用户名: 密码: 验证码:
阿托伐他汀上调PPARs表达及抑制心肌细胞肥大的作用
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的:心肌肥厚是临床上多种心血管疾病走向终末阶段的共同的病理生理基础。作为临床应用广泛的降低胆固醇药物,他汀已被证明有抑制心肌肥厚和抗炎的作用,但其机制尚未明确。近年研究证明,过氧化物酶体增殖物活化型受体(PPAR)α、β/δ和γ的活化可改善心肌肥厚,而且他汀在鼠的肝细胞和兔的脂肪细胞中上调PPARs。本研究旨在1.探讨阿托伐他汀在体外对AngⅡ介导的肥大心肌细胞的影响;2.分析PPARα、β/δ和γ在其中的可能作用;3.观察PPAR β/δ的特异激动剂(合成配体)GW0742使PPAR β/δ活化在心肌细胞肥大中所起的作用。
     方法:1.体外原代培养新生大鼠的心室肌细胞,用AngⅡ诱导建立心肌肥厚模型;在模型中加入不同浓度的阿托伐他汀(10、1、0.1μmol·L~(-1))或PPARβ/δ激动剂GW0742(10μmol·L~(-1))作用一定时间。2.测量心肌细胞肥大指标:用软件分析测量心肌细胞表面积,~3H-亮氨酸的掺入检测心肌细胞蛋白合成速率及使用RT-PCR半定量测定心肌肥厚的特征性指标ANP和BNP。3.在不同处理组,使用RT-PCR半定量测量观察炎性因子MMP9,MMP2,IL-1β和Corin mRNA的变化。4.使用RT-PCR观察PPARα、β/δ和γmRNA的表达变化,用激光共聚焦方法测定PPARs的蛋白水平的表达和活性变化及其在细胞内的定位。5.用报告基因载体转染法,检测阿托伐他汀是否对PPARs有活化作用。
     结果:1.阿托伐他汀抑制体外由AngⅡ介导的心肌细胞肥大:AngⅡ可使体外培养的心肌细胞面积(P<0.01)和~3H-亮氨酸的掺入增加(P<0.01),升高ANP、BNP与Corin(P<0.01)的表达,阿托伐他汀可逆转上述变化并呈剂量依赖性(P<0.01),而溶剂对照的DMSO对心肌肥厚无影响(P>0.05)。2.阿托伐他汀抑制肥大心肌细胞炎性因子的过度表达:在AngⅡ介导的心肌细胞肥大模型中,MMP9,MMP2,IL-1β表达明显增加(P<0.01),而阿托伐他汀可降
Objective: Cardiac hypertrophy is fundamental response of cardiac myocytes to various stimuli, which is associated with significantly increased risk of heart failure and malignant arrhythmia. Statins, HMG-CoA reductase inhibitors, are widely prescribed cholesterol-lowering drugs that also appear to play beneficial roles in the prevention of cardiac hypertrophy and inflammation, however, little is known about the mechanism. Recently, it has been proved that peroxisome peroliferator activated-receptors (PPARs) are involved in prevention of cardiac hypertrophy, and statins exert their effect on upregulation of PPARs in rat hepatocytes and rabbit adipocytes, respectively. The purposes of this study were: 1. to investigate the effects of atorvastatin on Ang II -induced cardiomyocytes hypertrophy and cytokines;2. to characterize the role of PPARs in regulating hypertrophic gene expressions in response to atorvastatin;3. to study the role of GW0742, a PPAR β/δ agonist, on Ang II-induced hypertrophic cardiomyocyte growth and gene expression of cytokines.Methods: 1 .Primary cultures of cardiac myocytes (MC) were prepared from the ventricles of Wistar rats (1-2d). Hypertrophy in neonatal rat cardiac MC was established with angiotensin II (Ang II) and treated with atorvastatin (10, 1, 0.1 μmol·L~(-1)) or GW0742 (10μmol·L~(-1)). 2. The characteristic parameters of hypertrophic myocytes include the surface area of MC which was analyzed by the aid of NIH Image J software, the synthetic rate of protein in MC that was detected by ~3H-leucine incorporation, and the mRNA expression of atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) was measured by reverse transcription-polymerase chain reaction (RT-PCR). 3. At mRNA level, the changes of MMP9,
    MMP2, IL-ip and Corin in all groups were measured by RT-PCR. 4. PPAR a, p/5, y subtypes were measured by RT-PCR at mRNA level, and detected by immuno-fluorescence staining at protein level, activation and intracellular localization. 5. A reporter gene assay was applied to evaluate the effect of atorvastatin on the transcriptional activities of PPARs.Results: 1. Atorvatatin inhibited Ang II -induced neonatal cardiacmyocytes hypertrophy in a dose-dependent manner, through decreasing surface area (PO.01), mRNA expression of ANP, BNP, Corin (PO.01), and 3H-leucine incorporation (PO.01) in MC. 2. Atorvastatin decreased the mRNA level of MMP9, MMP2 and IL-ip (PO.01) in hypertrophic myocytes. 3. The expression of PPARs was downregulated by Ang II at both mRNA and protein levels, otherwise, atorvastatin administration could reverse the changes above and improve activities of PPARs, but no effect was investaged on the cells exposed to DM SO, the solvent of atorvastatin. 4. To confirm PPARs-dependent pathway is involved in the inhibition of cardiac hypertrophy, a plasmid carrying PPRE derived from the promoter of rat acyl-CoA oxidase (ACO) gene was constructed. With it, HepG2 cells were transiently transfected with or without atorvastatin. Luciferase activities of transfected cells were measured using dual-luciferase reporter assay kit. The results indicated that the luciferase expression markedly increased in the atorvastatin-treated group compared with the vehicle group. 5. At lOumol'L"', GW0742, a highly selective PPAR p/5 activator, inhibited Ang II -induced protein synthesis (3H-leucine incorporation) (P<0.05), induction of the fetal-type gene ANP (PO.05), BNP (P<0.05) and cardiac myocyte size (P<0.05). Meanwhile, GW0742 has no such effect on normal myocytes (P > 0.05). 6. Induction of cardiac hypertrophy with Ang II also led to an augment at the transcription levels of MMP9, MMP2, IL-lp, and these changes were reversed by GW0742. 7. The agonist upregulated PPAR p/5 both at mRNA level (PO.05) and protein level (P<0.05), and increased PPAR p/5 activity, promote its translocation into cellular
    nucli.Conclusion: 1. Atorvastatin inhibits cardiac hypertrophy and has a potential role in the prevention of cytokine expressions induced by Angll in vitro;2. Atorvastatin upregulates and activates PPARs in hypertrophic myocytes;3. PPAR p/5 agonist GW0742 exerts anti-hypertophy and anti-inflammatory effect on myocytes stimulated by Angll. Furthermore, it increases the expression of PPAR p75 at mRNA and protein levels.In summary, the present investigation indicates that atorvastatin inhibits the Ang II -induced cardiomyocyte hypertrophy through activation of PPARs pathway partly. Our data may provide a novel insight into the development mechanism for the pathological process of cardiac hypertrophy and be helpful to further explore the novel pharmacyological effect of statins.
引文
1. Levy D, Garrison RJ, Savage DD, et al. Prognostic implications of echocardiographically determined left ventricular mass in the Framingham heart study. N Engl J Med. 1990, 322(22): 1561-1566.
    2. Morgan HE, Gordon EE, Kira Y, et al. Biochemical mechanisms of cardiac hypertrophy. Annu Rev Physiol. 1987, 49: 533-543.
    3. Lorell BH, Carabello BA. Left ventricular hypertrophy: pathogenesis, detection, and prognosis. Circulation. 2000, 102(4): 470-479
    4. Zou Y, Takano H, Akazawa H, et al. Molecular and cellular mechanisms of mechanical stress-induced cardiac hypertrophy. Endocr J. 2002, 49(1): 1-13.
    5. Igel M, Sudhop T, von Bergrnann K. Metabolism and drug interations of 3-hydroxy-3-methylglutaryl coenzyme A-reductase inhibitors (statins). Eur J Clin Pharmacol. 2001, 57(5): 357-364
    6. Scandinavian Simvastatin Survival Study Group. Randomised trial of cholesterol lowering in 4444 patients with coronary heart disease: the Scandinavian Simvastatin Survival Study (4S). Lancet. 1994, 344 (8934): 1383-1389.
    7. Ichiki T, Takeda T, Tokunou T, et al. Downregulation of angiotensin Ⅱ type 1 receptor by hydrophobic 3-hydroxy-3-methylglutaryl coenzyme reductase inhibitions in vascular smooth muscle cells. Artherioscler Thromb Vasc Biol. 2001, 21 (12): 1896-1901
    8. Laufs U, La Fata V, Plutzky J, et al. Upregulation of endothelial nireic oxide synthase by HMG-CoA reductase inhibtions. Circulation. 1998, 97(12): 1129-1135.
    9. Radomski MW, Rees DD, Dutra A, et al. S-nitroso-glutathione inhibits platelet activation in vitro and in vivo. Br J Pharmacol. 1992, 107(3): 745-749
    10. Nishimura T, Vaszar LT, Faul JL, et al. Simvastatin rescues rats from fatal pulmonary hypertension by inducing apoptosis of neointimal smooth muscle cells. Circulation. 2003, 108(13): 1640-1645.
    11. Hayashidani S, Tsutsui H, Shiomi T, et al. Fluvastatin, a 3-hydroxy-3-methyllglutaryl coenzyme A reductase inhibitor, attenuates left ventricular remodeling and failure after experimental myocardial infarction. Circulation. 2002, 105(7): 868-873.
    12. Porter KE, Turner NA, O'Regan D J, et al. Simvastatin reduces human atrial myofibroblast proliferation independently of cholesterol lowering via inhibition of Rho A. Cardiovascular Research. 2004, 61(4): 745-755
    13. Patel R, Nagueh SF, Tsybouleva N, et al. Simvastatin induces regression of cardiac hypertrophy and fibrosis and improves cardiac function in a transgenic rabbit model of human hypertrophic cardiomyopathy. Circulation. 2001, 104(3): 317-324.
    14. Issemann I, Green S. Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature. 1990, 347(6294): 645-650.
    15.叶平.过氧化体增殖物激活型受体对心肌能量代谢调控的病理生理机制.中国动脉硬化杂志.2003,11(1):81-83.
    16. Dreyer C, Krey G., Keller H, et al. Control of the peroxisomal beta-oxidation pathway by a novel family of nuclear hormone receptors. Cell. 1992, 68: 879-887.
    17. Djouadi F, Weinheimer CJ, Kelly DP. The role of PPAR alph as a "lipostat" transcription factor [J]. Adv Exp Med Biol. 1999, 466:211-220.
    18. Desvergne B, Wahli W. Peroxisome proliferator-activated receptors: nuclear control of metabolism [J]. Endocr Rev. 1999, 20(5): 649-688.
    19. Kliewer SA, Forman BM, Blumberg B, et al. Differential expression and activation of a family of murine peroxisome proliferator-activated receptors. Pro Natl Acad Sci USA. 1994, 91(1): 7355-7359
    20. Elbrecht A, Chen Y, Cullinan CA, et al. Molecular cloning, expression and characterization of human peroxisome proliferator activated receptors gamma 1 and gamma 2. Biochem Biophys Res Commun. 1996, 224(2): 431-437
    21. Kliewer SA, Sundseth SS, Jones SA, et al. Fatty acids and eicosanoids regulate gene expression through direct interactions with peroxisome proliferator-activated receptors alpha and gamma. Proe Natl Acad Sci USA. 1997, 94(9): 4318-4323.
    22. Irukayama-Tomobe Y, Miyauchi T, Sakai S, et al. Endothelin-l-induced cardiac hypertrophy is inhibited by activation of peroxisome proliferatoractivated receptor-or partly via blockade of blockade of c-Jun NH2-terminal kinase path-way. Circulation. 2004, 109(7): 904-910.
    23.叶平,方红,贺艳丽,等.非诺贝特和吡格列酮对心肌细胞肿瘤坏死因子-α表达的影响及机制探讨.中国药学杂志.2004,39:258-260。
    24.伍仕敏,叶平,周新,等.过氧化物酶体增殖物活化型受体γ在抑制大鼠心肌肥厚中的作用.中华心血管病杂志.2004,32(3):250-253。
    25. Asakawa M, Takamo H, Nagai T, et al. Peroxisome proliferator activated receptor gamma plays a critical role in inhibition of cardiac hypertrophy in vitro and in vivo. Circulation. 2002, 105(7): 1240-1246.
    26. Yamamoto K, Ohki R, Lee RT, et al. Peroxisome proliferators activated receptor gamma activators inhibit cardiac hypertrophy in cardiacmyocytes. Circulation. 2001, 104(14): 1670-1675.
    27. Ogata T, Miyauchi T, Sakai S, et al. Myocardial fibrosis and diastolic dysfunction in deoxycorticosterone acetate-salt hypertensive rats is ameliorated by the peroxisome proliferator-activated receptor-alpha activator fenofibrate, partly by suppressing inflammatory responses associated with the nuclear factor-kappa B pathway. J Am Coll Cardiol. 2004, 43(8): 1481-1488.
    28. Grip O, Janciauskiene S, Lindgren S. Atorvastatin activates PPAR gamma and attenuates the inflammatory response in human monocytes. Inflamm Res. 2002, 51(2): 58-62.
    29. Kabine M, El Kebbaj Z, Oaxaca-Castillo D. Peroxisome proliferator-activated receptors as regulators of lipid metabolism, tissue differential expression in adipose tissues during cold acclimatization and hibernation of jerboa (Jaculus orientalis). Biochimie. 2004, 86(11): 763-770.
    30. Landrier JF, Thomas C, Grober J, et al. Statin induction of liver fatty acidbinding protein (L-FABP) gene expression is peroxisome proliferatoractivated receptor-alpha-dependent. J Biol Chem. 2004, 279(44): 45512-45518.
    31.董丰,龚开政,张振刚,等.原代心肌细胞培养方法的探讨。大连医科大学学报.2001,23(4):207-208。
    32. Skrka Chlopcikova, Jitka Psotova, Petra Miketova. Neonatal rat cardiomyocytes- a model for the study of morphological, biochemical and edectrophy- siological characteristics of the heart. Biomed. 2001, 145(2):49-55.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700