Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Exp Eye Res ; 73(6): 753-64, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11846507

RESUMO

This study was undertaken to determine whether experimental retinal detachment produces changes in retinal localization of three isoforms of transforming growth factor beta (TGF-beta) and the type II receptor for this protein. Neural retinas of young adult cats were detached from the pigment epithelium. Survival times varied from 3 to 28 days to study the temporal course of TGF-beta localization during retinal degeneration. ELISA assay for TGF-beta1 and -beta2 was performed on samples of fluid from the vitreous chamber to determine whether active or inactive TGF-beta was present. Confocal microscopy was used to localize TGF-beta1, -beta2 and -beta3 and the type II TGF-beta receptor at the various detachment durations. Following experimental retinal detachment the levels of TGF-beta2 increased in the vitreous chamber but no changes in TGF-beta1 were detected. Levels were increased 3 days post-detachment and continued throughout the 28 day period studied. The most prominent changes in immunolocalization occurred in the TGF-beta1 and -beta2 isoforms. Increased immunolabeling was seen in Müller cells and ganglion cell bodies. Hypertrophied Müller cell processes formed periretinal membranes that were heavily labeled by the TGF-beta2 antibody. Some increased immunostaining for TGF-beta3 was observed in the ganglion cell bodies. Labeling for the TGF-beta type II receptor was seen in Müller cells, ganglion cells and the inner and outer plexiform layers in both normal and detached retinas. Changes in localization of the receptor after detachment paralleled the changes seen in TGF-beta protein localization. These results demonstrate that retinal detachment induces the synthesis and secretion of TGF-beta2. Growth factor and receptor immunolabeling were increased in Müller cells suggesting that this isoform is involved in the retinal gliotic response and may contribute to the development of proliferative vitreoretinopathy.


Assuntos
Descolamento Retiniano/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Western Blotting , Gatos , Ensaio de Imunoadsorção Enzimática , Microscopia Confocal , Isoformas de Proteínas/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Degeneração Retiniana/metabolismo , Corpo Vítreo/metabolismo
2.
Biochem Biophys Res Commun ; 266(2): 382-5, 1999 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-10600512

RESUMO

Angiotensin II (Ang II) appears to participate in the regulation of neovascularization processes in the retina. Migration of perimural cells such as pericytes plays a key role in regulation of angiogenesis. We hypothesize that Ang II stimulates migration of retina pericytes. For this we studied the effects of Ang II on migration of bovine retinal pericytes using modified Boyden chambers and collagen IV-covered polyester membranes. Ang II stimulated migration of pericytes by 54.8 +/- 9.7% (n = 10, p < 0.001). This effect was blocked by an AT(1) receptor antagonist (Losartan) but not by an AT(2) receptor antagonist (PD123319). We determined using checkerboard assays (n = 3) that Ang II induces migration of pericytes by chemotaxis (gradient-dependent), in opposition to chemokinesis (nondirected). Thus, Ang II via its AT(1) receptor acts as a chemotactic factor and stimulates migration of retina microvascular pericytes. This effect may contribute to Ang II-induced regulation of neovascularization processes in the retina.


Assuntos
Angiotensina II/farmacologia , Pericitos/efeitos dos fármacos , Retina/efeitos dos fármacos , Antagonistas de Receptores de Angiotensina , Animais , Bovinos , Movimento Celular/efeitos dos fármacos , Quimiotaxia/efeitos dos fármacos , Imidazóis/farmacologia , Losartan/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Pericitos/metabolismo , Piridinas/farmacologia
3.
Am J Physiol ; 276(5): H1664-71, 1999 05.
Artigo em Inglês | MEDLINE | ID: mdl-10330252

RESUMO

We studied in anesthetized rats whether aminopeptidase P (AMP) may be involved in bradykinin (BK) metabolism and responses. For this we inhibited AMP with the specific inhibitor apstatin (Aps). Studies were done with Aps alone or together with the angiotensin-converting enzyme inhibitor lisinopril (Lis). Aps increased the vasodepressor response to an intravenous bolus of BK (400 ng/kg): vehicle, -3.0 +/- 0.7 mmHg; Aps, -7.8 +/- 0.7 mmHg (P < 0.01 vs. vehicle); Lis, -23.8 +/- 1.8 mmHg; Aps + Lis, -37.5 +/- 1.9 mmHg (P < 0.01 vs. Lis). Aps did not affect the vasodepressor response to BK given into the descending aorta. Plasma BK increased only in Aps + Lis-treated rats (in pg/ml): control, 48.0 +/- 1.4; Lis, 57.5 +/- 7.6; Aps + Lis, 121. 8 +/- 30.6 (P < 0.05 vs. control or Lis), whereas in rats infused with BK (400 ng. kg-1. min-1 for 5 min), Aps increased plasma BK (in pg/ml): control, 51.9 +/- 2.5; Aps, 83.5 +/- 20.5; Lis, 725 +/- 225; Aps + Lis, 1,668 +/- 318 (P < 0.05, Aps vs. control and Lis vs. Aps + Lis). In rats with aortic coarctation hypertension, the acute antihypertensive effects of Aps plus Lis were greater than Lis alone (P < 0.01). Hoe-140, a BK B2-receptor antagonist, abolished the difference. We concluded that in the rat AMP contributes to regulation of BK metabolism and responses.


Assuntos
Aminopeptidases/metabolismo , Bradicinina/farmacologia , Peptidil Dipeptidase A/metabolismo , Vasoconstrição/efeitos dos fármacos , Acetilcolina/farmacologia , Aminopeptidases/antagonistas & inibidores , Angiotensina I/farmacologia , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Aorta/efeitos dos fármacos , Aorta/enzimologia , Coartação Aórtica/metabolismo , Pressão Sanguínea/fisiologia , Bradicinina/metabolismo , Hipertensão/metabolismo , Injeções Intra-Arteriais , Injeções Intravenosas , Lisinopril/farmacologia , Masculino , Peptídeos/farmacologia , Inibidores de Proteases/farmacologia , Ratos , Ratos Wistar , Vasoconstrição/fisiologia
4.
Jpn J Pharmacol ; 81(3): 279-85, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10622216

RESUMO

We studied the effect of chronic nitric oxide synthase (NOS) blockade in the brain on mean arterial pressure [MAP (mmHg)], heart rate [HR (bpm)] and baroreceptor reflex sensitivity [BRS (mean slope: bpm/mmHg)] in Wistar-Kyoto (WKY) and spontaneously hypertensive rats (SHR). Intracerebroventricular (i.c.v.) infusion of the nonselective NOS inhibitor N-Nitro-L-arginine-methylester (L-NAME) (50 microg/kg per day, 11-12 days) increased MAP in WKY and SHR (125+/-2.1 vs 118+/-1.1 controls, P<0.01 and 179+/-3.59 vs 156+/-4.0 controls, P<0.001, respectively) without affecting HR. In L-NAME-treated WKY, BRS to bradycardia was suppressed (-0.79+/-0.09 vs -1.76+/-0.17 controls, P=0.001), whereas in SHR, L-NAME did not affect BRS to bradycardia. BRS to tachycardia remained unaffected in either strain. In WKY, 7-nitroindazole (7-NI x Na+) (34 microg i.c.v./kg per day, 11-12 days), a selective nNOS inhibitor, did not affect MAP or HR, but BRS to bradycardia and tachycardia was decreased (-0.37+/-0.20 vs -0.97+/-0.41 controls, P<0.01 and -1.78+/-0.20 vs -2.52+/-0.40 controls, P=0.05, respectively). In SHR, the same dose of 7-NI x Na+ increased resting MAP (171+/-5.00 vs 150+/-7.00 controls, P<0.05) without affecting HR or BRS to bradycardia or tachycardia. Thus in WKY, BRS to acute changes in systemic blood pressure (BP) is regulated by NO produced by nNOS in the brain, serving as a neurotransmitter in sympathetic and parasympathetic efferent pathways. In SHR, systemic BP is regulated in part by NO released by the type I NOS isoenzyme in the brain.


Assuntos
Pressão Sanguínea/fisiologia , Óxido Nítrico Sintase/fisiologia , Pressorreceptores/fisiologia , Animais , Barorreflexo/efeitos dos fármacos , Barorreflexo/fisiologia , Pressão Sanguínea/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/farmacologia , Frequência Cardíaca/efeitos dos fármacos , Indazóis/administração & dosagem , Indazóis/farmacologia , Injeções Intraventriculares , NG-Nitroarginina Metil Éster/administração & dosagem , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase Tipo I , Ratos , Ratos Endogâmicos SHR
5.
Proc Assoc Am Physicians ; 110(5): 412-21, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9756092

RESUMO

Cell-surface expression of endothelial P-selectin increases adhesion and migration of leukocytes and thus may participate in the pathogenesis of reperfusion injury and atherosclerosis. Angiotensin II (Ang II) is also thought to be involved in such disease states. Nitric oxide (NO) downregulates P-selectin expression, and bradykinin (BK) is known to stimulate NO release from endothelial cells. The objective of this study was to determine the effects of 10-min stimulation of cultured human umbilical endothelial cells (HUVECs) with Ang II, BK, or both on P-selectin expression. Ang II (10(-9)-10(-5) M) stimulated P-selectin expression in a concentration-dependent manner, exhibiting a significant effect at 10(-7) M and reaching a plateau at 5 x 10(-5) M. Pretreatment of HUVECs with the AT1 antagonist losartan and the AT1/AT2 antagonist saralasin but not the AT2 antagonist PD123319 (all at 10(-5) M) markedly attenuated the effect of 10(-7) M Ang II. The effects of Ang II on P-selectin expression were not affected by the presence of the NO synthase inhibitor nitro-L-arginine (L-NA, 5 x 10(-4) M) but were abolished by pretreatment with superoxide dismutase (SOD). BK (10(-6) M) abolished the effects of 10(-7) M Ang II on P-selectin expression but did not affect P-selectin expression induced by desmopressin (0.01-10 microM). L-NA obliterated the blunting effect of BK on the Ang II-induced P-selectin membrane expression. BK alone slightly stimulated P-selectin expression, but in the presence of L-NA, BK markedly enhanced P-selectin expression. The effects of BK in the presence of NA were not altered by SOD, indicating that at difference with Ang II, it acts by a mechanism other than superoxide generation. Thus, Ang II acting on AT1 receptors stimulates superoxide generation, which, in turn, induces expression of P-selectin on the endothelial cell surface. BK inhibits the effects of Ang II, likely acting via NO. We conclude that the balance between Ang II, BK, and NO can regulate P-selectin expression on the endothelial cell membrane, an important component of the cascade leading to leukocyte adhesion to the vascular endothelium.


Assuntos
Angiotensina II/metabolismo , Bradicinina/metabolismo , Selectina-P/biossíntese , Angiotensina II/farmacologia , Antagonistas de Receptores de Angiotensina , Bradicinina/farmacologia , Membrana Celular/metabolismo , Células Cultivadas , Desamino Arginina Vasopressina/metabolismo , Desamino Arginina Vasopressina/farmacologia , Endotélio Vascular/citologia , Humanos , Nitroarginina/farmacologia , Espécies Reativas de Oxigênio , Receptor Tipo 1 de Angiotensina , Receptor Tipo 2 de Angiotensina
6.
J Pharmacol Exp Ther ; 286(1): 403-10, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9655885

RESUMO

Angiotensin 1-7 (Ang 1-7) has been reported to induce relaxation which is partially blocked by a kinin receptor antagonist. We investigated the relationship between kinins and angiotensin peptides with use of preconstricted isolated pig coronary arteries. Ang 1-7 alone (up to 10(-5) M) had no relaxant effect. Bradykinin (BK) (10(-10)-10(-7) M) induced transient relaxation, returning to basal tone, although BK remained in the bath. In these BK-stimulated rings, Ang 1-7 but not BK (both 5 x 10(-6) M) again relaxed the rings by approximately 50%. This relaxation was blocked by a BK B2 antagonist, a kininase, and a nitric oxide synthase inhibitor. Ang 1-7 inhibited purified angiotensin-converting enzyme (ACE) by 30 +/- 3.5% (n = 4) at 10(-6) M. However, in BK-pretreated rings, the ACE inhibitor ramiprilat did not induce relaxation, nor did it affect the relaxant response to Ang 1-7, which suggests that the effect of Ang 1-7 was not caused by ACE inhibition. Ang 1-7-induced vasodilation was reduced by 69.9 +/- 6.2% by an AT2 receptor blocker, PD-123319, and 29.3 +/- 7.3% by an AT1 antagonist, losartan. Neither the nonselective AT1/AT2 receptor antagonist sarthran nor saralasin inhibited the response to Ang 1-7. Ang II did not elicit relaxation either alone or in the presence of losartan, which suggests that activation of AT2 receptors does not cause relaxation. Thus, in the presence of bradykinin, Ang 1-7 relaxes pig coronary arteries via a PD-123319-sensitive mechanism involving nitric oxide, kinins and the BK B2 receptor. The kallikrein-kinin and renin-angiotensin systems may be linked through the interaction of Ang 1-7 and BK.


Assuntos
Angiotensina II/farmacologia , Bradicinina/farmacologia , Vasos Coronários/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Vasodilatação/efeitos dos fármacos , Angiotensina I , Animais , Vasos Coronários/fisiologia , Imidazóis/farmacologia , Técnicas In Vitro , Peptidil Dipeptidase A/metabolismo , Piridinas/farmacologia , Receptor B2 da Bradicinina , Receptores da Bradicinina/fisiologia , Substância P/farmacologia , Suínos
7.
Hypertension ; 30(2 Pt 1): 217-21, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9260983

RESUMO

Angiotensin-(1-7) [Ang-(1-7)] reportedly potentiates hypotensive responses to bradykinin. We studied whether increases in circulating bradykinin would alter responses to Ang-(1-7). In rats anesthetized with thiobutabarbital, bradykinin infusion (5 microg/kg per minute I.A.) resulted in a rapid decrease in mean arterial pressure (MAP) of about 20 mm Hg (P<.01, n=9), although MAP slowly increased by 10 mm Hg after 15 minutes. When Ang-(1-7) (20, 80, and 380 nmol per rat I.A.) was given during bradykinin infusion, it elicited hypotension at 80 and 380 nmol (deltaMAP: -15+/-2.7 and -21+/-3.3 mmHg, respectively; P<.001); this hypotension was not affected by the angiotensin type 1 antagonist L-158,809 (200 microg/kg I.A.), the angiotensin type 2 antagonist PD 123319 (10 mg/kg I.A.), saralasin, or sarthran (10 microg/kg per minute). The bradykinin type 2 receptor antagonist icatibant (30 microg per rat) eliminated the hypotensive responses to Ang-(1-7), which now increased MAP at all doses tested (P<.005). Thus in the presence of bradykinin, Ang-(1-7) induces hypotensive responses that are blocked by icatibant and unaffected by angiotensin receptor antagonists. Ang-(1-7) given to saline-infused rats elicited hypertensive responses at all doses (deltaMAP: 6.4+/-1.5, 12+/-1.6, and 16.3+/-2.7 mmHg, respectively; P<.01); these responses were abolished by L-158,809 and sarthran. In rats pretreated with saralasin, Ang-(1-7) induced hypotension at 80 and 380 nmol (deltaMAP: -7.7+/-2.3 and -9.5+/-2.7, respectively; P<.05), whereas icatibant abolished this response. Thus in the rat, Ang-(1-7) can decrease blood pressure by a mechanism involving the bradykinin type 2 receptor and participates with bradykinin in a vasodepressor pathway that may serve a counterregulatory role, modulating the vasoconstrictor effects of Ang II.


Assuntos
Angiotensina II/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Bradicinina/farmacologia , Hipotensão/fisiopatologia , Fragmentos de Peptídeos/farmacologia , Angiotensina I , Angiotensina II/antagonistas & inibidores , Antagonistas de Receptores de Angiotensina , Animais , Antagonistas dos Receptores da Bradicinina , Sinergismo Farmacológico , Masculino , Ratos , Ratos Sprague-Dawley
8.
Am J Physiol ; 272(5 Pt 1): C1578-85, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9176149

RESUMO

Submandibular enzymatic vasoconstrictor (SEV, rK9) induces vascular contraction and platelet aggregation by a mechanism requiring intact enzymatic activity. On the basis of a published report demonstrating growth-promoting enzymatic activity in an extract of the rat submandibular gland, we hypothesized that SEV would affect DNA synthesis. Recombinant SEV (rSEV), expressed in a baculovirus system, increased DNA synthesis 3- to 25-fold in Chinese hamster lung (CCL39) fibroblasts and in rabbit and rat vascular smooth muscle cells in a dose-dependent manner dose eliciting 50% of maximal response: 0.1-1 nM); this effect was inhibited by pertussis toxin (PTX). Inactive rSEV failed to enhance DNA synthesis. In CCL39 fibroblasts, rSEV increased total phosphoinositol (PI) formation (6- to 10-fold at 10 nM), which was inhibited 49% by PTX; it was also partially inhibited by the tyrosine kinase inhibitor genistein (33%) but was not affected by the protein kinase C inhibitor bisindolylmaleimide. These results show that rSEV increases DNA synthesis and PI formation in mesenchymal cells in a dose- and enzymatic activity-dependent manner through a pathway partially mediated by a PTX-sensitive G protein. Thus SEV can induce growth-associated responses, perhaps through a protease-activated receptor mechanism.


Assuntos
DNA/biossíntese , Fosfatos de Inositol/biossíntese , Pulmão/metabolismo , Músculo Liso Vascular/metabolismo , Serina Endopeptidases/farmacologia , Animais , Células Cultivadas , Cricetinae , Cricetulus , Fibroblastos/metabolismo , Pulmão/citologia , Músculo Liso Vascular/citologia , Coelhos , Ratos , Proteínas Recombinantes , Serina Endopeptidases/biossíntese , Serina Endopeptidases/isolamento & purificação
9.
J Hypertens ; 15(4): 421-9, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9211177

RESUMO

OBJECTIVE: In balloon-injured rat carotid arteries, angiotensin-converting enzyme inhibitors (ACEI) decrease neointima formation, and a kinin receptor antagonist partially reverses this inhibitory effect. We studied which of the events leading to neointima formation are involved in the effects of ACEI and kinins. METHODS: We administered 5 mg/kg per day ramipril, either alone or combined with the kinin receptor antagonist icatibant (Hoe 140), on the days each wave occurred and studied the effects on neointima formation 14 days after balloon injury. Ramipril alone or combined with icatibant had no effect on neointima formation when administered from 2 days before to 3 or 5 days after balloon injury. In contrast, ramipril inhibited neointima formation when administered from day 7 to day 14. Treatment with icatibant had a small effect, which was sufficient to abolish the effects of ramipril (control 0.11 +/- 0.01 mm2, ramipril 0.08 +/- 0.01 mm2; P < 0.05; ramipril plus icatibant 0.09 +/- 0.01 mm2; NS, ramipril plus icatibant versus control). Thus ramipril was not effective when treatment was stopped after 3 or 5 days, but was mildly effective when treatment was administered during the second week. The effect on migration was studied by counting the number of neointimal cells in rats treated from 2 days before to 4 days after injury. Ramipril decreased the number of cells by 93% compared with controls (control 65.0 +/- 13.5 cells/slice, ramipril 4.7 +/- 2.0 cells/slice; P < 0.001), and this effect was blunted significantly by icatibant (19.5 +/- 5.7 cells/slice; P < 0.009, versus ramipril; P < 0.007, versus controls). The influence of treatment on the rate of proliferation (the 5'-bromo-2'-deoxyuridine index) was studied in the media 3 days, and in the neointima 7 and 10 days after balloon injury. Although proliferation peaked in the neointima after 7 days, there were no differences among the groups at any time. Thus neither ramipril nor icatibant affected the rate of proliferation at the times sampled. Ramipril increased cell density (cells/mm2) in the neointima, and this effect was abolished by cotreatment with icatibant (P < 0.05). CONCLUSION: The ACEI needs to be present throughout the experimental period to be most effective. ACEI act on neointima formation in part by inhibiting migration; thus, because ramipril was mildly effective when administered from 7 to 10 days after injury, it is likely that vascular smooth muscle cell migration also occurs continuously. Kinins help mediate roughly 30% of the effect of ACEI on migration. In addition, ACEI, through kinins, affect a process that increases the density of the cells in the neointima, perhaps by decreasing extracellular matrix deposition.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Cininas/fisiologia , Músculo Liso Vascular/efeitos dos fármacos , Animais , Artérias Carótidas/efeitos dos fármacos , Cateterismo/efeitos adversos , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Masculino , Músculo Liso Vascular/patologia , Ramipril/farmacologia , Ratos , Ratos Sprague-Dawley
10.
Thromb Res ; 85(1): 33-44, 1997 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-8983123

RESUMO

Submandibular enzymatic vasoconstrictor (SEV), a member of the kallikrein family of enzymes, elicits biological effects by a proteolytically mediated mechanism. We studied 1) whether SEV is able to aggregate platelets and 2) whether SEV may activate a receptor other than the cloned thrombin receptor. SEV (10(-8)M) aggregated platelets, released ATP and increased intracellular Ca2+. Elastase treatment rendered human platelets unresponsive to SEV and thrombin (TH), but not to cathepsin G. In desensitization experiments performed with gamma-TH, after two successive additions of approximately 50 nM gamma-TH, a third dose elicited 15.8 +/- 3.4% of the initial response (n = 4), but platelets responded to approximately 20 nM SEV by 33.8 +/- 7.2% of control (p < 0.03 vs last response to gamma-TH). After desensitization to SEV (n = 4), the response to a third dose was 4 +/- 1.3% of control, but gamma-TH still induced 37.7 +/- 12.4% aggregation (p < 0.02 vs last response to SEV). Incubation of washed rabbit platelets with alpha-TH digested with elastase (10(-10) M TH added to 7 micrograms/ml elastase for 1 min) rendered them unresponsive to additional challenges with TH, but they still responded to an equipotent dose of SEV (2.7 x 10(-9) M) by 86 +/- 48% of control. In isolated rabbit aortic rings contracted with 10(-6) M norepinephrine (NE) to 42 +/- 3% of maximum. SEV (2.8 x 10(-8) M) caused further contraction to 87 +/- 4%. In contrast, alpha-TH (1.6 x 10(-7) M) tended to relax both NE- and SEV-contracted rings by 14 +/- 2 and 16.2 +/- 2%, respectively (n = 3 each). We concluded that part of the platelet-aggregating effect of SEV may be mediated by activation of a receptor(s) different from that of TH.


Assuntos
Plaquetas/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Glicoproteínas da Membrana de Plaquetas/metabolismo , Receptores de Superfície Celular/metabolismo , Serina Endopeptidases/farmacologia , Animais , Plaquetas/metabolismo , Plaquetas/patologia , Cálcio/metabolismo , Humanos , Coelhos
11.
J Vasc Surg ; 23(2): 314-22, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8637109

RESUMO

PURPOSE: Neointima formation after arterial injury is inhibited by increased blood flow. The object of this study was to determine whether nitric oxide mediates the effect of increased blood flow on neointima formation. METHOD: Balloon catheter-denuded rat carotid arteries were exposed to increased blood flow or control blood flow by ligation of the contralateral carotid artery. Beginning 2 days before balloon denudation, rats were given either saline vehicle alone or the nitric oxide synthase inhibitor N-nitro-L-arginine-methyl ester (L-NAME) at a dose of 10 mg/kg/day or 2 mg/kg/day intraperitoneally. The normalized neointima area was measured 14 days after denudation. RESULTS: Blood flow was significantly increased by ligation of the contralateral carotid artery for all drug treatments (p<0.008). In rats given saline vehicle only, normalized neointima area was significantly reduced after increased blood flow compared with control blood flow (0.33+/-0.04 compared with 0.48+/-0.03; p=0.006). Systolic blood pressure was significantly elevated by treatment with high-dose L-NAME (p=0.002 compared with vehicle), but was not altered by low-dose L-NAME (p=NS compared with vehicle). Normalized neointima area was not significantly reduced after increased carotid blood flow for rats treated with either dose of L-NAME (p=NS). CONCLUSION: The inhibition of neointima formation by increased blood flow was abolished with hypertensive and nonhypertensive doses of the nitric oxide synthase inhibitor L-NAME, which suggests that the L-NAME effects are independent of systemic hemodynamic alterations. It is concluded that flow-induced inhibition of neointima formation is mediated in part by nitric oxide.


Assuntos
Artérias Carótidas/patologia , Hemorreologia , Óxido Nítrico/fisiologia , Túnica Íntima/patologia , Animais , Arginina/análogos & derivados , Arginina/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Artérias Carótidas/efeitos dos fármacos , Lesões das Artérias Carótidas , Cateterismo/efeitos adversos , Cateterismo/instrumentação , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/lesões , Endotélio Vascular/patologia , Inibidores Enzimáticos/farmacologia , Injeções Intraperitoneais , Masculino , NG-Nitroarginina Metil Éster , Óxido Nítrico Sintase/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional , Cloreto de Sódio , Túnica Íntima/efeitos dos fármacos , Túnica Íntima/lesões
12.
J Card Surg ; 10(4 Suppl): 436-40, 1995 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7579839

RESUMO

The consequences of ischemia and reperfusion on endothelial dependent and independent coronary flow patterns following a variety of ischemic insults in isolated perfused rabbit hearts were studied. A blood perfused ex vivo model was developed that provided reliable and stable systolic performance comparable to crystalloid perfused hearts, but with a four to sevenfold decrease in resting coronary flow and a three to six fold increase in coronary flow reserve compared to Krebs' perfusion. Following incremental graded 37 degrees C ischemia of 10 to 45 minutes, blood perfused hearts had compromised systolic performance, but unaffected response to exogenous endothelial dependent and independent agonists whereas in crystalloid perfused hearts, the response to these same agonists was blunted prior to noting a decrement in systolic function. Further studies assessed the consequences of 30 and 45 minutes of ischemia on the regulatory role of basal nitric oxide released by the coronary endothelium. In both blood and crystalloid perfused hearts, basal nitric oxide secretion had a significant and persistent regulatory role on coronary vascular tonus over a tenfold range of coronary flow despite ischemic injury that severely depressed systolic performance. Finally, hearts were preserved in University of Wisconsin (UW) or St. Thomas' (ST) solutions for 4 hours at 4 degrees C. With crystalloid perfusion, ST results in impaired postischemic response to both endothelial dependent and independent agonists. After UW preservation and with all blood perfused hearts, postischemic flow patterns were unchanged. Using physiological blood perfusion protocols, the endothelium and arterial smooth muscle were found more resistant to ischemia-reperfusion injury than the myocyte.


Assuntos
Endotélio Vascular/fisiologia , Isquemia Miocárdica/fisiopatologia , Animais , Circulação Coronária , Reperfusão Miocárdica , Óxido Nítrico/fisiologia , Óxido Nítrico Sintase/antagonistas & inibidores , Coelhos
13.
Hypertension ; 25(5): 1008-13, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7737707

RESUMO

To test the hypothesis that renal kinins act as natriuretic and diuretic hormones, we examined the effect of inhibiting glandular kallikrein on renal function in normotensive unanesthetized rats during normal sodium intake. To inhibit kallikrein at both the luminal and basolateral sides of the distal nephron, we used Fab fragments of monoclonal antibodies to rat urinary kallikrein (Fab-kallikrein). Fab fragments have advantages over intact IgG: they are filtered through the glomerulus and reach the lumen of the distal nephron, where kallikrein is localized and urinary kinins are released. Furthermore, the Fab fragment-antigen complex does not activate the complement system, avoiding the side effects associated with intact antibodies. Fab-kallikrein effectively blocked generation of kinins in the nephron lumen, decreasing urinary kininogenase activity (kallikrein) by 74% to 85% and kinin excretion by 76% to 79%. Fab-kallikrein induced a 30% decrease in urine volume and a 20% to 40% decrease in urinary sodium excretion but did not alter blood pressure, glomerular filtration rate, or renal blood flow. Although urinary prostaglandin E2 excretion also tended to decrease, this change was slower and of lesser magnitude than those of kinin and kininogenase excretion and did not attain statistical significance after Bonferroni's correction. In controls injected with either vehicle or Fab fragments of monoclonal antibodies to ricin (a vegetable protein not present in mammals), none of these parameters decreased significantly. We conclude that renal kinins participate in the short-term regulation of water and sodium excretion in normotensive unanesthetized rats, acting as diuretic and natriuretic hormones.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Água Corporal/metabolismo , Dinoprostona/urina , Calicreínas/fisiologia , Rim/fisiologia , Sódio/urina , Animais , Anticorpos Monoclonais/imunologia , Fragmentos Fab das Imunoglobulinas/imunologia , Calicreínas/antagonistas & inibidores , Masculino , Ratos , Ratos Sprague-Dawley
14.
J Thorac Cardiovasc Surg ; 109(3): 466-72, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7877307

RESUMO

Mechanical function and coronary hemodynamics were assessed in 73 isolated rabbit hearts randomly subjected to 0, 10, 20, 30, or 45 minutes of 37 degrees C global ischemia and 45 minutes of reperfusion in either a modified Krebs buffer or homologous blood-perfused Langendorff mode (n = 7 to 9 hearts per group). Isovolumic developed pressure, resting coronary flow, and response to endothelium-dependent (bradykinin) and -independent (nitroglycerin) agonists were quantitated at defined preload and heart rate. Perfusate did not influence systolic performance, which was impaired after 30 minutes of ischemia and fell to 64% to 72% of preischemic values after 45 minutes of ischemia (p < 0.05). However, basal coronary flow was at least sixfold greater in crystalloid-perfused hearts. Moreover, coronary hyperemia (p < 0.05) persisted for Krebs-perfused hearts subjected to all but the longest ischemic interval. After equilibration, all postischemic blood-perfused hearts had basal flow unchanged from before ischemia. Bradykinin and nitroglycerin induced similar increases in coronary flow for each group before and after each ischemia interval. However, the magnitude of this increase was greater in blood-perfused hearts (p < 0.01) and was not attenuated by the ischemic times encompassed in this protocol. In contrast, endothelium-dependent and -independent coronary flow reserve was abolished after 20 minutes of ischemia or longer in Krebs-perfused hearts. These data suggest that the unphysiologic resting flow patterns of crystalloid-perfused isolated hearts obfuscate interpretation of the interaction between coronary flow reserve and ischemic injury.


Assuntos
Circulação Coronária/fisiologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Reperfusão Miocárdica/métodos , Animais , Sangue , Bradicinina/farmacologia , Circulação Coronária/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Técnicas In Vitro , Soluções Isotônicas , Nitroglicerina/farmacologia , Coelhos , Distribuição Aleatória , Sístole/efeitos dos fármacos
15.
Br J Pharmacol ; 114(1): 6-7, 1995 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-7712030

RESUMO

We examined whether a specific aminopeptidase P (APP) inhibitor, apstatin, increases vasodepressor responses to bradykinin in anaesthetized rats, and whether it would augment blood pressure responses further after treatment with the angiotensin-converting enzyme inhibitor (ACEi), lisinopril. Apstatin doubled the maximum blood pressure response to bradykinin. The area under the curve (AUC), which incorporates both peak blood pressure changes and duration of response, was doubled in apstatin-treated rats vs controls and in the apstatin+lisinopril group vs lisinopril alone. These data demonstrate that APP is an important kinase in vivo.


Assuntos
Aminopeptidases/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Bradicinina/farmacologia , Animais , Relação Dose-Resposta a Droga , Lisinopril/farmacologia , Masculino , Ratos , Ratos Wistar
17.
Hypertension ; 23(6 Pt 2): 865-8, 1994 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7515854

RESUMO

We examined the effect of non-antihypertensive doses of the angiotensin-converting enzyme inhibitor ramipril, kinins, and/or nitric oxide on left ventricular hypertrophy in rats with aortic coarctation. We investigated the effect of either HOE 140, a specific B2 receptor antagonist, or NG-nitro-L-arginine methyl ester (L-NAME), a nitric oxide synthase inhibitor, on the antihypertrophic effect of ramipril at non-antihypertensive doses (10 micrograms/kg per day) failed to alter left ventricular hypertrophy significantly, although a small decrease was obtained. Given at a dose of 1 mg/kg per day for 6 weeks, ramipril prevented increased blood pressure and left ventricular hypertrophy after aortic coarctation. Neither of these effects was blocked by simultaneous administration of HOE 140 (500 micrograms/kg per day). In rats with aortic coarctation treated with L-NAME, blood pressure increased further but left ventricular weight did not. Ramipril (1 mg/kg per day) significantly reduced left ventricular hypertrophy, although blood pressure was still higher than in rats given water alone. The slope of the correlation between left ventricular weight and blood pressure in rats that received L-NAME was significantly lower than in rats that did not (0.52 versus 1.29; P = .008). This suggests that for each 1 mm Hg that the blood pressure increased, the increase in left ventricular weight was less in the L-NAME groups. Thus, only antihypertensive doses of ramipril possessed antihypertrophic activity. Kinins did not participate in the chronic antihypertensive and antihypertrophic effects of ramipril. In hypertension induced or aggravated by chronic nitric oxide synthase, L-NAME partially impaired development of left ventricular hypertrophy for reasons that are unclear.


Assuntos
Cardiomegalia/prevenção & controle , Cininas/fisiologia , Óxido Nítrico/fisiologia , Ramipril/farmacologia , Aminoácido Oxirredutases/antagonistas & inibidores , Animais , Coartação Aórtica/complicações , Arginina/análogos & derivados , Arginina/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Cardiomegalia/etiologia , Relação Dose-Resposta a Droga , Ventrículos do Coração , Hipertensão/etiologia , Masculino , Miocárdio/patologia , NG-Nitroarginina Metil Éster , Óxido Nítrico Sintase , Tamanho do Órgão/efeitos dos fármacos , Ratos
18.
Hypertension ; 23(6 Pt 2): 919-23, 1994 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8206628

RESUMO

It has been reported that kinins mediate part of the beneficial cardiac effects induced by treatment with angiotensin-converting enzyme inhibitors in situations such as ischemia-reperfusion injury, myocardial infarction, and cardiac hypertrophy. However, it is not known whether the heart contains an independent kallikrein-kinin system. We measured kallikrein in tissue and in the incubation medium of heart slices. Heart slices released active and total (trypsin-activatable) kallikrein into the medium (46 +/- 5 and 380 +/- 18 pg bradykinin/mg, respectively, after 1 hour and 78 +/- 6 and 654 +/- 14 pg bradykinin/mg after 2 hours, n = 7). Release was not due to tissue damage because lactate dehydrogenase, a cytosolic marker, decreased from 8.9 +/- 2.9 to 2.9 +/- 1.0 U/mg per hour. Although kallikrein was released, total tissue kallikrein in the slices did not change (423 +/- 25 pg bradykinin/mg in nonincubated slices and 370 +/- 42 pg bradykinin/mg after 2 hours, P = NS), suggesting pool replenishment. Cardiac kallikrein activity was inhibited by incubation with anti-glandular kallikrein antibodies. Pretreatment with the protein synthesis inhibitor puromycin (10 mg IP) lowered release of active kallikrein from 78 +/- 6 to 22 +/- 4 pg bradykinin/mg and total kallikrein from 654 +/- 14 to 113 +/- 9 pg bradykinin/mg (P < .001). By using reverse transcription polymerase chain reaction with kallikrein family oligonucleotide primers and a specific kallikrein probe, we found that mRNA for tissue kallikrein is present in both atrial and ventricular RNA. Kallikrein activity was also detected in primary cultures of neonatal rat atrial and ventricular cardiocytes and their incubation medium.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Calicreínas/metabolismo , Cininas/metabolismo , Miocárdio/metabolismo , Animais , Cromatografia de Afinidade , Eletroforese em Gel de Poliacrilamida , Calicreínas/genética , Masculino , Miocárdio/citologia , Reação em Cadeia da Polimerase , Inibidores de Proteases/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Transcrição Gênica
20.
Hypertension ; 23(1 Suppl): I235-9, 1994 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8282366

RESUMO

N-[1-(R,S)-carboxyl-3-phenylpropyl]-Ala-Ala-Phe-p-aminobenzoate (cFP-AAF-pAB), an active-site-directed inhibitor of metalloendopeptidase-24.15, has been shown to lower blood pressure, increase cardiac output and renal blood flow, and potentiate the intravenous bradykinin-induced vasodepressor response. Because in vivo cFP-AAF-pAB can be converted to N-[1-(R,S)-carboxyl-3-phenylpropyl]-Ala-Ala (a compound with angiotensin converting enzyme inhibitory activity) by metalloendopeptidase-24.11, it is possible that some of its effects are due to angiotensin converting enzyme inhibition. In the present study, we questioned (1) whether cFP-AAF-pAB inhibits angiotensin converting enzyme in vivo and (2) whether cFP-AAF-pAB has renal effects that are independent of its conversion to an angiotensin converting enzyme inhibitor. cFP-AAF-pAB alone (3 mumol in 300 microL per rat) almost abolished the blood pressure response to angiotensin I, suggesting that in vivo it inhibits angiotensin converting enzyme. In rats pretreated with a high dose of enalaprilat (1 mg/kg), cFP-AAF-pAB had no further effect on blood pressure, renal blood flow, or potentiation of the vasodepressor response to bradykinin but still increased glomerular filtration rate by 44 +/- 9% (P < .01); urine volume increased by 118 +/- 10% (P < .001), urinary sodium excretion by 230 +/- 31% (P < .001), urinary potassium excretion by 68 +/- 14% (P < .01), and urinary cyclic GMP by 55 +/- 18% (P < .01). All of these changes were significant compared with enalaprilat/vehicle-treated rats. Fractional excretion of sodium and potassium did not differ from controls.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Taxa de Filtração Glomerular/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Rim/fisiologia , Metaloendopeptidases/antagonistas & inibidores , Oligopeptídeos/farmacologia , Circulação Renal/efeitos dos fármacos , Angiotensina I/farmacologia , Angiotensina II/farmacologia , Animais , Sítios de Ligação , Pressão Sanguínea/efeitos dos fármacos , Bradicinina/farmacologia , Rim/irrigação sanguínea , Rim/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley , Valores de Referência , Fluxo Sanguíneo Regional/efeitos dos fármacos , Micção/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...