Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-38323905

RESUMO

Cardiac fibrosis is considered as unbalanced extracellular matrix (ECM) production and degradation, contributing to heart failure. Short-chain acyl-CoA dehydrogenase (SCAD) negatively regulates pathological cardiac hypertrophy. The purpose of this study was to investigate the possible role of SCAD in cardiac fibrosis. In-vivo experiments were performed on spontaneously hypertensive rats (SHR) and SCAD knockout mice. The cardiac tissues of hypertensive patients with cardiac fibrosis were used for measurement of SCAD expression. In-vitro experiments, with angiotensin II (Ang II), SCAD siRNA and adenovirus-SCAD (Ad-SCAD) were performed using cardiac fibroblasts (CFs). SCAD expression was significantly decreased in the left ventricles of SHR. Notably, swim training ameliorated cardiac fibrosis in SHR in association with the elevation of SCAD. The decrease in SCAD protein and mRNA expression levels in SHR CFs were in accordance with those in the left ventricular myocardium of SHR. In addition, SCAD expression was downregulated in CFs treated with Ang II in vitro, and SCAD siRNA interference induced the same changes in cardiac fibrosis as Ang II-treated CFs, while Ad-SCAD treatment significantly reduced the Ang II-induced CFs proliferation, α-SMA and collagen expression. In SHR infected with Ad-SCAD, the cardiac fibrosis of the left ventricle was significantly decreased. On the other hand, cardiac fibrosis occurred in conventional SCAD knockout mice. SCAD immunofluorescence intensity of cardiac tissue in hypertensive patients with cardiac fibrosis was lower than that of healthy subjects. All together, the current experimental outcomes indicate that SCAD has a negative regulatory effect on cardiac fibrosis and support its potential therapeutic target for suppressing cardiac fibrosis.

2.
Br J Pharmacol ; 180(23): 3024-3044, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37377111

RESUMO

BACKGROUND AND PURPOSE: Our recent studies have shown that flavin adenine dinucleotide (FAD) exerts cardiovascular protective effects by supplementing short-chain acyl-CoA dehydrogenase (SCAD). The current study aimed to elucidate whether riboflavin (the precursor of FAD) could improve heart failure via activating SCAD and the DJ-1-Keap1-Nrf2 signalling pathway. EXPERIMENTAL APPROACH: Riboflavin treatment was given to the mouse transverse aortic constriction (TAC)-induced heart failure model. Cardiac structure and function, energy metabolism and apoptosis index were assessed, and relevant signalling proteins were analysed. The mechanisms underlying the cardioprotection by riboflavin were analysed in the cell apoptosis model induced by tert-butyl hydroperoxide (tBHP). KEY RESULTS: In vivo, riboflavin ameliorated myocardial fibrosis and energy metabolism, improved cardiac dysfunction and inhibited oxidative stress and cardiomyocyte apoptosis in TAC-induced heart failure. In vitro, riboflavin ameliorated cell apoptosis in H9C2 cardiomyocytes by decreasing reactive oxygen species (ROS). At the molecular level, riboflavin significantly restored FAD content, SCAD expression and enzymatic activity, activated DJ-1 and inhibited the Keap1-Nrf2/HO1 signalling pathway in vivo and in vitro. SCAD knockdown exaggerated the tBHP-induced DJ-1 decrease and Keap1-Nrf2/HO1 signalling pathway activation in H9C2 cardiomyocytes. The knockdown of SCAD abolished the anti-apoptotic effects of riboflavin on H9C2 cardiomyocytes. DJ-1 knockdown hindered SCAD overexpression anti-apoptotic effects and regulation on Keap1-Nrf2/HO1 signalling pathway in H9C2 cardiomyocytes. CONCLUSIONS AND IMPLICATIONS: Riboflavin exerts cardioprotective effects on heart failure by improving oxidative stress and cardiomyocyte apoptosis via FAD to stimulate SCAD and then activates the DJ-1-Keap1-Nrf2 signalling pathway.


Assuntos
Butiril-CoA Desidrogenase , Insuficiência Cardíaca , Animais , Camundongos , Butiril-CoA Desidrogenase/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Flavina-Adenina Dinucleotídeo/metabolismo , Flavina-Adenina Dinucleotídeo/farmacologia , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/metabolismo , Estresse Oxidativo , Apoptose , Miócitos Cardíacos/metabolismo
3.
Eur J Pharmacol ; 954: 175849, 2023 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-37331684

RESUMO

Short-chain acyl-CoA dehydrogenase (SCAD), the rate-limiting enzyme for fatty acid ß-oxidation, has a negative regulatory effect on pathological cardiac hypertrophy and fibrosis. FAD, a coenzyme of SCAD, participates in the electron transfer of SCAD-catalyzed fatty acid ß-oxidation, which plays a crucial role in maintaining the balance of myocardial energy metabolism. Insufficient riboflavin intake can lead to symptoms similar to short-chain acyl-CoA dehydrogenase (SCAD) deficiency or flavin adenine dinucleotide (FAD) gene abnormality, which can be alleviated by riboflavin supplementation. However, whether riboflavin can inhibit pathological cardiac hypertrophy and fibrosis remains unclear. Therefore, we observed the effect of riboflavin on pathological cardiac hypertrophy and fibrosis. In vitro experiments, riboflavin increased SCAD expression and the content of ATP, decreased the free fatty acids content and improved PE-induced cardiomyocytes hypertrophy and AngⅡ-induced cardiac fibroblasts proliferation by increasing the content of FAD, which were attenuated by knocking down the expression of SCAD using small interfering RNA. In vivo experiments, riboflavin significantly increased the expression of SCAD and the energy metabolism of the heart to improve TAC induced pathological myocardial hypertrophy and fibrosis in mice. The results demonstrate that riboflavin improves pathological cardiac hypertrophy and fibrosis by increasing the content of FAD to activate SCAD, which may be a new strategy for treating pathological cardiac hypertrophy and fibrosis.


Assuntos
Butiril-CoA Desidrogenase , Flavina-Adenina Dinucleotídeo , Animais , Camundongos , Butiril-CoA Desidrogenase/genética , Butiril-CoA Desidrogenase/metabolismo , Flavina-Adenina Dinucleotídeo/farmacologia , Riboflavina/farmacologia , Cardiomegalia/patologia , Ácidos Graxos não Esterificados , Fibrose
4.
J Hypertens ; 41(5): 775-793, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36883465

RESUMO

OBJECTIVES: Short-chain acyl-CoA dehydrogenase (SCAD), a key enzyme in the fatty acid oxidation process, is not only involved in ATP synthesis but also regulates the production of mitochondrial reactive oxygen species (ROS) and nitric oxide synthesis. The purpose of this study was to investigate the possible role of SCAD in hypertension-associated vascular remodelling. METHODS: In-vivo experiments were performed on spontaneously hypertensive rats (SHRs, ages of 4 weeks to 20 months) and SCAD knockout mice. The aorta sections of hypertensive patients were used for measurement of SCAD expression. In-vitro experiments with t-butylhydroperoxide (tBHP), SCAD siRNA, adenovirus-SCAD (MOI 90) or shear stress (4, 15 dynes/cm 2 ) were performed using human umbilical vein endothelial cells (HUVECs). RESULTS: Compared with age-matched Wistar rats, aortic SCAD expression decreased gradually in SHRs with age. In addition, aerobic exercise training for 8 weeks could significantly increase SCAD expression and enzyme activity in the aortas of SHRs while decreasing vascular remodelling in SHRs. SCAD knockout mice also exhibited aggravated vascular remodelling and cardiovascular dysfunction. Likewise, SCAD expression was also decreased in tBHP-induced endothelial cell apoptosis models and the aortas of hypertensive patients. SCAD siRNA caused HUVEC apoptosis in vitro , whereas adenovirus-mediated SCAD overexpression (Ad-SCAD) protected against HUVEC apoptosis. Furthermore, SCAD expression was decreased in HUVECs exposed to low shear stress (4 dynes/cm 2 ) and increased in HUVECs exposed to 15 dynes/cm 2 compared with those under static conditions. CONCLUSION: SCAD is a negative regulator of vascular remodelling and may represent a novel therapeutic target for vascular remodelling.


Assuntos
Butiril-CoA Desidrogenase , Hipertensão , Ratos , Animais , Camundongos , Humanos , Recém-Nascido , Butiril-CoA Desidrogenase/genética , Butiril-CoA Desidrogenase/metabolismo , Remodelação Vascular , Ratos Endogâmicos SHR , Ratos Wistar , Células Endoteliais da Veia Umbilical Humana/metabolismo , RNA Interferente Pequeno/metabolismo , Camundongos Knockout
5.
Life Sci ; 258: 118156, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-32735886

RESUMO

AIMS: Flavin adenine dinucleotide (FAD), participates in fatty acid ß oxidation as a cofactor, which has been confirmed to enhance SCAD activity and expression. However, the role of FAD on hypertensive vascular remodeling is unclear. In this study, we investigated the underlying mechanisms of FAD on vascular remodeling and endothelial homeostasis. MAIN METHODS: Morphological examination of vascular remodeling were analyzed with hematoxylin and eosin (HE) staining, Verhoeff's Van Gieson (EVG) staing, Dihydroethidium (DHE) staining and Sirius red staining. HUVECs apoptotic rate was detected by flow cytometry and HUVECs reactive oxygen species (ROS) was detected by DHE-probe. Enzymatic reactions were used to detect SCAD enzyme activity. The protein level was detected by Western Blots, the mRNA level was detected by quantitative real-time PCR. KEY FINDINGS: In vivo experiments, FAD significantly decreased blood pressure and ameliorated vascular remodeling by increasing SCAD expression, Nitric Oxide (NO) production and reducing ROS production. In vitro experiments, FAD protected against the tBHP induced injury in HUVEC, by increasing the activity of SCAD, increasing the elimination of free fatty acid (FFA), scavenging ROS, reducing apoptotic rate, thereby improving endothelial cell function. SIGNIFICANCE: FAD has a new possibility for preventing and treating hypertensive vascular remodeling.


Assuntos
Acil-CoA Desidrogenases/metabolismo , Ativadores de Enzimas/uso terapêutico , Flavina-Adenina Dinucleotídeo/uso terapêutico , Hipertensão/tratamento farmacológico , Remodelação Vascular/efeitos dos fármacos , Animais , Pressão Sanguínea/efeitos dos fármacos , Ativadores de Enzimas/farmacologia , Flavina-Adenina Dinucleotídeo/farmacologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Masculino , Ratos Endogâmicos SHR , Ratos Wistar
6.
Biochem Pharmacol ; 178: 114100, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32540485

RESUMO

Short-chain acyl-CoA dehydrogenase (SCAD), the rate-limiting enzyme for fatty acid ß-oxidation, has a negative regulatory effect on pathological cardiac hypertrophy and fibrosis. Furthermore, flavin adenine dinucleotide (FAD) can enhance the expression and enzyme activity of SCAD. However, whether FAD can inhibit pathological cardiac hypertrophy and fibrosis remains unclear. Therefore, we observed the effect of FAD on pathological cardiac hypertrophy and fibrosis. FAD significantly inhibited PE-induced cardiomyocyte hypertrophy and AngII-induced cardiac fibroblast proliferation. In addition, FAD ameliorated pathological cardiac hypertrophy and fibrosis in SHR. FAD significantly increased the expression and enzyme activity of SCAD. Meanwhile, ATP content was increased, the content of free fatty acids and reactive oxygen species were decreased by FAD in vivo and in vitro. In addition, molecular dynamics simulations were also used to provide insights into the structural stability and dynamic behavior of SCAD. The results demonstrated that FAD may play an important structural role on the SCAD dimer stability and maintenance of substrate catalytic pocket to increase the expression and enzyme activity of SCAD. In conclusion, FAD can inhibit pathological cardiac hypertrophy and fibrosis through activating SCAD, which may be a novel effective treatment for pathological cardiac hypertrophy and fibrosis, thus prevent them from developing into heart failure.


Assuntos
Butiril-CoA Desidrogenase/genética , Cardiomegalia/prevenção & controle , Cardiotônicos/farmacologia , Fibroblastos/efeitos dos fármacos , Flavina-Adenina Dinucleotídeo/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Trifosfato de Adenosina/biossíntese , Animais , Sítios de Ligação , Butiril-CoA Desidrogenase/metabolismo , Cardiomegalia/enzimologia , Cardiomegalia/genética , Cardiomegalia/patologia , Proliferação de Células/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Estabilidade Enzimática , Ácidos Graxos não Esterificados/antagonistas & inibidores , Ácidos Graxos não Esterificados/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose , Insuficiência Cardíaca/prevenção & controle , Masculino , Simulação de Dinâmica Molecular , Miocárdio/enzimologia , Miocárdio/patologia , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Ratos , Ratos Endogâmicos SHR , Ratos Wistar , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo
7.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 31(6): 756-761, 2019 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-31315737

RESUMO

OBJECTIVE: To observe the changes of short-chain acyl-CoA dehydrogenase (SCAD) expression on human umbilical vein endothelial cell (HUVEC) apoptosis and investigate its relationship with apoptosis. METHODS: The HUVEC was cultured normally for 2-3 days. The apoptotic model of HUVEC was established by tert-butyl hydrogen peroxide (tBHP). The HUVEC was treated by different concentrations of tBHP (0, 10, 20, 30, 40, 50 µmol/L) for 12 hours and different time (0, 3, 6, 9, 12 hours) with 50 µmol/L tBHP to establish the apoptotic model of HUVEC. The cell viability was detected by methyl thiazolyl tetrazolium (MTT), the mRNA expression of SCAD was determined by real-time polymerase chain reaction (PCR), the protein expression of SCAD was achieved by Western Blot. The best concentrate and time were determined to interfere the HUVEC to achieve the apoptotic model of HUVEC. The SCAD gene of HUVEC was knocked down by RNA interference sequence (siRNA274, siRNA414, siRNA679). The mRNA expression of SCAD, the protein expression of SCAD and the activity of SCAD enzyme were detected to achieve the best RNA interference sequence. The HUVEC was intervened by the best RNA interference sequence and tBHP. The cell activity and apoptosis rate, the enzyme activity of SCAD, the mRNA and protein expression of SCAD, the contents of reactive oxygen species (ROS), aderosine triphosphate (ATP) and free fatty acid (FFA) were detected to observe the effect of SCAD on apoptosis of HUVEC. RESULTS: (1) The cell viability, the mRNA expression and the protein expression of SCAD were decreased gradually in a concentration and time dependent manner with the increase of tBHP concentration and the prolongation of intervention time. The decline was most significant in the group of the 50 µmol/L tBHP to interfere HUVEC for 12 hours. (2) The siRNA679 transfection was the most significant in reducing SCAD mRNA and protein expressions among the three interference sequences (siRNA274, siRNA414, siRNA679). (3) Compare with blank control group, the cell viability was significantly decreased in the siRNA679 group (A value: 0.48±0.09 vs. 1.00±0.09, P < 0.01), the apoptotic rate of HUVEC was significantly increased [(29.96±2.09)% vs. (2.90±1.90)%, P < 0.01], the expression of SCAD mRNA and SCAD protein, the activity of SCAD enzyme and the content of ATP were significantly decreased [SCAD mRNA (2-ΔΔCt): 0.50±0.16 vs. 1.34±0.12, SCAD/α-Tubulin: 0.67±0.11 vs. 1.00±0.06, the activity of SCAD enzyme (kU/g): 0.38±0.04 vs. 0.53±0.04, the content of ATP (µmol/g): 0.14±0.02 vs. 0.19±0.01, all P < 0.05], the contents of FFA and ROS were significantly increased [FFA (nmol/g): 0.84±0.07 vs. 0.47±0.04, ROS (average fluorescence intensity): 647.5±23.7 vs. 434.2±46.5, both P < 0.01]. Meanwhile, SCAD siRNA treatment triggered the same apoptosis as HUVEC treated with tBHP. CONCLUSIONS: Down-regulation of SCAD may play an important role in HUVEC apoptosis. Increase in the expression of SCAD may become an important part in intervening HUVEC apoptosis.


Assuntos
Apoptose , Butiril-CoA Desidrogenase/metabolismo , Células Endoteliais da Veia Umbilical Humana , Sobrevivência Celular , Humanos , Espécies Reativas de Oxigênio
8.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 31(2): 172-177, 2019 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-30827304

RESUMO

OBJECTIVE: To Study the changes of short-chain acyl-CoA dehydrogenase (SCAD) in heart failure (HF) after myocardial infarction (MI), and the effect of aerobic exercise on SCAD. METHODS: Healthy male Sprague-Dawley (SD) rats were divided into sham operation group (Sham group), sham operation swimming group (Sham+swim group), HF model group (LAD group) and HF swimming group (LAD+swim group) by random number table method, with 9 rats in each group. The left anterior descending branch of coronary artery (LAD) was ligated to establish a rat model of HF after MI. In Sham group, only one loose knot was threaded under the left coronary artery, and the rest operations were the same as those in LAD group. Rats in Sham+swim group and LAD+swim group were given swimming test for 1 week after operation (from 15 minutes on the 1st day to 60 minutes on the 5th day). Then they were given swimming endurance training (from the 2nd week onwards, 60 minutes daily, 6 times weekly, 10 weeks in a row). Tail artery systolic pressure (SBP) was measured before swimming endurance training and every 2 weeks until the end of the 10th week. Ten weeks after swimming training, echocardiography was performed to measure cardiac output (CO), stroke volume (SV), left ventricular ejection fraction (LVEF), shortening fraction (FS), left ventricular end-systolic diameter (LVESD), left ventricular end-diastolic diameter (LVEDD), left ventricular end-systolic volume (LVESV), and left ventricular end-diastolic volume (LVEDV). Morphological changes of heart were observed by Masson staining. Apoptosis of myocardial cells was detected by transferase-mediated deoxyuridine triphosphate-biotin nick end labeling stain (TUNEL) and apoptosis index (AI) was calculated. Reverse transcription-polymerase chain reaction (RT-PCR) and Western Blot were used to detect the mRNA and protein expression of myocardial SCAD respectively. In addition, the enzyme activity of SCAD, the content of adenosine triphosphate (ATP) and free fatty acid (FFA) in serum and myocardium were detected according to the kit instruction steps. RESULTS: Compared with Sham group, Sham+swim group showed SBP did not change significantly, with obvious eccentric hypertrophy and increased myocardial contractility, and LAD group showed persistent hypotension, obvious MI, thinning of left ventricle, and decreased myocardial systolic/diastolic function. Compared with LAD group, SBP, systolic/diastolic function and MI in LAD+swim group were significantly improved [SBP (mmHg, 1 mmHg = 0.133 kPa): 119.5±4.4 vs. 113.2±4.5 at 4 weeks, 120.3±4.0 vs. 106.5±3.7 at 6 weeks, 117.4±1.3 vs. 111.0±2.3 at 8 weeks, 126.1±1.6 vs. 119.4±1.9 at 10 weeks; CO (mL/min): 59.10±6.31 vs. 33.19±4.76, SV (µL): 139.42±17.32 vs. 84.02±14.26, LVEF: 0.523±0.039 vs. 0.309±0.011, FS: (28.17±2.57)% vs. (15.93±3.64)%, LVEDD (mm): 8.80±0.19 vs. 9.35±0.30, LVESD (mm): 5.90±0.77 vs. 7.97±0.60, LVEDV (µL): 426.57±20.84 vs. 476.24±25.18, LVESV (µL): 209.50±25.18 vs. 318.60±16.10; AI: (20.4±1.4)% vs. (31.2±4.6)%; all P < 0.05]. Compared with Sham group, the mRNA and protein expression of myocardium SCAD, the activity of SCAD in Sham+swim group were significantly increased, the content of ATP was slightly increased, the content of serum FFA was significantly decreased, and the content of myocardial FFA was slightly decreased; conversely, the mRNA and protein expression of myocardium SCAD, the activity of SCAD and the content of ATP in LAD group were significantly decreased, the content of serum and myocardial FFA were significantly increased. Compared with LAD group, the mRNA and protein expression of myocardium SCAD, the content of ATP were significantly increased in LAD+swim group [SCAD mRNA (2-ΔΔCt): 0.52±0.16 vs. 0.15±0.01, SCAD/GAPDH (fold increase from Sham group): 0.94±0.08 vs. 0.60±0.11, ATP content (µmol/g): 52.8±10.1 vs. 14.7±6.1, all P < 0.05], the content of serum and myocardial FFA were significantly decreased [serum FFA (nmol/L): 0.11±0.03 vs. 0.29±0.04, myocardial FFA (nmol/g): 32.7±8.2 vs. 59.7±10.7, both P < 0.05], and the activity of SCAD was slightly increased (kU/g: 12.3±4.3 vs. 8.9±5.8, P > 0.05). CONCLUSIONS: The expression of SCAD in HF was significantly down-regulated, and the expression was significantly up-regulated after aerobic exercise intervention, indicating that swimming may improve the severity of HF by up-regulating the expression of SCAD.


Assuntos
Butiril-CoA Desidrogenase/metabolismo , Insuficiência Cardíaca/metabolismo , Animais , Insuficiência Cardíaca/etiologia , Masculino , Infarto do Miocárdio/complicações , Condicionamento Físico Animal , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley
9.
Oncol Lett ; 16(1): 467-474, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29928434

RESUMO

Glutathione S-transferase A1 (GSTA1) is a phase II detoxification enzyme and serves a crucial role in anti-cancer drug resistance. In our previous study, GSTA1 was identified to be highly expressed in various subtypes of non-small-cell lung cancer cell lines compared with human embryonic lung fibroblast cell line MRC-5. The aim of the present study was to investigate the effect of GSTA1 expression on the proliferation and apoptosis of A549 cells. GSTA1 expression was knocked down or with overexpressed using lentivirus particles. Western blot analysis and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were used to assess the protein, and mRNA levels of GSTA1 in A549 cells, respectively. The effect of GSTA1 manipulation on cell proliferation and apoptosis were investigated in vitro using MTT assays, Hoechst 33258 staining and flow cytometry, and in vivo using A549 cell line xenografts in nude mice. The results of the western blot analysis and RT-qPCR revealed that stable cell models of GSTA1 knockdown, and overexpression were established. The data of the MTT assay indicated that the downregulation of GSTA1 significantly inhibited cell proliferation compared with si-control-transfected cells. These si-GSTA1 A549 cells exhibited typical morphological changes of apoptosis, including chromatin condensation and shrunken nuclei compared with the si-control counterparts. An AnnexinV-fluorescein isothiocyanate assay verified that the downregulation of GSTA1 significantly induced cell apoptosis in vitro. In addition, overexpression of GSTA1 significantly promoted tumor growth in vivo. Accordingly, downregulation of GSTA1 suppressed tumor growth. In conclusion, GSTA1 plays an important role in regulation of cell proliferation and cell apoptosis in A549 cell line.

10.
J Cell Mol Med ; 20(7): 1381-91, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26989860

RESUMO

Short-chain acyl-CoA dehydrogenase (SCAD), a key enzyme of fatty acid ß-oxidation, plays an important role in cardiac hypertrophy. However, its effect on the cardiomyocyte apoptosis remains unknown. We aimed to determine the role of SCAD in tert-butyl hydroperoxide (tBHP)-induced cardiomyocyte apoptosis. The mRNA and protein expression of SCAD were significantly down-regulated in the cardiomyocyte apoptosis model. Inhibition of SCAD with siRNA-1186 significantly decreased SCAD expression, enzyme activity and ATP content, but obviously increased the content of free fatty acids. Meanwhile, SCAD siRNA treatment triggered the same apoptosis as cardiomyocytes treated with tBHP, such as the increase in cell apoptotic rate, the activation of caspase3 and the decrease in the Bcl-2/Bax ratio, which showed that SCAD may play an important role in primary cardiomyocyte apoptosis. The changes of phosphonate AMP-activated protein kinase α (p-AMPKα) and Peroxisome proliferator-activated receptor α (PPARα) in cardiomyocyte apoptosis were consistent with that of SCAD. Furthermore, PPARα activator fenofibrate and AMPKα activator AICAR treatment significantly increased the expression of SCAD and inhibited cardiomyocyte apoptosis. In conclusion, for the first time our findings directly demonstrated that SCAD may be as a new target to prevent cardiomyocyte apoptosis through the AMPK/PPARα/SCAD signal pathways.


Assuntos
Apoptose , Butiril-CoA Desidrogenase/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/enzimologia , Proteínas Quinases Ativadas por AMP/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Cardiotônicos/farmacologia , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fenofibrato/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , PPAR alfa/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Ribonucleotídeos/farmacologia , terc-Butil Hidroperóxido/farmacologia
11.
J Cell Mol Med ; 19(7): 1672-88, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25753319

RESUMO

This study was designed to investigate the expression of short-chain acyl-CoA dehydrogenase (SCAD), a key enzyme of fatty acid ß-oxidation, during rat heart development and the difference of SCAD between pathological and physiological cardiac hypertrophy. The expression of SCAD was lowest in the foetal and neonatal heart, which had time-dependent increase during normal heart development. In contrast, a significant decrease in SCAD expression was observed in different ages of spontaneously hypertensive rats (SHR). On the other hand, swim-trained rats developed physiological cardiac hypertrophy, whereas SHR developed pathological cardiac hypertrophy. The two kinds of cardiac hypertrophy exhibited divergent SCAD changes in myocardial fatty acids utilization. In addition, the expression of SCAD was significantly decreased in pathological cardiomyocyte hypertrophy, however, increased in physiological cardiomyocyte hypertrophy. SCAD siRNA treatment triggered the pathological cardiomyocyte hypertrophy, which showed that the down-regulation of SCAD expression may play an important role in pathological cardiac hypertrophy. The changes in peroxisome proliferator-activated receptor α (PPARα) was accordant with that of SCAD. Moreover, the specific PPARα ligand fenofibrate treatment increased the expression of SCAD and inhibited pathological cardiac hypertrophy. Therefore, we speculate that the down-regulated expression of SCAD in pathological cardiac hypertrophy may be responsible for 'the recapitulation of foetal energy metabolism'. The deactivation of PPARα may result in the decrease in SCAD expression in pathological cardiac hypertrophy. Changes in SCAD are different in pathological and physiological cardiac hypertrophy, which may be used as the molecular markers of pathological and physiological cardiac hypertrophy.


Assuntos
Butiril-CoA Desidrogenase/metabolismo , Cardiomegalia/enzimologia , Coração/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Pressão Sanguínea/efeitos dos fármacos , Butiril-CoA Desidrogenase/genética , Cardiomegalia/diagnóstico por imagem , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Modelos Animais de Doenças , Ácidos Graxos/metabolismo , Fenofibrato/farmacologia , Coração/efeitos dos fármacos , Coração/fisiopatologia , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Fator de Crescimento Insulin-Like I/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Tamanho do Órgão/efeitos dos fármacos , Oxirredução/efeitos dos fármacos , PPAR alfa/metabolismo , Fenilefrina/farmacologia , Interferência de RNA/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos Endogâmicos SHR , Ratos Wistar , Especificidade por Substrato/efeitos dos fármacos , Sístole/efeitos dos fármacos , Fatores de Tempo , Ultrassonografia
12.
Life Sci ; 124: 41-9, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25636810

RESUMO

AIMS: Short-chain acyl-CoA dehydrogenase (SCAD) is a key enzyme in fatty acid oxidation. In the present study we aim to investigate the changes in SCAD between pathological and physiological cardiomyocyte hypertrophy. We also explore the different signaling pathways of pathological and physiological cardiomyocyte hypertrophy. MAIN METHODS: After neonatal rat cardiomyocytes were treated as setups, cell surface area, expression of SCAD, PPARα, phospho-ERK1/2, activity of SCAD, free fatty acid content and ATP content in the cardiomyocytes were measured. KEY FINDINGS: Neonatal rat cardiomyocytes treated by PE showed an increased cell surface area and free fatty acid content, increased ERK1/2 phosphorylation, decreased expression of PPARα, decreased expression and activity of SCAD and decreased levels of ATP. Neonatal rat cardiomyocytes treated by IGF-1 showed the reverse effects except for the cell surface area. PPARα inhibitor GW6471 and PPARα activator Fenofibrate treatments abrogated the effects induced by IGF-1 and PE in cardiomyocytes respectively, as well as ERK1/2 activator EGF and ERK1/2 inhibitor PD98059. SIGNIFICANCE: SCAD has different changes between pathological and physiological cardiomyocyte hypertrophy. The ERK1/2/PPARα/SCAD signaling pathways play different roles in pathological and physiological cardiomyocyte hypertrophy. SCAD may be used as a new target to prevent the development of pathological cardiac hypertrophy.


Assuntos
Butiril-CoA Desidrogenase/metabolismo , Cardiomegalia/patologia , Ácidos Graxos/metabolismo , Miócitos Cardíacos/patologia , PPAR alfa/metabolismo , Animais , Animais Recém-Nascidos , Cardiomegalia/tratamento farmacológico , Modelos Animais de Doenças , Fenofibrato/farmacologia , Flavonoides/farmacologia , Fator de Crescimento Insulin-Like I/farmacologia , Sistema de Sinalização das MAP Quinases/fisiologia , Miócitos Cardíacos/efeitos dos fármacos , Oxazóis/farmacologia , Fenilefrina/farmacologia , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Tirosina/análogos & derivados , Tirosina/farmacologia
13.
Asian Pac J Cancer Prev ; 15(20): 8631-5, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25374180

RESUMO

Glutathione S-transferase A1 (GSTA1) appears to be primarily involved in detoxification processes, but possible roles in lung cancer remain unclear. The objective of this study was to investigate the expression and function of GSTA1 in lung cancer cells. Real-time PCR and Western blotting were performed to assess expression in cancer cell lines and the normal lung cells, then verify the A549 cells line with stable overexpression. Localization of GSTA1 proteins was assessed by cytoimmunofluorescence. Three double-strand DNA oligoRNAs (SiRNAs) were synthesized prior to being transfected into A549 cells with Lipofectamine 2000, and then the most efficient SiRNA was selected. Expression of the GSTA1 gene in the transfected cells was determined by real-time PCR and Western blotting. The viability of the transfected cells were assessed by MTT. Results showed that the mRNA and protein expression of A549 cancer cells was higher than in MRC-5 normal cells. Cytoimmunofluorescence demonstrated GSTA1 localization in the cell cytoplasm and/or membranes. Transfection into A549 cells demonstrated that down-regulated expression could inhibit cell viability. Our data indicated that GSTA1 expression may be a target molecule in early diagnosis and treatment of lung cancer.


Assuntos
Detecção Precoce de Câncer/métodos , Regulação Neoplásica da Expressão Gênica , Marcadores Genéticos/genética , Glutationa Transferase/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Análise de Variância , Western Blotting , Sobrevivência Celular/genética , Imunofluorescência , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/terapia , RNA Interferente Pequeno/análise , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real/métodos , Sensibilidade e Especificidade , Transfecção , Células Tumorais Cultivadas , Regulação para Cima
14.
Nan Fang Yi Ke Da Xue Xue Bao ; 33(8): 1137-40, 2013 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-23996752

RESUMO

OBJECTIVE: To study the effect of apigenin on the proliferation and apoptosis of human lung cancer cell line NCI-H460. METHODS: NCI-H460 cells were cultured with different concentrations of apigenin, and MTT assay was used to evaluate the cell inhibition rates. Apoptosis of NCI-H460 cells was observed under a fluorescence microscope with Hoechst 33258 staining and quantified by flow cytometry using annexin V-FITC/PI stain. The expressions of apoptosis-related proteins Bax, Bcl-2 and caspase-3 were analyzed by Western blotting. RESULTS: Apigenin causes concentration- and time-dependent inhibition of the proliferation of the cells. NCI-H460 cells treated with apigenin showed significant morphological changes of apoptosis, and the cell apoptotic rates increased as apigenin concentration increased. Western blotting demonstrated that apigenin increased the protein levels of Bax and caspase-3 and reduced the protein expression of Bcl-2. CONCLUSION: Apigenin can inhibit the proliferation and induce apoptosis of NCI-H460 cells possibly by up-regulating expression of Bax and caspase-3 and down-regulating the expression of Bcl-2.


Assuntos
Apigenina/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Caspase 3/metabolismo , Linhagem Celular Tumoral , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais , Proteína X Associada a bcl-2/metabolismo
15.
J Ethnopharmacol ; 145(1): 152-61, 2013 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-23164764

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: We have recently reported that tanshinone IIA attenuated cardiac fibrosis in two-kidney, two-clip renovascular hypertensive rats via inhibiting NAD(P)H oxidase. However, little is known about the cellular and molecular mechanisms of tanshinone IIA mediated anti-fibrotic effects in cardiac fibroblasts after H(2)O(2) stimulation. The present study was performed to investigate whether H(2)O(2) may increase collagen synthesis in cardiac fibroblasts by affecting the expression and activity of NAD(P)H oxidase and whether the effects of H(2)O(2) on cardiac fibroblasts can be blocked by treatment of tanshinone IIA. MATERIALS AND METHODS: Cardiac fibroblasts were treated with H(2)O(2) (100 µmol/L) in the presence or absence of tanshinone IIA (1 µmol/L), NAD(P)H oxidase inhibitors diphenyleneiodonium (10 µmol/L), siRNA-p47phox, siRNA-Nox2 and siRNA-Nox4. Collagen synthesis was measured by [(3)H]proline incorporation, O(2)(-) production were determined by flow cytometry and DHE fluorescence microscopy. NAD(P)H oxidase activity was measured by lucigenin-enhanced chemiluminescence. RESULTS: H(2)O(2) induced the activity of NAD(P)H oxidase, O(2)(-) production, collagen synthesis and fibronectin expression in cardiac fibroblasts, and DPI abolished this induction. Exposure of adult rat cardiac fibroblasts to H(2)O(2) had time-dependent increase in the expression of p47phox, Nox2 and Nox4 oxidases. In addition, tanshinone IIA significantly inhibited H(2)O(2)-induced collagen synthesis via attenuation of O(2)(-) generation and NAD(P)H oxidase activity. Moreover, siRNA-mediated knockdown of p47phox, Nox2 and Nox4 inhibited H(2)O(2)-induced NADPH oxidase activity. H(2)O(2)-induced collagen synthesis and fibronectin expression were also inhibited by p47phox, Nox2 and Nox4 knock down. CONCLUSIONS: Our data show that NAD(P)H oxidase plays a significant role in regulating collagen synthesis in H(2)O(2)-stimulated cardiac fibroblasts. Inhibition of NAD(P)H oxidase with tanshinone IIA completely blocked the H(2)O(2)-stimulated collagen production, which will raise the experimental basis for using tanshinone IIA to cardiac fibrosis in clinic.


Assuntos
Abietanos/farmacologia , Cardiotônicos/farmacologia , Colágeno/biossíntese , Peróxido de Hidrogênio/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Animais , DNA/biossíntese , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibronectinas/biossíntese , Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes/métodos , Coração/efeitos dos fármacos , Peróxido de Hidrogênio/antagonistas & inibidores , Masculino , Glicoproteínas de Membrana/biossíntese , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/biossíntese , NADPH Oxidases/metabolismo , Estresse Oxidativo/fisiologia , Cultura Primária de Células/métodos , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
16.
Pharmazie ; 66(7): 517-24, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21812327

RESUMO

Tanshinone IIA is one of major constituents of Salvia miltiorrhiza Bunge known as Danshen. Our and others' studies have shown that Tan IIA could protect cardimyocyte against apoptosis; however the effect of Tan IIA on cardiac remodeling disease is still unknown. In this study, we investigated the effects of Tan IIA on cardiac hypertrophy and fibrosis in two-kidney, two-clip (2K2C) hypertensive rats and by which, if any, mechanisms. Administration of 2K2C hypertensive rats with Tan IIA attenuated cardiac dysfunction and fibrosis. However Tan IIA treatment had no effects on BP control. Further studies revealed that Tan IIA inhibited the increased NAD(P)H oxidase activity and expression as well as O2*- production in 2K2C hypertensive rats. Our results indicated that Tan IIA significantly improved cardiac function and attenuated fibrosis in 2K2C hypertensive rats. The protective action of Tan IIA is likely mediated by its antioxidant effect, independent of BP control, partially via inhibiting NADPH oxidase.


Assuntos
Abietanos/farmacologia , Cardiomegalia/prevenção & controle , Cardiotônicos , Inibidores Enzimáticos , Hipertensão Renovascular/patologia , NADPH Oxidases/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Animais , Western Blotting , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Circulação Coronária/efeitos dos fármacos , Fibrose , Cardiopatias/diagnóstico por imagem , Cardiopatias/prevenção & controle , Hemodinâmica/efeitos dos fármacos , Hipertensão Renovascular/metabolismo , Masculino , Miocárdio/patologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Volume Sistólico/efeitos dos fármacos , Ultrassonografia , Disfunção Ventricular Esquerda/patologia , Disfunção Ventricular Esquerda/prevenção & controle
17.
Cytokine ; 52(3): 210-4, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20667748

RESUMO

To examine the roles of intracellular calcium in RANKL-induced bone marrow macrophages (BMMs) differentiation, the effects of intracellular calcium chelator BAPTA-AM on RANKL-induced BMMs differentiation, and the activation of its relating signal proteins (MAPKs, and the PI3K/Akt) were studied. BMMs were cultured with various concentrations of BAPTA-AM in the presence of M-CSF (25 ng/ml) and RANKL (25 ng/ml) for 7 days, osteoclastogenic ability, cytosolic free Ca(2+) concentration, osteoclast survival and the expression of phosphorylated ERK1/2, SAPK/JNK, Akt and p38 MAPK were measured by TRAP staining, spectrofluorometer and Western blotting. BAPTA-AM inhibited osteoclastogenesis and osteoclast survival of BMMs by RANKL induction. In osteoclasts without the pretreatment of BAPTA-AM, the increased response of [Ca(2+)](i) was observed within 15 min and the maximum was about 1.2 times that of control. This response was sustained for 30 min and returned to the control level at 1h after RANKL-inducing, and the increased response of [Ca(2+)](i) was completely abolished and sustained to at least 8h by BAPTA-AM. Although immunoblotting data revealed that RANKL could activate the phosphorylation of ERK1/2, SAPK/JNK, Akt and p38 MAPK, the expression of ERK1/2, Akt and p38 MAPK phosphorylation was inhibited by BAPTA-AM dose-dependently. These results revealed that BAPTA-AM inhibit osteoclastogenic ability of BMMs via suppressing the increase of [Ca(2+)](i) which lead to inhibit RANKL-induced the phosphorylation of ERK, Akt and p38 MAPK, but not JNK. This finding may be useful in the development of an osteoclastic inhibitor that targets intracellular signaling factors.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Cálcio/química , Diferenciação Celular/efeitos dos fármacos , Quelantes/farmacologia , Ácido Egtázico/análogos & derivados , Macrófagos/efeitos dos fármacos , Proteínas Quinases/metabolismo , Ligante RANK/fisiologia , Animais , Western Blotting , Células da Medula Óssea/citologia , Quelantes/química , Ácido Egtázico/química , Ácido Egtázico/farmacologia , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Fosforilação , Espectrometria de Fluorescência
18.
Phytomedicine ; 18(1): 58-64, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20638255

RESUMO

The adaptive changes that develop in the pressure-overloaded left ventricular myocardium include cardiac hypertrophy and interstitial fibrosis. The objectives of the present study were to evaluate the effects of Tanshinone II-A, a bioactive diterpene quinone isolated from Danshen, on cardiac fibrosis and collagen metabolism in rats with renovascular hypertension. Male Sprague-Dawley rats were subjected to two-kidney two-clip (2K2C) or sham operation (sham) and treated with Valsartan (Val, 26.7 mg/kg/d), Tanshinone II-A (Tsn, 70, 35 mg/kg/d) or vehicle. Six weeks later, systolic blood pressure (BP), LV weight, collagen abundance, cardiac function parameters, hydroxyproline content and mRNA levels of matrix metalloproteinase (MMP)-2, MMP-9, tissue inhibitor of metalloproteinase (TIMP)-1 and TIMP-2 were evaluated. Both high-dose (Tsn-H, 70 mg/kg/d) and low-dose (Tsn-L, 35 mg/kg/d) of Tsn failed to attenuate 2K2C-induced BP elevation but significantly attenuated the attendant interstitial fibrosis. Val suppressed elevations of BP and left ventricular systolic pressure (LVSP) in 2K2C rats. Val and Tsn-H exerted comparable suppressive effects on the gene expression of MMP-9 and TIMP-1, while Val decreased the MMP-2 mRNA level without affecting the transcript levels of TIMP-2. Both Val and Tsn-H attenuated cardiac dysfunction, while Tsn-L showed slight improvement. These data demonstrate for the first time, that Tsn prevented cardiac fibrosis and improved cardiac function in a rat model of renovascular hypertensive independent of hypotensive effect. Tsn conferred its beneficial effects on the collagen metabolism probably through its regulation of transcript levels of the MMPs/TIMPs balance.


Assuntos
Abietanos/uso terapêutico , Fármacos Cardiovasculares/uso terapêutico , Colágeno/metabolismo , Fibrose/prevenção & controle , Hipertensão Renovascular/tratamento farmacológico , Fitoterapia , Salvia miltiorrhiza/química , Abietanos/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Fármacos Cardiovasculares/farmacologia , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Expressão Gênica , Coração/efeitos dos fármacos , Hipertensão Renovascular/metabolismo , Hipertensão Renovascular/patologia , Masculino , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Miocárdio/patologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Tetrazóis/farmacologia , Tetrazóis/uso terapêutico , Inibidor Tecidual de Metaloproteinase-1/genética , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Valina/análogos & derivados , Valina/farmacologia , Valina/uso terapêutico , Valsartana
19.
Mol Cell Biochem ; 309(1-2): 99-107, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18008142

RESUMO

Human essential hypertension is a complex polygenic trait with underlying genetic components that remain unknown. The spontaneously hypertensive rat (SHR) is a well-characterized experimental model for essential hypertension. By comparative proteomics, we previously identified glutathione S-transferase, mu 2 (GSTM2), a protein involved in detoxification of reactive oxygen species, which had a significant reduction in left ventricles of 16-week-old SHR compared with WKY rats. In parallel, Western blotting and RT-PCR showed a similar reduction of GSTM2 in left ventricles and aortas of 4-, 8-, and 16-week-old SHR, which is before the onset of hypertension. This suggests that differential expression is not attributable to long-term changes in blood pressure. Meanwhile, the activities of GSTM2 were significantly decreased in different ages old SHR. Conversely, there was an enhanced generation of superoxide anion and activation of NADPH oxidase in SHR, which was accompanied by an increase in the protein expression of p47phox, a subunit of NADPH oxidase. These data suggest that it maybe a reduction in antioxidant defenses, evident by a reduced expression and activity of GSTM2, in the left ventricles and aortas of SHR that leads to increased levels of superoxide anion and activation of NADPH oxidase.


Assuntos
Glutationa Transferase/metabolismo , Estresse Oxidativo , Envelhecimento , Animais , Aorta/enzimologia , Aorta/fisiopatologia , Pressão Sanguínea , Ecocardiografia , Regulação Enzimológica da Expressão Gênica , Glutationa Transferase/genética , Ventrículos do Coração/enzimologia , Ventrículos do Coração/fisiopatologia , Hipertrofia , Immunoblotting , Masculino , Miocárdio/enzimologia , Miocárdio/patologia , NADPH Oxidases/metabolismo , Subunidades Proteicas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos SHR , Superóxidos/metabolismo , Sístole
20.
Arch Biochem Biophys ; 460(1): 25-32, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17346664

RESUMO

Peroxisome proliferator-activated receptors (PPARalpha, beta/delta and gamma) are nuclear receptors and PPARgamma activation was previously reported to inhibit collagen expression in the heart, but whether PPARbeta/delta also regulates collagen expression in the heart remains unclear. In this study, we investigated the effect of PPARbeta/delta activation on angiotensin II (Ang II)-induced collagen type I expression in adult rat cardiac fibroblasts. The results showed that PPARbeta/delta was expressed at the moderate level in cardiac fibroblasts. GW501516, a selective PPARbeta/delta agonist, depressed Ang II-stimulated collagen type I expression and collagen synthesis in cardiac fibroblasts in a concentration-dependent manner. Furthermore, these inhibitory effects of GW501516 were completely reversed by the knockdown of PPARbeta/delta via RNA interference. In summary, we find that PPARbeta/delta is present in cardiac fibroblasts and PPARbeta/delta activation inhibits Ang II-induced collagen type I expression at least in part via decreasing collagen synthesis. PPARbeta/delta may be a promising therapeutic target for myocardial fibrosis.


Assuntos
Angiotensina II/antagonistas & inibidores , Colágeno Tipo I/metabolismo , Fibroblastos/metabolismo , Miocárdio/citologia , PPAR delta/metabolismo , PPAR beta/metabolismo , Angiotensina II/farmacologia , Animais , Relação Dose-Resposta a Droga , Fibroblastos/efeitos dos fármacos , Masculino , PPAR delta/agonistas , PPAR beta/agonistas , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Tiazóis/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...