用户名: 密码: 验证码:
二甲基精氨酸—二甲胺水解酶对内皮祖细胞衰老的调控与糖尿病血管功能障碍
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
研究背景
     内皮祖细胞(endothelial progenitor cells, EPCs)是一类能分化为成熟的内皮细胞及心肌细胞,促进血管新生和改善心功能的干细胞。EPCs参与血管修复和血管新生,为治疗缺血性疾病提供了新的策略。
     血管内皮生长因子(vascular endothelial growth factor, VEGF)是调节EPCs功能最强的因子。VEGF与2型VEGF受体(KDR)相互作用而调节EPCs功能,包括内皮修复与血管新生。
     二甲基精氨酸-二甲胺水解酶(dimethylarginine dimethylaminohydrolase, DDAH)能特异性的水解L-精氨酸的同类物非对称二甲基精氨酸(asymmetric dimethylarginine, ADMA)。ADMA是一种主要的内源性一氧化氮合酶(nitric oxide synthase, NOS)抑制物,能竞争性抑制NOS,减少一氧化氮(nitric oxide, NO)合成。研究发现,内皮细胞衰老和血管新生涉及DDAH/ADMA途径。
     人类沉默信息调节因子2(Silent information regulator 2, Sir2) SIRT1是Ⅲ型组蛋白去乙酰化蛋白,能引起基因表达沉默。研究发现,SIRT1通过DDAH/ADMA途径抑制内皮细胞衰老。文献报道,高糖诱导EPCs衰老涉及SIT1途径。
     基于DDAH/ADMA与SIRT1参与血管新生及细胞衰老的调节以及SIRT1介导DDAH/ADMA途径的效应,我们推测DDAH/ADMA可能通过VEGF/KDR途径参与EPCs分化与功能的调控,该作用可能涉及SIRT1途径。因此,本实验在体外培养人外周血来源EPCs研究了DDAH/ADMA对EPCs功能与衰老的影响。
     方法
     EPCs培养与鉴定:梯度离心加选择性粘附分离健康人外周血EPCs,用内皮基础培养基EBM-2培养EPCs。培养7天后鉴定,包括乙酰化低密度脂蛋白(Dil-acLDL)与荆豆凝集素1(FITC-UEA-1)双染鉴定分化EPCS;流式细胞仪检测细胞CD133、CD34、CD45、KDR、vWF等标记物表达。
     DDAH对EPCs分化及功能调控及机制:提取分化第7天的EPCs,检测DDAH1和DDAH2表达;提取第7和17天EPCs,检测DDAH2、SIRT1表达。为了探讨DDAH2对EPCs功能的调控,运用pGCSIL-GFP-hDDAH2慢病毒转染EPCs沉默DDAH2表达;为了探讨SIRT1对DDAH2表达的调控,运用pGCSIL-GFP-hSIRT1慢病毒转染EPCs沉默SIRT1表达。提取细胞mRNA, Real-time PCR检测DDAH1、DDAP2、SIRT1、VEGF和KDR mRNA表达;流式细胞仪和Western Blot检测DDAH2和SIRT1蛋白表达;高效液相色谱法(high performance liquid chromatography, HPLC)检测细胞上清ADMA;β-半乳糖苷酶活性评价EPCs衰老程度;tube小管形成实验评价EPCs血管形成能力;纤维连接蛋白检测EPCs粘附能力。
     结果
     人外周血EPCs主要以DDAH2亚型表达为主,且随着EPCs分化时间延长而增加。干扰DDAH2表达能诱导EPCs衰老并抑制血管形成和粘附能力,抑制VEGF/KDR的mRNA表达。SIRT1能随着EPCs分化而表达增加,干扰SIRT1表达能下调DDAH2、VEGF/KDR表达,但对细胞上清ADMA浓度没有影响。
     结论
     DDAH2参与EPCs分化过程,并通过调节VEGF/KDR途径抑制EPCs的衰老,其作用机制涉及SIRT1途径。
     研究背景
     血管病变是糖尿病主要并发症之一。临床与动物实验发现糖尿病时,EPCs衰老率显著增加、功能受损。高糖能诱导EPCs衰老、血管形成能力减弱,导致糖尿病血管功能减弱、侧枝循环建立减少,引发心血管疾病。
     ADMA是一种新的心血管疾病预测因子,是内源性NOS抑制物,能竞争性抑制NOS,减少NO的合成。糖尿病时,细胞DDAH2表达降低、活性减弱,引起ADMA浓度增加并抑制NO生成。临床与动物实验证明,糖尿病时EPCs功能受损与NO浓度降低有关。细胞实验证明,ADMA能直接损伤EPCs功能。
     糖尿病或高糖孵育EPCs时,EPCs SIRT1表达下调,同时伴随细胞衰老和血管形成功能减退。内皮细胞实验发现,SIRT1可上调DDAH2表达,增加活性,促进ADMA水解,抑制内皮细胞自然衰老过程。
     本实验在2型糖尿病患者与培养EPCs细胞探讨了EPCs衰老与DDAH/ADMA之间的关系,以及该过程是否涉及SIRT1途径。
     方法
     临床研究:对照组与2型糖尿病患者组各40例。取外周静脉血,分离血浆与细胞。HPLC检测血浆ADMA浓度;梯度离心加粘附分离细胞EPCs,β-半乳糖苷酶活性评价细胞衰老,Real-time PCR法检测细胞DDAH2、SIRT1 mRNA表达。
     细胞实验:外源性给予葡萄糖(10,20,30 mmol/L)孵育EPCs建立高糖损伤EPCs细胞模型,以甘露醇(10,20,30 mmol/L)作为渗透压对照组,探讨高糖诱导EPCs衰老;构建pGC-FU-hDDAH2慢病毒探讨DDAH2抑制高糖诱导EPCs衰老;外源性给予ADMA(0, 0.3,0.6, 1pmol/L)探讨ADMA诱导EPCs衰老作用;运用SIRT1激动剂白藜芦醇(resveratrol, RSV)探讨SIRT1调节高糖诱导细胞衰老作用机制。
     梯度离心加粘附分离正常人外周血EPCs。细胞处理48h后,β-半乳糖苷酶活性评价细胞衰老。其他指标的检测为细胞处理24h时,Real-time PCR与Western Blot检测DDAH2、SIRT1 mRNA与蛋白表达,HPLC检测细胞上清ADMA水平,Griess法检测上清NO水平。
     结果
     2型糖尿病患者外周血EPCs衰老率显著升高,DDAH2与SIRT1 mRNA表达下调,同时伴随ADMA水平升高。
     葡萄糖能浓度依赖性地诱导健康人外周血EPCs衰老,下调EPCs DDAH2 mRNA和蛋白表达,增加培养上清ADMA水平并减少NO生成。渗透压对照组对细胞没有明显影响。过表达hDDAH2能抑制高糖诱导的EPCs衰老,并降低培养液中ADMA水平。外源性ADMA能浓度依赖性的诱导EPCs衰老。高糖能下调SIRT1 mRNA和蛋白表达。SIRT1激动剂RSV能显著抑制高糖诱导EPCs衰老,并且逆转高糖所致的DDAH2 mRNA表达下调及ADMA浓度升高。
     结论
     DDAH2/ADMA参与了高糖诱导的EPCs衰老,其作用涉及SIRT1途径。
     研究背景
     白藜芦醇(resveratrol, RSV)是一种多酚类的SIRT1天然激动剂。SIRT1参与糖代谢和胰岛素分泌的调节。糖尿病大鼠实验证明,RSV能上调VEGF与eNOS表达,抑制氧化应激反应,提高糖尿病大鼠骨髓细胞血管形成能力。细胞实验证明,RSV促进人外周血EPCs增殖、迁移以及血管新生。此外,RSV也能抑制TNF-α所致EPCs损伤。
     (E)-3,5,4'-三甲氧基-1,2-二苯乙烯((E)-3,5,4'-trimethoxystilbene, BTM-0512)是RSV的甲基化衍生物,其口服吸收率与半衰期均优于RSV。
     依据RSV对血管内皮和EPC保护作用以及SIRT1-DDAH2/ADMA对EPCs功能及衰老的调节作用,本实验在高脂饮食加单次腹腔注射链脲佐菌素(Streptozotocin, STZ)诱发的2型糖尿病大鼠模型与培养大鼠骨髓EPCs细胞,探讨RSV衍生物BTM-0512能否改善胰岛素抵抗,以及对EPCs衰老的影响及机制。
     方法
     动物实验:高脂饮食加单次腹腔注射STZ (35mg/kg)建立2型糖尿病大鼠模型。实验分为正常对照组、模型组、BTM-0512低剂量组(10mg/kg)和BTM-0512高剂量组(40mg/kg)。大鼠连续灌胃3周。
     取大鼠全血,分离血浆检测大鼠空腹血糖(FBG)、糖化血红蛋白(Hb1AC)、葡萄糖耐量(OGTT)、HOME.IR、胰岛素敏感指数(IAI)、胰岛素分泌指数(IS)以及ADMA浓度。切取胸主动脉观测内皮依赖性舒张反应,免疫组化检测血管内皮DDAH2、SIRT1蛋白表达。采用梯度离心加选择性粘附分离大鼠骨髓EPCs细胞,β-半乳糖苷酶活性评价细胞衰老程度,Real-time PCR检测EPCs DDAH2、SIRT1 mRNA表达,tube小管形成实验检测EPCs血管形成能力,transwell小室评价EPCs迁移能力,纤维连接蛋白粘附法评价EPCs粘附能力。
     细胞实验:高糖处理EPCs,探讨BTM-0512 (0.3,1,3μM)抑制高糖诱导EPCs衰老及可能机制,给予SIRT1特异性阻断剂splitomicin (50μM)探讨SIRT1在BTM-0512对EPCs保护中作用。β-半乳糖苷酶活性评价细胞衰老,Real-time PCR检测DDAH2、SIRT1 mRNA表达,Western Blot检测DDAH2蛋白表达;HPLC检测细胞上清ADMA水平。
     结果
     BTM-0512能剂量依赖性降低2型糖尿病大鼠FBG和HblAC,改善HOME.IR和IAI,但对OGTT与IS没有影响。BTM-0512能剂量依赖性改善2型糖尿病大鼠血管舒张能力,抑制骨髓EPCs衰老,保护EPCs迁移、粘附及血管形成功能;BTM-0512能上调血管内皮及EPCs DDAH2、SIRT1表达。
     在培养EPCs, BTM-0512能剂量依赖性抑制高糖诱导细胞衰老,上调细胞DDAH2、SIRT1 mRNA表达并降低培养上清中ADMA浓度。SIRT1抑制剂splitomicin (50μmol/L)能取消BTM-0512对高糖诱导的EPCs衰老与EPCs DDAH2 mRNA表达下调,以及上清ADMA浓度升高的逆转作用。
     结论
     1.BTM-0512可降低2型糖尿病大鼠血糖,改善IR,并能改善血管舒张功能。
     2.BTM-0512能减轻高糖所致EPCs衰老,其机制涉及SIRT1-DDAH2/ADMA途径。
Endothelial progenitor cells (EPCs), which can differentiate into mature endothelial cells and myocytes, promote neovascularization and improve cardiac function. EPCs participate in the reparation of vessles and angiogenesis, and are used for the treatment of ischemic diseases.
     Vascular endothelial growth factor (VEGF) is a potent factor regulating EPCs functions, including reendothelization and angiogenesis. The effects of VEGF have been shown to mediated by type 2 VEGF receptor (KDR).
     It is know that dimethylarginine dimethylaminohydrolase (DDAH) metabolizes asymmetric dimethylarginine (ADMA), which is endogenous inhibitor of nitric oxide synthase (NOS) activity and inhibits nitric oxide (NO) production. It has been demonstrated that the DDAH/ADMA pathway is involed in senescence of endothelial cell and angiogesis.
     SIRT1, human silent information regulator 2 (Sir2), is a potent NAD+-dependent protein deacetylase and plays an important role in the maintenance of gene silencing. SIRT1 inhibites senescence of endothelial cells through the DDAH2/ADMA pathway. It has been shown that SIRT1 is involved in the senescence of EPCs induced by high glucose.
     Based on the regulatory effect of DDAH/ADMA and SIRT1 on angiogenesis and cell senescence, and SIRT1-mediated the effect of DDAH/ADMA, therefore, in the present study we tested the effect of DDAH/ADMA and SIRT1 on senescenc and fucntion of EPCs.
     METHODS
     EPCs were isolated by density gradient centrifugation from human peripheral blood mononuclear cells, and cultured in endothelial basal medium-2 (EBM-2) supplemented with EGM-2 Single-Quots. On the 7th day, EPCs were characterized by dual staining for Dil-acetylated low-density lipoprotein (Dil-acLDL) and FITC-ulex europaeus agglutinin-1 (FITC-UEA-1), and by flow cell markers including CD 133, CD34, KDR, CD45 and von willebrand factor (vWF).
     To test the effect of DDAH on the differentiation and function of EPCs, mRNA expressions of DDAH1 and 2 were detected at 7th day, and the expressions of DDAH2 and SIRT1 mRNA and protein were detected at 7th and 17th days. To study the regulatory effect of DDAH2 on senescence and fucntion of EPCs, cells were transfected with pGCSIL-GFP-hDDAH2 shRNA to interrupt expression of DDAH2 protein. To study the regulatory effect of SIRT1 on DDAH2, cells were transfected with pGCSIL-GFP-hSIRT1 shRNA to interrupt expression of SIRT1 protein. The levels of DDAH1, DDAH2, VEGF, KDR and SIRT1 mRNA were analyzed by Real-time PCR. Protein expressions of DDAH2 and SIRT1 were detected by flow cytometry or Western Blotting. The level of ADMA was detected by high performance liquid chromatography (HPLC). Cell senescence, angiogenesis and adhesion were determined by senescence-associatedβ-galactosidase (SA-β-gal) activity assay, tube formation and fibronectin.
     RESULTS
     Peripheral blood EPCs predominantly expressed DDAH2 which was increased with EPCs differentiation. Interrupting DDAH2 expression induced EPCs senecence and dysfunction, including angiogenesis and adhesion. The expressions of VEGF and KDR mRNA were down-regulated. The expression of SIRT1 was increased with EPCs differentiation. Interrupting SIRT1 inhibited the expressions of DDAH2, VEGF and KDR, but had no effect on the level of ADMA.
     CONCLUSION
     DDAH2 participated in the differentiation of EPCs and regulated the senescence and fucntion of EPCs through the VEGF/KDR pathway by the activation of SIRT1.
     Vascular disease is a main complication of diabetes mellitus. Clinical and animal researches have showed that diabetes accelareted EPCs senescence and attenuated EPCs function. And high glucose induced EPCs senescence and impaired angiogenesis, resulting in dysfunction of vessels and reduction of collateral circulation. These results suggest that EPCs senescence may be related with the development of cardiovascular diseases.
     ADMA, a major endogenous inhibitor of NOS, has been considered as a novel risk factor for cardiovascular diseases. It has been demonstrated that the plasma level of ADMA is elevated and NO concentration is decreased concomitantly with a reduction of expression and acitvity of DDAH in patients with diabetes. Clinical and animal researches have showed that dysfunction of EPCs is related with reduction of NO level in diabetes. Exogenous ADMA directly induced the damage of EPCs.
     Studies in vivo or in vitro have documented that high glucose treatment induced senescence and angiogenesis impairment of EPCs and down-regulation of SIRT1 expression. It has been reported that the regulatory effect of SIRT1 on endothelial cells senescnece is mediated by the DDAH2/ADMA pathway.
     In the present experiment, we explored the regulatory effect of DDAH2/ADMA on sensecence of EPCs in diabetic rats or EPCs treated with high glucose. Since SIRT1 plays an important role in the development of senescence of EPCs, we also tested the effect of SIRT1 on the DDAH2/ADMA pathway.
     METHODS
     Forty control people and 40 T2DM patients were recruied, and control people were age and sex matched with T2DM patients. The level of ADMA in plasma was detected by HPLC. EPCs were isolated by density gradient centrifugation from human peripheral blood mononuclear cells. EPCs senescence was evaluated by SA-β-gal activity and the expression of DDAH2, SIRT1 mRNA was measured by Real-time PCR.
     EPCs were treated with glucose (10,20 or 30 mmol/L) for inducing senescence, and mannitol(10,20,30 mM) was used as osmotic pressure control. To study the effect of DDAH2 on senescence of EPCs, EPCs were transfected with pGC-FU-hDDAH2. ADMA(0,0.3,0.6,1μmol/L) was used to directly induce senscence of EPCs. The effect of resveratrol (RSV), a agonist of SIRT1, on senscence of EPCs was observed.
     Peripheral blood mononuclear cells were collected for isolating EPCs. EPCs senescence was evaluated by SA-β-gal activity. The expressions of DDAH2 and SIRT1 mRNA or protein were measured by Real-time PCR and by Wetern Blot, respectively. The level of ADMA was detected by HPLC, and the level of NO was detected by Griess.
     RESULTS
     In T2DM patients, the percentage of EPCs senescence was increased, while the expressions of both DDAH2 and SIRT1 mRNA were down-regulated concomitantly with an increase of concentrations of ADMA in plasma.
     High glucose induced senescenc of EPCs in a dose-dependent manner. The mRNA and protein expression of DDAH2 were down-regulated, and level of NO was decreased while level of ADMA was increased in the presence of high glucose. Overexpression of DDAH2 reversed the effect of glucose, but did not affect NO concentration in cultured EPCs. ADMA induced the senescence of EPCs in a dose-dependent manner. High glucose treatment down-regulated the mRNA and protein of SIRT1, which an effect was attenuated by RSV.
     CONCLUSION
     DDAH2/ADMA pathway participats in senescence of EPCs induced by high glucose, which is related with activation of SIRT1.
     Resveratrol (trans-3,5,4'-trihydroxystilbene) is a natural activator of SIRT1. Growing evidence suggests that SIRT1 regulates glucose and insulin secretion. It has been showed that in diabetic rats, RSV exerts multiple benificial effects including upregulating expression of VEGF and eNOS, inhibiting oxidative stress and mobilizing angiogenesis. In cultured EPCs, RSV can induce cell proliferation, migration and angiogenesis. Others have found that RSV reduced the injury of EPCs by TNF-a.
     (E)-3,5,4'-trimethoxystilbene (BTM-0512) is the methylated derivative of RSV. The absorbance rate of BTM-0512 is higher than RSV, and its half-life is extended.
     Based on the protective effect of RSV on senescence of endothelial cells and EPCs and the regulatory effect of SIRT1-DDAH2/ADMA pathway on EPCs function, therefore, we tested the influence of BTM-0512 on insulin resistance and senescence of EPCs in type 2 diabetic rats and cultured EPCs.
     METHODS
     Type 2 diabetic rat were established by bonus injection of streptozotocin (STZ,35 mg/kg, intraperitonealy) and feeding high-fat/high-sucrose diet. Rats were divided into 4 groups:control, T2DM, T2DM plus BTM-0512(10 mg/kg) and T2DM plus BTM-0512(40 mg/kg). The animals were treated with drugs for 3 weeks.
     The concentrations of fasting blood glucose (FBG) and glycosylated hemoglobin (Hb1AC), the level of fast insulin (FIns) and oral glucose tolerance (OGTT) were detected. The values of HOME.IR, insulin activity index (IAI) and insulin secretion index (IS) were calculated to evaluate insulin resistance (IR). The level of ADMA was measured. Vasodilator responses to acetylcholine in aortic rings and the protein expression of expression of DDAH2 and SIRT1 in the isolated aortia were measured. EPCs were isolated by density gradient centrifugation from rat bone marrow. Cell senescence, angiogenesis, adhesion and migration were determined by SA-β-gal activity assay, tube formation, fibronectin and transwell assay respectively. The mRNA expressions of DDAH2 and SIRT1 were measured by Real-time PCR.
     EPCs from rat bone marrow were characterized by dual staining for Dil-acLDL and UEA-1 on the 7th day. EPCs were treated with glucose (30 mmol/L) for inducing senescence. To study the effect of BTM-0512 on senescence of EPCs, EPCs were pretreated with BTM-0512 (0-3 μmol/L). To study the role of SIRT1 in BTM-0512 effect, EPCs were pretreated with splitomicin (50μmol/L), the special inhibitor of SIRT1.
     EPCs senescence was evaluated by examining SA-β-gal activity. The mRNA expressions of DDAH2 and SIRT1 were measured by Real-time PCR. Protein expression was detected by Wetern Blot. The level of ADMA was detected by HPLC. The level of NO was detected by Griess.
     RESULTS
     BTM-0512 significantly decreased the level of FBG and Hb1 AC in a dose-dependent manner. BTM-0512 improved the HOME.IR and IAI, but had no effect on OGTT or IS. BTM-0512 improved vasodilator responses to acetylcholine in diabetic rats. BTM-0512 inhibited senescence of EPCs and up-regulated the expression of DDAH2 and SIRT1 protein of endothelial cells as well as the expression of DDAH2 and SIRT1 mRNA of EPCs.
     BTM-0512 attenuated senescence of EPCs induced by high-glucose in a dose-dependent manner. BTM-0512 also reversed high glucose induced the downregulation of SIRT1 and DDAH2 mRNA expression with increased the level of ADMA, which were reversed by splimtomicin (50μM), the sepecial inhibiotr of SIRT1.
     CONCLUSION
     1. BTM-0512 reduced fasting blood glucose and improved insulin resistance in type 2 diabetic rats. BTM-0512 also improved endothelial function in diabetic rats.
     2. BTM-0512 exerted the beneficial effects on high glucose induced EPCs senescence, and the effects of BTM-0512 was related to activation of SIRT1-DDAH2/ADMA pathway.
引文
[1]Asahara T, Murohara T, Sullivan A, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science,1997,275(5302):964-967
    [2]Rafii S, Shapiro F, Rimarachin J, et al. Isolation and characterization of human bone marrow microvascular endothelial cells:hematopoietic progenitor cell adhesion. Blood,1994,84(1):10-19
    [3]Xue S, Zhang HT, Zhang P, et al. Functional endothelial progenitor cells derived from adipose tissue show beneficial effect on cell therapy of traumatic brain injury. Neurosci Lett,2010 (in press)
    [4]Strehlow K, Werner N, Berweiler J, et al. Estrogen increases bone marrow-derived endothelial progenitor cell production and diminishes neointima formation. Circulation,2003,107(24):3059-3065
    [5]Hur J, Yoon CH, Kim HS, et al. Characterization of two types of endothelial progenitor cells and their different contributions to neovasculogenesis. Arterioscler Thromb Vasc Biol,2004,24(2):288-293
    [6]Iwaguro H, Yamaguchi J, Kalka C, et al. Endothelial progenitor cell vascular endothelial growth factor gene transfer for vascular regeneration. Circulation, 2002,105(6):732-738
    [7]Kalka C, Tehrani H, Laudenberg B, et al. VEGF gene transfer mobilizes endothelial progenitor cells in patients with inoperable coronary disease. Ann Thorac Surg,2000,70(3):829-834
    [8]Ziegler BL, Valtieri M, Porada GA, et al. KDR receptor:a key marker defining hematopoietic stem cells. Science,1999,285(5433):1553-1558
    [9]Ito A, Tsao PS, Adimoolam S, et al. Novel mechanism for endothelial dysfunction:dysregulation of dimethylarginine dimethylaminohydrolase. Circulation,1999,99(24):3092-3095
    [10]Boger RH, Bode-Boger SM. Asymmetric dimethylarginine, derangements of the endothelial nitric oxide synthase pathway, and cardiovascular diseases. Semin Thromb Hemost,2000,26(5):539-545
    [11]Ueda S, Kato S, Matsuoka H, et al. Regulation of cytokine-induced nitric oxide synthesis by asymmetric dimethylarginine:role of dimethylarginine dimethylaminohydrolase. Circ Res,2003,92(2):226-233
    [12]Jacobi J, Sydow K, von Degenfeld G, et al. Overexpression of dimethylarginine dimethylaminohydrolase reduces tissue asymmetric dimethylarginine levels and enhances angiogenesis. Circulation,2005,111(11):1431-1438
    [13]Leiper JM, Santa Maria J, Chubb A, et al. Identification of two human dimethylarginine dimethylaminohydrolases with distinct tissue distributions and homology with microbial arginine deiminases. Biochem J,1999,343 Pt 1:209-214
    [14]Hasegawa K, Wakino S, Tanaka T, et al. Dimethylarginine dimethylaminohydrolase 2 increases vascular endothelial growth factor expression through Spl transcription factor in endothelial cells. Arterioscler Thromb Vase Biol,2006,26(7):1488-1494
    [15]Xiao J, Xiao ZJ, Liu ZG, et al. Involvement of dimethylarginine dimethylaminohydrolase-2 in visfatin-enhanced angiogenic function of endothelial cells. Diabetes Metab Res Rev,2009,25(3):242-249
    [16]Grozinger CM, Chao ED, Blackwell HE, et al. Identification of a class of small molecule inhibitors of the sirtuin family of NAD-dependent deacetylases by phenotypic screening. J Biol Chem,2001,276(42):38837-38843
    [17]Milne JC, Lambert PD, Schenk S, et al. Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes. Nature,2007, 450(7170):712-716
    [18]Huang J, Gan Q, Han L, et al. SIRT1 overexpression antagonizes cellular senescence with activated ERK/S6k1 signaling in human diploid fibroblasts. PLoS One,2008,3(3):e1710
    [19]Mattagajasingh I, Kim CS, Naqvi A, et al. SIRT1 promotes endothelium-dependent vascular relaxation by activating endothelial nitric oxide synthase. Proc Natl Acad Sci U S A,2007,104(37):14855-14860
    [20]Ota H, Akishita M, Eto M, et al. Sirtl modulates premature senescence-like phenotype in human endothelial cells. J Mol Cell Cardiol,2007,43(5):571-579
    [21]Potente M, Ghaeni L, Baldessari D, et al. SIRT1 controls endothelial angiogenic functions during vascular growth. Genes Dev,2007, 21(20):2644-2658
    [22]Menghini R, Casagrande V, Cardellini M, et al. MicroRNA 217 modulates endothelial cell senescence via silent information regulator 1. Circulation,2009, 120(15):1524-1532
    [23]Chen D, Lin Y, Zhang H. Characterization and expression of two amphioxus DDAH genes originating from an amphioxus-specific gene duplication. Gene, 2008,410(1):75-81
    [24]Balestrieri ML, Rienzo M, Felice F, et al. High glucose downregulates endothelial progenitor cell number via SIRT1. Biochim Biophys Acta,2008, 1784(6):936-945
    [25]Scalera F, Fulge B, Martens-Lobenhoffer J, et al. Red wine decreases asymmetric dimethylarginine via SIRT1 induction in human endothelial cells. Biochem Biophys Res Commun,2009,390(3):703-709
    [26]Kawamoto A, Tkebuchava T, Yamaguchi J, et al. Intramyocardial transplantation of autologous endothelial progenitor cells for therapeutic neovascularization of myocardial ischemia. Circulation,2003,107(3):461-468
    [27]Takeda Y, Uemura S, Iwama H, et al. Treatment with recombinant placental growth factor (PlGF) enhances both angiogenesis and arteriogenesis and improves survival after myocardial infarction. Circ J,2009,73(9):1674-1682
    [28]Yan J, Tie G, Park B, et al. Recovery from hind limb ischemia is less effective in type 2 than in type 1 diabetic mice:roles of endothelial nitric oxide synthase and endothelial progenitor cells. J Vasc Surg,2009,50(6):1412-1422
    [29]Salven P, Mustjoki S, Alitalo R, et al. VEGFR-3 and CD 133 identify a population of CD34+ lymphatic/vascular endothelial precursor cells. Blood, 2003,101(1):168-172
    [30]Shantsila E, Watson T, Tse HF, et al. New insights on endothelial progenitor cell subpopulations and their angiogenic properties. J Am Coll Cardiol,2008, 51(6):669-671
    [31]Tran CT, Fox MF, Vallance P, et al. Chromosomal localization, gene structure, and expression pattern of DDAH1:comparison with DDAH2 and implications for evolutionary origins. Genomics,2000,68(1):101-105
    [32]Thum T, Tsikas D, Stein S, et al. Suppression of endothelial progenitor cells in human coronary artery disease by the endogenous nitric oxide synthase inhibitor asymmetric dimethylarginine. J Am Coll Cardiol,2005, 46(9):1693-1701
    [33]Achan V, Tran CT, Arrigoni F, et al. all-trans-Retinoic acid increases nitric oxide synthesis by endothelial cells:a role for the induction of dimethylarginine dimethylaminohydrolase. Circ Res,2002,90(7):764-769
    [34]Smith CL, Birdsey GM, Anthony S, et al. Dimethylarginine dimethylaminohydrolase activity modulates ADMA levels, VEGF expression, and cell phenotype. Biochem Biophys Res Commun,2003,308(4):984-989
    [35]Tomikawa J, Fukatsu K, Tanaka S, et al. DNA methylation-dependent epigenetic regulation of dimethylarginine dimethylaminohydrolase 2 gene in trophoblast cell lineage. J Biol Chem,2006,281(17):12163-12169
    [36]Bai S, Ghoshal K, Datta J, et al. DNA methyltransferase 3b regulates nerve growth factor-induced differentiation of PC 12 cells by recruiting histone deacetylase 2. Mol Cell Biol,2005,25(2):751-766.
    [37]Marin-Husstege M, Muggironi M, Liu A, et al. Histone deacetylase activity is necessary for oligodendrocyte lineage progression. J Neurosci,2002, 22(23):10333-10345
    [38]Hamdollah Zadeh MA, Glass CA, Magnussen A, et al. VEGF-mediated elevated intracellular calcium and angiogenesis in human microvascular endothelial cells in vitro are inhibited by dominant negative TRPC6. Microcirculation,2008,15(7):605-614
    [39]Namiecinska M, Marciniak K, Nowak JZ. VEGF as an angiogenic, neurotrophic, and neuroprotective factor. Postepy Hig Med Dosw,2005, 59:573-583
    [40]Bahlmann FH, De Groot K, Spandau JM, et al. Erythropoietin regulates endothelial progenitor cells. Blood,2004,103(3):921-926
    [41]Heissig B, Hattori K, Dias S, et al. Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand. Cell,2002,109(5):625-637
    [42]Masoro EJ. Role of sirtuin proteins in life extension by caloric restriction. Mech Ageing Dev,2004,125(9):591-594
    [43]Liu T, Liu PY, Marshall GM. The critical role of the class III histone deacetylase SIRT1 in cancer. Cancer Res,2009,69(5):1702-1705
    [44]Nemoto S, M M Fergusson, T Finkel. SIRT1 functionally interacts with the metabolic regulator and transcriptional coactivator PGC-1{alpha}. J Biol Chem, 2005,280(16):16456-16460
    [45]Rodgers JT, Puigserver P. Fasting-dependent glucose and lipid metabolic response through hepatic sirtuin 1. Proc Natl Acad Sci U S A,2007, 104(31):12861-12866
    [46]van der Horst A, Tertoolen LG, de Vries-Smits LM, et al. FOXO4 is acetylated upon peroxide stress and deacetylated by the longevity protein hSir2(SIRT1). J Biol Chem,2004,279(28):28873-28879
    [47]Furukawa A, Tada-Oikawa S, Kawanishi S, et al. H2O2 accelerates cellular senescence by accumulation of acetylated p53 via decrease in the function of SIRT1 by NAD+ depletion. Cell Physiol Biochem,2007,20(1-4):45-54
    [48]Tepper OM, Galiano RD, Capla JM, et al. Human endothelial progenitor cells from type II diabetics exhibit impaired proliferation, adhesion, and incorporation into vascular structures. Circulation,2002,106(22):2781-2786
    [49]Loomans CJ, de Koning EJ, F J Staal, et al. Endothelial progenitor cell dysfunction:a novel concept in the pathogenesis of vascular complications of type 1 diabetes. Diabetes,2004,53(1):195-199
    [50]Fadini GP, Miorin M, Facco M, et al. Circulating endothelial progenitor cells are reduced in peripheral vascular complications of type 2 diabetes mellitus. J Am Coll Cardiol,2005,45(9):1449-1457
    [51]Chen MC, Sheu JJ, Wang PW, et al. Complications impaired endothelial progenitor cell function in Type 2 diabetic patients with or without critical leg ischaemia:implication for impaired neovascularization in diabetes. Diabet Med, 2009,26(2):134-141
    [52]Khoo CP, Valorani MG, Brittan M, et al. Characterization of endothelial progenitor cells in the NOD mouse as a source for cell therapies. Diabetes Metab Res Rev,2009,25(l):89-93
    [53]Krankel N, Adams V, Linke A, et al. Hyperglycemia reduces survival and impairs function of circulating blood-derived progenitor cells. Arterioscler Thromb Vasc Biol,2005,25(4)::698-703
    [54]Yamagishi S, Ueda S, Nakamura K, et al. Role of asymmetric dimethylarginine (ADMA) in diabetic vascular complications. Curr Pharm Des,2008, 14(25):2613-2618
    [55]Lin KY, Ito A, Asagami T, et al. Impaired nitric oxide synthase pathway in diabetes mellitus:role of asymmetric dimethylarginine and dimethylarginine dimethylaminohydrolase. Circulation,2002,106(8):987-992
    [56]Sorrenti V, Mazza F, Campisi A, et al. High glucose-mediated imbalance of nitric oxide synthase and dimethylarginine dimethylaminohydrolase expression in endothelial cells. Curr Neurovasc Res,2006,3(1):49-54
    [57]Scalera F, Borlak J, Beckmann B, et al. Endogenous nitric oxide synthesis inhibitor asymmetric dimethyl L-arginine accelerates endothelial cell senescence. Arterioscler Thromb Vasc Biol,2004,24(10):1816-1822
    [58]Jiang DJ, Jia SJ, Dai Z, et al. Asymmetric dimethylarginine induces apoptosis via p38 MAPK/caspase-3-dependent signaling pathway in endothelial cells. J Mol Cell Cardiol,2006,40(4):529-539
    [59]Napoli C, Balestrieri ML, Sica V, et al. Beneficial effects of low doses of red wine consumption on perturbed shear stress-induced atherogenesis. Heart Vessels,2008,23(2):124-133
    [60]Rosso A, Balsamo A, Gambino R, et al. p53 Mediates the accelerated onset of senescence of endothelial progenitor cells in diabetes. J Biol Chem,2006, 281(7):4339-4347
    [61]Scalera F, Fulge B, Martens-Lobenhoffer J, et al. Red wine decreases asymmetric dimethylarginine via SIRT1 induction in human endothelial cells. Biochem Biophys Res Commun,2009,297(5):H1876-1881
    [62]Wang CY, Wen MS, Wang HW, et al. Increased vascular senescence and impaired endothelial progenitor cell function mediated by mutation of circadian gene Per2. Circulation,2008,118(21):2166-2173
    [63]Kuki S, Imanishi T, Kobayashi K, et al. Hyperglycemia accelerated endothelial progenitor cell senescence via the activation of p38 mitogen-activated protein kinase. Circ J,2006,70(8):1076-1081
    [64]Surdacki A, Marewicz E, Wieczorek-Surdacka E, et al. Synergistic effects of asymmetrical dimethyl-L-arginine accumulation and endothelial progenitor cell deficiency on renal function decline during a 2-year follow-up in stable angina. Nephrol Dial Transplant,2009 (in press)
    [65]Chen YH, Lin SJ, Lin FY, et al. High glucose impairs early and late endothelial progenitor cells by modifying nitric oxide-related but not oxidative stress-mediated mechanisms. Diabetes,2007,56(6):1559-1568
    [66]Liang C, Ren Y, Tan H, et al. Rosiglitazone via upregulation of Akt/eNOS pathways attenuates dysfunction of endothelial progenitor cells, induced by advanced glycation end products. Br J Pharmacol,2009,158(8):1865-1873
    [67]Jiang JL, Wang S, Li NS, et al. The inhibitory effect of simvastatin on the ADMA-induced inflammatory reaction is mediated by MAPK pathways in endothelial cells. Biochem Cell Biol,2007,85(1):66-77
    [68]Scalera F, Martens-Lobenhoffer J, Tager M, et al. Effect of L-arginine on asymmetric dimethylarginine (ADMA) or homocysteine-accelerated endothelial cell aging. Biochem Biophys Res Commun,2006, 345(3):1075-1082
    [69]Bode-Boger SM, Martens-Lobenhoffer J, Tager M, et al. Aspirin reduces endothelial cell senescence. Biochem Biophys Res Commun,2005, 334(4):1226-1232
    [70]Wang M, Crisostomo PR, Herring C, et al. Human progenitor cells from bone marrow or adipose tissue produce VEGF, HGF, and IGF-I in response to TNF by a p38 MAPK-dependent mechanism. Am J Physiol Regul Integr Comp Physiol,2006,291(4):R880-884
    [71]Paiva H, Lehtimaki T, Laakso J, et al. Plasma concentrations of asymmetric-dimethyl-arginine in type 2 diabetes associate with glycemic control and glomerular filtration rate but not with risk factors of vasculopathy. Metabolism,2003,52(3):303-307
    [72]Xiong Y, Fu YF, Fu SH, et al. Elevated levels of the serum endogenous inhibitor of nitric oxide synthase and metabolic control in rats with streptozotocin-induced diabetes. J Cardiovasc Pharmacol,2003,42(2):191-196.
    [73]Bode-Boger SM, Scalera F, Martens-Lobenhoffer J. Asymmetric dimethylarginine (ADMA) accelerates cell senescence. Vasc Med,2005,10 Suppl 1:S65-71
    [74]Jia S J, Jiang DJ, Hu CP, et al. Lysophosphatidylcholine-induced elevation of asymmetric dimethylarginine level by the NADPH oxidase pathway in endothelial cells. Vascul Pharmacol,2006,44(3):143-148
    [75]Zhang GG, Shi RZ, Jiang DJ, et al. Involvement of the endothelial DDAH/ADMA pathway in nitroglycerin tolerance:the role of ALDH-2. Life Sci,2008,82(13-14):699-707
    [76]Brunet A, Sweeney LB, Sturgill JF, et al. Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science,2004, 303(5666):2011-2015
    [77]Yamakuchi M, Lowenstein. CJ MiR-34, SIRT1 and p53:the feedback loop. Cell Cycle,2009,8(5):712-715
    [78]Ungvari ZI, Labinskyy N, Mukhopadhyay P, et al. Resveratrol attenuates mitochondrial oxidative stress in coronary arterial endothelial cells. Am J Physiol Heart Circ Physiol,2009,297(5):H1876-1881
    [79]Csiszar A, Labinskyy N, Pinto JT, et al. Resveratrol induces mitochondrial biogenesis in endothelial cells. Am J Physiol Heart Circ Physiol,2009, 297(1):H13-20
    [80]Orimo M, Minamino T, Miyauchi H, et al. Protective role of SIRT1 in diabetic vascular dysfunction. Arterioscler Thromb Vasc Biol,2009,29(6):889-894
    [81]Maddux BA, See W, Lawrence JC, Jr., et al. Protection against oxidative stress-induced insulin resistance in rat L6 muscle cells by mircomolar concentrations of alpha-lipoic acid. Diabetes,2001,50(2):404-410
    [82]Singh I, Carey AL, Watson N, et al. Oxidative stress-induced insulin resistance in skeletal muscle cells is ameliorated by gamma-tocopherol treatment. Eur J Nutr,2008,47(7):387-392
    [83]Dokken BB, Saengsirisuwan V, Kim JS, et al. Oxidative stress-induced insulin resistance in rat skeletal muscle:role of glycogen synthase kinase-3. Am J Physiol Endocrinol Metab,2008,294(3):E615-621
    [84]Liang F, Kume S, Koya D. SIRT1 and insulin resistance. Nat Rev Endocrinol, 2009,5(7):367-373
    [85]Kobayashi Y, Furukawa-Hibi Y, Chen C, et al. SIRT1 is critical regulator of FOXO-mediated transcription in response to oxidative stress. Int J Mol Med, 2005,16(2):237-243
    [86]Grogaard HK, Sigurjonsson OE, Brekke M, et al. Cardiac accumulation of bone marrow mononuclear progenitor cells after intracoronary or intravenous injection in pigs subjected to acute myocardial infarction with subsequent reperfusion. Cardiovasc Revasc Med,2007,8(1):21-27
    [87]Babin-Ebell J, Sievers HH, Charitos EI, et al. Transmyocardial laser revascularization combined with intramyocardial endothelial progenitor cell transplantation in patients with intractable ischemic heart disease ineligible for conventional revascularization:preliminary results in a highly selected small patient cohort. Thorac Cardiovasc Surg,58(1):11-16
    [88]Kaeberlein M, McDonagh T, Heltweg B, et al. Substrate-specific activation of sirtuins by resveratrol. J Biol Chem,2005,280(17):17038-17045
    [89]刘立,王世辉,胡春,等.茋类化合物抗肿瘤活性研究进展.中国药物化学杂志,2007,17(05):321-326
    [90]Csiszar A, Labinskyy N, Podlutsky A, et al. Vasoprotective effects of resveratrol and SIRT1:attenuation of cigarette smoke-induced oxidative stress and proinflammatory phenotypic alterations. Am J Physiol Heart Circ Physiol, 2008,294(6):H2721-2735
    [91]Penumathsa SV, Maulik N. Resveratrol:a promising agent in promoting cardioprotection against coronary heart disease. Can J Physiol Pharmacol,2009, 87(4):275-286
    [92]Xu Q, Hao X, Yang Q, et al. Resveratrol prevents hyperglycemia-induced endothelial dysfunction via activation of adenosine monophosphate-activated protein kinase. Biochem Biophys Res Commun,2009,388(2):389-394
    [93]Gan L, Matsuura H, Ichiki T, et al. Improvement of neovascularization capacity of bone marrow mononuclear cells from diabetic mice by ex vivo pretreatment with resveratrol. Hypertens Res,2009,32(7):542-547
    [94]Wang XB, Huang J, Zou JG, et al. Effects of resveratrol on number and activity of endothelial progenitor cells from human peripheral blood. Clin Exp Pharmacol Physiol,2007,34(11):1109-1115
    [95]Kelley DE, He J, Menshikova EV, et al. Dysfunction of mitochondria in human skeletal muscle in type 2 diabetes. Diabetes,2002,51(10):2944-2950
    [96]Roberts CK, Barnard RJ, Sindhu RK, et al. Oxidative stress and dysregulation of NAD(P)H oxidase and antioxidant enzymes in diet-induced metabolic syndrome. Metabolism,2006,55(7):928-934
    [97]Tripathy D, Aljada A, Dandona P. Free fatty acids (FFA) and endothelial dysfunction; role of increased oxidative stress and inflammation.-to:Steinberg et al. (2002) Vascular function, insulin resistance and fatty acids. Diabetologia, 2003,46(2):300-301
    [98]Ogihara T, Asano T, Katagiri H, et al. Oxidative stress induces insulin resistance by activating the nuclear factor-kappa B pathway and disrupting normal subcellular distribution of phosphatidylinositol 3-kinase. Diabetologia, 2004,47(5):794-805
    [99]Ceriello A. Oxidative stress and glycemic regulation. Metabolism,2000,49(2 Suppl 1):27-29
    [100]Laight DW, Carrier MJ, Anggard EE. Antioxidants, diabetes and endothelial dysfunction. Cardiovasc Res,2000,47(3):457-464
    [101]Da Ros R, Assaloni R, Ceriello A. The preventive anti-oxidant action of thiazolidinediones:a new therapeutic prospect in diabetes and insulin resistance. Diabet Med,2004,21(11):1249-1252
    [102]Senti M, Tomas M, Fito M, et al. Antioxidant paraoxonase 1 activity in the metabolic syndrome. J Clin Endocrinol Metab,2003,88(11):5422-5426
    [103]Leonard SS, Xia C, Jiang BH, et al. Resveratrol scavenges reactive oxygen species and effects radical-induced cellular responses. Biochem Biophys Res Commun,2003,309(4):1017-1026
    [104]Yu HP, Hwang TL, Yen CH, et al. Resveratrol prevents endothelial dysfunction and aortic superoxide production after trauma hemorrhage through estrogen receptor-dependent hemeoxygenase-1 pathway. Crit Care Me,2010 (in press)
    [105]Thirunavukkarasu M, Penumathsa SV, Koneru S, et al. Resveratrol alleviates cardiac dysfunction in streptozotocin-induced diabetes:Role of nitric oxide, thioredoxin, and heme oxygenase. Free Radic Biol Med,2007,43(5):720-729
    [106]Sutra T, Oiry C, Azay-Milhau J, et al. Preventive effects of nutritional doses of polyphenolic molecules on cardiac fibrosis associated with metabolic syndrome: involvement of osteopontin and oxidative stress. J Agric Food Chem,2008, 56(24):11683-11687
    [107]Deng JY, Hsieh PS, Huang JP, et al. Activation of estrogen receptor is crucial for resveratrol-stimulating muscular glucose uptake via both insulin-dependent and-independent pathways. Diabetes,2008,57(7):1814-1823
    [108]Silan C. The effects of chronic resveratrol treatment on vascular responsiveness of streptozotocin-induced diabetic rats. Biol Pharm Bull,2008,31(5):897-902
    [109]Sun C, Zhang F, Ge X, et al. SIRT1 improves insulin sensitivity under insulin-resistant conditions by repressing PTP1B. Cell Metab,2007, 6(4):307-319
    [110]Pfluger PT, Herranz D, Velasco-Miguel S, et al. Sirtl protects against high-fat diet-induced metabolic damage. Proc Natl Acad Sci U S A,2008, 105(28):9793-9798
    [111]Baur JA, Pearson KJ, Price NL, et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature,2006,444(7117):337-342
    [112]Gonzalez-Rodriguez A, Mas Gutierrez JA, Sanz-Gonzalez S, et al. Inhibition of PTP1B restores IRS 1-mediated hepatic insulin signaling in IRS2-deficient mice. Diabetes,2009,59(3):588-599
    [113]Chabert P, Fougerousse A, Brouillard R. Anti-mitotic properties of resveratrol analog (Z)-3,5,4'-trimethoxystilbene. Biofactors,2006,27(1-4):37-46
    [114]Matsuda H, Tewtrakul S, Morikawa T, et al. Anti-allergic activity of stilbenes from Korean rhubarb (Rheum undulatum L.): structure requirements for inhibition of antigen-induced degranulation and their effects on the release of TNF-alpha and IL-4 in RBL-2H3 cells. Bioorg Med Chem,2004, 12(18):4871-4876
    [115]Guzik TJ, Mussa S, Gastaldi D, et al. Mechanisms of increased vascular superoxide production in human diabetes mellitus:role of NAD(P)H oxidase and endothelial nitric oxide synthase. Circulation,2002,105(14):1656-1662
    [116]Sasaki N, Yamashita T, Takaya T, et al. Augmentation of vascular remodeling by uncoupled endothelial nitric oxide synthase in a mouse model of diabetes mellitus. Arterioscler Thromb Vasc Biol,2008,28(6):1068-1076
    [117]Altinova AE, Arslan M, Sepici-Dincel A, et al. Uncomplicated type 1 diabetes is associated with increased asymmetric dimethylarginine concentrations. J Clin Endocrinol Metab,2007,92(5):1881-1885
    [118]Yasuda S, Miyazaki S, Kanda M, et al. Intensive treatment of risk factors in patients with type-2 diabetes mellitus is associated with improvement of endothelial function coupled with a reduction in the levels of plasma asymmetric dimethylarginine and endogenous inhibitor of nitric oxide synthase. Eur Heart J,2006,27(10):1159-1165
    [119]Jiang DJ, Hu GY, Jiang JL, et al. Relationship between protective effect of xanthone on endothelial cells and endogenous nitric oxide synthase inhibitors. Bioorg Med Chem,2003,11(23):5171-5177
    [120]Jiang DJ, Jiang JL, Zhu HQ, et al. Demethylbellidifolin preserves endothelial function by reduction of the endogenous nitric oxide synthase inhibitor level. J Ethnopharmacol,2004,93(2-3):295-306
    [121]Wang S, Jiang JL, Hu CP, et al. Relationship between protective effects of rosiglitazone on endothelium and endogenous nitric oxide synthase inhibitor in streptozotocin-induced diabetic rats and cultured endothelial cells. Diabetes Metab Res Rev,2007,23(2):157-164
    [122]Ungvari Z, Labinskyy N, Mukhopadhyay P, et al. Resveratrol attenuates mitochondrial oxidative stress in coronary arterial endothelial cells. Am J Physiol Heart Circ Physiol,2009,297(5):H1876-1881.
    [123]Hu Z, Zhang F, Yang Z, et al. Low-dose aspirin promotes endothelial progenitor cell migration and adhesion and prevents senescence. Cell Biol Int, 2008,32(7):761-768
    [124]Imanishi T, Hano T, Sawamura T, et al. Oxidized low-density lipoprotein induces endothelial progenitor cell senescence, leading to cellular dysfunction. Clin Exp Pharmacol Physiol,2004,31(7):407-413
    [125]Assmus B, Urbich C, Aicher A, et al. HMG-CoA reductase inhibitors reduce senescence and increase proliferation of endothelial progenitor cells via regulation of cell cycle regulatory genes. Circ Res,2003,92(9):1049-1055
    [126]Spyridopoulos I, Haendeler J, Urbich C, et al. Statins enhance migratory capacity by upregulation of the telomere repeat-binding factor TRF2 in endothelial progenitor cells. Circulation,2004,110(19):3136-3142
    [127]Imanishi T, Kobayashi K, Kuroi A, et al. Pioglitazone inhibits angiotensin II-induced senescence of endothelial progenitor cell. Hypertens Res,2008, 31(4):757-765
    [128]Sorrentino SA, Bahlmann FH, Besler C, et al. Oxidant stress impairs in vivo reendothelialization capacity of endothelial progenitor cells from patients with type 2 diabetes mellitus:restoration by the peroxisome proliferator-activated receptor-gamma agonist rosiglitazone. Circulation,2007,116(2):163-173
    [129]Xia L, Wang XX, Hu XS, et al. Resveratrol reduces endothelial progenitor cells senescence through augmentation of telomerase activity by Akt-dependent mechanisms. Br J Pharmacol,2008,155(3):387-394
    [1]Vaziri H, Dessain SK, Eaton ENg, et al. hSIR2(SIRT1) functions as an NAD-dependent p53 deacetylase. Cell,2001,107(2):149-159
    [2]Chua KF, Mostoslavsky R, Lombard DB, et al. Mammalian SIRT1 limits replicative life span in response to chronic genotoxic stress. Cell Metab,2005, 2(1):67-76
    [3]Orimo M, Minamino T, Miyauchi H, et al. Protective role of SIRT1 in diabetic vascular dysfunction. Arterioscler Thromb Vasc Biol,2009,29(6):889-894
    [4]Chen CJ, Yu W, Fu YC, et al. Resveratrol protects cardiomyocytes from hypoxia-induced apoptosis through the SIRT1-FoxO1 pathway. Biochem Biophys Res Commun,2009,378(3):389-393
    [5]Menghini R, Casagrande V, Cardellini M, et al. MicroRNA 217 modulates endothelial cell senescence via silent information regulator 1. Circulation,2009, 120(15):1524-1532
    [6]Brunet A, Sweeney LB, Sturgill JF, et al. Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science,2004, 303(5666):2011-2015
    [7]van der Horst A, Tertoolen LG, de Vries-Smits LM, et al. FOXO4 is acetylated upon peroxide stress and deacetylated by the longevity protein hSir2(SIRT1). J Biol Chem,2004,279(28):28873-28879
    [8]Bordone L, Motta MC, Picard F, et al. Sirtl regulates insulin secretion by repressing UCP2 in pancreatic beta cells. PLoS Biol,2006,4(2):e31
    [9]Nemoto S, Fergusson MM, Finkel T. SIRT1 functionally interacts with the metabolic regulator and transcriptional coactivator PGC-1{alpha}. J Biol Chem, 2005,280(16):16456-16460
    [10]Picard F, Kurtev M, Chung N, et al. Sirtl promotes fat mobilization in white adipocytes by repressing PPAR-gamma. Nature,2004,429(6993):771-776
    [11]Qiao L, Shao J. SIRT1 regulates adiponectin gene expression through Foxol-C/enhancer-binding protein alpha transcriptional complex. J Biol Chem, 2006,281(52):39915-39924
    [12]Gomez-Valades AG, Mendez-Lucas A, Vidal-Alabro A, et al. Pckl gene silencing in the liver improves glycemia control, insulin sensitivity, and dyslipidemia in db/db mice. Diabetes,2008,57(8): 2199-2210
    [13]Sulaiman M, Matta MJ, Sunderesan NR, et al. Resveratrol, an activator of SIRT1, upregulates sarcoplasmic calcium ATPase and improves cardiac function in diabetic cardiomyopathy. Am J Physiol Heart Circ Physiol,2010, 298(3):H833-843
    [14]Shen Z, Ajmo JM, Rogers CQ, et al. Role of SIRT1 in regulation of LPS-or two ethanol metabolites-induced TNF-alpha production in cultured macrophage cell lines. Am J Physiol Gastrointest Liver Physiol,2009,296(5):G1047-1053
    [15]Sun C, Zhang F, Ge X, et al. SIRT1 improves insulin sensitivity under insulin-resistant conditions by repressing PTP1B. Cell Metab,2007, 6(4):307-319
    [16]Pfluger PT, Herranz D, Velasco-Miguel S, et al. Sirtl protects against high-fat diet-induced metabolic damage. Proc Natl Acad Sci U S A,2008, 105(28):9793-9798
    [17]Moynihan KA, Grimm AA, Plueger MM, et al. Increased dosage of mammalian Sir2 in pancreatic beta cells enhances glucose-stimulated insulin secretion in mice. Cell Metab,2005,2(2):105-117
    [18]Rutanen J, Yaluri N, Modi S, et al. SIRT1 mRNA expression may be associated with energy expenditure and insulin sensitivity. Diabetes,2010 (in press)
    [19]Yoshizaki T, Schenk S, Imamura T, et al. SIRT1 inhibits inflammatory pathways in macrophages and modulates insulin sensitivity. Am J Physiol Endocrinol Metab,2009,298(3):E419-428.
    [20]Tanno M, Sakamoto J, Miura T, et al. Nucleocytoplasmic shuttling of the NAD+-dependent histone deacetylase SIRT1. J Biol Chem,2007, 282(9):6823-6832
    [21]Tanno M, Kuno A, Yano T, et al. Induction of manganese superoxide dismutase by nuclear translocation and activation of SIRT1 promotes cell survival in chronic heart failure. J Biol Chem,2010,285(11):8375-8382
    [22]Rane S, He M, Sayed D, et al. Downregulation of miR-199a derepresses hypoxia-inducible factor-1 alpha and Sirtuin 1 and recapitulates hypoxia preconditioning in cardiac myocytes. Circ Res,2009,104(7):879-886
    [23]Alcendor RR, Gao S, Zhai P, et al. Sirtl regulates aging and resistance to oxidative stress in the heart. Circ Res,2007,100(10):1512-1521
    [24]Mattagajasingh I, Kim CS, Naqvi A, et al. SIRT1 promotes endothelium-dependent vascular relaxation by activating endothelial nitric oxide synthase. Proc Natl Acad Sci U S A,2007,104(37):14855-14860
    [25]Ota H, Akishita M, Eto M, et al. Sirtl modulates premature senescence-like phenotype in human endothelial cells. J Mol Cell Cardiol,2007,43(5):571-579
    [26]Arunachalam G, Yao H, Sundar IK, et al. SIRT1 regulates oxidant-and cigarette smoke-induced eNOS acetylation in endothelial cells:Role of resveratrol. Biochem Biophys Res Commun,2010,393(l):66-72
    [27]Dekker RJ, van Thienen JV, Rohlena J, et al. Endothelial KLF2 links local arterial shear stress levels to the expression of vascular tone-regulating genes. Am J Pathol,2005,167(2):609-618.
    [28]Parmar KM, Larman HB, Dai G, et al. Integration of flow-dependent endothelial phenotypes by Kruppel-like factor 2. J Clin Invest,2006, 116(1):49-58
    [29]Gracia-Sancho J, Villarreal G, Zhang Y, et al. Activation of SIRT1 by resveratrol induces KLF2 expression conferring an endothelial vasoprotective phenotype. Cardiovasc Res,2010,85(3):514-519
    [30]Potente M, Ghaeni L, Baldessari D, et al. SIRT1 controls endothelial angiogenic functions during vascular growth. Genes Dev,2007, 21(20):2644-2658
    [31]Cardellini M, Menghini R, Martelli E, et al. TIMP3 is reduced in atherosclerotic plaques from subjects with type 2 diabetes and increased by SirTl. Diabetes, 2009,58(10):2396-2401
    [32]Miyazaki R, Ichiki T, Hashimoto T, et al. SIRT1, a longevity gene, downregulates angiotensin Ⅱ type 1 receptor expression in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol,2008,28(7):1263-1269
    [33]Chen WY, Wang DH, Yen RC, et al. Tumor suppressor HIC1 directly regulates SIRT1 to modulate p53-dependent DNA-damage responses. Cell,2005, 123(3):437-448
    [34]Deltour S, Pinte S, Guerardel C, et al. The human candidate tumor suppressor gene HIC1 recruits CtBP through a degenerate GLDLSKK motif. Mol Cell Biol, 2002,22(13):4890-4901
    [35]Stankovic-Valentin N, Verger A, Deltour-Balerdi S, et al. A L225A substitution in the human tumour suppressor HIC1 abolishes its interaction with the corepressor CtBP. FEBS J,2006; 273(13):2879-2890
    [36]Zhang Q, Wang SY, Fleuriel C, et al. Metabolic regulation of SIRT1 transcription via a HIC1:CtBP corepressor complex. Proc Natl Acad Sci U S A, 2007,104(3):829-833
    [37]Stankovic-Valentin N, Deltour S, Seeler J, et al. An acetylation/deacetylation-SUMOylation switch through a phylogenetically conserved psiKXEP motif in the tumor suppressor HIC1 regulates transcriptional repression activity. Mol Cell Biol,2007,27(7):2661-2675
    [38]Chen W, Cooper TK, Zahnow CA, et al. Epigenetic and genetic loss of Hicl function accentuates the role of p53 in tumorigenesis. Cancer Cell,2004, 6(4):387-398
    [39]Lee J, Padhye A, Sharma A, et al. A pathway involving FXR and shp positively regulates hepatic SIRT1 levels via MIR-34A inhibition. J Biol Chem,2010 (in press)
    [40]Yamakuchi M, Ferlito M, Lowenstein CJ. miR-34a repression of SIRT1 regulates apoptosis. Proc Natl Acad Sci U S A,2008,105(36):13421-13426
    [41]Fujita Y, Kojima K, Hamada N, et al. Effects of miR-34a on cell growth and chemoresistance in prostate cancer PC3 cells. Biochem Biophys Res Commun, 2008,377(1):114-119
    [42]Strum JC, Johnson JH, Ward J, et al. MicroRNA 132 regulates nutritional stress-induced chemokine production through repression of SirTl. Mol Endocrinol,2009,23(11):1876-1884
    [43]Chen L, Feng Y, Zhou Y, et al. Dual role of Zn2+ in maintaining structural integrity and suppressing deacetylase activity of SIRT1. J Inorg Biochem,2010, 104(2):180-185
    [44]Nemoto S, Fergusson MM, Finkel T. Nutrient availability regulates SIRT1 through a forkhead-dependent pathway. Science,2004,306(5704):2105-2108
    [45]Borradaile NM, Pickering JG Polyploidy impairs human aortic endothelial cell function and is prevented by nicotinamide phosphoribosyltransferase. Am J Physiol Cell Physiol,2010,298(1):C66-74
    [46]Guo X, Williams JG, Schug TT, et al. DYRK1A and DYRK3 promote cell survival through phosphorylation and activation of SIRT1. J Biol Chem,2010 (in press)
    [47]Kang H, Jung JW, Kim MK, et al. CK2 is the regulator of SIRT1 substrate-binding affinity, deacetylase activity and cellular response to DNA-damage. PLoS One,2009,4(8):e6611
    [48]Puca R, Nardinocchi L, Starace G, et al. Nox1 is involved with p53 deacetylation and suppression of its transcriptional activity and apoptosis. Free Radic Biol Med,2010 (in press)
    [49]Milne JC, Lambert PD, Schenk S, et al. Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes. Nature,2007, 450(7170):712-716
    [50]Ota H, Eto M, Kano MR, et al. Cilostazol inhibits oxidative stress-induced premature senescence via upregulation of Sirtl in human endothelial cells. Arterioscler Thromb Vasc Biol,2008,28(9):1634-1639

© 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