Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Colloids Surf B Biointerfaces ; 228: 113402, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37331193

RESUMO

Nanocarriers are utilized to deliver bioactive substances in the treatment of neurodegenerative diseases such as Alzheimer's. In this work, we prepared donepezil hydrochloride-loaded molybdenum disulfide modified thermo-responsive polymer as the thermo-responsive nanocarrier. Then, glycine was grafted to the surface of the polymer to improve the targeting and sustained release. The morphology, crystallinity, chemical bonding, and thermal behavior of nanoadsorbent were fully characterized by field emission scanning electron microscopes, energy dispersive X-ray, X-ray diffraction, Fourier-transform infrared spectroscopy, and thermo-gravimetric measurement. Response surface methodology with the central composite design was applied to optimize the sorption key factors such as pH solution (A: 5-9), contact time (B: 10-30 min), and temperature (C: 30-50 °C). Non-linear isotherm modeling confirmed that the sorption of the drug follows the Ferundlich model based on higher correlation coefficient values (R2 =0.9923) and lower errors values (root means square errors: 0.16 and Chi-square: 0.10), suggesting a heterogeneous multilayer surface sorption. The non-linear sorption kinetic modeling revealed that the pseudo-second-order kinetic model well-fitted the sorption data of the drug on the nanoadsorbent surface based on higher R2 values (R2 =0.9876) and lower errors values (root means square errors: 0.05 and Chi-square: 0.02). The in vitro drug release experiment of donepezil hydrochloride shown that about 99.74 % of drug release was found to be occurred at pH= 7.4 (T = 45 °C) within 6 h, whereas about 66.32 % of drug release occurred at pH= 7.4 (T = 37 °C). The release of donepezil hydrochloride from as prepared drug delivery system has shown a sustained release profile, which was fitted to Korsmeyer-Peppas kinetics.


Assuntos
Sistemas de Liberação de Medicamentos , Polímeros , Polímeros/química , Donepezila , Preparações de Ação Retardada , Concentração de Íons de Hidrogênio , Espectroscopia de Infravermelho com Transformada de Fourier
2.
Int J Mol Sci ; 21(21)2020 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-33147762

RESUMO

Genetic mutations and aberrant epigenetic alterations are the triggers for carcinogenesis. The emergence of the drugs targeting epigenetic aberrations has provided a better outlook for cancer treatment. Histone deacetylases (HDACs) are epigenetic modifiers playing critical roles in numerous key biological functions. Inappropriate expression of HDACs and dysregulation of PI3K signaling pathway are common aberrations observed in human diseases, particularly in cancers. Histone deacetylase inhibitors (HDACIs) are a class of epigenetic small-molecular therapeutics exhibiting promising applications in the treatment of hematological and solid malignancies, and in non-neoplastic diseases. Although HDACIs as single agents exhibit synergy by inhibiting HDAC and the PI3K pathway, resistance to HDACIs is frequently encountered due to activation of compensatory survival pathway. Targeted simultaneous inhibition of both HDACs and PI3Ks with their respective inhibitors in combination displayed synergistic therapeutic efficacy and encouraged the development of a single HDAC-PI3K hybrid molecule via polypharmacology strategy. This review provides an overview of HDACs and the evolution of HDACs-based epigenetic therapeutic approaches targeting the PI3K pathway.


Assuntos
Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinogênese/genética , Epigênese Genética , Humanos , Camundongos , Morfolinas/farmacologia , Polifarmacologia , Pirimidinas/farmacologia
3.
Int J Mol Sci ; 20(12)2019 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-31197106

RESUMO

Vascular remodeling is a characteristic feature of cardiovascular diseases. Altered cellular processes of vascular smooth muscle cells (VSMCs) is a crucial component in vascular remodeling. Histone deacetylase inhibitor (HDACI), butyrate, arrests VSMC proliferation and promotes cell growth. The objective of the study is to determine the mechanism of butyrate-induced VSMC growth. Using proliferating VSMCs exposed to 5 mM butyrate, immunoblotting studies are performed to determine whether PI3K/Akt pathway that regulates different cellular effects is a target of butyrate-induced VSMC growth. Butyrate inhibits phosphorylation-dependent activation of PI3K, PDK1, and Akt, eliciting differential effects on downstream targets of Akt. Along with previously reported Ser9 phosphorylation-mediated GSK3 inactivation leading to stability, increased expression and accumulation of cyclin D1, and epigenetic histone modifications, inactivation of Akt by butyrate results in: transcriptional activation of FOXO1 and FOXO3 promoting G1 arrest through p21Cip1/Waf1 and p15INK4B upregulation; inactivation of mTOR inhibiting activation of its targets p70S6K and 4E-BP1 impeding protein synthesis; inhibition of caspase 3 cleavage and downregulation of PARP preventing apoptosis. Our findings imply butyrate abrogates Akt activation, causing differential effects on Akt targets promoting convergence of cross-talk between their complimentary actions leading to VSMC growth by arresting proliferation and inhibiting apoptosis through its effect on dual targets, HDAC activity and PI3K/Akt pathway network.


Assuntos
Butiratos/farmacologia , Histona Desacetilases/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Animais , Apoptose , Proliferação de Células , Células Cultivadas , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos
4.
Biochem Cell Biol ; 95(4): 482-490, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28376311

RESUMO

Oxidative stress and inflammation are major contributors to acrolein toxicity. Peroxisome proliferator activated receptor gamma (PPARγ) has antioxidant and anti-inflammatory effects. We investigated the contribution of PPARγ ligand GW1929 to the attenuation of oxidative stress in acrolein-induced insult. Male gp91phox knock-out (KO) mice were treated with acrolein (0.5 mg·(kg body mass)-1 by intraperitoneal injection for 7 days) with or without GW1929 (GW; 0.5 mg·(kg body mass)-1·day-1, orally, for 10 days). The livers were processed for further analyses. Acrolein significantly increased 8-isoprostane and reduced PPARγ activity (P < 0.05) in the wild type (WT) and KO mice. GW1929 reduced 8-isoprostane (by 32% and 40% in WT and KO mice, respectively) and increased PPARγ activity (by 81% and 92% in WT and KO, respectively). Chemokine activity was increased (by 63%) in acrolein-treated WT mice, and was reduced by GW1929 (by 65%). KO mice exhibited higher xanthine oxidase (XO). Acrolein increased XO and COX in WT mice and XO in KO mice. GW1929 significantly reduced COX in WT and KO mice and reduced XO in KO mice. Acrolein significantly reduced the total antioxidant status in WT and KO mice (P < 0.05), which was improved by GW1929 (by 75% and 74%). The levels of NF-κB were higher in acrolein-treated WT mice. GW1929 reduced NF-κB levels (by 51%) in KO mice. Acrolein increased CD36 in KO mice (by 43%), which was blunted with GW1929. Data confirms that the generation of free radicals by acrolein is mainly through NAD(P)H, but other oxygenates play a role too. GW1929 may alleviate the toxicity of acrolein by attenuating NF-κB, COX, and CD36.


Assuntos
Acroleína/farmacologia , Inflamação/tratamento farmacológico , PPAR gama/metabolismo , Receptores Imunológicos/deficiência , Transdução de Sinais/efeitos dos fármacos , Animais , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo
5.
Acta Pharmacol Sin ; 35(4): 476-82, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24562305

RESUMO

AIM: Peroxisome proliferator activated receptors (PPARs) are nuclear transcription factors that regulate numerous genes influencing blood pressure. The aim of this study was to examine the effects of clofibrate, a PPARα ligand, on blood pressure in spontaneously hypertensive rats (SHR). METHODS: Wistar-Kyoto (WKY) and spontaneously hypertensive rats (SHR), 8-9 weeks old, were randomly allocated into groups treated with vehicle or clofibrate (250 mg·kg(-1)·d(-1), ip for 21 d). Systolic blood pressure (SBP) was measured before and after the study period using tail-cuff plethysmography. Rats were sacrificed under anesthesia and blood, urine and tissue samples were processed for subsequent analysis. RESULTS: SHR rats showed significantly higher SBP compared with WKY rats (198±6 mmHg vs 93±7 mmHg), and a 3-fold increase in urinary protein excretion. Clofibrate treatment reduced SBP by 26%±2% and proteinuria by 43%±9% in SHR but not in WKY rats. The urinary nitrite/nitrate excretion in SHR rats was nearly 2-fold greater than that in WKY, and was further increased by 30%±4% and 48%±3%, respectively, following clofibrate treatment. In addition, PPARα protein expression and PPARα activity were significantly lower in SHR than that in WKY rats. Clofibrate treatment significantly increased PPARα protein expression and PPARα activity in SHR rats, but not in WKY rats. Moreover, the vasoconstrictor response of aortic ring was markedly increased in SHRs, which was blunted after clofibrate treatment. CONCLUSION: PPARα contributes to regulation of blood pressure and vascular reactivity in SHR, and clofibrate-mediated reduction in blood pressure and proteinuria is probably through increased NO production.


Assuntos
Anti-Hipertensivos/farmacologia , Aorta/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Clofibrato/farmacologia , Hipertensão/prevenção & controle , PPAR alfa/agonistas , Vasoconstrição/efeitos dos fármacos , Animais , Aorta/metabolismo , Aorta/fisiopatologia , Biomarcadores/urina , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Ligantes , Masculino , Óxido Nítrico/metabolismo , PPAR alfa/metabolismo , Proteinúria/metabolismo , Proteinúria/prevenção & controle , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Transdução de Sinais/efeitos dos fármacos , Vasoconstritores/farmacologia
6.
Ren Fail ; 36(4): 567-74, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24479849

RESUMO

Increased NAD(P)H oxidase-dependent free radical generation has been proposed to be a mechanism in glycerol-induced acute renal failure (ARF). Previously, we showed a PPARγ-mediated regulation of free radical generation in ARF. In this study, we examined NAD(P)H oxidase-dependent pathology in ARF and its connection with PPARγ using both Sprague-Dawley rats and gp91phox (+/-) mice. Male gp91phox (+/-) or wild type (+/+) mice were distributed into vehicle and ARF group (50% glycerol; 8 mL/kg bw; i.m.). Animals were placed in metabolic cages for 24 hr and were sacrificed under pentobarbital anesthesia. Urine, plasma and kidneys were processed for biochemical and molecular analysis. Glycerol doubled proteinuria in (+/+) mice (68 ± 4 mg/24 hr) but not in (+/-) mice (43 ± 9 mg/24 hr). This was associated with a markedly reduced creatinine excretion in (+/+) mice (Con: 0.6 ± 0.03 & ARF: 0.37 ± 0.02). Basal plasma and urinary NO was higher in (+/-) mice than the (+/+) type while plasma 8-isoprostane level was lower in (+/-) mice (WT: 165 ± 20; KO: 100 ± 15 pg/mL). Glycerol reduced UNOXV in both (+/+) and (+/-) mice although plasma NO was unchanged. Glycerol also doubled 8-isoprostane in (+/+) (363 ± 22 pg/mL) but not in (+/-) mice (152 ± 20 pg/mL) and this was associated with an increased NAD(P)H oxidase activity in the (+/+) mice. In ARF, PPARγ expression was reduced in (+/+) mice but increased in (+/-) mice. PPARγ activity was also reduced in (+/+) mice but was unchanged in (+/-) mice. We conclude that gp91phox contributes to NAD(P)H oxidase-mediated increased free radical generation in ARF and this may be via reduced PPARγ.


Assuntos
Injúria Renal Aguda/metabolismo , Glicoproteínas de Membrana/metabolismo , NADPH Oxidases/metabolismo , PPAR gama/metabolismo , Injúria Renal Aguda/induzido quimicamente , Animais , Dinoprosta/análogos & derivados , Dinoprosta/sangue , Radicais Livres/metabolismo , Glicerol , Masculino , Camundongos , Camundongos Knockout , NADPH Oxidase 2 , Óxido Nítrico/metabolismo , PPAR gama/genética , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Tiazolidinedionas/farmacologia , Transcrição Gênica
7.
J Health Care Poor Underserved ; 24(4 Suppl): 118-31, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24241266

RESUMO

An essential component of NAD(P)H, gp91phox, maintains the functionality of the enzyme in producing oxygen radicals. NAD(P)H oxidase plays an important role in oxidative stress but its precise contribution in acrolein-induced toxicity was not explored. We examined the involvement of NAD(P)H oxidase and other oxidant system in acrolein toxicity using gp91phox knockout mice. Male gp91phox knockout (KO) mice (20-25 gm) or wild type (WT) controls were treated with acrolein (0.5 µg/kg; 1 week). Animals were sacrificed and the liver was used to determine biochemical parameters. Knockout mice generated low (1.43 ±.02 pg/µg protein) free radicals as evident in 8-Isoprostane compared with the WT mice (2.19 ± 0.1). Acrolein increased 8-Isoprostane in WT (P<.05) and KO (p<.05) mice. Xanthine Oxidase (XO) activity was higher (p<.05) in KO (0.56 ± 0.06 µ unit/µg protein) than WT mice. Acrolein increased XO in KO mice, but significantly increased it only in WT. Cycloxygenase (COX) activity was not different between WT and KO mice, although acroelin increased COX in WT. Knockout mice exhibited a significantly low (2.1 ± 0.2 µmol/mg protein) total antioxidant status (TAS) compared with the WT (3.5 ± 0.3). Acrolein reduced TAS in both WT and KO mice equally. Baseline NFκB was significantly higher in KO mice, although acrolein increased NFκB in WT but not in KO. CD36 was higher (p<.05) in KO mice than the WT and acrolein increased (p<.05) CD36 further in KO but not in WT mice. These data suggest that NAD(P)H oxidase contributes significantly in acrolein-induced oxidative stress. We also suggests that in the absence of NAD(P)H oxidase XO plays a definitive role together with reduced antioxidant ability to compound the toxic effects of acrolein. We propose that in absence of NAD(P)H oxidase a different signaling process may involve that utilizes CD36 besides NFκB.


Assuntos
Acroleína/toxicidade , Antígenos CD36/metabolismo , Poluentes Ambientais/toxicidade , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Dinoprosta/análogos & derivados , Dinoprosta/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , NADPH Oxidases/genética , Xantina Oxidase/metabolismo
8.
J Basic Clin Physiol Pharmacol ; 24(4): 287-97, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23893683

RESUMO

BACKGROUND: Modulation of inflammatory signaling has been elucidated in several disease models. Acrolein, an environmental pollutant, has been linked to diseases such as atherosclerosis and to the inflammatory process involving nuclear factor κB (NFκB). Serum response factor (SRF), a transcription factor, regulates cell development, differentiation and proliferation through signaling molecules such as extracellular signal-regulated kinase 1/2 (ERK1/2) and CD36. We hypothesized that acrolein toxicity involves SRF in the process of activating NFκB and may involve CD36/ERK1/2. METHODS: Vascular smooth muscle cells (VSMCs) were exposed to acrolein (0.5 µg/mL) in the presence or absence of 10 nM QNZ (NFκB inhibitor), 300 nM CCG1423 (SRF inhibitor) and 50 µM PD98059 (ERK1/2 inhibitor). Protein and RNA were isolated. Changes in expression were determined by Western blot and polymerase chain reaction (PCR) array. RESULTS: Subtoxic doses of acrolein increased ERK1/2, SRF and NFκB protein expression, whereas CD36 expression was unchanged. Increase in NFκB expression was accompanied by an increase in activity. ERK1/2 inhibition only blunted SRF expression. SRF inhibition blunted NFκB expression but not that of ERK1/2. CD36 expression was unchanged in the presence of either inhibitor. PCR array analysis indicated up-regulation of nine genes (>4- to 50-fold) and down-regulation of six genes (>4- to 12-fold) involved in inflammatory signaling. CONCLUSIONS: We propose that SRF is required in acrolein activation of NFκB and is ERK1/2 dependent.


Assuntos
Acroleína/toxicidade , Poluentes Ambientais/toxicidade , Inflamação/induzido quimicamente , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Fator de Resposta Sérica/imunologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Inflamação/genética , Inflamação/imunologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Músculo Liso Vascular/imunologia , Miócitos de Músculo Liso/imunologia , NF-kappa B/genética , NF-kappa B/imunologia , Ratos , Fator de Resposta Sérica/antagonistas & inibidores , Fator de Resposta Sérica/genética
9.
Ren Fail ; 32(3): 384-90, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20370457

RESUMO

Neutral endopeptidase (NEP: EC 3.4.24.11) is involved in the degradation of peptides such as atrial natriuretic peptide, angiotensin II (AngII), and endothelin-1 (ET-1). In this study we propose that NEP inhibition provides protection in glycerol-induced acute renal failure (ARF). Renal vascular responses were evaluated in ARF rats where ARF was induced by injecting 50% glycerol in candoxatril, a NEP inhibitor (30 mg/kg, orally; for 3 weeks) pretreated rats. AngII and U46619 (a TxA2 mimetic) vasoconstriction was increased (2- to 4-fold) in ARF while ET-1 vasoconstriction was surprisingly reduced (23+/-3%; p<0.05). In ARF, candoxatril paradoxically enhanced ET-1 response (60+/-20%; p<0.05) but reduced AngII vasoconstriction (51+/-11%; p<0.05) without affecting U46619 response. However, candoxatril treatment was without effect on plasma ET-1 and TxB2 levels in ARF. Candoxatril reduced plasma AngII by 34+/-4% (p<0.05) in ARF which was approximately 3.5-fold higher compared to control. Candoxatril doubled the nitrite excretion in control but was without effect on proteinuria or nitrite excretion in ARF. Candoxatril enhanced Na+ and creatinine excretion in ARF by 73+/-9% and 33+/-2%, respectively. These results suggest that NEP inhibition may confer protection in glycerol-induced ARF by stimulating renal function but without a consistent effect on renal production and renal vascular responses to endogenous vasoconstrictors.


Assuntos
Injúria Renal Aguda/fisiopatologia , Rim/irrigação sanguínea , Neprilisina/antagonistas & inibidores , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Injúria Renal Aguda/metabolismo , Administração Oral , Angiotensina II/metabolismo , Angiotensina II/farmacologia , Animais , Anti-Hipertensivos/farmacologia , Creatinina/sangue , Endotelina-1/metabolismo , Endotelina-1/farmacologia , Indanos/farmacologia , Masculino , Natriurese/efeitos dos fármacos , Neprilisina/farmacologia , Nitratos/urina , Propionatos/farmacologia , Proteinúria , Ratos , Ratos Sprague-Dawley , Tromboxano A2/metabolismo , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia
10.
Pharmacol Ther ; 125(3): 436-45, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20117134

RESUMO

Oxidative stress has been reported to play a critical role in the pathology of acute renal failure (ARF). An interaction between different reactive species and/or their sources have been the focus of extensive studies. The exact sources of reactive species generated in biological systems under different disease states are always elusive because they are also a part of physiological processes. Exaggerated involvement of different oxidation pathways including NAD(P)H oxidase has been proposed in different models of ARF. An interaction between oxygen species and nitrogen species has drawn extensive attention because of the deleterious effects of peroxynitrite and their possible effects on antioxidant systems. Recent advances in molecular biology have allowed us to understand glomerular function more precisely, especially the organization and importance of the slit diaphragm. Identification of slit diaphragm proteins came as a breakthrough and a possibility of therapeutic manipulation in ARF is encouraging. Transcriptional regulation of the expression of slit diaphragm protein is of particular importance because their presence is crucial in the maintenance of glomerular function. This review highlights the involvement of oxidative stress in ARF, sources of these reactive species, a possible interaction between different reactive species, and involvement of PPARgamma, a nuclear transcription factor in this process.


Assuntos
Injúria Renal Aguda/metabolismo , Óxido Nítrico/metabolismo , Estresse Oxidativo/genética , PPAR gama/metabolismo , Injúria Renal Aguda/genética , Animais , Radicais Livres/metabolismo , Regulação da Expressão Gênica , Membrana Basal Glomerular/metabolismo , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Modelos Biológicos , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo
11.
Am J Nephrol ; 30(3): 201-8, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19420904

RESUMO

BACKGROUND: Peroxisome proliferator-activated receptor gamma (PPARgamma), a nuclear transcription factor, modulates the expression/activity of G protein-coupled receptors (GPCRs), but its role in GPCR signaling is not clear. Increased GPCR kinase-2 (GRK-2) activity and receptor desensitization have been reported in hypertension. METHOD: In this study we investigated the role of GRK-2 in PPARgamma-mediated blood pressure regulation in hypertension. SHR or WKY rats were treated with GW1929, a selective PPARgamma ligand (0.5 mg/kg/day), or vehicle for 2 months. Systolic blood pressure (tail cuff plethysmography), whole kidney perfusion (laser scanner) and renal vascular reactivity (isolated perfused kidney) was determined. RESULTS: GW1929 significantly reduced blood pressure (20 +/- 1%) and increased renal perfusion (61 +/- 3%) in SHR compared to WKY rats. Vasoconstriction to phenylephrine (100 microg) in the isolated perfused kidney was greater in SHRs (29 +/- 1%) compared to WKY rats and this was abolished by GW1929. GW1929 enhanced acetylcholine-induced (30-300 microg) and sodium nitroprusside-induced vasodilatation in SHR by 46 +/- 2% (p < 0.05) and 33 +/- 2% (p < 0.05), respectively. Isoprenalin-induced (5-30 microg) vasodilatation was 43 +/- 2% lower in SHR compared to WKY and GW1929 enhanced this vasodilatation by 55 +/- 2%. In SHR kidney, GW1929 enhanced expression of PPARgamma mRNA (34 +/- 1%) but reduced that of GRK-2 (31 +/- 3%). CONCLUSION: We suggest that downregulation of PPARgamma but upregulation of GRK-2 increases blood pressure and impaired renal vascular reactivity in SHR and that PPARgamma-mediated improvement in hypertension may involve transcriptional regulation of GRK-2 function.


Assuntos
Pressão Sanguínea/fisiologia , Quinase 2 de Receptor Acoplado a Proteína G/fisiologia , Hipertensão/fisiopatologia , Rim/irrigação sanguínea , PPAR gama/fisiologia , Animais , Masculino , Ratos , Ratos Endogâmicos SHR
12.
FEBS J ; 274(22): 5962-78, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17961182

RESUMO

Vascular smooth muscle cell (VSMC) proliferation is an important etiological factor in vascular proliferative diseases such as primary atherosclerosis, hypertension, arterial and in-stent restenosis, and transplant vasculopathy. Our studies established that butyrate, a bacterial fermentation product of dietary fiber and a chromatin modulator, is a potent inhibitor of VSMC proliferation. The cardiovascular health benefits of a high-fiber diet, the principle source of butyrate in the body, have been known for a long time, however, very little is known about the antiatherogenic potential of butyrate. Because oxidative stress plays an important role in the pathogenesis of atherosclerosis, we examined involvement of the glutathione/glutathione S-transferase (GST) antioxidant system in butyrate's inhibition of VSMC proliferation. Treatment of proliferating VSMCs with butyrate leads to the induction of several GSTs. Interestingly, our study also demonstrated the nuclear localization of GST-P1 (GST-7-7), which is considered to be a cytosolic protein; this was demonstrated using immunostaining and was corroborated by western blotting. Also, the butyrate-induced antiproliferative action, and the induction of GST-P1 and its nuclear localization are downregulated when butyrate is withdrawn. Furthermore, assessment of intracellular glutathione levels reveals their augmentation by butyrate. Conversely, butyrate treatment reduces the levels of reactive oxygen species in VSMCs. Collectively, the butyrate-treatment-related increase in glutathione content, the reduction in reactive oxygen species, the upregulation of GST and the nuclear localization of GST-P1 in growth-arrested VSMCs imply that butyrate's antiproliferative action involves modulation of the cellular redox state. Thus, induction of the glutathione/GST antioxidant system appears to have other regulatory role(s) besides detoxification and regulation of the cellular redox state, for example, cell-cycle control and cell proliferation, which are both critical to atherogenesis.


Assuntos
Antioxidantes/metabolismo , Ácido Butírico/farmacologia , Proliferação de Células/efeitos dos fármacos , Glutationa Transferase/metabolismo , Glutationa/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Animais , Sequência de Bases , Primers do DNA , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo
13.
J Pharmacol Exp Ther ; 322(2): 461-8, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17494862

RESUMO

Peroxisome proliferator-activated receptor gamma (PPARgamma), a nuclear transcription factor, modulates vascular responses to angiotensin II (AII) or thromboxane A(2) (TxA(2)) via regulation of their gene/receptor. Increased vasoconstriction and deteriorating renal function in glycerol-induced acute renal failure (ARF) may be attributed to down-regulation of PPARgamma. In this study, we investigated the effect of ciglitazone (CG), a PPARgamma inducer, on AII and TxA(2) production and activity in glycerol-induced ARF. Vascular responses to AII or 9,11-dideoxy-11alpha,9alpha-epoxymethano prostaglandin F(2alpha) (U46619), a TxA(2) mimetic, were determined in preglomerular vessels following induction of ARF with glycerol. Renal damage and function were assessed in CG-treated (9 nmol/kg for 21 days) rats. PPARgamma protein expression and activity, which were significantly lower in ARF rats, were enhanced by CG (26 and 30%). CG also increased PPARgamma mRNA by 67 +/- 6%, which was reduced in ARF. In ARF, there was significant tubular necrosis and apoptosis, a 5-fold increase in proteinuria and a 2-fold enhancement in vasoconstriction to AII and U46619. CG reduced proteinuria (49 +/- 3%), enhanced Na(+) (124 +/- 35%) and creatinine excretion (92 +/- 25%), markedly diminished tubular necrosis, and reduced ARF-induced increase in AII (40 +/- 3%) and TxA(2) (39 +/- 2%) production, the attending increase in vasoconstriction to AII (36 +/- 2%) and U46619 (50 +/- 11%), and the increase in angiotensin receptor-1 (AT(1)) (23 +/- 3%) or thromboxane prostaglandin (TP) receptor (13 +/- 1%). CG reduced free radical generation by 55 +/- 14% while elevating nitrite excretion (65 +/- 13%). Our results suggest that enhanced activity of AII and TxA(2), increased AT(1) or TP receptor expression, and renal injury in glycerol-induced ARF are consequent to down-regulation of PPARgamma gene. CG ameliorated glycerol-induced effects through maintaining PPARgamma gene.


Assuntos
Injúria Renal Aguda/prevenção & controle , Angiotensina II/metabolismo , Glomérulos Renais/efeitos dos fármacos , Tiazolidinedionas/farmacologia , Tromboxano A2/metabolismo , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/metabolismo , Angiotensina II/sangue , Angiotensina II/farmacologia , Animais , Creatinina/urina , Dinoprosta/análogos & derivados , Dinoprosta/sangue , Expressão Gênica/efeitos dos fármacos , Glicerol/toxicidade , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Masculino , Óxido Nítrico/metabolismo , Nitritos/urina , PPAR gama/agonistas , PPAR gama/genética , PPAR gama/metabolismo , Proteinúria/prevenção & controle , Proteinúria/urina , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/metabolismo , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo , Artéria Renal/efeitos dos fármacos , Artéria Renal/fisiologia , Sódio/urina , Tiazolidinedionas/uso terapêutico , Tromboxano A2/agonistas , Tromboxano B2/urina , Vasoconstrição/efeitos dos fármacos
14.
J Cardiovasc Pharmacol ; 48(3): 88-94, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17031261

RESUMO

Vascular aging is characterized by endothelial dysfunction that is primarily attributed to increased superoxide production, the exact source of which remains ambiguous. This study compared the NAD(P)H and xanthine oxidase (XO) systems as sources of superoxide and impaired vascular function in aging. Male Sprague Dawley rats, 4-months-old (young) and 18-months-old (Aging), were used. Systolic blood pressure was higher (36 +/- 3%) in the aging group compared with young rats, and this was accompanied by reduced acetylcholine-induced renal vasodilatation. Urinary excretion of nitrite was lower in the aging rats (P < 0.05), and this was associated with reduced nitric oxide synthase (NOS) activity and reduced eNOS and iNOS protein expression in the aorta. Aged rats showed a n approximately twofold increase in free radical generation, as evident by increased plasma 8-isoprostane level, and an approximately fourfold increase in proteinuria compared with the young rats. Vascular NADP(H) oxidase was unchanged between both groups, as was the expression of p67phox or p47phox components of NAD(P)H oxidase. However, XO activity was increased (19 +/- 1%; P < 0.05) as well as XO expression in the aorta of aging rats. These results suggest that increased free radical generation-associated increase in SBP in aging rats is XO but not NAD(P)H oxidase-dependent.


Assuntos
Envelhecimento/fisiologia , Aorta/enzimologia , Estresse Oxidativo , Xantina Oxidase/metabolismo , Acetilcolina/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Radicais Livres/sangue , Masculino , NADPH Oxidases/metabolismo , Óxido Nítrico Sintase/metabolismo , Proteinúria/urina , Ratos , Ratos Sprague-Dawley
15.
Pharmacol Res ; 54(3): 234-40, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16820301

RESUMO

Peroxisome proliferator activated receptor-gamma (PPAR-gamma), a nuclear transcription factor, modulates angiotensin II (AII) or thromboxane A(2) (TxA(2)) response in the vasculature via transcriptional regulation of their gene or receptor expression. Increased AII or TxA(2) vasoconstriction and deteriorating renal function observed in glycerol-induced acute renal failure (ARF) may be attributed to a down-regulated PPAR-gamma expression/activity probably via an increased free radical generation. In this study, we investigated the effect of PPAR-gamma induction in glycerol-induced ARF by examining renal vascular reactivity to AII and TxA(2) and by renal expression/activity of PPAR-gamma. Vascular responses to AII or U46619, a TxA(2) mimetic were determined in rat isolated perfused kidney following induction of ARF with glycerol (50%, v/v, i.m.). Extent of renal damage and function were assessed with or without pre-treatment with ciglitazone (9 nmol kg(-1) x 21 days), a PPAR-gamma inducer. In ARF, vasoconstriction was enhanced to AII (three-fold; p<0.05) and U46619 (82%; p<0.05). Ciglitazone reduced AII and U46619 vasoconstriction by 59+/-1% (p<0.05) and 56+/-1% (p<0.05), respectively. Ciglitazone reduced proteinuria (38+/-3%) which was two-fold higher in ARF. Similarly, ciglitazone enhanced Na(+) excretion by 1.5 times while reducing BUN by 49+/-6%. On the contrary, ciglitazone did not change plasma creatinine which was significantly higher in ARF rats. Ciglitazone reduced free radical generation by 30+/-3% while elevating nitrite excretion approximately 2-fold. PPAR-gamma expression and activity were significantly lower in ARF rats and ciglitazone enhanced PPAR-gamma protein expression and activity by 45+/-3% and 52+/-4%, respectively. Data from this study suggest that reduced PPAR-gamma expression and activity may be involved in the pathology of glycerol-induced ARF and induction of PPAR-gamma by ciglitazone confers protection through reduced AII and TxA(2) vasoconstriction and/or enhanced renal function via reducing free radical generation.


Assuntos
Injúria Renal Aguda/metabolismo , PPAR gama/biossíntese , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/fisiopatologia , Angiotensina II/sangue , Animais , Ácido Ascórbico/farmacologia , Clofibrato/farmacologia , Regulação para Baixo , Radicais Livres/metabolismo , Glicerol , Rim/irrigação sanguínea , Rim/metabolismo , Masculino , Óxido Nítrico/metabolismo , Nitrilas/urina , Ratos , Ratos Sprague-Dawley , Circulação Renal/efeitos dos fármacos , Tiazolidinedionas/farmacologia , Tromboxano A2/sangue , Vasoconstrição/efeitos dos fármacos
16.
Mol Cell Biochem ; 254(1-2): 21-36, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14674679

RESUMO

Excessive proliferation of vascular smooth muscle cells (VSMCs) is a critical element in the development of several vascular pathologies, particularly in atherosclerosis and in restenosis due to angioplasty. We have shown that butyrate, a powerful antiproliferative agent, a strong promoter of cell differentiation and an inducer of apoptosis inhibits VSMC proliferation at physiological concentrations with no cytotoxicity. In the present study, we have used cDNA array technology to unravel the molecular basis of the antiproliferative effect of butyrate on VSMCs. To assess the involvement of gene expression in butyrate-inhibited VSMC proliferation, proliferating VSMCs were exposed to 5 mmol/l butyrate 1 through 5 days after plating. Expression profiles of 1.176 genes representing different functional classes in untreated control and butyrate treated VSMCs were compared. A total of 111 genes exhibiting moderate (2.0-5.0 fold) to strong (> 5.0 fold) differential expression were identified. Analysis of these genes indicates that butyrate treatment mainly alters the expression of four different functional classes of genes, which include: 43 genes implicated in cell growth and differentiation, 13 genes related to stress response, 11 genes associated with vascular function and 8 genes normally present in neuronal cells. Examination of differentially expressed cell growth and differentiation related genes indicate that butyrate-inhibited VSMC proliferation appears to involve down-regulation of genes that encode several positive regulators of cell growth and up-regulation of some negative regulators of growth or differentiation inducers. Some of the down-regulated genes include proliferating cell nuclear antigen (PCNA), retinoblastoma susceptibility related protein p130 (pRb), cell division control protein 2 homolog (cdc2), cyclin B1, cell division control protein 20 homolog (p55cdc), high mobility group (HMG) 1 and 2 and several others. Whereas the up-regulated genes include cyclin D1, p21WAF1, p141NK4B/p15INK5B, Clusterin, inhibitor of DNA binding 1 (ID1) and others. On the other hand, butyrate-responsive stress-related genes include some of the members of heat shock protein (HSP), glutathione-s-transferase (GST), glutathione peroxidase (GSH-PXs) and cytochrome P450 (CYP) families. Additionally, several genes related to vascular and neuronal function are also responsive to butyrate treatment. Although involvement of genes that encode stress response, vascular and neuronal functional proteins in cell proliferation is not clear, cDNA expression array data appear to suggest that they may play a role in the regulation of cell proliferation. However, cDNA expression profiles indicate that butyrate-inhibited VSMC proliferation involves combined action of a proportionally large number of both positive and negative regulators of growth, which ultimately causes growth arrest of VSMCs. Furthermore, these butyrate-induced differential gene expression changes are not only consistent with the antiproliferative effect of butyrate but are also in agreement with the roles that these gene products play in cell proliferation.


Assuntos
Butiratos/farmacologia , Regulação da Expressão Gênica , Músculo Liso Vascular/metabolismo , Animais , Aorta/citologia , Northern Blotting , Ácido Butírico/metabolismo , Divisão Celular , Células Cultivadas , DNA/metabolismo , DNA Complementar/metabolismo , Regulação para Baixo , Glutationa Transferase/metabolismo , Neurônios/metabolismo , Hibridização de Ácido Nucleico , Análise de Sequência com Séries de Oligonucleotídeos , Ligação Proteica , RNA/metabolismo , RNA Mensageiro/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Regulação para Cima
17.
Clin Exp Hypertens ; 25(6): 335-47, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12940471

RESUMO

These studies examined the interactions of neutral endopeptidase (NEP), endothelin-1 (ET-1), and nitric oxide (NO) in deoxycorticosterone acetate (DOCA)-induced hypertension. Male Sprague-Dawley rats (n = 35) were uninephrectomized (UNx) or uninephrectomized and treated with DOCA (25 mg pellet implanted subcutaneously). Candoxatril (30 mg/kg day(-1)), a NEP inhibitor, was given orally for 3 weeks in UNx or DOCA rats. Sham nephrectomized rats (SHAM) served as controls. Except SHAM, all other groups received 1% NaCl in drinking water ad libitum. Measurements were taken of systolic blood pressure (SBP), left ventricle (LV), and aortic weight (AW), plasma ET-1, and urinary excretion of nitrite and Na+. Whole body vascular hypertrophy and morphometric analysis of histological sections of the heart were also determined. In DOCA rats, SBP increased from 113 +/- 5 to 170 +/- 5 mmHg without significant changes in body weight (BW). Candoxatril reduced the increase in SBP to 135 +/- 9 mmHg (P < 0.05), abolished the increased LV wall thickness (P < 0.05), and increased the reduced LV lumen diameter (P < 0.05) in DOCA-salt rats. Candoxatril also reduced plasma ET-1 by 88 +/- 9% and 89 +/- 17% (P < 0.05) in UNx and DOCA rats, respectively, and elicited increases in urinary excretion of nitrite. These effects were accompanied by a marked increase in urinary excretion of Na+ (U(Na)V) (P < 0.05) and a blunting of the proteinuria (32 +/- 5%; P < 0.05) in DOCA rats. We conclude that endopeptidase inhibition in DOCA-salt hypertension reduced the increase in blood pressure and the attendant tissue hypertrophy and renal injury. These effects suggest a correlation between endopeptidase-related reduction in ET-1 production and protection in DOCA-salt hypertension.


Assuntos
Hipertensão/enzimologia , Indanos/farmacologia , Neprilisina/antagonistas & inibidores , Propionatos/farmacologia , Animais , Anti-Hipertensivos/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Desoxicorticosterona , Endotelina-1/sangue , Hipertensão/induzido quimicamente , Hipertensão/metabolismo , Rim/efeitos dos fármacos , Masculino , Modelos Animais , Óxido Nítrico/metabolismo , Ratos , Ratos Sprague-Dawley , Sódio/metabolismo
18.
Mol Cell Biochem ; 240(1-2): 83-98, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12487375

RESUMO

Acrolein, a major component of cigarette smoke, an environmental pollutant and an endogenous lipid peroxidation product, has been implicated in the development of atherosclerosis. Although a link between vascular injury and acrolein has been indicated, the exact molecular mechanism of acrolein-induced toxicity to vasculature is unknown. In an effort to elucidate the molecular basis of acrolein-induced vascular toxicity, the possibility of the intracellular signaling system as one of the targets of acrolein-induced toxicity is investigated in the present study. Exposure of cultured rat vascular smooth muscle cells (VSMCs) to different doses of acrolein not only causes cytotoxicity but also alters cellular morphology in a concentration and time-dependent manner. VSMCs exhibit cytotoxicity to a narrow concentration range of 5-10 microg/ml and display no toxicity to 2 microg/ml acrolein even after 24 h of exposure. Furthermore, exposure to acrolein results in activation of members of the mitogen-activated protein kinase (MAPK) family and protein tyrosine kinases. The extracellular signal-regulated kinases 1 and 2 (ERK1/2), stress-activated protein kinases/c-jun NH2-terminal kinases (SAPK/JNK) and p38MAPK are effectively and transiently activated by acrolein in a concentration and time-dependent fashion. While all three MAPKs exhibit significant activation within 5 min of exposure to acrolein, maximum activation (ERK1/2 and p38MAPK) or close to maximum activation (SAPK/JNK) occurs on exposure to 5 microg/ml acrolein for 2 h. Acrolein-induced activation of MAPKs is further substantiated by the activation of transcription factors, c-jun and activator transcription factor-2 (ATF-2), by acrolein-activated SAPK/JNK and p38MAPK, respectively. Additionally several cellular proteins exhibit spectacular protein tyrosine phosphorylation, particularly in response to 2 and 5 microg/ml of acrolein. Interestingly, the acrolein-induced activation of MAPKs precedes acrolein-stimulated protein tyrosine phosphorylation, which occurs after 2 h of exposure to acrolein. However, the time course of maximum protein tyrosine phosphorylation profile corresponds to the peak activation profile of MAPKs. The activation of MAPKs and protein tyrosine phosphorylation by acrolein appears to be independent of acrolein-induced toxicity. VSMCs exposed to 2 microg/ml acrolein exhibit no toxicity but stimulates significant activation of MAPKs and protein tyrosine phosphorylation. Although acrolein-induced VSMC toxicity is not blocked by MAPK inhibitors, PD98059, an inhibitor of MAPK kinase and SB203580, an inhibitor of p38MAPK, eitheralone or in combination, each MAPK responds differently to the inhibitors. Most prominently, although SB203580, an inhibitor of both SAPK/JNK and p38MAPK, significantly inhibited acrolein-induced activation of p38MAPK, it also stimulated SAPK/JNK activation by acrolein alone and in combination with PD98059. These results provide the first evidence that the activation of both growth-regulated (ERK1/2) and stress-regulated (SAPK/JNK and p38MAPK) MAPKs as well as tyrosine kinases are involved in the mediation of acrolein-induced effects on VSMC, which may play a crucial role in vascular pathogenesis due to environmentally and endogenously produced acrolein.


Assuntos
Acroleína/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Aorta , Ativação Enzimática/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Estresse Oxidativo , Fosforilação/efeitos dos fármacos , Proteínas Quinases/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio , Proteínas Quinases p38 Ativadas por Mitógeno
19.
Blood Press ; 11(2): 110-5, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12035871

RESUMO

The present study was designed to evaluate the possible antioxidant effect of pergolide, a DA-2 receptor agonist, in deoxycorticosterone acetate (DOCA)-salt hypertension and its role in endogenous endothelin-1 (ET- 1) production and organ hypertrophy. Male Sprague-Dawley rats were uninephrectomized (UNx) or uninephrectomized, and received subcutaneous implants of DOCA and drank 1% sodium chloride (DOCA). DOCA rats were treated daily for 3 weeks with pergolide (1 mg/kg, i.p.) or vitamin C (1 mg/rat, orally). DOCA-salt treatment increased systolic blood pressure (SBP) in UNx rats by 45 +/- 2 mmHg from 117 +/- 5 to 162 +/- 10 mmHg (p < 0.05), an effect blunted by pergolide and vitamin C. Superoxide generation was not increased in DOCA rats; however, both pergolide and vitamin C significantly reduced superoxide generation by 49 +/- 7% and 52 +/- 13%, respectively (p < 0.05). Plasma ET-1 levels increased twofold in UNx rats but was reduced to 42 +/- 7% (p < 0.05) in DOCA compared to UNx rats. Pergolide and vitamin C reduced plasma ET-1 levels further by 43 +/-10% (p < 0.05) and 46 +/- 8% (p < 0.05), respectively. Pergolide increased urinary Na+ excretion but did not alter urinary protein excretion or the left ventricular and aortic hypertrophy in DOCA rats. These data suggest that the reduction of SBP by pergolide in DOCA-salt hypertension may be attributed to its natriuretic ability, not its ability to reduce superoxide generation or ET- 1 production.


Assuntos
Antioxidantes/uso terapêutico , Hipertensão/tratamento farmacológico , Pergolida/uso terapêutico , Animais , Ácido Ascórbico/uso terapêutico , Pressão Sanguínea/efeitos dos fármacos , Desoxicorticosterona/toxicidade , Agonistas de Dopamina/uso terapêutico , Endotelina-1/biossíntese , Hipertensão/patologia , Hipertensão/fisiopatologia , Rim/efeitos dos fármacos , Rim/patologia , Rim/fisiopatologia , Masculino , Natriurese/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Superóxidos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...