Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Science ; 272(5262): 728-31, 1996 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-8614835

RESUMO

The vascular complications of diabetes mellitus have been correlated with enhanced activation of protein kinase C (PKC). LY333531, a specific inhibitor of the beta isoform of PKC, was synthesized and was shown to be a competitive reversible inhibitor of PKC beta 1 and beta 2, with a half-maximal inhibitory constant of approximately 5 nM; this value was one-fiftieth of that for other PKC isoenzymes and one-thousandth of that for non-PKC kinases. When administered orally, LY333531 ameliorated the glomerular filtration rate, albumin excretion rate, and retinal circulation in diabetic rats in a dose-responsive manner, in parallel with its inhibition of PKC activities.


Assuntos
Diabetes Mellitus Experimental/complicações , Angiopatias Diabéticas/prevenção & controle , Inibidores Enzimáticos/farmacologia , Indóis/farmacologia , Isoenzimas/antagonistas & inibidores , Maleimidas/farmacologia , Proteína Quinase C/antagonistas & inibidores , Administração Oral , Albuminúria/prevenção & controle , Animais , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Experimental/fisiopatologia , Angiopatias Diabéticas/enzimologia , Angiopatias Diabéticas/etiologia , Diglicerídeos/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Inibidores Enzimáticos/química , Taxa de Filtração Glomerular/efeitos dos fármacos , Humanos , Indóis/administração & dosagem , Indóis/química , Isoenzimas/metabolismo , Glomérulos Renais/metabolismo , Masculino , Maleimidas/administração & dosagem , Maleimidas/química , Músculo Liso Vascular/enzimologia , Fosforilação/efeitos dos fármacos , Proteína Quinase C/metabolismo , Proteína Quinase C beta , Ratos , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional/efeitos dos fármacos , Fluxo Plasmático Renal/efeitos dos fármacos , Retina/metabolismo , Vasos Retinianos/fisiopatologia , Especificidade por Substrato
2.
Kidney Int ; 73(10): 1114-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18272958

RESUMO

Diabetic retinopathy and diabetic nephropathy are common microvascular complications of diabetes. The kallikrein-kinin system (KKS) has been implicated in the development of both conditions, and, in particular, bradykinin and its receptors have been shown to exert angiogenic and proinflammatory actions. Several of the key processes that underlie the development of diabetic retinopathy, such as increased vascular permeability, edema, neovascularization, and inflammatory changes, have been associated with the KKS, and recent work has shown that components of the KKS, including plasma kallikrein, factor XIIa, and high-molecular-weight kininogen, are present in the vitreous of people with diabetic retinopathy. The role of the KKS in the development of diabetic nephropathy is controversial, with both adverse and protective effects of bradykinin and its receptors reported. The review examines the role of the KKS in pathways central to the development of diabetic retinopathy and compares this with reported actions of this system in diabetic nephropathy. The possibility of therapeutic intervention targeting bradykinin and its receptors as treatment for diabetic microvascular conditions is considered.


Assuntos
Nefropatias Diabéticas/etiologia , Retinopatia Diabética/etiologia , Sistema Calicreína-Cinina/fisiologia , Humanos
3.
Acta Physiol (Oxf) ; 224(1): e13060, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29489063

RESUMO

AIM: Recent work has demonstrated that activation of the epithelial sodium channel (ENaC) by aberrantly filtered serine proteases causes sodium retention in nephrotic syndrome. The aim of this study was to elucidate a potential role of plasma kallikrein (PKLK) as a candidate serine protease in this context. METHODS: We analysed PKLK in the urine of patients with chronic kidney disease (CKD, n = 171) and investigated its ability to activate human ENaC expressed in Xenopus laevis oocytes. Moreover, we studied sodium retention in PKLK-deficient mice (klkb1-/- ) with experimental nephrotic syndrome induced by doxorubicin injection. RESULTS: In patients with CKD, we found that PKLK is excreted in the urine up to a concentration of 2 µg mL-1 which was correlated with albuminuria (r = .71) and overhydration as assessed by bioimpedance spectroscopy (r = .44). PKLK increased ENaC-mediated whole-cell currents, which was associated with the appearance of a 67 kDa γ-ENaC cleavage product at the cell surface consistent with proteolytic activation. Mutating a putative prostasin cleavage site in γ-ENaC prevented channel stimulation by PKLK. In a mouse model for nephrotic syndrome, active PKLK was present in nephrotic urine of klkb1+/+ but not of klkb1-/- mice. However, klkb1-/- mice were not protected from ENaC activation and sodium retention compared to nephrotic klkb1+/+ mice. CONCLUSION: Plasma kallikrein is detected in the urine of proteinuric patients and mice and activates ENaC in vitro involving the putative prostasin cleavage site. However, PKLK is not essential for volume retention in nephrotic mice.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Rim/enzimologia , Natriurese , Síndrome Nefrótica/enzimologia , Calicreína Plasmática/metabolismo , Equilíbrio Hidroeletrolítico , Adulto , Idoso , Animais , Composição Corporal , Estudos de Casos e Controles , Modelos Animais de Doenças , Doxorrubicina , Canais Epiteliais de Sódio/genética , Feminino , Humanos , Rim/fisiopatologia , Masculino , Potenciais da Membrana , Camundongos Knockout , Pessoa de Meia-Idade , Síndrome Nefrótica/genética , Síndrome Nefrótica/fisiopatologia , Síndrome Nefrótica/urina , Estado de Hidratação do Organismo , Calicreína Plasmática/genética , Calicreína Plasmática/urina , Estudos Prospectivos , Eliminação Renal , Insuficiência Renal Crônica/enzimologia , Insuficiência Renal Crônica/fisiopatologia , Insuficiência Renal Crônica/urina , Xenopus laevis
4.
J Clin Invest ; 95(3): 1353-62, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7883982

RESUMO

Angiotensin II (AII)- and Arg8-vasopressin (AVP)-regulated gene expression in vascular cells has been reported to contribute to vascular homeostasis and hypertrophy. In this report, AVP-induced expression of plasminogen activator inhibitor (PAI)-2 mRNA in rat microvessel endothelial (RME) cells was identified using differential mRNA display. Further characterization of vasoactive peptide effects on PAI expression revealed that AII stimulated a 44.8 +/- 25.2-fold and a 12.4 +/- 3.2-fold increase in PAI-2 mRNA in RME cells and rat aortic smooth muscle cells (RASMC), respectively. AII also stimulated a 10- and 48-fold increase in PAI-1 mRNA in RME cells and RASMC, respectively. These AII effects were inhibited by either Sar1, Ile8-angiotensin or the AT1 antagonist DuP 735, but were not significantly altered in the presence of the AT2 antagonist PD123319. AII stimulation of RASMC and RME cells also significantly increased both PAI-1 protein and PAI activity released to the culture medium. Inhibition of protein kinase C completely blocked PMA-stimulated induction of PAI-2 mRNA in both cell types and inhibited the AII-stimulated increase in RASMC by 98.6 +/- 2.8%. In contrast, protein kinase C inhibition only partially decreased the AII-stimulated PAI-2 expression in RME cells by 68.8 +/- 11.1%, suggesting that a protein kinase C-independent mechanism contributes to a 6.9 +/- 1.5-fold AII induction of PAI-2 expression in endothelial cells. AII and PMA also stimulated protein tyrosine phosphorylation in RME cells, and the tyrosine kinase inhibitor genistein partially blocked their induction of PAI-2 mRNA. These findings suggest that AII may regulate plasminogen activation in the vasculature by inducing both PAI-1 and PAI-2 expression.


Assuntos
Angiotensina II/farmacologia , Endotélio Vascular/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Inibidor 1 de Ativador de Plasminogênio/biossíntese , Inibidor 2 de Ativador de Plasminogênio/biossíntese , Antagonistas de Receptores de Angiotensina , Animais , Aorta/citologia , Sequência de Bases , Sondas de DNA , DNA Complementar/genética , Relação Dose-Resposta a Droga , Microcirculação/citologia , Dados de Sequência Molecular , Plasminogênio/metabolismo , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 2 de Ativador de Plasminogênio/genética , Proteína Quinase C/metabolismo , Proteínas Tirosina Quinases/metabolismo , Ratos , Receptores de Angiotensina/metabolismo , Saralasina/farmacologia , Transdução de Sinais
5.
J Clin Invest ; 100(9): 2158-69, 1997 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-9410892

RESUMO

To investigate potential interactions between angiotensin II (AII) and the insulin signaling system in the vasculature, insulin and AII regulation of insulin receptor substrate-1 (IRS-1) phosphorylation and phosphatidylinositol (PI) 3-kinase activation were examined in rat aortic smooth muscle cells. Pretreatment of cells with AII inhibited insulin-stimulated PI 3-kinase activity associated with IRS-1 by 60%. While AII did not impair insulin-stimulated tyrosine phosphorylation of the insulin receptor (IR) beta-subunit, it decreased insulin-stimulated tyrosine phosphorylation of IRS-1 by 50%. AII inhibited the insulin-stimulated association between IRS-1 and the p85 subunit of PI 3-kinase by 30-50% in a dose-dependent manner. This inhibitory effect of AII on IRS-1/PI 3-kinase association was blocked by the AII receptor antagonist saralasin, but not by AT1 antagonist losartan or AT2 antagonist PD123319. AII increased the serine phosphorylation of both the IR beta-subunit and IRS-1. In vitro binding experiments showed that autophosphorylation increased IR binding to IRS-1 from control cells by 2.5-fold versus 1.2-fold for IRS-1 from AII-stimulated cells, suggesting that AII stimulation reduces IRS-1's ability to associate with activated IR. In addition, AII increased p85 serine phosphorylation, inhibited the total pool of p85 associated PI 3-kinase activity, and decreased levels of the p50/p55 regulatory subunit of PI 3-kinase. These results suggest that activation of the renin-angiotensin system may lead to insulin resistance in the vasculature.


Assuntos
Angiotensina II/fisiologia , Insulina/fisiologia , Músculo Liso Vascular/fisiologia , Fosfosserina/metabolismo , Animais , Aorta/fisiologia , Células Cultivadas , Proteínas Substratos do Receptor de Insulina , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Fosfotirosina/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor de Insulina/metabolismo , Receptores de Angiotensina/fisiologia , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Acetato de Tetradecanoilforbol/farmacologia
6.
J Clin Invest ; 96(4): 1759-67, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7560067

RESUMO

Primary cardiac abnormalities have been frequently reported in patients with diabetes probably due to metabolic consequences of the disease. Approximately 2,000 mRNA species from the heart of streptozotocin-induced diabetic and control rats were compared by the mRNA differential display method, two of eight candidate clones thus isolated (DH1 and 13) were confirmed by Northern blot analysis. The expression of clone 13 was increased in the heart by 3.5-fold (P < 0.05) and decreased in the aorta by twofold (P < 0.05) in diabetes as compared to control. Sequence analysis showed that clone 13 is a rat mitochondrial gene. DH1 was predominantly expressed in the heart with an expression level 6.8-fold higher in the diabetic rats than in control (P < 0.001). Insulin treatment significantly (P < 0.001) normalized the expression of DH1 in the hearts of diabetic rats. DH1 expression was observed in cultured rat cardiomyocytes, but not in aortic smooth muscle cells or in cardiac derived fibroblasts. The expression in cardiomyocytes was regulated by insulin and glucose concentration of culture media. The full length cDNA of DH1 had a single open-reading frame with 85 and 92% amino acid identity to human and mouse UDP-GlcNAc:Gal beta 1-3GalNAc alpha R beta 1-6 N-acetylglucosaminyltransferase (core 2 GlcNAc-T), respectively, a key enzyme determining the structure of O-linked glycosylation. Transient transfection of DH1 cDNA into Cos7 cells conferred core 2 GlcNAc-T enzyme activity. In vivo, core 2 GlcNAc-T activity was increased by 82% (P < 0.05) in diabetic hearts vs controls, while the enzymes GlcNAc-TI and GlcNAc-TV responsible for N-linked glycosylation were unchanged. These results suggest that core 2 GlcNAc-T is specifically induced in the heart by diabetes or hyperglycemia. The induction of this enzyme may be responsible for the increase in the deposition of glycoconjugates and the abnormal functions found in the hearts of diabetic rats.


Assuntos
DNA Complementar/isolamento & purificação , Diabetes Mellitus Experimental/enzimologia , Hiperglicemia/enzimologia , Miocárdio/enzimologia , N-Acetilglucosaminiltransferases/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Células Cultivadas , Glicosilação , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos NOD , Dados de Sequência Molecular , Ratos , Ratos Sprague-Dawley
7.
J Clin Invest ; 104(4): 447-57, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10449437

RESUMO

Both insulin resistance and hyperinsulinemia have been reported to be independent risk factors for cardiovascular diseases. However, little is known regarding insulin signaling in the vascular tissues in insulin-resistant states. In this report, insulin signaling on the phosphatidylinositol 3-kinase (PI 3-kinase) and mitogen-activated protein (MAP) kinase pathways were compared in vascular tissues of lean and obese Zucker (fa/fa) rats in both ex vivo and in vivo studies. Ex vivo, insulin-stimulated tyrosine phosphorylation of insulin receptor beta subunits (IRbeta) in the aorta and microvessels of obese rats was significantly decreased compared with lean rats, although the protein levels of IRbeta in the 2 groups were not different. Insulin-induced tyrosine phosphorylation of insulin receptor substrates 1 and 2 (IRS-1 and IRS-2) and their protein levels were decreased in the aorta of obese rats compared with lean rats. The association of p85 subunit to the IRS proteins and the IRS-associated PI 3-kinase activities stimulated by insulin in the aorta of obese rats were significantly decreased compared with the lean rats. In addition, insulin-stimulated serine phosphorylation of Akt, a downstream kinase of PI 3-kinase pathway, was also reduced significantly in isolated microvessels from obese rats compared with the lean rats. In euglycemic clamp studies, insulin infusion greatly increased tyrosine phosphorylation of IRbeta- and IRS-2-associated PI 3-kinase activity in the aorta of lean rats, but only slight increases were observed in obese rats. In contrast, insulin stimulated tyrosine phosphorylation of MAP kinase (ERK-1/2) equally in isolated microvessels of lean and obese rats, although basal tyrosine phosphorylation of ERK-1/2 was higher in the obese rats. To our knowledge, these data provided the first direct measurements of insulin signaling in the vascular tissues, and documented a selective resistance to PI 3-kinase (but not to MAP kinase pathway) in the vascular tissues of obese Zucker rats.


Assuntos
Resistência à Insulina/fisiologia , Insulina/fisiologia , Obesidade/fisiopatologia , Animais , Aorta/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Técnicas In Vitro , Insulina/farmacologia , Proteínas Substratos do Receptor de Insulina , Peptídeos e Proteínas de Sinalização Intracelular , Fígado/metabolismo , Masculino , Microcirculação/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Ratos , Ratos Zucker , Receptor de Insulina/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Tirosina/metabolismo
8.
Circulation ; 101(6): 676-81, 2000 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-10673261

RESUMO

BACKGROUND: The vasodilatory effect of insulin can be acute or increase with time from 1 to 7 hours, suggesting that insulin may enhance the expression of endothelial nitric oxide synthase (eNOS) in endothelial cells. The objective of the present study was to characterize the extent and signaling pathways by which insulin regulates the expression of eNOS in endothelial cells and vascular tissues. METHODS AND RESULTS: Physiological concentrations of insulin (10(-10) to 10(-7) mmol/L) increased the levels of eNOS mRNA, protein, and activity by 2-fold after 2 to 8 hours of incubation in cultured bovine aortic endothelial cells. Insulin enhanced eNOS gene expression in microvessels isolated from Zucker lean rats but not from insulin-resistant Zucker fatty rats. Inhibitors of phosphatidylinositol-3 kinase (PI-3 kinase) decreased the effect of insulin on eNOS gene expression, but a general protein kinase C (PKC) inhibitor, GF109203X or PKCbeta isoform inhibitor, LY333531 enhanced eNOS expression. In contrast, PKC activators inhibited both the activation by insulin of PI-3 kinase and eNOS mRNA levels. Overexpression of PKCbeta isoform in endothelial cells inhibited the stimulation by insulin of eNOS expression and PI-3 kinase activities in parallel. CONCLUSIONS: Insulin can regulate the expression of eNOS gene, mediated by the activation of PI-3 kinase, in endothelial cells and microvessels. Thus, insulin may chronically modulate vascular tone. The activation of PKC in the vascular tissues as in insulin resistance and diabetes may inhibit PI-3 kinase activity and eNOS expression and may lead to endothelial dysfunctions in these pathological states.


Assuntos
Endotélio Vascular/enzimologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Óxido Nítrico Sintase/biossíntese , Animais , Bovinos , Células Cultivadas , Diabetes Mellitus/enzimologia , Inibidores Enzimáticos/farmacologia , Indóis/farmacologia , Maleimidas/farmacologia , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo III , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Quinase C/metabolismo , Ratos , Ratos Zucker , Transdução de Sinais/efeitos dos fármacos
9.
Diabetes ; 42(1): 80-9, 1993 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-7678404

RESUMO

Hyperglycemia has been implicated in the pathogenesis of both micro- and macrovascular complications in diabetes. Little is known, however, about glucose transporters and their regulation in the vascular system. In this study, the regulation of glucose transporters by glucose was examined in cultured BAECs and BSMCs, and in human arterial smooth muscle cells. Both BAECs and BSMCs transported glucose via the facilitated diffusion transport system. Glucose-transport activity in vascular smooth muscle cells was inversely and reversibly regulated by glucose. Exposure of BSMCs and HSMCs to high glucose decreased Vmax for 2DG and 3-O-MG uptake, whereas Km remained unchanged. The hexose-transport system of BAECs exhibited lower 2DG and 3-O-MG uptake compared with BSMCs and showed little or no adaptation to changes in ambient glucose. Northern blot analysis demonstrated that GLUT1 mRNA levels in BAECs and BSMCs were unaffected by the concentration of glucose in the medium. GLUT2-5 mRNA could not be detected by Northern blot analysis. GLUT1 protein, quantified by Western blot analysis, was more abundant in BSMCs than in BAECs and was decreased by approximately 50% when medium glucose was elevated from 1.2 to 22 mM for 24 h. The alterations in the level of GLUT1 protein correlated with the changes observed in transport activity. These observations suggest differential regulation of glucose transporter in response to glucose between smooth muscle and endothelial cells. The sites of autoregulation may involve translational control and/or the stability of the protein in the smooth muscle cells.(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Desoxiglucose/metabolismo , Endotélio Vascular/metabolismo , Glucose/farmacologia , Metilglucosídeos/metabolismo , Proteínas de Transporte de Monossacarídeos/metabolismo , Músculo Liso Vascular/metabolismo , 3-O-Metilglucose , Animais , Aorta Abdominal/efeitos dos fármacos , Aorta Abdominal/metabolismo , Transporte Biológico/efeitos dos fármacos , Northern Blotting , Bovinos , Membrana Celular/metabolismo , Células Cultivadas , Endotélio Vascular/efeitos dos fármacos , Cinética , Proteínas de Transporte de Monossacarídeos/biossíntese , Monossacarídeos/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , RNA/genética , RNA/isolamento & purificação , RNA Mensageiro/metabolismo , Trítio
10.
Diabetes ; 48(5): 1120-30, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10331419

RESUMO

Diminished insulin action in the vasculature may contribute to the development of cardiovascular diseases in diabetes. We have studied insulin's effects on the phosphatidylinositol (PI) 3-kinase pathway in arterial smooth muscle cells (SMCs) and its inhibition by endothelin (ET)-1, a potent vasoactive hormone reported to be elevated in insulin resistance and other vascular diseases. ET-1 increased the level of serine phosphorylation of insulin receptor beta subunit but increased both tyrosine and serine phosphorylation of insulin receptor substrate (IRS)-2. Pretreatment of cells with ET-1 (10 nmol/l) inhibited insulin-stimulated PI 3-kinase activity associated with IRS-2 by 50-60% and inhibited the association of p85 subunit of PI 3-kinase to IRS-2. The inhibition of insulin-stimulated PI 3-kinase activity by ET-1 was prevented by BQ-123, a selective ET(A) receptor antagonist, but was not affected by pertussis toxin. Treatment of cells with phorbol 12-myristate 13-acetate, an activator of protein kinase C (PKC), reduced both insulin-stimulated PI 3-kinase activity by 57% and the association of IRS-2 to the p85 subunit of PI 3-kinase by 40%, whereas GF109203X, a specific inhibitor of PKC, partially prevented the inhibitory effect of ET-1 on insulin-induced PI 3-kinase activity. These results suggested that ET-1 could interfere with insulin signaling in SMCs by both PKC-dependent and -independent pathways.


Assuntos
Endotelina-1/farmacologia , Insulina/farmacologia , Músculo Liso Vascular/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Animais , Antagonistas dos Receptores de Endotelina , Inibidores Enzimáticos/farmacologia , Proteínas de Ligação ao GTP/fisiologia , Humanos , Proteínas Substratos do Receptor de Insulina , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Peptídeos Cíclicos/farmacologia , Toxina Pertussis , Inibidores de Fosfoinositídeo-3 Quinase , Fosfoproteínas/metabolismo , Fosfosserina/metabolismo , Proteína Quinase C/metabolismo , Ratos , Ratos Zucker , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Virulência de Bordetella/farmacologia
11.
Diabetes ; 50(2): 444-54, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11272159

RESUMO

Systemic hypertension exacerbates diabetic retinopathy and other coexisting ocular disorders through mechanisms that remain largely unknown. Increased vascular permeability and intraocular neovascularization characterize these conditions and are complications primarily mediated by vascular endothelial growth factor (VEGF). Because systemic hypertension increases vascular stretch, we evaluated the expression of VEGF, VEGF-R2 (kinase insert domain-containing receptor [KDR]), and VEGF-R1 (fms-like tyrosine kinase [Flt]) in bovine retinal endothelial cells (BRECs) undergoing clinically relevant cyclic stretch and in spontaneously hypertensive rat (SHR) retina. A single exposure to 20% symmetric static stretch increased KDR mRNA expression 3.9 +/- 1.1-fold after 3 h (P = 0.002), with a gradual return to baseline within 9 h. In contrast, BRECs exposed to cardiac-profile cyclic stretch at 60 cpm continuously accumulated KDR mRNA in a transcriptionally mediated, time-dependent and stretch-magnitude-dependent manner. Exposure to 9% cyclic stretch increased KDR mRNA expression 8.7 +/- 2.9-fold (P = 0.011) after 9 h and KDR protein concentration 1.8 +/- 0.3-fold (P = 0.005) after 12 h. Stretched-induced VEGF responses were similar. Scatchard binding analysis demonstrated a 180 +/- 40% (P = 0.032) increase in high-affinity VEGF receptor number with no change in affinity. Cyclic stretch increased basal thymidine uptake 60 +/- 10% (P < 0.001) and VEGF-stimulated thymidine uptake by 2.6 +/- 0.2-fold (P = 0.005). VEGF-NAb reduced cyclic stretch-induced thymidine uptake by 65%. Stretched-induced KDR expression was not inhibited by AT1 receptor blockade using candesartan. Hypertension increased retinal KDR expression 67 +/- 42% (P < 0.05) in SHR rats compared with normotensive WKY control animals. When hypertension was reduced using captopril or candesartan, retinal KDR expression returned to baseline levels. VEGF reacted similarly, but Flt expression did not change. These data suggest a novel molecular mechanism that would account for the exacerbation of diabetic retinopathy by concomitant hypertension, and may partially explain the principal clinical manifestations of hypertensive retinopathy itself. Furthermore, these data imply that anti-VEGF therapies may prove therapeutically effective for hypertensive retinopathy and/or ameliorating the deleterious effects of coexistent hypertension on VEGF-associated disorders such as diabetic retinopathy.


Assuntos
Hipertensão/fisiopatologia , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Vasos Retinianos/metabolismo , Animais , Anti-Hipertensivos/farmacologia , Benzimidazóis/farmacologia , Compostos de Bifenilo , Pressão Sanguínea/efeitos dos fármacos , Captopril/farmacologia , Bovinos , Células Cultivadas , Angiopatias Diabéticas/complicações , Angiopatias Diabéticas/fisiopatologia , Retinopatia Diabética/complicações , Retinopatia Diabética/fisiopatologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Hipertensão/complicações , Hipertensão/tratamento farmacológico , Hipertensão/patologia , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Receptores Proteína Tirosina Quinases/genética , Receptores de Fatores de Crescimento/genética , Receptores de Fatores de Crescimento do Endotélio Vascular , Vasos Retinianos/patologia , Estresse Mecânico , Tetrazóis/farmacologia
12.
Arterioscler Thromb Vasc Biol ; 20(10): 2297-302, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11031218

RESUMO

Although the renin-angiotensin system has been implicated in increasing plasminogen activator inhibitor-1 (PAI-1) expression, the role of the angiotensin type 1 (AT(1)) receptor is controversial. This report examines the effects of angiotensin peptides, angiotensin-converting enzyme inhibition, and AT(1) antagonism on rat aortic and cardiac PAI-1 gene expression. In vitro, angiotensin (Ang) I, Ang II, and angiotensin Arg(2)-Phe(8) (Ang III) were potent agonists of PAI-1 mRNA expression in rat aortic smooth muscle cells (RASMCs), and stimulation of PAI-1 by these peptides was blocked by the AT(1) antagonist candesartan. Angiotensin Val(3)-Phe(8) (Ang IV) and angiotensin Asp(1)-Pro(7) (Ang [1-7]) did not affect PAI-1 expression in RASMCs. In neonatal rat cardiomyocytes, Ang II increased PAI-1 mRNA expression by 4-fold (P<0.01), and this response was completely blocked by AT(1) receptor antagonism. Continuous intrajugular infusion of Ang II into Sprague-Dawley rats for 3 hours increased aortic and cardiac PAI-1 mRNA expression by 17- and 9 fold, respectively, and these Ang II responses were completely blocked by coinfusion with candesartan. Aortic and cardiac PAI-1 expressions were compared in spontaneously hypertensive rats and Wistar-Kyoto rats. PAI-1 expression in the aorta and heart from spontaneously hypertensive rats was 5.8-fold and 2-fold higher, respectively, than in control Wistar-Kyoto rats (P<0.05). Candesartan treatment for 1 week reduced aortic and cardiac PAI-1 expression in spontaneously hypertensive rats by 94% and 72%, respectively (P<0.05), but did not affect vascular PAI-1 levels in Wistar-Kyoto rats. These results demonstrate a role for the AT(1) receptor in mediating the effects of Ang II on aortic and cardiac PAI-1 gene expression.


Assuntos
Aorta/metabolismo , Miocárdio/metabolismo , Inibidor 1 de Ativador de Plasminogênio/biossíntese , Receptores de Angiotensina/metabolismo , Angiotensina II/farmacologia , Antagonistas de Receptores de Angiotensina , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Animais Recém-Nascidos , Anti-Hipertensivos/farmacologia , Aorta/efeitos dos fármacos , Benzimidazóis/farmacologia , Compostos de Bifenilo , Células Cultivadas , Expressão Gênica/efeitos dos fármacos , Ventrículos do Coração/efeitos dos fármacos , Infusões Intravenosas , Masculino , Músculo Liso Vascular/metabolismo , Inibidor 1 de Ativador de Plasminogênio/genética , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina , Receptor Tipo 2 de Angiotensina , Tetrazóis/farmacologia
13.
Diabetes Care ; 22(8): 1245-51, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10480765

RESUMO

OBJECTIVE: To determine the effectiveness of vitamin E treatment in normalizing retinal blood flow and renal function in patients with <10 years of type 1 diabetes. RESEARCH DESIGN AND METHODS: An 8-month randomized double-masked placebo-controlled crossover trial evaluated 36 type 1 diabetic and 9 nondiabetic subjects. Subjects were randomly assigned to either 1,800 IU vitamin E/day or placebo for 4 months and followed, after treatment crossover, for a further 4 months. Retinal blood flow was measured using video fluorescein angiography, and renal function was assessed using normalized creatinine clearance from timed urine collections. RESULTS: After vitamin E treatment, serum levels of vitamin E were significantly elevated (P<0.01) in both type 1 diabetic and control patients. Hemoglobin A1c was not affected by vitamin E treatment. Diabetic patient baseline retinal blood flow (29.1+/-7.5 pixel2/s) was significantly (P = 0.030) decreased compared with that of nondiabetic subjects (35.2+/-7.2 pixel2/s). After vitamin E treatment, diabetic patient retinal blood flow (34.5+/-7.8 pixel2/s) was significantly increased (P<0.001) and was comparable with that of nondiabetic subjects. Additionally, vitamin E treatment significantly (P = 0.039) normalized elevated baseline creatinine clearance in diabetic patients. CONCLUSIONS: Oral vitamin E treatment appears to be effective in normalizing retinal hemodynamic abnormalities and improving renal function in type 1 diabetic patients of short disease duration without inducing a significant change in glycemic control. This suggests that vitamin E supplementation may provide an additional benefit in reducing the risks for developing diabetic retinopathy or nephropathy.


Assuntos
Creatinina/metabolismo , Diabetes Mellitus Tipo 1/tratamento farmacológico , Suplementos Nutricionais , Vasos Retinianos/efeitos dos fármacos , Vitamina E/uso terapêutico , Adolescente , Adulto , Análise de Variância , Estudos Cross-Over , Diabetes Mellitus Tipo 1/metabolismo , Retinopatia Diabética/tratamento farmacológico , Retinopatia Diabética/metabolismo , Relação Dose-Resposta a Droga , Método Duplo-Cego , Feminino , Humanos , Masculino , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Inibidor 1 de Ativador de Plasminogênio/sangue , Fluxo Sanguíneo Regional/efeitos dos fármacos , Resultado do Tratamento , Vitamina E/efeitos adversos
14.
Exp Clin Endocrinol Diabetes ; 107(2): 133-9, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10320054

RESUMO

Insulin resistance and hypertension commonly occur together. Pharmacological inhibition of the renin-angiotensin system has been found to reduce not only hypertension, but also insulin resistance. This raises the possibility that the renin-angiotensin system may interact with insulin signalling. We have investigated the relationship between insulin and angiotensin II (AII) intracellular signalling in vivo using an intact rat heart model, and in vitro using rat aorta smooth muscle cells (RASMC). Results generated in the in vivo studies indicate that, like insulin, AII stimulates tyrosine phosphorylation of the insulin receptor substrates IRS-1 and IRS-2. This leads to binding of IRS-1 and IRS-2 to PI3-kinase. However, in contrast to the effect of insulin. IRS-1- and IRS-2-associated PI3-kinase activity is inhibited by AII in a dose-dependent manner. Moreover, AII inhibits insulin-stimulated IRS-1/IRS-2-associated PI3-kinase activity. The in vivo effects of AII are mediated via the AT1 receptor. The results of the in vitro studies indicate that AII inhibits insulin-stimulated, IRS-1-associated PI3-kinase activity by interfering with the docking of IRS-1 with the p85 regulatory subunit of PI3-kinase. It appears that AII achieves this effect by stimulating serine phosphorylation of the insulin receptor beta-subunit IRS-1, and the p85 regulatory subunit of PI3-kinase. These actions result in the inhibition of normal interactions between the insulin signalling pathway components. Thus, we believe that AII negatively modulates insulin signalling by stimulating multiple serine phosphorylation events in the early components of the insulin signalling cascade. Overactivity of the renin-angiotensin system is likely to impair insulin signalling and contribute to insulin resistance observed in essential hypertension.


Assuntos
Angiotensina II/fisiologia , Insulina/fisiologia , Receptor Cross-Talk , Transdução de Sinais , Animais , Humanos , Proteínas Substratos do Receptor de Insulina , Fosfoproteínas/fisiologia , Fosfosserina/metabolismo , Receptor de Insulina/fisiologia
15.
Heart Fail Monit ; 1(3): 74-82, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-12634871

RESUMO

Diabetes mellitus is associated with an increased risk of cardiovascular disease (CVD), even in the presence of intensive glycemic control. Substantial clinical and experimental evidence suggests that both diabetes and insulin resistance cause a combination of endothelial dysfunctions, which may diminish the anti-atherogenic role of the vascular endothelium. Endothelial dysfunctions that have been described include decreased endothelium-dependent vasorelaxation, increased leukocyte-endothelial cell adhesion and vascular permeability, and the altered production of a variety of vasoactive substances, which affect coagulation, extracellular matrix homeostasis, and smooth muscle physiology. The primary mechanisms that contribute to these endothelial dysfunctions in diabetes appear to involve the activation of protein kinase C (PKC) pathways, increased non-enzymatic glycation, increased oxidant stress, and reduced endothelial insulin action. In addition, many of the adverse effects of these abnormalities associated with hyperglycemia and insulin resistance are mediated and amplified by potent vasoactive hormones including angiotensin II, transforming growth factor-beta, and vascular endothelial growth factor. Multiple interventions have been shown to improve endothelial dysfunction in diabetes, including PKC inhibition, infusion of soluble receptors for advanced glycation end-products, antioxidant and insulin supplementation, and angiotensin-converting enzyme inhibition. These findings are consistent with a model involving a combination of factors contributing to the etiology of the endothelial dysfunctions in diabetes. Further work is needed to determine whether endothelial function can be used as a therapeutic target to reduce CVD and improve clinical outcomes.


Assuntos
Diabetes Mellitus/fisiopatologia , Endotélio Vascular/fisiopatologia , Animais , Permeabilidade Capilar/fisiologia , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/fisiopatologia , Diabetes Mellitus/epidemiologia , Humanos , Fatores de Risco , Vasodilatação/fisiologia
16.
Thromb Haemost ; 110(3): 434-41, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23676986

RESUMO

Plasma kallikrein (PK) is a serine protease generated from plasma prekallikrein, an abundant circulating zymogen expressed by the Klkb1 gene. The physiological actions of PK have been primarily attributed to its production of bradykinin and activation of coagulation factor XII, which promotes inflammation and the intrinsic coagulation pathway. Recent genetic, molecular, and pharmacological studies of PK have provided further insight into its role in physiology and disease. Genetic analyses have revealed common Klkb1 variants that are association with blood metabolite levels, hypertension, and coagulation. Characterisation of animal models with Klkb1 deficiency and PK inhibition have demonstrated effects on inflammation, vascular function, blood pressure regulation, thrombosis, haemostasis, and metabolism. These reports have also identified a host of PK substrates and interactions, which suggest an expanded physiological role for this protease beyond the bradykinin system and coagulation. The review summarises the mechanisms that contribute to PK activation and its emerging role in diabetes and metabolism.


Assuntos
Diabetes Mellitus/sangue , Regulação da Expressão Gênica , Calicreína Plasmática/fisiologia , Adipogenia , Animais , Coagulação Sanguínea , Pressão Sanguínea , Bradicinina/metabolismo , Fator XII/metabolismo , Variação Genética , Glucose/metabolismo , Hemostasia , Humanos , Hipertensão/metabolismo , Inflamação , Camundongos , Pré-Calicreína/metabolismo , Trombose/metabolismo
17.
Arterioscler Thromb Vasc Biol ; 18(11): 1771-9, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9812917

RESUMO

Increased expression of plasminogen activator inhibitor-1 (PAI-1) has been reported in atherosclerotic and balloon-injured vessels. Little is known regarding the factors and mechanisms that may negatively regulate PAI-1 expression. In this report, the effect of cGMP-coupled vasoactive hormones, including natriuretic factors and nitric oxide, on the regulation of PAI-1 expression in vascular smooth muscle cells was examined. Atrial natriuretic factor 1-28 (ANF) and C-type natriuretic factor-22 (CNP) reduced angiotensin II (Ang II)- and platelet-derived growth factor-stimulated PAI-1 mRNA expression in rat aortic smooth muscle cells by 50% to 70%, with corresponding reductions in PAI-1 protein release. Treatment of human aortic smooth muscle cells with CNP similarly inhibited both platelet-derived growth factor-induced PAI-1 mRNA expression and PAI-1 protein release by 50%. Dose-response studies revealed that the inhibitory effects of CNP and ANF on PAI-1 expression were concentration dependent, with IC50s of approximately 1 nmol/L for both natriuretic peptides. Ang II-stimulated PAI-1 expression was also inhibited by the nitric oxide donor S-nitroso-N-acetylpenicillamine. The membrane-permeant cGMP analogue 8-Br-cGMP reduced Ang II-stimulated PAI-1 expression by 60%, and an inhibitor of soluble guanylyl cyclase (1H-[1,2,4]oxadiazolo[4, 3-a]quinoxalin-1-one) significantly impaired the inhibitory effects of S-nitroso-N-acetylpenicillamine on Ang II-stimulated PAI-1 expression. Studies of PAI-1 mRNA stability in cells treated with actinomycin D showed that ANF did not alter PAI-1 mRNA half-life, suggesting that natriuretic factors reduce PAI-1 transcription. These data show that natriuretic factors and nitric oxide, via a cGMP-dependent mechanism, inhibit PAI-1 synthesis in vascular smooth muscle cells. Thus, cGMP-coupled vasoactive hormones may play an important role in suppressing vascular PAI-1 expression.


Assuntos
Fator Natriurético Atrial/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Peptídeo Natriurético Tipo C/farmacologia , Doadores de Óxido Nítrico/farmacologia , Inibidor 1 de Ativador de Plasminogênio/genética , Animais , Células Cultivadas , Depressão Química , Meia-Vida , Humanos , Músculo Liso Vascular/citologia , Penicilamina/análogos & derivados , Penicilamina/farmacologia , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley
18.
J Biol Chem ; 265(31): 18780-5, 1990 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-2229041

RESUMO

Whereas there is biological evidence that the reductive cleavage of disulfide bonds is critical for the activation of endocytosed macromolecules such as toxins, immunotoxins, and other drug carriers, virtually nothing is known about the specifics of this cleavage. To study this process, a model compound was synthesized in which a radioiodinated tyramine was linked through a disulfide bond to an undegradable carrier, poly(D-lysine), known to be efficiently endocytosed. Cultured Chinese hamster ovary cells were pulse-labeled with this probe, and the disulfide cleavage was measured as released acid-soluble radioactivity at different times of chase. Pulse-labeled cells were also subjected to subcellular fractionation to identify intracellular structures associated with disulfide cleavage. Cleavage began without lag, amounted to about approximately 7% of the initial cell-bound radioactivity in the first hour and continued for more than 6 h. It was abolished in the presence of N-ethylmaleimide. When sulfhydryl groups present at the cell surface were blocked with cell-impermeant sulfhydryl reagent, the initial phase of disulfide cleavage was inhibited, indicating that cleavage began at the cell surface. A long-lasting intracellular phase of disulfide cleavage began after about approximately 30 min of chase. Subcellular fractionation and kinetic analysis indicated that neither lysosomes nor endosomes were participating in that phase, leaving the Golgi apparatus as the most probable site of endocytic disulfide cleavage.


Assuntos
Dissulfetos/metabolismo , Endocitose , Lisossomos/metabolismo , Metotrexato/análogos & derivados , Organelas/metabolismo , Polilisina/análogos & derivados , Tiramina/análogos & derivados , Animais , Linhagem Celular , Cistamina/farmacologia , Hidrólise , Cinética , Metotrexato/metabolismo , Polilisina/metabolismo , Fatores de Tempo , Tiramina/metabolismo
19.
J Biol Chem ; 267(20): 13811-4, 1992 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-1321126

RESUMO

A number of protein-tyrosine phosphatase(s) (PTPases) have been shown to dephosphorylate the insulin receptor in vitro; however, it is not known whether any individual PTPase has specificity for certain phosphotyrosine residues of the receptor that regulate its intrinsic tyrosine kinase activity. We evaluated the deactivation of the insulin receptor kinase by three candidate enzymes that are expressed in insulin-sensitive rat tissues, including the receptor-like PTPases LAR and LRP, and the intracellular enzyme, PTPase1B. Purified insulin receptors were activated by insulin and receptor dephosphorylation, and kinase activity was quantitated after incubation with recombinant PTPases from an Escherichia coli expression system. When related to the level of overall receptor dephosphorylation, LAR deactivated the receptor kinase 3.1 and 2.1 times more rapidly than either PTPase1B or LRP, respectively (p less than 0.03). To assess whether these effects were associated with preferential dephosphorylation of the regulatory (Tyr-1150) domain of the receptor beta-subunit, we performed tryptic mapping of the insulin receptor beta-subunit after dephosphorylation by PTPases. Relative to the rate of initial loss of 32P from receptor C-terminal sites, LAR dephosphorylated the Tris-phosphorylated Tyr-1150 domain 3.5 and 3.7 times more rapidly than either PTPase1B or LRP, respectively (p less than 0.01). The accelerated deactivation of the insulin receptor kinase by LAR and its relative preference for regulatory phosphotyrosine residues further support a potential role for this transmembrane PTPase in the physiological regulation of insulin receptors in intact cells.


Assuntos
Glicoproteínas/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Receptor de Insulina/metabolismo , Receptores de Superfície Celular , Animais , Clonagem Molecular , Escherichia coli/genética , Cinética , Proteínas Tirosina Fosfatases/genética , Ratos , Proteínas Tirosina Fosfatases Classe 4 Semelhantes a Receptores , Proteínas Recombinantes/metabolismo
20.
Am J Physiol ; 267(3 Pt 1): E369-79, 1994 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-7943217

RESUMO

We have reported that membranous protein kinase C (PKC) activities and total diacylglycerol (DAG) levels are increased in the heart and aorta of diabetic rats, which cannot be easily reversed by euglycemic control. However, insulin treatment, which achieved euglycemia, can prevent the increase in PKC activities and DAG levels. Chronic exposure to elevated glucose levels (5.5 vs. 22 mM) increased DAG levels in cultured bovine and rat aortic endothelial cells and smooth muscle cells by 31, 140, and 143%, respectively, only after 3 days of incubation. Glyceraldehyde, which can stimulate the de novo synthesis of DAG, significantly increased DAG levels by 7.1 +/- 0.6-fold after only 16 h of incubation. Elevated glucose levels did not affect labeled DAG when all of the vascular cells were incubated with [3H]arachidonate, [3H]glycerol, or [3H]phosphatidylcholine, whereas [3H]palmitate- and [3H]oleic acid-labeled DAG levels were significantly increased, indicating that the glucose-stimulated increase in DAG is derived partially from the de novo synthesis pathway. Immunoblotting studies showed increases only in PKC isoform beta II but not alpha in aortic smooth muscle cells. The phosphorylation level of MARCKS protein, an intracellular substrate of PKC, was also increased, consistent with the PKC activity increase. These findings showed that diabetic and hyperglycemia-induced increases in PKC activity and DAG levels in the heart and aorta are preventable by insulin treatment.


Assuntos
Aorta/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diglicerídeos/metabolismo , Glucose/farmacologia , Insulina/farmacologia , Miocárdio/metabolismo , Proteína Quinase C/metabolismo , Animais , Bovinos , Células Cultivadas , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA