprotective effects. The cardioprotective effects were also abolished by inhibition of nitric oxide synthase (NOS), extracellular signal-regulated kinases 1/2 (ERK1/2), p38 mitogen-activated protein kinase (p38 MAPK), and K(ATP) channels in vitro. Furthermore, parstatin increased coronary flow and decreased perfusion pressure in the isolated heart. The vasodilatory properties of parstatin were confirmed in rat coronary arterioles. CONCLUSION: A single treatment of parstatin administered prior to ischaemia confers immediate cardioprotection by recruiting the Gi-protein activation pathway including p38 MAPK, ERK1/2, NOS, and K(ATP) channels. Parstatin exerts effects on both the cardiomyocytes and the coronary circulation to induce cardioprotection. This suggests a potential therapeutic role of parstatin in the treatment of cardiac injury resulting from ischaemia and reperfusion.


Asunto(s)
Cardiotónicos/farmacología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Fragmentos de Péptidos/farmacología , Receptor PAR-1/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Circulación Coronaria/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Guanilato Ciclasa/metabolismo , Frecuencia Cardíaca/efectos de los fármacos , Canales KATP/efectos de los fármacos , Canales KATP/metabolismo , Masculino , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares/metabolismo , Transducción de Señal/efectos de los fármacos , Guanilil Ciclasa Soluble , Factores de Tiempo , Vasodilatación/efectos de los fármacos , Función Ventricular Izquierda/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Am J Physiol Cell Physiol ; 294(5): C1215-26, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18367587

RESUMEN

Thrombin has been reported to play a pivotal role in the initiation of angiogenesis by indirectly regulating and organizing a network of angiogenic molecules. In addition, it has been proposed that thrombin can directly activate endothelial cell proliferation. However, in this report it was shown that thrombin is a poor growth factor for human endothelial cells, and its modest mitogenic activity is mediated indirectly by the release of heparin-binding epidermal growth factor, subsequent to proteinase-activated receptor 1 (PAR1) activation. On the other hand, it was demonstrated that thrombin is a potent anti-apoptotic factor for endothelial cells, pointing to a novel role of thrombin in vascular protection. Analysis by annexin V-propidium iodide double staining revealed that thrombin, specifically, promoted survival of serum-starved endothelial cells in a concentration-dependent manner. In contrast to its mitogenic effect, the anti-apoptotic effect of thrombin was largely independent of its catalytic activity and was mediated through interaction with alphanubeta3 and alpha5beta1 integrins, whereas the involvement of PAR1 was limited. These results provide new insights in understanding the role of thrombin in endothelial cell signaling and vascular biology.


Asunto(s)
División Celular/fisiología , Supervivencia Celular/fisiología , Endotelio Vascular/citología , Trombina/fisiología , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Adhesión Celular/efectos de los fármacos , Células Cultivadas , Receptores ErbB/efectos de los fármacos , Receptores ErbB/fisiología , Citometría de Flujo , Humanos , Venas Umbilicales
15.
Semin Thromb Hemost ; 33(7): 680-7, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18000795

RESUMEN

Angiogenesis, the growth of new blood vessels from preexisting ones, is a necessary component of embryogenesis, wound healing, and the proliferative phase of the female reproductive cycle. Angiogenesis also plays a critical role in important pathologic processes such as cancer and cardiovascular complications. In addition, clinical, laboratory, and pharmacologic evidence has shown a link between angiogenesis, coagulation, hemostasis, and thrombosis in the settings of these pathologies. Recent studies in our laboratory revealed that thrombin has a significant stimulatory effect on angiogenesis. This effect of thrombin is independent of fibrin formation and can be attributed mainly to the activation of protease-activated receptor-1 (PAR-1). PAR-1 is widely expressed in vascular cells and is involved in cardiovascular complications such as atherosclerosis, restenosis, and neointimal formation. It is also expressed in many cancer cells contributing to induction of tumor growth and metastasis. In this review, we will summarize our present-day understanding of the role of thrombin and PAR-1 in angiogenesis and the potential therapeutic utility of targeting PAR-1 in angiogenesis-related disease, such as atherosclerosis, restenosis, and cancer.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Neoplasias/metabolismo , Neovascularización Patológica/prevención & control , Neovascularización Fisiológica/efectos de los fármacos , Receptor PAR-1/antagonistas & inhibidores , Receptor PAR-1/metabolismo , Animales , Apoptosis , Coagulación Sanguínea , Vasos Sanguíneos , Enfermedades Cardiovasculares/complicaciones , Enfermedades Cardiovasculares/patología , Endotelio Vascular/metabolismo , Humanos , Metástasis de la Neoplasia , Neoplasias/irrigación sanguínea , Neoplasias/patología , Trombina/metabolismo , Tromboplastina/metabolismo
16.
J Pharmacol Exp Ther ; 318(1): 246-54, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16595737

RESUMEN

Many studies support the notion that protease-activated receptor (PAR)-1 plays a pivotal role in angiogenesis. However, direct evidence and understanding the molecular mechanisms involved were limited because PAR-1-specific antagonists have been developed only recently. In the present study, we evaluated the effects of two well characterized PAR-1 antagonists, SCH79797 ((N-3-cyclopropyl-7-{[4-(1-methylethyl)phenyl]-methyl}-7H-pyrrolo[3,2-f]quinazoline-1,3-diamine)) and RWJ56110 [(alphaS)-N-[(1S)-3-amino-1-[[(phenylmethyl)amino]carbonyl]propyl]-alpha-[[[[[1-(2,6-dichlorophenyl)methyl]-3-(1-pyrrolidinylmethyl)-1H-indol-6-yl]amino]carbonyl]amino]-3,4-difluorobenzenepropanamide], in the angiogenic cascade. These antagonists suppressed both the basic angiogenesis and that stimulated by thrombin in the chick chorioallantoic membrane model in vivo. PAR-1 antagonists also abrogated tube formation in the in vitro Matrigel system. These inhibitory effects were dose-dependent and well correlated with the inhibitory effects of SCH79797 and RWJ56110 on primary endothelial cell proliferation and on the initiation of apoptosis. PAR-1 blockage resulted in inhibition of endothelial cell growth by increasing the sub-G0/G1 fraction and reducing the percentage of cells in the S phase. Consistent with this, PAR-1 antagonists reduced incorporation of [3H]thymidine in endothelial cells and blocked the phosphorylation of extracellular signal-regulated kinases in a fashion depending specifically on PAR-1 activation. Analysis by annexin V/propidium iodide staining and poly(ADP-ribose) polymerase cleavage revealed that PAR-1 blockage increased apoptotic cell death by a mechanism involving caspases. These results provide further evidence that PAR-1 is a key receptor that mediates angiogenesis and suggest PAR-1 as target for developing antiangiogenic agents with potential therapeutic application in cancer and other angiogenesis-related diseases.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Apoptosis/fisiología , Células Endoteliales/fisiología , Neovascularización Fisiológica/fisiología , Receptor PAR-1/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Embrión de Pollo , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Pirroles/farmacología , Quinazolinas/farmacología , Receptor PAR-1/fisiología
17.
J Biol Chem ; 280(33): 29393-6, 2005 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-15998637

RESUMEN

Previous studies have suggested that thrombin interacts with integrins in endothelial cells through its RGD (Arg-187, Gly-188, Asp-189) sequence. All existing crystal structures of thrombin show that most of this sequence is buried under the 220-loop and therefore interaction via RGD implies either partial unfolding of the enzyme or its proteolytic digestion. Here, we demonstrate that surface-absorbed thrombin promotes attachment and migration of endothelial cells through interaction with alpha(v)beta(3) and alpha(5)beta(1) integrins. Using site-directed mutants of thrombin we prove that this effect is mediated by the RGD sequence and does not require catalytic activity. The effect is abrogated when residues of the RGD sequence are mutated to Ala and is not observed with proteases like trypsin and tissue-type plasminogen activator, unless the RGD sequence is introduced at position 187-189. The potent inhibitor hirudin does not abrogate the effect, suggesting that thrombin functions through its RGD sequence in a non-canonical conformation. A 1.9-Angstroms resolution crystal structure of free thrombin grown in the presence of high salt (400 mm KCl) shows two molecules in the asymmetric unit, one of which assumes an unprecedented conformation with the autolysis loop shifted 20 Angstroms away from its canonical position, the 220-loop entirely disordered, and the RGD sequence exposed to the solvent.


Asunto(s)
Oligopéptidos/fisiología , Trombina/química , Trombina/fisiología , Movimiento Celular , Células Endoteliales/fisiología , Humanos , Conformación Proteica , Relación Estructura-Actividad
18.
Semin Thromb Hemost ; 30(1): 63-9, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15034798

RESUMEN

Clinical, laboratory, histopathological, and pharmacological evidence support the notion that the coagulation system, which is activated in most cancer patients, plays an important role in tumor biology. Our laboratory has provided evidence that thrombin activates angiogenesis, a process which is essential in tumor growth and metastasis. This event is independent of fibrin formation. At the cellular level many actions of thrombin can contribute to activation of angiogenesis: (1). Thrombin decreases the ability of endothelial cells to attach to basement membrane proteins. (2). Thrombin greatly potentiates vascular endothelial growth factor- (VEGF-) induced endothelial cell proliferation. This potentiation is accompanied by up-regulation of the expression of VEGF receptors (kinase insert domain-containing receptor [KDR] and fms-like tyrosine kinase [Flt-1]). (3). Thrombin increases the mRNA and protein levels of alpha (v)beta (3) integrin and serves as a ligand to this receptor. Furthermore, thrombin increases the secretion of VEGF and enhances the expression and protein synthesis of matrix metalloprotease-9 and alpha (v)beta (3) integrin in human prostate cancer PC-3 cells. These results could explain the angiogenic and tumor-promoting effect of thrombin and provide the basis for development of thrombin receptor mimetics or antagonists for therapeutic application.


Asunto(s)
Neoplasias/patología , Neovascularización Patológica/etiología , Trombina/fisiología , Endotelio Vascular/patología , Humanos , Receptores de Trombina/fisiología
19.
Am J Physiol Cell Physiol ; 283(5): C1501-10, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12372811

RESUMEN

Thrombin has been reported to be a potent angiogenic factor both in vitro and in vivo, and many of the cellular effects of thrombin may contribute to activation of angiogenesis. In this report we show that thrombin-treatment of human endothelial cells increases mRNA and protein levels of alphavbeta3-integrin. This thrombin-mediated effect is specific, dose dependent, and requires the catalytic site of thrombin. In addition, thrombin interacts with alphavbeta3 as demonstrated by direct binding of alphavbeta3 protein to immobilized thrombin. This interaction of thrombin with alphavbeta3-integrin, which is an angiogenic marker in vascular tissue, is of functional significance. Immobilized thrombin promotes endothelial cells attachment, migration, and survival. Antibody to alphavbeta3 or a specific peptide antagonist to alphavbeta3 can abolish all these alphavbeta3-mediated effects. Furthermore, in the chick chorioallantoic membrane system, the antagonist peptide to alphavbeta3 diminishes both basal and the thrombin-induced angiogenesis. These results support the pivotal role of thrombin in activation of endothelial cells and angiogenesis and may be related to the clinical observation of neovascularization within thrombi.


Asunto(s)
Hemostáticos/farmacología , Neovascularización Patológica/fisiopatología , Receptores de Vitronectina/metabolismo , Trombina/farmacología , Alantoides/citología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Embrión de Pollo , Endotelio Vascular/citología , Expresión Génica/efectos de los fármacos , Hemostáticos/metabolismo , Humanos , ARN Mensajero/análisis , Receptores de Vitronectina/genética , Trombina/metabolismo , Venas Umbilicales/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA