Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Bioorg Med Chem Lett ; 23(9): 2595-7, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23528297

RESUMO

By screening directed libraries of serine hydrolase inhibitors using the cell surface form of endothelial lipase (EL), we identified a series of carbamate-derived (EL) inhibitors. Compound 3 raised plasma HDL-C levels in the mouse, and a correlation was found between HDL-C and plasma compound levels. Spectroscopic and kinetic studies support a covalent mechanism of inhibition. Our findings represent the first report of EL inhibition as an effective means for increasing HDL-C in an in vivo model.


Assuntos
HDL-Colesterol/sangue , Endotélio Vascular/enzimologia , Inibidores Enzimáticos/química , Lipase/antagonistas & inibidores , Tiocarbamatos/química , Animais , Endotélio Vascular/efeitos dos fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Concentração de Íons de Hidrogênio , Lipase/metabolismo , Lipoproteínas HDL/deficiência , Lipoproteínas HDL/genética , Lipoproteínas HDL/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tiocarbamatos/síntese química , Tiocarbamatos/farmacologia
2.
Biotech Histochem ; 85(3): 195-204, 2010 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-20109096

RESUMO

We previously reported a sensitive, quantitative immunohistochemical assay using formalin fixed, paraffin embedded rat testicular tissues to assess the degree of proliferation-related toxicity. An indexing scheme was devised based on the percentage of PCNA-positive cells positioned as a single layer along the basement membrane at the perimeter of similarly staged seminiferous tubules (PCNA index). We observed significant decreases in the PCNA index in testes of rats treated with an experimental compound that has been shown to produce testicular histopathology. This relatively simple assay provided a more quantitative and sensitive assessment of early testicular toxicity. A separate investigation of the rates of apoptosis in adjacent serial sections of affected rat seminiferous tubules showed that the incidence of apoptosis increased as the rate of proliferation of spermatogonial cells in the tubules decreased. Therefore, we developed a simultaneous PCNA immunohistochemical and TUNEL histochemical assay not only to reduce preparation and analysis time but also to allow determination of the relation between effects of various compounds producing testicular toxicity on the two cellular processes within the same tissue section. We show that an experimental compound known to cause testicular toxicity produced a concurrent reduction of proliferation and increase in apoptosis in seminiferous tubules. In dose-response studies, we show that increased apoptosis was apparent at lower doses that did not show a significant decrease in PCNA, thus indicating the greater sensitivity of the TUNEL indexing assay to detect early evidence of toxicity. Detailed analyses show the presence of TUNEL-positive cells in tubules with normal PCNA labeling, which suggests that an effect on apoptosis occurs prior to significant changes in cell proliferation in the meiotic pathway for this particular testicular toxicant. This single assay employing the simultaneous dual labeling of apoptosis and proliferation has potential utility for detecting early testicular toxicity of experimental compounds in preclinical development and shedding light on potential cellular mechanisms for toxicity, which should help identify compounds with reduced testicular toxicity.


Assuntos
Antígeno Nuclear de Célula em Proliferação/toxicidade , Túbulos Seminíferos/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Marcação In Situ das Extremidades Cortadas , Masculino , Ratos , Ratos Sprague-Dawley
3.
Biotech Histochem ; 83(5): 211-20, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19016366

RESUMO

It is well known that toxicants such as cyclophosphamide and ethanol can have deleterious effects on normal spermatogenesis. End points such as testis weight and sperm counts have been used widely to assess gross structural and functional changes in testes resulting from toxicant exposure. Histopathological assessments are more sensitive measures of testicular health, but generally they are neither quantitative nor sensitive enough to detect early toxicity. Recently, immunolabeling cells with proliferating cell nuclear antigen (PCNA) has been used to identify proliferating spermatogonia; however, there have been no systematic attempts to quantify these changes. We have developed a sensitive, reliable and quantitative assay using immunohistochemistry on formalin fixed, paraffin embedded rat testes to assess the degree of proliferation-related toxicity. An indexing scheme was derived based on the determination of radially positioned PCNA-positive cells within similarly staged seminiferous tubules presenting a single layer of PCNA-positive cells along the basement membrane of the basal tubular compartment. An average of 60 tubules in the testes were counted per animal. Our results show significant decreases in the PCNA index in rats treated with an experimental compound that has been shown to produce testicular histopathology. The analysis provides a quick, reliable, sensitive, and quantitative means for assessing early testicular toxicity. The assay has potential utility as an in vivo biomarker for detecting early testicular toxicity of experimental compounds in preclinical development as well as for refining follow-up compounds with reduced testicular toxicity.


Assuntos
Biomarcadores/análise , Avaliação Pré-Clínica de Medicamentos , Antígeno Nuclear de Célula em Proliferação/análise , Testículo/citologia , Animais , Imuno-Histoquímica , Masculino , Ratos , Ratos Sprague-Dawley , Túbulos Seminíferos/química , Sensibilidade e Especificidade , Espermatogônias/química , Espermatogônias/citologia
4.
Br J Pharmacol ; 150(4): 415-23, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17211454

RESUMO

BACKGROUND AND PURPOSE: While investigating the effects of systemic urotensin II (U-II), a potent vasoactive peptide acting at the UT receptor, we observed ear pinna flushing after systemic administration to conscious rats. In the present study, U-II-induced ear flushing was quantified in terms of ear pinna temperature change and potential mechanisms were explored. EXPERIMENTAL APPROACH: U-II-induced ear flushing was quantified by measuring lateral ear pinna temperature changes and compared to that of calcitonin gene-related peptide (CGRP), a known cutaneous vasodilator. Further, the effects of a variety of pharmacological agents on U-II-induced ear flushing were explored. KEY RESULTS: Subcutaneous injection of U-II (9 microg kg(-1))produced localized ear pinna flushing with an onset of approximately 15 min, a duration of approximately 30 min and a maximal temperature change of 9 degrees C. In contrast, CGRP caused cutaneous flushing within multiple cutaneous beds including the ear pinna with a shorter onset and greater duration than U-II. A potent UT receptor antagonist, urantide, blocked U-II-induced ear flushing but did not affect CGRP-induced ear flushing. Pretreatment with indomethacin or L-Nomega-nitroarginine methylester (L-NAME) abolished U-II-induced ear flushing. Mecamylamine or propranolol did not affect this response to U-II. Direct intracerebroventricular injection studies suggested that the ear flushing response to U-II was not mediated directly by the CNS. CONCLUSION AND IMPLICATIONS: Our results suggest that U-II-induced ear flushing and temperature increase is mediated by peripheral activation of the UT receptor and involves prostaglandin- and nitric oxide-mediated vasodilation of small capillary beds in the rat ear pinna.


Assuntos
Orelha Externa/irrigação sanguínea , Rubor/induzido quimicamente , Urotensinas/farmacologia , Antagonistas Adrenérgicos beta/farmacologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Temperatura Corporal/efeitos dos fármacos , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Inibidores Enzimáticos/farmacologia , Indometacina/farmacologia , Injeções Subcutâneas , Masculino , Mecamilamina/farmacologia , NG-Nitroarginina Metil Éster/farmacologia , Antagonistas Nicotínicos/farmacologia , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Propranolol/farmacologia , Ratos , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional/efeitos dos fármacos , Urotensinas/administração & dosagem , Urotensinas/antagonistas & inibidores , Vasodilatadores/farmacologia
5.
Biochemistry (Mosc) ; 67(1): 56-64, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11841340

RESUMO

The serine protease thrombin is well recognized as being pivotal to the maintenance of hemostasis under both normal and pathological conditions. Its cellular actions are mediated through a unique family of protease-activated receptors (PARs). These receptors represent a novel family of G protein-coupled receptors that undergo proteolytic cleavage of their amino terminus and subsequent autoactivation by a tethered peptide ligand. This paper reviews the consequences of PAR activation in thrombosis, vascular injury, inflammation, tissue injury, and within the tumor microenvironment.


Assuntos
Endopeptidases/metabolismo , Receptores de Trombina/metabolismo , Trombina/metabolismo , Animais , Humanos , Modelos Biológicos , Neoplasias/metabolismo , Receptor PAR-1 , Trombose
6.
Thromb Res ; 104(2): 113-26, 2001 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-11672755

RESUMO

RWJ-53308 is a novel nonpeptide glycoprotein IIb/IIIa (GPIIb/IIIa) antagonist that inhibits fibrinogen binding to GPIIb/IIIa with an IC(50) of 0.4+/-0.3 nM. RWJ-53308 inhibits thrombin-induced platelet aggregation in human gel-filtered platelets (IC(50)=60+/-12 nM) and platelet aggregation in human platelet-rich plasma (PRP) in response to collagen, arachidonic acid, ADP, and SFLLRN-NH(2) (IC(50)=60+/-10, 150+/-30, 70+/-4, and 160+/-80 nM, respectively). The potency of RWJ-53308 in dog and guinea pig PRP is similar to human PRP. RWJ-53308 inhibits ex vivo collagen- and ADP-induced platelet aggregation in conscious dogs for up to 4 h following 0.3 mg/kg iv, and through 4 and 6 h following 1 and 3 mg/kg po. Oral bioavailability is 16+/-7%. RWJ-53308 reduces thrombus weight in a canine arteriovenous (AV) shunt model following intravenous (0.01-0.1 mg/kg) and oral (3 mg/kg) administration. In a guinea pig carotid artery pinch-injury model, RWJ-53308 completely suppresses thrombus-induced cyclic flow reductions (CFR) at 0.7 mg/kg iv. RWJ-53308 also blocks thrombus formation in photoactivation- and ferric chloride-induced models of thrombosis in guinea pigs at 0.3 and 1 mg/kg iv, respectively. In summary, RWJ-53308 is a potent orally active GPIIb/IIIa antagonist that may be useful for both acute and chronic treatment of arterial thrombotic disorders.


Assuntos
Ácidos Nipecóticos/farmacocinética , Inibidores da Agregação Plaquetária/farmacocinética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Piridinas/farmacocinética , Animais , Disponibilidade Biológica , Modelos Animais de Doenças , Cães , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Fibrinogênio/efeitos dos fármacos , Fibrinogênio/metabolismo , Cobaias , Humanos , Concentração Inibidora 50 , Ácidos Nipecóticos/administração & dosagem , Ácidos Nipecóticos/toxicidade , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/administração & dosagem , Inibidores da Agregação Plaquetária/toxicidade , Ligação Proteica/efeitos dos fármacos , Piridinas/administração & dosagem , Piridinas/toxicidade , Ratos , Trombose/tratamento farmacológico
7.
Bioorg Med Chem Lett ; 11(19): 2619-22, 2001 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-11551763

RESUMO

Conformationally constrained analogues of the GPIIb/IIIa antagonist elarofiban (RWJ-53308) have been synthesized and biologically evaluated. The 1,2,4-triazolo[3,4-a]pyridine scaffold provided potent antagonists with favorable pharmacodynamic and pharmacokinetic attributes in dogs. Compounds 12a and 13a exhibited enhancements in oral bioavailability, t(1/2), and ex vivo duration of action (inhibition of ADP-induced platelet aggregation) relative to elarofiban.


Assuntos
Ácidos Nipecóticos/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Piridinas/química , Piridinas/farmacologia , Triazóis/química , Animais , Cães , Ácidos Nipecóticos/síntese química , Ácidos Nipecóticos/química , Ácidos Nipecóticos/farmacocinética , Inibidores da Agregação Plaquetária/síntese química , Inibidores da Agregação Plaquetária/química , Inibidores da Agregação Plaquetária/farmacocinética , Piridinas/síntese química , Piridinas/farmacocinética
8.
J Pharmacol Exp Ther ; 298(1): 34-42, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11408522

RESUMO

Human platelets possess two distinct thrombin-activated receptors, PAR-1 (protease-activated receptor-1) and PAR-4, whereas human vascular smooth muscle cells possess only PAR-1. Although such thrombin receptors have been studied extensively in vitro, their physiological roles are still rather ill-defined. We have now employed a potent, selective PAR-1 antagonist, RWJ-58259, to probe the in vivo significance of PAR-1 in thrombosis and vascular injury. RWJ-58259 was examined in two thrombosis models in guinea pigs: the arteriovenous (A-V) shunt assay (monitoring thrombus weight) and the Rose Bengal intravascular photoactivation assay (monitoring time to occlusion). Administration of RWJ-58259 (10 mg/kg, total i.v. dose) did not inhibit thrombus formation in either thrombosis model, although local, intrashunt delivery in the A-V shunt model did elicit a modest antithrombotic effect (thrombus weight reduction from 35 +/- 2 to 24 +/- 4 mg). These results are consistent with the presence of more than one thrombin-sensitive receptor on guinea pig platelets, in analogy with human platelets. Indeed, we were able to establish that guinea pig platelets express three thrombin receptors, PAR-1, PAR-3, and PAR-4. We also examined RWJ-58259 in a vascular restenosis model involving balloon angioplasty in rats. Perivascular administration of RWJ-58259 (10 mg) significantly reduced neointimal thickness (77 +/- 5 microm to 45 +/- 5 microm, P < 0.05), clearly demonstrating an important role for PAR-1 in vascular injury. From these results, it is evident that a PAR-1 antagonist is not especially effective for treating platelet-dependent thrombosis; however, it could well be beneficial for treating restenosis attendant to arterial injury.


Assuntos
Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/metabolismo , Oclusão de Enxerto Vascular/tratamento farmacológico , Indazóis/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Receptores de Trombina/antagonistas & inibidores , Ureia/farmacologia , Angioplastia com Balão , Animais , Artérias Carótidas/fisiologia , Artérias Carótidas/cirurgia , Técnicas de Cultura de Células , Cobaias , Humanos , Indazóis/química , Indazóis/uso terapêutico , Masculino , Agregação Plaquetária/fisiologia , Ratos , Ratos Sprague-Dawley , Receptor PAR-1 , Receptores de Trombina/fisiologia , Trombose/tratamento farmacológico , Ureia/análogos & derivados , Ureia/química , Ureia/uso terapêutico
10.
Thromb Haemost ; 86(5): 1320-6, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11816724

RESUMO

The response to vascular injury is a complex wound healing response involving cell proliferation, migration, remodeling and inflammation. In the present studies we employed a rat balloon angioplasty model of vascular injury to investigate the potential role of sphingolipid signaling in the response to vascular injury. The enzyme serine palmitoyltransferase (SPT) catalyzes the first committed step in de novo sphingolipid biosynthesis. We observed marked upregulation of expression of both SPT subunits in actively proliferating cells in injured vessels. This enhanced SPT expression occurs in de-differentiated fibroblasts and proliferating vascular smooth muscle cells. The upregulation is particularly apparent in the proliferating luminal edge of the neointima and the adventitial de-differentiated fibroblasts and may serve as a hallmark of this process. The possible functional consequences of this enzyme upregulation and its role in the response to vascular injury are suggested but remain to be determined.


Assuntos
Aciltransferases/metabolismo , Angioplastia com Balão/efeitos adversos , Lesões das Artérias Carótidas/enzimologia , Animais , Fibroblastos/enzimologia , Imuno-Histoquímica , Masculino , Modelos Animais , Músculo Liso Vascular/citologia , Músculo Liso Vascular/enzimologia , Ratos , Ratos Sprague-Dawley , Serina C-Palmitoiltransferase
11.
Arterioscler Thromb Vasc Biol ; 19(12): 3014-24, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10591683

RESUMO

Expression of protease-activated receptor-1 (PAR-1), a cell-surface receptor for thrombin, is increased in balloon-injured rat carotid artery and human atherosclerotic tissue. To examine the role of PAR-1 in vascular injury, we compared vascular injury responses in wild-type (WT) and PAR-1-deficient (PAR-1(-/-)) mice. Arterial injury was induced by inserting a flexible guidewire into the common carotid artery and withdrawing it 6 times with rotation. Bromodeoxyuridine, delivered subcutaneously by osmotic minipump, was used to measure cellular proliferation. Mice were perfusion-fixed at 1, 2, 5, 10, and 14 days after injury. Extensive endothelial damage, mural thrombosis, platelet adherence, and medial smooth muscle cell loss and necrosis were apparent at day 1 in both WT and PAR-1(-/-) mice. The incidence of thrombosis or platelet deposition in WT and PAR-1(-/-) mice declined from 100% at day 1 to 25% and 21%, respectively, at 14 days. Endothelial disruption, as assessed by Evan's blue uptake, was maximum at day 1 and declined by day 14. This apparent endothelial regrowth was similar in WT and PAR-1(-/-) mice. Significant medial thickening at 14 days after injury was similar in WT (from 22.8+/-1.7 to 30.7+/-1.9 microm) and PAR-1(-/-) (from 23.2+/-2.1 to 30.5+/-2.2 microm) mice. Medial area also increased in response to injury but to a lesser extent in PAR-1(-/-) mice (from 0.0250+/-0.0044 to 0.0312+/-0.0047 mm(2)) than in WT mice (from 0.0266+/-0.0040 to 0.0398+/-0.0050 mm(2)). Neointima was variable and occurred in 6 of 13 WT and 5 of 12 PAR-1(-/-) mice. However, intimal area tended to be less in PAR-1(-/-) mice (0. 0016+/-0.0007 mm(2)) compared with WT mice (0.0082+/-0.0032 mm(2)), although this difference did not achieve statistical significance (P=0.06). Cell density was significantly greater in normal carotids from PAR-1(-/-) (6.4+/-0.5 x 10(3)/mm(2)) compared with WT (4.3+/-0. 8 x 10(3)/mm(2)) mice and remained elevated after injury. Vessel and lumen diameters tended to increase in WT mice after injury, whereas vessel diameter was unchanged and lumen diameter actually decreased in PAR-1(-/-) mice. Cell proliferation in injured carotid arteries was similar in PAR-1(-/-) and WT mice. These data suggest that PAR-1(-/-) may play a role in vascular injury responses in this mouse model via possible effects on extracellular matrix regulation.


Assuntos
Angioplastia com Balão/efeitos adversos , Lesões das Artérias Carótidas/fisiopatologia , Receptores de Trombina/genética , Animais , Artérias Carótidas/química , Artérias Carótidas/patologia , Artérias Carótidas/fisiopatologia , Lesões das Artérias Carótidas/patologia , Modelos Animais de Doenças , Endotélio Vascular/química , Endotélio Vascular/lesões , Endotélio Vascular/patologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor PAR-1 , Receptores de Trombina/análise , Trombina/fisiologia , Trombose/fisiopatologia , Fatores de Tempo , Túnica Íntima/química , Túnica Íntima/lesões , Túnica Íntima/patologia
12.
J Med Chem ; 42(25): 5254-65, 1999 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-10602710

RESUMO

Although intravenously administered antiplatelet fibrinogen receptor (GPIIb/IIIa) antagonists have become established in the acute-care clinical setting for the prevention of thrombosis, orally administered drugs for chronic use are still under development. Herein, we present details from our exploration of structure-activity surrounding the prototype fibrinogen receptor antagonist RWJ-50042 (racemate of 1), which was derived from a unique approach involving the gamma-chain of fibrinogen (Hoekstra et al. J. Med. Chem. 1995, 38, 1582). Our analogue studies culminated in the discovery of RWJ-53308 (2), a potent, orally active GPIIb/IIIa antagonist. To progress from RWJ-50042 to a suitable candidate for clinical development, we conducted a series of optimization cycles that employed solid-phase parallel synthesis for the rapid, efficient preparation of nearly 250 analogues, which were assayed for fibrinogen receptor affinity and inhibition of platelet aggregation induced by four different activators. This strategy produced several promising analogues for advanced study, including 3-(3,4-methylenedioxybenzene)-beta-amino acid analogue 3 (significant improved in vivo potency) and 3-(3-pyridyl)-beta-amino acid 2 (significantly improved potency, oral absorption, and duration of action). In dogs, 2 displayed significant ex vivo antiplatelet activity on oral administration at 1.0 mg/kg, 16% systemic oral bioavailability, minimal metabolic transformation, and an excellent safety profile. Additionally, 2 was found to be efficacious in three in vivo thrombosis models: canine arteriovenous (AV) shunt (0.01-0.1 mg/kg, iv), guinea pig photoactivation-induced injury (0.3-3 mg/kg, iv), and guinea pig ferric chloride-induced injury (0.3-1 mg/kg, iv). On the basis of its noteworthy preclinical data, RWJ-53308 (2) was selected for clinical evaluation.


Assuntos
Ácidos Nipecóticos/química , Ácidos Nipecóticos/farmacologia , Inibidores da Agregação Plaquetária/química , Inibidores da Agregação Plaquetária/farmacologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Prolina/análogos & derivados , Piridinas/farmacologia , Administração Oral , Animais , Área Sob a Curva , Disponibilidade Biológica , Cães , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Ácidos Nipecóticos/farmacocinética , Inibidores da Agregação Plaquetária/farmacocinética , Piridinas/química , Piridinas/farmacocinética , Relação Estrutura-Atividade
13.
Biotech Histochem ; 74(4): 172-80, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10555857

RESUMO

We designed an effective quadruple staining protocol that combines histochemistry (HC) and double-labeling immunohistochemistry (IHC: IHC) to stain simultaneously several different morphological features and cell types in vascular lesions. Morphometric image analysis to quantitate vascular wall thickening, lumen area, and proliferating smooth muscle cells on consecutive serial sections is adequate, but morphometric precision and dependable cellular characterization and co-localization could be obtained if analyses are performed on one tissue section. The development of a neointima in the rat carotid artery was induced by angioplasty with a balloon catheter. Tissues were stained for elastin by a modified van Gieson method, then processed for double-labeling IHC:IHC for proliferating cell nuclear antigen and smooth muscle actin followed by hematoxylin staining. The four resulting tissue stains labeled elastin filaments black, proliferating nuclei brown, smooth muscle actin red and nonproliferating nuclei blue. Our staining protocol improved the descriptive and quantitative analysis of relation between smooth muscle cell proliferation and protein expression. Also, neointimal thickening could be measured to analyze its relation to cellular proliferation. Providing one slide with four stains maximizes the information from a single slice of tissue, reduces slide preparation and analysis time, and overcomes the restriction of tissue sample availability. This technique can be applied to a wide spectrum of morphologic and morphometric studies.


Assuntos
Biomarcadores/análise , Artérias Carótidas/patologia , Estenose das Carótidas/patologia , Endotélio Vascular/patologia , Imuno-Histoquímica/métodos , Actinas/análise , Actinas/metabolismo , Fosfatase Alcalina/química , Angioplastia com Balão/efeitos adversos , Angioplastia Coronária com Balão/efeitos adversos , Animais , Artérias Carótidas/metabolismo , Estenose das Carótidas/etiologia , Estenose das Carótidas/metabolismo , Modelos Animais de Doenças , Elastina/análise , Elastina/metabolismo , Endotélio Vascular/metabolismo , Hematoxilina/química , Indicadores e Reagentes/química , Masculino , Fibras Musculares Esqueléticas/metabolismo , Músculo Liso Vascular/patologia , Antígeno Nuclear de Célula em Proliferação/análise , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ratos , Ratos Sprague-Dawley , Coloração e Rotulagem/métodos
14.
Thromb Haemost ; 81(5): 808-14, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10365757

RESUMO

Protease-induced cell signaling is mediated by specific receptors such as the emerging family of protease activated receptors (PARs). Since proteases are involved in various aspects of vascular injury, we assessed expression of PAR-2, a protease-activated receptor closely related to the thrombin receptor (PAR-1) but activated by an unknown protease, in vascular injury. Rat carotids were subjected to balloon-catheter injury and perfusion fixed at 1, 3, 7 or 14 days after injury. Sections of injured and normal carotid arteries were immunohistochemically labeled with a polyclonal antibody raised against the N-terminal residues 37-53 of human PAR-2. Sections were also labeled with antibodies to factor VIII-related antigen, smooth muscle actin and a proliferating cell nuclear antigen (PCNA). In normal vessels, PAR-2 labeling was diffuse and patchy in medial smooth muscle and endothelium. At one and three days after injury, before appearance of neointima, PAR-2 labeling increased in cells adjacent to damaged or necrotic smooth muscle cells. In addition, proliferating adventitial myofibroblasts labeled strongly for PAR-2. At 7 and 14 days after injury, the media and neointima of injured vessels had increased PAR-2 labeling which was most intense at the luminal edge of the neointima. Double immunohistochemical labeling confirmed the greatest expression of PAR-2 in areas with the greatest density of PCNA-positive cells. In addition, PAR-2 mRNA localization using in situ hybridization paralleled PAR-2 expression. The data suggest an upregulation of PAR-2 in response to vascular injury which is associated with medial smooth muscle damage, proliferating adventitial myofibroblasts and smooth muscle cells of the neointima, particularly those at the proliferating luminal edge of the neointima. Possible functional consequences of this receptor upregulation and its role in the response to vascular injury remain to be determined.


Assuntos
Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Músculo Liso Vascular/metabolismo , Receptores de Trombina/biossíntese , Animais , Cateterismo , Divisão Celular , Movimento Celular , Humanos , Masculino , Músculo Liso Vascular/patologia , Ratos , Ratos Sprague-Dawley , Receptor PAR-2 , Transdução de Sinais , Regulação para Cima
15.
J Pharmacol Exp Ther ; 288(2): 671-8, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9918574

RESUMO

We developed mice deficient in protease-activated receptor-2 (PAR-2) or PAR-1 to explore the pathophysiological functions of these receptors. In this report, we evaluated mean arterial pressure and heart rate (HR) changes in response to PAR-1 or PAR-2 activation in anesthetized wild-type (WT), PAR-1-deficient (PAR-1(-/-)), and PAR-2-deficient (PAR-2(-/-)) mice. In WT mice, TFLLRNPNDK, a PAR-1 selective activating peptide, caused hypotension and HR decreases at 1 micromol/kg. TFLLRNPNDK also caused secondary hypertension following L-NAME pretreatment. These responses were absent in PAR-1(-/-) mice. In WT mice, SLIGRL, a PAR-2 selective activating peptide, caused hypotension without changing HR at 0.3 micromol/kg. SLIGRL did not induce hypertension following Nomega-nitrol-arginine-methyl ester-HCl (L-NAME). The response to SLIGRL was absent in PAR-2(-/-) mice. SFLLRN, a nonselective receptor activating peptide caused hypotension and HR decreases in WT mice at 0.3 micromol/kg, as well as secondary hypertension following L-NAME. SFLLRN still induced hypotension in PAR-1(-/-) mice, but HR decrease and secondary hypertension following L-NAME were absent. The hypotensive and bradycardic responses to SFLLRN and TFLLRNPNDK in PAR-2(-/-) mice were accentuated compared with WT mice. By using mouse strains deficient in either PAR-1 or PAR-2, we confirmed the in vivo specificity of TFLLRNPNDK and SLIGRL as respective activating peptides for PAR-1 and PAR-2, and the distinct hemodynamic responses mediated by activation of PAR-1 or PAR-2. Moreover, the accentuated response to PAR-1 activation in PAR-2-deficient mice suggests a compensatory response and potential receptor cross-talk.


Assuntos
Pressão Sanguínea/fisiologia , Frequência Cardíaca/fisiologia , Receptores de Trombina/fisiologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Feminino , Frequência Cardíaca/efeitos dos fármacos , Masculino , Camundongos , Oligopeptídeos/farmacologia , Receptor PAR-2 , Receptores de Trombina/deficiência , Receptores de Trombina/efeitos dos fármacos , Especificidade por Substrato
16.
Can J Physiol Pharmacol ; 76(1): 16-25, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9564545

RESUMO

Vascular expression and cellular functions of the thrombin receptor (PAR-1) and protease activated receptor 2 (PAR-2) suggest similar but distinct vascular regulatory roles. The vascular actions of PAR-1 and PAR-2 in vivo were differentiated by monitoring mean arterial pressure (MAP) and heart rate (HR) of anesthetized mice in response to intravenous SFLLRN (0.1, 0.3, and 1 mumol/kg) and SLIGRL (0.1, 0.3, and 1 mumol/kg), the respective receptor-activating sequences for PAR-1 and PAR-2, and TFLLRNPNDK (0.3, 1, and 3 mumol/kg), a synthetic peptide selective for PAR-1. All peptides dose dependently decreased MAP (order of potency: SLIGRL > SFLLRN > TFLLRNPNDK). SLIGRL induced a more prolonged hypotension with a slow return to baseline, whereas SFLLRN- and TFLLRNPNDK-induced hypotension was followed by a rapid return towards baseline and a sustained moderate hypotension. SFLLRN and TFLLRNPNDK, but not SLIGRL, decreased HR. N omega-Nitro-L-arginine methyl ester HCl (L-NAME), an inhibitor of nitric oxide synthesis, attenuated the cumulative hypotensive response to SLIGRL but had no effect on the SFLLRN and TFLLRNPNDK hypotension. However, L-NAME revealed a rebound hypertension in response to SFLLRN and TFLLRNPNDK but not SLIGRL. In conclusion, activation of either PAR-1 or PAR-2 in vivo results in hypotension. In addition, only PAR-1 activation induced hypertension following L-NAME, reflecting concurrent PAR-1-mediated vasoconstriction. Thus, these different hemodynamic responses in vivo suggest distinct physiological or pathophysiological roles for PAR-1 and PAR-2 in local vascular regulation.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Oligopeptídeos/farmacologia , Receptores de Trombina/agonistas , Acetilcolina/farmacologia , Animais , Relação Dose-Resposta a Droga , Masculino , Camundongos , Oligopeptídeos/administração & dosagem , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/farmacologia , Receptor PAR-1 , Receptor PAR-2
17.
J Pharmacol Exp Ther ; 279(3): 1365-78, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8968361

RESUMO

Thrombin's cellular actions are mediated by a novel G-protein coupled transmembrane receptor. We infused SFLLRN, a peptide that directly activates thrombin receptors, into the left circumflex coronary artery (CFX) of anesthetized dogs to evaluate the cardiovascular effects of thrombin receptor activation in vivo. Intracoronary SFLLRN, 0.9, 9 and 90 nmol/min, produced transient, dose-related increases in CFX blood flow, followed by sustained decreases in CFX and left anterior descending (LAD) blood flow. SFLLRN also decreased positive and negative dP/dtmax, arterial pressure, cardiac output and heart rate. Peripheral vascular resistance transiently decreased and then increased. SFLLRN decreased systolic wall thickening (WT) and increased ST segment level within the CFX perfusion area. In contrast, WT was increased, and ST segment was unchanged in the LAD perfusion area. CFX flow, but not LAD flow, increased transiently above control after SFLLRN infusion. FSLLRN, a peptide that does not activate thrombin receptors, had no effect at 90 nmol/min. The response to intravenous SFLLRN was greatly attenuated when compared with intracoronary infusion, and regional changes in coronary flow and function were absent. Decreases in arterial pressure, heart rate, coronary blood flow, and positive and negative dP/dtmax, were inhibited after bilateral vagotomy. Moreover, arterial pressure and peripheral resistance increased in response to SFLLRN after vagotomy. Initial CFX flow increase, regional dysfunction, ST level changes and hyperemic response were comparable but attenuated after vagotomy. Ex vivo platelet function was not affected by SFLLRN up to 100 microM. We conclude that regional myocardial ischemia and cardiac dysfunction result from thrombin receptor-mediated local coronary vasoconstriction. Thus, thrombin generation at a site of vascular injury or thrombus may significantly affect vascular tone and myocardial perfusion.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Débito Cardíaco/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Receptores de Trombina/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Animais , Cães , Eletrocardiografia , Injeções Intravenosas , Masculino , Fragmentos de Peptídeos/administração & dosagem , Agregação Plaquetária/efeitos dos fármacos , Receptores de Trombina/metabolismo , Vagotomia
18.
Thromb Haemost ; 76(6): 860-6, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8972001

RESUMO

The thrombin receptor (ThrR) is a membrane-bound, G-protein-coupled receptor for the serine protease thrombin. This receptor is expressed in a wide variety of cells and tissues, and elicits a range of physiological responses associated with tissue injury, inflammation, and wound repair. To achieve a better understanding of the physiological role of the ThrR, we have employed homologous recombination to create mice with a disrupted ThrR gene. Following heterozygous (+/-) intercrosses, a total of 351 surviving offspring were genotyped. Only 7% of these offspring were identified as homozygous (-/-) for the disrupted allele, indicating a profound effect on embryonic development. Paradoxically, adult ThrR-/- mice appeared to be normal by anatomical and histological analysis, including their platelet number and function. Similarly, ThrR deficiency had no detectable effect in adult ThrR-/- mice on basal heart rate, arterial blood pressure, vasomotor responses to angiotensin II and acetycholine, and coagulation parameters, even though the ThrR is expressed in many cardiovascular tissue types. In addition, the loss of ThrR function in the peripheral vasculature of adult ThrR-/- mice was confirmed by the absence of various standard hemodynamic effects of the ThrR-activating peptides SFLLRN-NH2 and TFLLRNPNDK-NH2. Our results indicate that ThrR deficiency has a strong impact on fetal development; however. ThrR-/- mice that proceed to full development display surprisingly little change in phenotype compared to the wild-type.


Assuntos
Hemodinâmica/fisiologia , Receptores de Trombina/deficiência , Animais , Regulação da Expressão Gênica , Camundongos , Camundongos Mutantes , Fenótipo , Receptores de Trombina/genética
19.
Am J Physiol ; 270(5 Pt 2): H1585-96, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-8928863

RESUMO

alpha-Thrombin alters vascular tone via a cell surface receptor. We used isolated guinea pig hearts perfused with buffer at constant flow to assess the effects of thrombin-receptor activation on coronary perfusion pressure, left ventricular function, and electrocardiogram. alpha-Thrombin produced concentration-dependent (0.03-1 U/ml), transient decreases in perfusion pressure followed by sustained increases. Concurrently, alpha-thrombin markedly reduced ventricular function. SFLLRN, a peptide that directly activates thrombin receptors, had qualitatively similar effects, except that it was less potent (0.1-30 microM). FSLLRN, a structurally similar peptide that does not activate thrombin receptors, had no effect. alpha-Thrombin and SFLLRN also changed S-T segment level and T-wave morphology. Previous alpha-thrombin exposure markedly inhibited the response to a alpha-thrombin but only moderately attenuated the response to SFLLRN. However, previous SFLLRN exposure did not alter subsequent response to alpha-thrombin or SFLLRN. Pretreatment with hirudin (3 U/ml), an inhibitor of thrombin's proteolytic action, prevented alpha-thrombin but not SFLLRN responses. Cromakalim (0.5 microM), a coronary vasodilator, reversed the effects of alpha-thrombin and SFLLRN on ventricular function, suggesting that depression of ventricular function resulted, in part, from vasoconstriction-induced myocardial perfusion deficit. Our results show that alpha-thrombin at physiologically relevant concentrations, has marked effects on coronary vascular resistance and ventricular function in isolated guinea pig hearts that are mediated by the proteolytically activated thrombin receptor.


Assuntos
Vasos Coronários/efeitos dos fármacos , Coração/efeitos dos fármacos , Receptores de Trombina/efeitos dos fármacos , Trombina/farmacologia , Animais , Benzopiranos/farmacologia , Vasos Coronários/metabolismo , Cromakalim , Cobaias , Hirudinas/farmacologia , Técnicas In Vitro , Masculino , Fragmentos de Peptídeos/farmacologia , Perfusão , Inibidores de Proteases/farmacologia , Pirróis/farmacologia , Receptores de Trombina/fisiologia , Vasodilatadores/farmacologia , Função Ventricular Esquerda
20.
J Cardiovasc Pharmacol ; 23(2): 300-10, 1994 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7511761

RESUMO

RWJ-29009, (6S)-trans(-)-1-(6,7-dihydro-6-hydroxy-5,5-dimethyl-2-nitro-5 H-thieno[3,2-b]pyran-7-yl)-2-piperidinone, is a structurally novel and extremely potent potassium channel activator that may be useful for treatment of hypertension and ischemic heart disease. We assessed the cardiovascular profile of RWJ 29009 in anesthetized and conscious dogs. RWJ 29009 (0.1-2 micrograms/kg intravenously, i.v.) dose-relatedly increased coronary blood flow (CBF) and decreased arterial pressure in anesthetized dogs. Total peripheral resistance and coronary vascular resistance were concurrently reduced without significant changes in heart rate (HR) or cardiac output (CO). Left ventricular (LV) dP/dtmax and myocardial contractile force were decreased only at the highest dose of 10 micrograms/kg. Cromakalim (3-100 micrograms/kg), although much less potent, had a qualitatively similar profile. Glyburide pretreatment (5 mg/kg i.v.) shifted the dose response of RWJ 29009 for increasing CBF and decreasing arterial pressure to the right. The dose responses of cromakalim were similarly shifted to the right, whereas the effects of nifedipine on CBF and arterial pressure were not affected by glyburide. RWJ 29009 (0.3 and 1 microgram/kg) had no effect on myocardial O2 consumption (MVO2) except for a transient increase immediately after administration of 1 microgram/kg. MVO2 returned to control 15 min after dosing, although CBF remained significantly increased. In conscious dogs, RWJ 29009 (0.3-10 micrograms/kg, i.v. and orally, p.o.) produced dose-related increases in CBF and decreases in arterial pressure similar to those produced in anesthetized dogs, except that HR was increased concurrently. The i.v. and p.o. potency of RWJ 29009 were comparable, indicating high oral bioavailability. Thus, RWJ 29009 is an extremely potent coronary and peripheral vasodilator with a cardiovascular profile similar to that of other potassium channel activators. Like those of other potassium channel activators, its mechanism of action appears to involve activation of ATP-regulated potassium channels.


Assuntos
Hemodinâmica/efeitos dos fármacos , Piperidonas/farmacologia , Canais de Potássio/efeitos dos fármacos , Piranos , Tiofenos , Vasodilatadores/farmacologia , Administração Oral , Anestesia , Animais , Circulação Cerebrovascular/efeitos dos fármacos , Circulação Coronária/efeitos dos fármacos , Cães , Relação Dose-Resposta a Droga , Feminino , Frequência Cardíaca/efeitos dos fármacos , Injeções Intravenosas , Masculino , Miocárdio/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Piperidonas/administração & dosagem , Vasodilatadores/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA