绿茶多酚对高脂诱导的代谢紊乱和血管内皮功能障碍的影响
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
第一部分绿茶多酚对高脂诱导代谢紊乱的影响
     目的:观察高脂膳食对大鼠代谢指标的影响和肝脏脂肪沉积以及肝脏糖脂代谢相关基因表达的影响,并观察绿茶多酚对高脂膳食诱导的代谢改变、肝脏脂肪沉积和糖脂代谢基因表达改变的干预效果。
     方法:体重40-60g断乳雄性Wistar大鼠50只,适应性喂养一周后,分为5组,对照组给予标准饲料,高脂组和三个绿茶多酚干预组给予高脂饲料,实验开始时所有大鼠饮用去离子水,绿茶多酚干预组在达到成年体重后改为饮用不同浓度的绿茶多酚水溶液,第26周进行口服糖耐量试验(OGTT),26周末,处死大鼠,分离血清。用ELISA检测血清中胰岛素的含量;用生化试剂盒检测血糖、血脂水平,计算HOMA-IR指数;精确称量肝脏、睾周脂肪和肾周脂肪的重量,计算内脏脂肪系数和肝脏系数;取部分肝脏组织冰冻切片,进行油红O染色。采用实时荧光定量PCR方法检测肝脏组织中脂肪合成酶(FAS)、羟甲基戊二酸单酰辅酶A还原酶(HMG CoAR)、过氧化物酶体增殖物激活受体α (PPARα)、固醇调节元件结合蛋白-1c(SREBP-1c)、葡萄糖6磷酸酶(G6Pase)、磷酸烯醇式丙酮酸羧激酶(PEPCK)和葡萄糖转运蛋白2(GLUT2)的mRNA表达水平。
     结果:对照组大鼠进食量显著高于其它组,但是各组大鼠能量摄入量没有显著差异。自第9周开始,高脂组大鼠体重显著高于对照组,这种差异持续到实验结束。实验结束时,高脂组大鼠体重和内脏脂肪系数都显著高于对照组,与高脂组相比,绿茶多酚干预组大鼠的体重和内脏脂肪系数都显著降低。与对照组相比,高脂组大鼠空腹血糖水平、空腹胰岛素水平,胰岛素抵抗指数和OGTT试验曲线下面积都显著升高,绿茶多酚干预降低了高脂饲养大鼠的血糖水平,胰岛素抵抗指数和OGTT试验曲线下面积,但是对胰岛素水平没有明显影响。高脂饲养导致了大鼠血脂紊乱,其中血清甘油三酯(TG),低密度脂蛋白胆固醇(LDL-C)和总胆固醇(TC)水平与对照组相比显著升高,高密度脂蛋白胆固醇(HDL-C)水平显著下降,LDL-C/HDL-C值显著上升,绿茶多酚干预降低了高脂饲养大鼠血清TG、TC、LDL-C、LDL-C/HDL-C的水平,但是对HDL-C的水平没有明显影响。高脂饲养引起了大鼠肝脏脂肪沉积,肝脏系数升高,并引起了肝脏中糖脂代谢相关基因的表达改变,其中脂肪合成相关的基因FAS、HMG-CoAR和SREBP-1和糖异生关键限速酶PEPCK和G6Pase的mRNA表达水平显著升高,脂肪酸氧化相关的基因PPARα和肝脏中主要的葡萄糖转运体GLUT2的mRNA表达水平显著降低。绿茶多酚干预降低了高脂饲养大鼠的肝脏系数和肝脏脂肪沉积,并抑制了高脂诱导的大鼠肝脏糖脂代谢基因表达改变。
     结论:高脂饲养诱导大鼠发生代谢紊乱,肝脏脂肪沉积和糖脂代谢相关基因的表达异常,这些改变与能量摄入无关;绿茶多酚干预能够预防和改善高脂诱导的代谢紊乱和肝脏脂肪沉积。
     第二部分绿茶多酚对脂联素水平的影响及细胞信号机制
     目的:绿茶多酚的代谢调节机制到目前为止尚不清楚。脂联素被认为是肥胖和胰岛素抵抗治疗的新靶点。本部分主要观察绿茶多酚对高脂饲养大鼠脂联素水平的影响并探索相关机制。
     方法:动物饲养与干预同第一部分,血清脂联素水平采用ELISA方法检测,用实时荧光定量PCR检测大鼠内脏脂肪组织中脂联素和PPARγ的mRNA表达水平,采用免疫印迹法检测PPARγ,磷酸化PPARγ,细胞外信号调节激酶ERK1/2和磷酸化ERK1/2的蛋白表达水平。采用高糖DMEM培养基体外培养大鼠内脏脂肪组织,并进行绿茶多酚进行干预和ERK1/2的特异性抑制剂PD98059进行预处理。干预24h后,收集培养液,检测其中的脂联素水平,收集培养的内脏脂肪组织检测脂联素和PPARγ的mRNA表达水平以及PPARγ,磷酸化PPARγ,ERK1/2和磷酸化ERK1/2的蛋白表达水平。
     结果:高脂组大鼠脂联素mRNA水平和血清脂联素水平都显著低于对照组,而绿茶多酚干预显著升高了脂联素水平。与对照组相比,高脂组大鼠内脏脂肪中ERK1/2的激活和PPARγ的磷酸化显著升高,而PPARγ的mRNA水平和蛋白表达水平显著降低,与高脂组相比,绿茶多酚干预组ERK1/2和PPARγ磷酸化水平显著降低,而PPARγ表达水平显著升高。采用高糖体外培养内脏脂肪组织也显著降低了内脏脂肪组织中脂联素mRNA水平,分泌到培养液中的脂联素水平和PPARγ的表达水平,而上调了ERK1/2和PPARγ的磷酸化水平。而绿茶多酚干预和ERK1/2抑制剂处理均能抑制高糖培养诱导的这些改变。
     结论:上调脂联素水平是绿茶多酚抑制高脂饮食诱导代谢紊乱的重要机制之一。绿茶多酚通过抑制ERK1/2的激活,降低PPARγ的磷酸化水平和上调PPARγ表达水平来上调脂联素的水平。
     第三部分绿茶多酚对血管内皮高通透性的影响及机制
     目的:观察高脂饲料饲养大鼠主动脉内皮通透性的改变和绿茶多酚的干预效果,并在体内、体外细胞实验中探索绿茶多酚调控通透性改变的机制。
     方法:动物饲养与干预同第一部分,牛主动脉内皮细胞(BAECs)采用高糖培养基培养,分别采用绿茶多酚,二苯基碘(DPI)、SU5416(VEGF受体2抑制剂)、rhVEGF和β环糊精(胞膜窖结构抑制剂)进行干预后,检测单层BAECs通透性。大鼠主动脉内皮通透性采用伊文思蓝注射法测定,单层BAECs通透性利用穿过细胞的异硫氰酸荧光素(FITC)-右旋糖酐测定。采用二氢乙啶(DHE)荧光探针检测大鼠主动脉活性氧(ROS)水平,采用2',7'-二氯荧光素二乙酸酯(DCFH-DA)荧光探针检测细胞中的ROS水平。采用实时定量PCR方法检测VEGF和窖蛋白1的mRNA表达水平,采用ELISA检测大鼠血清和细胞培养液中的VEGF水平,采用蛋白免疫印迹法检测培养的BAECs中的cav-1蛋白表达水平。
     结果:与对照组相比,高脂组大鼠主动脉通透性显著升高,与高脂组相比,绿茶多酚干预组主动脉通透性显著降低。高脂膳食显著上调了大鼠的血清VEGF水平、主动脉中VEGF的mRNA表达水平和主动脉中的ROS水平,而绿茶多酚干预显著抑制了高脂诱导VEGF和ROS水平的升高。采用高糖培养基体外培养BAECs,显著上调了单层BAECs的通透性、VEGF的表达和分泌水平、以及细胞内ROS水平,同时还上调了NADPH氧化酶亚基p22phox和p67phox亚基的表达水平,并诱导了窖蛋白1表达水平升高,而绿茶多酚显著改善了高糖诱导的BAECs通透性、VEGF的表达和分泌水平、ROS、NADPH氧化酶亚基和窖蛋白1表达水平的升高。与高糖培养类似,rhVEGF预处理显著升高了普通培养基培养的单层BAECs的的通透性,而VEGF受体抑制剂SU5416预处理、NADPH氧化酶抑制剂DPI预处理和检测前采用胞膜窖结构抑制剂β环糊精孵育都显著降低了高糖培养单层BAECs的通透性,SU5416预处理还显著降低了窖蛋白1的表达水平。
     结论:高脂膳食能够诱导大鼠主动脉内皮通透性升高,这种效应能够被绿茶多酚有效改善。绿茶多酚调节高脂饲养大鼠内皮通透性的可能信号途径为:下调NADPH氧化酶表达水平,降低ROS产生,进而下调VEGF水平,降低窖蛋白1的表达水平,减少胞膜窖介导的大分子转运,改善内皮高通透性。
Part1The effect of GTPs on high-fat diet induced metabolic disorders
     Objective: The metabolic abnormalities associated with metabolic syndrome includeobesity, hyperglycemia, dyslipidemia, elevated blood pressure and decreased insulinsensitivity. Liver is the biggest and most important organ that regulates metabolism,and fatty liver is considered to be a performance of the metabolic syndrome in liver.This part of the study investigated the effect of high-fat diet on metabolic markers inrats, liver fat deposition and expression of genes related to glucose and lipidmetabolism in the liver, the effects of GTPs on high-fat diet-induced metabolicchanges was focused.
     Methods: Male SPF Wistar rats after weaning and weight between40-60g were used.After acclimation for a week, the rats were randomly divided into the following fivegroups: the control group, fed on standard chow and drank deionized water; the highfat group, fed on high fat diet, drank deionized water; three GTPs treated groups,fed on high fat diet, and the deionized drinking water was replaced with differentconcentrations of tea polyphenols solution (0.8g/L,1.6g/L and3.2g/L) when therats weights over180g. The oral glucose tolerance test (OGTT) was taken at the26thweek, and then the rats were sacrificed. The serum was separated for measurement ofserum insulin levels by ELISA and determination of blood sugar levels and bloodlipid levels by commencial kit. HOMA-IR index was calculated according to theformula. The liver, epididymal fat and perirenal fat weight, were accurately weighedand the visceral fat coefficient and liver coefficient were calculated. The fatdeposition in liver was measured by oil red O staining on frozen sections. Real-timequantitative PCR was used to detect the mRNA expression of fatty acid synthase(FAS),3-hydroxy-3-methylglutaryl-CoA reductase (HMGCoAR), peroxisomeproliferator activated receptor α,(PPARα), sterol regulatory element bindingprotein-1c (SREBP-1c), glucose-6-phosphatase (G6Pase), phosphoenolpyruvatecarboxykinase (PEPCK) and glucose transporter2(GLUT2) in the liver..
     Results: The food intake of the control group is higher than that of the other groups;however, the energy intake is equal in all groups due to the different energy density ofthe standard chow and the high fat chow. From the9th week on, the weight of high fatfed rats was significantly higher than that of the control group. At the end of theexperiment, body weight and visceral fat coefficients of high fat fed rats weresignificantly higher than that of the control group. Compared with the high-fat group,the body weight and visceral fat coefficients were significantly reduced in GTPs treated groups. High fat fed rats exhibited significantly higher blood glucose levelsthan the control group; the blood glucose of GTPs treated groups was significantlylower than that of the high-fat group. The glucose tolerance in high-fat rats was lowerand the insulin HOMA-IR index was higher than that in the control group, and GTPsintervention improved the glucose tolerance and down-regulated the HOMA-IR indexin high-fat fed rats. The serum insulin level in high-fat group is significantly higherthan that in the control group, GTPs treatment didn’t change the insulin level in highfat fed rats. High fat diet induced dyslipidemia in rats. Compare to the control group,the serum triglyceride (TG), low density lipoprotein cholesterol (LDL-C) and totalcholesterol (TC) level was significantly increased. The serum high-density lipoproteincholesterol (HDL-C) was significantly decreased, and the LDL-C/HDL-C rationincreased significantly, all these alterations except for HDL-C levels were alleviatedby GTPs treatment to different extents. In the liver, high-fat diet induced fatdeposition, increased mRNA expression of genes related to fat synthesis (the FAS,HMG-CoAR and SREBP-1), and reduced mRNA expression of fatty acid oxidationgene, PPARα. The alterations in fat deposition and expression of genes were allalleviated by GTPs treatment. High-fat diet also significantly raised the mRNAexpression of the key and rate-limiting enzymes of gluconeogenesis (PEPCK andG6Pase), and reduced the mRNA expression of the major glucose transporter in theliver (GLUT2), these alterations in hepatic glucose metabolism gene expressionsinduced by high fat diet were also mitigated by GTPs.
     Conclusion: The high-fat diet induced metabolic syndrome, fat deposition in the liverand dysregulated the hepatic mRNA expression of genes related to fat and glucosemetabolism in rats, these changes are irrelative to total energy intake. GTPs alleviatedthe high-fat diet-induced metabolic syndrome, fat deposition in the liver and abnormalhepatic mRNA expressions of glucose and lipid metabolism related genes.
     Part2The Effect and cell signaling mechanism of GTPs on adiponectinproduction
     Objective: The mechanism underlying the metabolism regulating effects of GTPsremains unclear. Hypoadiponectinemia plays an important role in the development ofobesity, metabolic syndrome and related diseases, and adiponectin had beenconsidered to be a new therapeutic target for treatment of obesity and insulinresistance. In this section we observed the effects of GTPs on adiponectin levels inhigh-fat fed rats and explored the possible mechanism.
     Methods: Animal management was stated in part1. The serum adiponectin levelswere determined by ELISA, The mRNA expression of adiponectin and PPARγ invisceral fat of the rats were determined by real-time quantitative PCR. The proteinexpression of PPARγ, the phosphorylation of PPARγ, and protein expressionextracellular signal-regulated kinase extracellular signal regulated kinase (ERK)1/2and phosphorylated ERK1/2levels were detected by Western blot. Rat visceraladipose tissue were cultured in vitro with high glucose DMEM medium, and treated with GTPs or pretreated with PD98059, a specific inhibitor of ERK1/2pathway. After24hours intervention, the culture medium was collected and the adiponectin levels init were determined by ELISA, the cultured visceral adipose tissue were collected andthe mRNA expression of adiponectin and PPARγand protein expressions PPARγ,phosphorylated PPARγ, ERK1/2and phosphorylated ERK1/2were determined byreal-time quantitative PCR and western blot respectively.
     Results: The adiponectin mRNA levels and serum adiponectin level was significantlylower in the high fat group than those in the control group, and GTPs treatmentsignificantly mitigated the decrease in adiponectin in high fat fed rats. Compared withthe control group, the ERK1/2activation and PPARγ phosphorylation in the visceralfat were increased significantly. Meanwhile, the PPARγ mRNA levels and proteinlevels were decreased significantly. Compared with the high-fat group, the activationof ERK1/2and PPARγ phosphorylation were significantly lower, and PPARγexpression levels were significantly increased. High glucose cultured visceral adiposetissue also exhibited significantly reduced mRNA and secretion levels of adiponectin,up-regulated ERK1/2and PPARγ phosphorylation levels and decreased PPARγexpression levels. GTPs intervention and ERK1/2inhibitor pretreatment bothinhibited the ERK1/2activation, PPARγphosphorylation, and raised the PPARγexpression and adiponectin expression and secretion in high glucose cultured ratvisceral adipose tissue.
     Conclusion: The regulation of adiponectin levels is an important mechanismunderlying the preventive and mitigative effects of GTPs on high-fat diet-inducedmetabolic disorders. GTPs increase the adiponectin level by inhibition of ERK1/2activation, decrease the PPARγ phosphorylation and increase the level of PPARγexpression.
     Part3The effects and mechanism of GTPs on vascular endothelialhyperpermeability
     Objective: Atherosclerotic cardiovascular diseases, which make biggest contributionto morbidity and mortality of cardiovascular diseases, are metabolic syndromerelated diseases. Endothelial hyperpermeability, referring to increased transport oflarge molecules including AGE and lipoproteins to the subendothelial space, is theprimary reaction of endothelium to damage factors like hyperglycemia orhyperlipidemia, an early marker and one of the major performances of endothelialdysfunction, and the primary change in the development of atherosclerosis. Theobjective of this part is to observe the effect of high-fat diet on endothelialpermeability in rat aorta with the focus on effects of GTPs intervention, and toexplore the mechanisms underneath both in vivo and in vitro.
     Methods: Animal management and intervention were same with part1, bovine aorticendothelial cells (BAECs) were cultured with high glucose culture medium, andtreated with GTPs (0.4, or4μg/mL.24h), or pretreated with DPI (10μM) or SU5416(10μM) pre-incubated for30min or incubated with rhVEGF (8ng/mL), beta cyclodextran intervention (8μM) for30min before the test of permeability inmonolayer bovine aortic endothelial cells. The endothelial permeability in rat aorticwas measured using Evans blue injection, and the permeability in monolayer bovineaortic endothelial cells was measured by the fluorescein isothiocyanate (FITC)-glucan assay. Dihydro-ethidium (DHE) fluorescent probe was used to detect thereactive oxygen species (ROS) level in rat aortic, the2',7'-dichloro-fluoresceindiacetate (DCFH-DA) fluorescent probe to detect ROS levels in cultured BAECs. Thereal-time quantitative PCR was used to detect the mRNA levels of vascularendothelial growth factor (VEGF) and caveolin-1. VEGF levels in serum and cellculture medium were measured by ELISA.Western blot assay was used to detect theprotein levels of cav-1, p22phox and p67phox in cultured BAECs.
     Results: Compared with the control group, the high fat diet increased the endothelialpermeability in rat aorta, comparing with the high-fat group, the permeability in aortaof GTPs treated groups were significantly lower. Meanwhile, high-fat dietsignificantly increased the ROS level and the mRNA level of vascular endothelialgrowth factor (VEGF), a strong permeability factor, in the aorta and upregulated theserum level of VEGF, GTPs treatment significantly reduced the high fat diet inducedincrease in VEGF and ROS. In high glucose cultured BAECs, the endothelialpermeability, VEGF expression and secretion, ROS level were all increased. Highglucose culture also increase the expression of p22phox and p67phox subunits ofNADPH oxidase and the expression of caveolin-1, GTPs mitigated the elevatedVEGF expression and secretion, ROS increase, up-regulated expression of p22phox,p67phox and caveolin-1induced by high glucose incubation. Similar to high glucoseculture, rhVEGF treatment increased the endothelial permeability in monolayerBAECs cultured in normal DMEM. Preincubation with SU5416(VEGF receptorinhibitor), DPI (NADPH oxidase inhibitor) or treatment with β cyclodextran(structural inhibitor of caveolae) all down-regulated the endothelial permeability inhigh glucose cultured monolayer BAECs. SU5416also down-regulated the cavelin-1expression in BAECs cultured in high glucose DMEM.
     Conclusion: High fat diet could induce endothelial hyperpermeability in rat aorta, andthis variation could be mitigated by GTPs treatment. GTPs regulated the endothelialpermeability in high fat rats by down-regulating NADPH oxidase, ROS production,thus decreased the VEGF levels and the expression of cav-1, which induceddown-regulation of caveolae mediated large molecule transport and alleviate theendothelial hyperpermeability.
引文
[1] Sargin M, Uygur-Bayramicli O, Sargin H, et al. Association of nonalcoholicfatty liver disease with insulin resistance: is OGTT indicated in nonalcoholicfatty liver disease? J Clin Gastroenterol,2003,37:399~402
    [2] Ford ES. Prevalence of the metabolic syndrome defined by the internationaldiabetes federation among adults in the U.S. Diabetes Care,2005,28:2745~2749
    [3] Ervin RB. Prevalence of metabolic syndrome among adults20years of age andover, by sex, age, race and ethnicity, and body mass index: Unitedstates,2003~2006. Natl Health Stat Rep,2009:1~7
    [4] Lakka HM, Laaksonen DE, latkka TA, et al. The metabolic syndrome and totaland cardiovascular disease mortality in middle-aged men. JAMA,2002,288(21):2709~2716
    [5] Asia Pacific Cohort Studies Collaboration. The burden of overweight and obesityin the Asia-Pacific region. Obes Rev,2007,8(3):191~196
    [6] Damjanovic M, Barton M. Curr Fat intake and cardiovascular response. HypertensRep.2008,10(1):25~31
    [7] Mundy AL, Haas E, Bhattacharya I, et al. Fat intake modifies vascularresponsiveness and receptor expression of vasoconstrictors: implications fordiet-induced obesity.Cardiovasc Res,2007,73(2):368~375
    [8] Pories WJ. Bariatric surgery: risks and rewards. J Clin Endocrinol Metab,2008,93:S89~96.
    [9] Kennedy MJ, Jellerson KD, Snow MZ, et al. Challenges in the PharmacologicManagement of Obesity and Secondary Dyslipidemia in Children andAdolescents. Paediatr Drugs.2013,[Epub ahead of print]
    [10] Pories WJ. Bariatric surgery: risks and rewards. J Clin Endocrinol Metab,2008,93: S89~96
    [11] Filippatos TD, Derdemezis CS, Gazi IF, et al. Orlistat-associated adverse effectsand drug interactions: a critical review. Drug Saf,2008,31:53~65
    [12] Daskalopoulou SS, Mikhailidis DP, Elisaf M. Prevention and treatment of themetabolic syndrome. Angiology,2004,55:589~612
    [13] Balentine DA, Wiseman SA, Bouwens LC. The chemistry of tea flavonoids. CritRev Food Sci Nutr,1997,37:693~704
    [14] Park HJ, Dinatale DA, Chung MY, et al. Green tea extract attenuates hepaticsteatosis by decreasing adipose lipogenesis and enhancing hepatic antioxidantdefenses in ob/ob mice. J Nutr Biochem,2011,22(4):393~400
    [15] Ikeda I, Hamamoto R, Uzu K, et al. Dietary gallate esters of tea catechins reducedeposition of visceral fat, hepatic triacylglycerol, and activities of hepaticenzymes related to fatty acid synthesis in rats. Biosci Biotechnol Biochem,2005,69:1049~1053
    [16] Murase T, Nagasawa A, Suzuki J, et al. Beneficial effects of tea catechins ondiet-induced obesity: stimulation of lipid catabolism in the liver. Int J Obes RelatMetab Disord,2002,26:1459~1464
    [17] Matsuzawa Y, Funahashi T, Kihara S, et al. Adiponectin and metabolic syndrome.Arterioscler Thromb Vasc Biol,2004,24(1):29~33
    [18] Oberthuer A, D nmez F, Oberh user F, et al. Hypoadiponectinemia in extremelylow gestational age newborns with severe hyperglycemia-a matched-pairedanalysis. PLoS One,2012,7(6): e38481
    [19] Kumada M, Kihara S, Sumitsuji S, et al. Association of hypoadiponectinemiawith coronary artery disease in men. Arterioscler Thromb Vasc Biol,2003,23(1):85~89
    [20] Baratta R, Amato S, Degano C, et al. Adiponectin Relationship with LipidMetabolism Is Independent of Body Fat Mass: Evidence from BothCross-Sectional and Intervention Studies. J Clin Endocrinol Metab,2004,89(6):2665~2671
    [21] Hotta K, Funahashi T, Arita Y, et al. Plasama concentration of a novel,adipose-specific protein, adiponectin, in type2diabetic patients. J ClicEndocrinol Metab,2001,86:1930~1935
    [22] Marinou K, Tousoulis D, Antonopoulos AS, et al.Obesity and cardiovasculardisease: from pathophysiology to risk stratification.Int J Cardiol,2010,138(1):3~8
    [23] Fruebis J, Tsao TS, Javorschi S, et al. Proteolytic cleavage product of30-kDaadipocyte complement-related protein increases fatty acid oxidation in muscleand causes weight loss in mice. Proc Natl Acad Sci U S A,2001,98(4):2005~2010
    [24] Yamauchi T, Kamon J, Waki H, et al. The fat-derived hormone adiponectinreverses insulin resistance associated with both lipoatrophy and obesity. NatMed,2001,7(8):941~946
    [25] Kubota N, Teranchi Y, Yamanchi T, et al. Disruption of adiponectin causesinsulin resistance and neointimal formation. J Biol Chem,2002,277(29):25863~25866
    [26] Gilardini L, McTernan PG, Girola A, et al. Adiponectin is a candidate marker ofmetabolic syndrome in obese children and adolescents.Atherosclerosis,2006,189(2):401~407
    [27] Kamoda T, Saitoh H, Saito M, et al. Serum adiponectin concentrations innewborn infants in early postnatal life. Pediatr Res,2004,56:690~693
    [28] Retnakaran R, Zinman B, Connelly PW, et al. Nontraditional cardiovascular riskfactors in pediatric metabolic syndrome. J Pediatr,2006,148:149~151
    [29] Shulman, G.I. Cellular mechanisms of insulin resistance. J Clin Invest,2000,106(2):171~176
    [30] Kadowaki T and Yamauchi T. Adiponectin and adiponectin receptors. Endocrinereviews,2005,26(3):439~451
    [31] Kaser S, Moschen A, Cayon A, et al. Adiponectin and its receptors innon-alcoholic steatohepatitis.Gut,2005,54(1):117~121
    [32] De Clercq V, Stringer D, Hunt R, et al. Adipokine Production by Adipose Tissue:A novel target for treating metabolic syndrome and its sequelae. In: Wang M(ed). Metabolic Syndrome: Underlying mechanisms and drug therapies.Hoboken, NJ: John Wiley&Sons, Inc.2011,73~131
    [33] SundaramR, Naresh R, Shanthi P, et al. Modulatory effect of green tea ectract onhepatic key enzymes of glucose metabolism in streptozotocin and high fat dietinduced diabetic rats. Phytomedicine,2013,20(7):577~584
    [34] Venables MC, Hulston CJ, Cox HR,et al.Green tea extract ingestion, fatoxidation, and glucose tolerance in healthy humans.Am J Clin Nutr,2008,87(3):778~784
    [35] Potenza MA, Marasciulo FL, Tarquinio M, et al. EGCG, a green tea polyphenol,improves endothelial function and insulin sensitivity, reduces blood pressure,and protects against myocardial I/R injury in SHR. Am J Physiol EndocrinolMetab,2007,292(5):E1378~1387
    [36] Cuevas AM, Guasch V, Castillo O, et al. A high-fat diet induces and red winecounteracts endothelial dysfunction in human volunteers. Lipids,2000,35(2):143~148
    [37] Cotez-Pinto HH, Camilo ME, Baptista AA, et al. Non alcoholic fatty liver:Anather feature of the metabolic syndrome? Clin Nutr,1999,18(6):353~358
    [38] Livak KJ, Schmittgen TD. Analysis of relative gene expression data usingreal-time quantitative PCR and the2(-Delta Delta C(T)) Method. Methods,2001,25:402~408
    [39] Enomoto M, Adachi H, Hirai Y, et al. LDL-C/HDL-C Ratio Predicts CarotidIntima-Media Thickness Progression Better Than HDL-C or LDL-C Alone. JLipids,2011,2011:549137
    [40] Kawamoto R, Tabara Y, Kohara K,et al. Low-density lipoprotein cholesterol tohigh-density lipoprotein cholesterol ratio is the best surrogate marker for insulinresistance in non-obese Japanese adults. Lipids Health Dis,2010,7(9):138
    [41] Kuriyama S, Shimazu T, Ohmori K, et al. Green tea consumption and mortalitydue to cardiovascular disease, cancer, and all causes in Japan. JAMA,2006,296(10):1255~1265
    [42] Bose M, Lambert JD, Ju J, et al. The major green tea polyphenol,(-)–epigallocatechin-3-gallate, inhibits obesity, metabolic syndrome, and fatty liverdisease in high-fat–fed mice. J Nutr,2008,138(9):1677~1683
    [43] Brown AL, Lane J, Holyoak C, et al. Health effects of green tea catechins inoverweight and obese men: a randomised controlled cross-over trial. Br J Nutr.,2011,106(12):1880~1889
    [44] Nascimento AR, Machado M, de Jesus N,et al. Structural and functionalmicrovascular alterations in a rat model of metabolic syndrome induced by ahigh-fat diet. Obesity (Silver Spring),2013[Epub ahead of print]
    [45] Bogdanski P, Suliburska J, Szulinska M, et al. Green tea extract reduces bloodpressure, inflammatory biomarkers, and oxidative stress and improvesparameters associated with insulin resistance in obese, hypertensivepatients.Nutr Res,2012,32(6):421~427
    [46] Ihm SH, Jang SW, Kim OR, et al. Decaffeinated green tea extract improveshypertension and insulin resistance in a rat model of metabolicsyndrome.Atherosclerosis,2012,224(2):377~383
    [47] Zheng XX, Xu YL, Li SH, et al. Effects of green tea catechins with or withoutcaffeine on glycemic control in adults: a meta-analysis of randomized controlledtrials.Am J Clin Nutr,2013,97(4):750~762
    [48] Suliburska J, Bogdanski P, Szulinska M, et al. Effects of green teasupplementation on elements, total antioxidants, lipids, and glucose values in theserum of obese patients. Biol Trace Elem Res,2012,149(3):315~322
    [49] Anandh Babu PV, Sabitha KE, Shyamaladevi CS. Green tea extract impedesdyslipidaemia and development of cardiac dysfunction in streptozotocin-diabeticrats.Clin Exp Pharmacol Physiol,2006,33(12):1184~1189
    [50] Maron DJ, Lu GP, Cai NS et al. Cholesterol-lowering effect of atheaflavin-enriched green tea extract: a randomized controlled trial.Arch InternMed,2003,163(12):1448~1453
    [51] Biddinger SB, Kahn CR. From mice to men: insights into the insulin resistancesyndromes. Annu Rev Physiol,2006,68:123~158
    [52] Smith S, Witkowski A, Joshi AK. Structural and functional organization of theanimal fatty acid synthase. Prog lipid res,2003,42(4):289~317
    [53] Berk PD. Regulatable fatty acid transport mechanisms are central to thepathophysiology of obesity, fatty liver, and metabolic syndrome. Hepatology,2008,48(5):1362~1376
    [54] Francis GA, Fayard E, Picard F, et al. Nuclear receptors and the control ofmetabolism. Annu rev physiol,2003,65(1):261~311
    [55] Roglans N, Vila L, Farre M, et al. Impairment of hepatic Stat-3activation andreduction of PPARalpha activity in fructose-fed rats. Hepatology,2007,45(3):778~788
    [56] Shrestha S, Ehlers SJ, Lee JY, et al. Dietary Green Tea Extract Lowers Plasmaand Hepatic Triglycerides and Decreases the Expression of Sterol RegulatoryElement-Binding Protein-1c mRNA and Its Responsive Genes in Fructose-Fed,Ovariectomized Rats. J Nutr,2009,139(4):640~645
    [57] Coleman RA, Lee DP. Enzymes of triacylglycerol synthesis and their regulation.Prog lipid res,2004,43(2):134~176
    [58] Jin ES, Beddow SA, Malloy CR, et al. Hepatic glucose production pathwaysafter three days of a high-fat diet. Metabolism,2013,62(1):152~162
    [59] Yasui K, Miyoshi N, Tababe H, et al. Effects of oolong tea on gene expression ofgluconeogenic enzymes in the mouse liver and in rat hepatoma H4IIE cells. JMed Food,2011,14(9):930~938
    [60] Cao H, Hininger-Favier I, Kelly MA, et al. Green tea polyphenol extractregulates the expression of genes involved in glucose uptake and insulinsignaling in rats fed a high fructose diet. J Agric Food Chem,2007,55(15):6372~6378
    [61] Renaldi O, Pramono B, Sinorita H, et al. Hypoadiponectinemia: a risk factor formetabolic syndrome. Acta Med Indones,2009,41(1):20~24
    [62] Lara-Castro C, Fu Y, Chung BH, et al. Adiponectin and the metabolic syndrome:mechanisms mediating risk for metabolic and cardiovascular disease. Curr. Opin.Lipidol,2007,18(3):263~270
    [63] Diez JJ, Iglesias P. The role of the novel adipocyte-derived hormone adiponectinin human disease. Eur J Endocrinol,2003,148(3):293~300
    [64] Ahima RS. Adipose tissue as an endocrine organ. Obesity,2006,14:242S~249S
    [65] Combs TP, Berg AH, Obici S, et al. Endogenous glucose production is inhibitedby the adipose-derived protein Acrp30. J Clin Invest,2001,108:1875~1881
    [66] Yamauchi T, Kamon J, Waki H, et al. The fat-derived hormone adiponectinreverses insulin resistance associated with both lipoatrophy and obesity. NatMed,2001,7(8):941~946
    [67] Shimada M, Mochizuki K, Sakurai N, et al. Dietary supplementation withepigallocatechin gallate elevates levels of circulating adiponectin in non-obesetype-2diabetic Goto-Kakizaki rats. Biosci Biotechnol Biochem,2007,71(8):2079~2082
    [68] Iwaki M, Matsuda M, Maeda N, et al. Induction of adiponectin, a fat-derivedantidiabetic and antiatherogenic factor, by nuclear receptors. Diabetes,2003,52(7):1655~1663
    [69] Yang WS, Jeng CY, Wu TJ, et al. Synthetic peroxisome proliferation activatedreceptor γ agonist, rosiglitone, increase plasma levels of adiponectin in type2diabetes patients. Diabetes Care,2002,25(2):376~380
    [70] Combs TP, Wagner JA, Berger J, et al. Induction of adipocyte complementrelated protein of30kilodaltons by PPARγ agonists: a potential mechanism ofinsulin sensitization. Endocrinology,2002,143(3):998~1007
    [71] Park KS, Ciaraldi TP, Abrams-Carter L, et al. PPAR-gamma gene expression iselevated in skeletal muscle of obese and type II diabetic subjects. Diabetes,1997,46:1230~1234
    [72] Inoue M, Ohtake T, Motomura W, et al. Increased expression of PPARgamma inhigh fat diet-induced liver steatosis in mice.Biochem Biophys Res Commun,2005,336(1):215~222
    [73] Gayet C, Lerayl V, Saito M, et al. The effects of obesity-associated insulinresistance on mRNA expression of peroxisome proliferator-activated receptor-gtarget genes, in dogs. Br J Nutr,2007,98:497~503
    [74] Liu LR, Lin SP, Chen CC, et al. Serum Amyloid A Induces Lipolysis byDownregulating Perilipin Through ERK1/2and PKA Signaling Pathways.Obesity,2011,19:2301~2309
    [75] Kaplan JM, Hake PW, Denenberg A, et al. Phosphorylation of ExtracellularSignal-Regulated Kinase(ERK)-1/2Is Associated with the Downregulation ofPeroxisome Proliferator–Activated Receptor (PPAR)-γ during PolymicrobialSepsis. Mol Med,2010,16(11-12):491~497
    [76] Adams M, Reginato M.J, Shao D, et al.Transcriptional Activation by PeroxisomeProliferator-activated Receptor g Is Inhibited by Phosphorylation at a ConsensusMitogen-activated Protein Kinase Site. J Bio Chem,1997,272(8):5128~5132
    [77] Kursawe R, Narayan D, Cali AM, et al. Downregulation of ADIPOQ andPPARgamma2gene expression in subcutaneous adipose tissue of obeseadolescents with hepatic steatosis. Obesity (Silver Spring),2010,18(10):1911~1917
    [78] Catalano PM, Nizielski SE, Shao J, et al. Downregulated IRS-1andPPARgamma in obese women with gestational diabetes: relationship to FFAduring pregnancy. Am J Physiol Endocrinol Metab,2002,282(3): E522~533
    [79] Matsuzawa Y. Adiponectin: a key player in obesity related disorders.Curr PharmDes,2010,16(17):1896~1901
    [80] Christa B, Josef W, and Markus N. Adiponectin, a key adipokine in obesityrelated liver diseases.World J Gastroenterol,2010,17(23):2801~2811
    [81] Toshimitsu Y, Shoichiro N, Ataru T, et al. Hypoadiponectinemia is associatedwith visceral fat accumulation and insulin resistance in Japanese men with type2diabetes mellitus. Metabolism,2003,52(10):1274~1278
    [82] Weyer C, Funahashi T, Tanaka S, et al. Hypoadiponectinemia in obesity and type2diabetes: close association with insulin resistance and hyperinsulinemia. J ClinEndocrinol Metab,2001,86(5):1930~1935
    [83] Milan G, Granzotto M, Scarda A, et al.Resistin and adiponectin expression invisceral fat of obese rats: effect of weight loss. Obes Res,2002,10(11):1095~1103
    [84] Panagopoulou P, Stamna E, Tsolkas G, et al. Adiponectin gene polymorphisms inobese Greek youth. Pediatr Endocrinol Metab,2009,22(10):955~959
    [85] González-Sánchez JL, Zabena CA, Martínez-Larrad MT, et al. An SNP in theadiponectin gene is associated with decreased serum adiponectin levels and riskfor impaired glucose tolerance.Obes Res,2005,13(5):807~812
    [86] Dastani Z, Hivert MF, Timpson N, et al. Novel loci for adiponectin levels andtheir influence on type2diabetes and metabolic traits: a multi-ethnicmeta-analysis of45,891individuals.PLoS Genet,2012,8(3):e1002607
    [87] Mousavinasab F, Tahtinen T, Jokelainen J, et al., Common polymorphisms(single-nucleotide polymorphisms SNP+45and SNP+276) of the adiponectingene regulate serum adiponectin concentrations and blood pressure in youngFinnish men. Mol Genet Metab,2006,87(2):147~151
    [88] Qi Y, Takahashi N, Hileman SM, et al. Adiponectin acts in the brain to decreasebody weight. Nat Med,2004,10(5):524~529
    [89] Kasim-Karakas SE, Tsodikov A, Singh U, et al. Responses of inflammatorymarkers to a low-fat, high-carbohydrate diet: effects of energy intake. Am J ClinNutr,2006,83(4):774~779
    [90] Qi L, Rimm E, Liu S, et al. Dietary glycemic index, glycemic load, cereal fiber,and plasma adiponectin concentration in diabetic men.Diabetes Care,2005,28(5):1022~1028
    [91] Combs TP, Berg AH, Rajala MW, et al. Sexual differentiation, pregnancy, calorierestriction, and aging affect the adipocyte-specific secretory protein adiponectin.Diabetes,2003,52(2):268~276
    [92] Escrivá F, Gavete ML, Fermín Y, et al. Effect of age and moderate foodrestriction on insulin sensitivity in Wistar rats: role of adiposity. J Endocrinol,2007,194(1):131~141
    [93] Kriketos AD, Gan SK, Poynten AM, et al. Exercise increases adiponectin levelsand insulin sensitivity in humans. Diabetes Care,2004,27(2):629~630
    [94] Zeng Q, Fu L, Takekoshi K,et al. Effects of short-term exercise on adiponectinand adiponectin receptor levels in rats. J Atheroscler Thromb,2007,14(5):261~265
    [95] Hsu CH, Tsai TH, Kao YH, et al. Effect of green tea extract on obese women: Arandomized, double-blind, placebo-controlled clinical trial. Clin Nutr,2008,27(3):363~370
    [96] Sharabi Y, Oron-Herman M, Kamari Y, et al. Effect of PPAR-gamma agonist onadiponectin levels in the metabolic syndrome: lessons from the high fructose fedrat model. Am J Hypertens,2007,20(2):206~210
    [97] Yamamoto Y, Hirose H, Miyashita K, et al. PPAR gamma2gene Pro12Alapolymorphism may influence serum level of an adipocyte-derived protein,adiponectin, in the Japanese population. Metabolism,2002,51(11):1407~1409
    [98] Catalano1PM, Nizielski SE, Shao J, et al. Downregulated IRS-1and PPARγ inobese women with gestational diabetes: relationship to FFA during pregnancy.AJP–Endo,2002,282(3): E522~533
    [99] Choi JH, Banks AS, Estall JL, et al. Anti-diabetic drugs inhibit obesity-linkedphosphorylation of PPARgamma by Cdk5. Nature,2010,466(7305):451~456
    [100] Ohshima T, Koga H, Shimotohno K. Transcriptional activity of peroxisomeproliferator-activated receptor gamma is modulated by SUMO-1modification. JBiol Chem,2004,279(28):29551~29557
    [101] Schupp M, Clemenz M, Gineste R, et al. Molecular characterization of newselective peroxisome proliferator-activated receptor gamma modulators withangiotensin receptor blocking activity. Diabetes,2005,54(12):3442~3452
    [102] Rangwala SM, Rhoades B, Shapiro JS, et al. Genetic modulation ofPPARgamma phosphorylation regulates insulin sensitivity. Dev Cell,2003,5(4):657~663
    [103] Burgermeister E, Seger R. MAPK Kinases as Nucleo-Cytoplasmic Shuttles forPPARγ. Cell Cycle,2007,6(13):1539~1548
    [104] Burns KA, Vanden Heuvel JP. Modulation of PPAR activity via phosphorylation.Biochim Biophys Acta,2007,1771(8):952~960
    [105] Hosooka T, Noguchi T, Kotani K, et al. Dok1mediates high-fat diet-inducedadipocyte hypertrophy and obesity through modulation of PPAR-gammaphosphorylation. Nat Med,2008,14(2):188~193
    [106] Herouvi D, Karanasios E, Karayianni C, et al. Cardiovascular disease inchildhood: the role of obesity. Eur J Pediatr.2013.[Epub ahead of print]
    [107] Mundy AL, Haas E, Bhattacharya I, et al. Fat intake modifies vascularresponsiveness and receptor expression of vasoconstrictors: implications fordiet-induced obesity.Cardiovasc Res,2007,73(2):368~375
    [108] Komarova Y, Malik AB. Regulation of endothelial permeability via paracellularand transcellular transport pathways. Annu Rev Physiol,2010,72:463~493
    [109] Sun SW, Zu XY, Tuo QH, et al. Caveolae and caveolin-1mediate endocytosisand transcytosis of oxidized low density lipoprotein in endothelial cells. ActaPharmacol Sin,2010,31(10):1336~1342
    [110] Fernandez-Hernando C, Yu J, Davalos A, et al. Endothelial-specificoverexpression of caveolin-1accelerates atherosclerosis in apolipoproteinE-deficient mice. Am J Pathol,2010,177(2):998~1003
    [111] Fernandez-Hernando C, Yu J, Suarez Y, Rahner C, et al. Genetic evidencesupporting a critical role of endothelial caveolin-1during the progression ofatherosclerosis. Cell Metab,2009,10(1):48~54
    [112] Kim NH, Jung HH, Cha DR, et al. Expression of vascular endothelial growthfactor in response to high glucose in rat mesangial cells. J Endocrinol,2000,165(3):617~624
    [113] Cukiernik M, Hileeto D, Evans T, et al. Vascular endothelial growth factor indiabetes induced early retinal abnormalities. Diabetes Res Clin Pract,2004,65(3):197~208
    [114] Feng Y, Venema VJ, Venema RC, et al. VEGF-induced permeability increase ismediated by caveolae. Invest Ophthalmol Vis Sci,1999,40(1):157~167
    [115] Zhao LN, Yang ZH, Liu YH, et al. Vascular endothelial growth factor increasespermeability of the blood-tumor barrier via caveolae-mediated transcellularpathway. J Mol Neurosci,2011,44(2):122~129
    [116] Moon EJ, Sonveaux P, Porporato PE, et al. NADPH oxidase-mediated reactiveoxygen species production activates hypoxia-inducible factor-1(HIF-1) via theERK pathway after hyperthermia treatment. Proc Natl Acad Sci U S A,2010,107(47):20477~20482.
    [117] Maura C, Giuseppina C, Elisa G, et al. Time-Dependent Stabilization ofHypoxia Inducible Factor-1α by Different Intracellular Sources of ReactiveOxygen Species. PLoS One,2012,7(10): e38388
    [118] Meng D, Mei A, Liu J,et al. NADPH oxidase4mediates insulin-stimulatedHIF-1α and VEGF expression, and angiogenesis in vitro. PLoS One,2012,7(10):e48393
    [119] Hood ED, Greineder CF, Dodia C, et al. Antioxidant protection byPECAM-targeted delivery of a novel NADPH-oxidase inhibitor to theendothelium in vitro and in vivo. J Control Release,2012,163(2):161-169
    [120] Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxidases:physiology and pathophysiology. Physiol Rev,2007,87(1):245~313
    [121] Pavlides S, Gutierrez-Pajares JL, Danilo C, et al. Atherosclerosis, caveolae andcaveolin-1.Adv Exp Med Biol,2012,729:127~144
    [122] Costa RR, Villela NR, Souza Md, et al. High fat diet induces central obesity,insulin resistance and microvascular dysfunction in hamsters. Microvasc Res.2011,82(3):416~422
    [123] Minshall RD, Tiruppathi C, Vogel SM, et al. Endothelial cell-surface gp60activates vesicle formation and trafficking via G(i)-coupled Src kinase signalingpathway. J Cell Biol,2000,150:1057~1070
    [124] Fernandez-Hernando C, Yu J, Suarez Y, et al, Genetic evidence supporting acritical role of endothelial caveolin-1during the progression of atherosclerosis.Cell Metab,2009,10(1):48~54
    [125] DeMaio L, Antonetti DA, Scaduto RC Jr, et al. VEGF increases paracellulartransport without altering the solvent-drag reflection coefficient. Microvasc Res,2004,68(3):295~302
    [126] Argaw AT, Gurfein BT, Zhang Y, et al. VEGF-mediated disruption ofendothelial CLN-5promotes blood-brain barrier breakdown. Proc Natl Acad SciU S A,2009,106(6):1977~1982
    [127] Chen J, Braet F, Brodsky S, et al. VEGF-induced mobilization of caveolaeand increase in permeability of endothelial cells. Am J Physiol CellPhysiol,2002,282(5):C1053~1063
    [128] Damjanovic M, Barton M. Curr Fat intake and cardiovascular response.Hypertens Rep,2008,10(1):25~31
    [129] Han J, Shuvaev VV, Muzykantov VR. Catalase and superoxide dismutaseconjugated with platelet-endothelial cell adhesion molecule antibody distinctlyalleviate abnormal endothelial permeability caused by exogenous reactiveoxygen species and vascular endothelial growth factor.J Pharmacol Exp Ther,2011,338(1):82~91
    [130] Guzik TJ, West NE, Black E, et al. Vascular superoxide production byNAD(P)H oxidase: association with endothelial dysfunction and clinical riskfactors. Circ Res,2000,86: E85~90
    [131] Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxidases:physiology and pathophysiology. Physiol Rev,2007,87:245~313
    [132] Soccio M, Toniato E, Evangelista V, et al. Oxidative stressand cardiovascularrisk: the role of vascular NAD(P)H oxidase and its genetic variants. Eur J ClinInvest,2005,35:305~314
    [133] Talior I, Tennenbaum T, Kuroki T,et al.PKC-delta-dependent activation ofoxidative stress in adipocytes of obese and insulin-resistant mice: role forNADPH oxidase.Am J Physiol Endocrinol Metab,2005,288(2):E405~411
    [134] Xi G, Shen X, Maile LA, et al. Hyperglycemia enhances IGF-I-stimulated Srcactivation via increasing Nox4-derived reactive oxygen species in aPKCζ-dependent manner in vascular smooth muscle cells.Diabetes,2012,61(1):104~113
    [135] Ying CJ, Xu JW, Ikeda K, et al. Tea polyphenols regulate nicotinamide adeninedinucleotide phosphate oxidase subunit expression and ameliorate angiotensinII-induced hyperpermeability in endothelial cells. Hypertens Res,2003,26(10):823~828
    [1]王梅,刘克军,王德江,等.中国脑出血疾病的直接费用负担现状及其问题.中国卫生经济,2005,7:43~46
    [2] Chen CM. Overview of obesity in Mainland China. Obes Rev,2005,9(Suppl1):14~21
    [3]陈春明,孔灵芝,闻芝梅,等.中国成人超重和肥胖症预防与控制指南.第1版.北京:人民卫生出版社,2006:9~14
    [4] Flegal KM, Carroll MD, Ogden CL, et al. Prevalence and trends in obesityamong US adults,1999–2000. JAMA,2002,288:1723~1727
    [5]武阳丰,马冠生,胡永华,等.中国居民的超重和肥胖流行现状.中华预防学杂志,2005,9(5):316~320
    [6] Tomoda K, Kubo K, Nishii Y, et al. Changes of ghrelin and leptin levels in plasmaby cigarette smoke in rats. J Toxicol Sci,2012,37(1):131~138
    [7] V h miko S, Isolauri E, Laitinen K. Weight status and dietary intake determineserum leptin concentrations in pregnant and lactating women and their infants. BrJ Nutr,2013,22:1~9[Epub ahead of print]
    [8] Barnea M, Shamay A, Stark AH, et al. A high-fat diet has a tissue-specific effecton adiponectin and related enzyme expression. Obesity,2006,14(12):2145~2153
    [9] EscrivaF, Gavete ML, FerminY, et al. Effect of age and moderate food restrictionon insulin sensitivity in Wistar rats: role of adiposity. J Endocrinol,2007,194(1):131~141
    [10] Kriketos AD, Gan SK, Poynten AM, et al. Exercise increases adiponectin levelsand insulin sensitivity in humans. Diabetes Care,2004,27(2):629~630
    [11] Kidani T, Kamei S, Miyawaki J, et al. Bisphenol A downregulates Akt signalingand inhibits adiponectin production and secretion in3T3-L1adipocytes.JAtheroscler Thromb,31,2010,17(8):834~843.
    [12] Zhao HY, Bi YF, Ma LY, et al. The effects of bisphenol A (BPA) exposure on fatmass and serum leptin concentrations have no impact on bone mineral densities innon-obese premenopausal women.Clin Biochem,2012,45(18):1602~1606
    [13] Ptak A, Gregoraszczuk EL. Bisphenol A induces leptin receptor expression,creating more binding sites for leptin, and activates the JAK/Stat, MAPK/ERKand PI3K/Akt signalling pathways in human ovarian cancer cell.Toxicol Lett.5,2012,210(3):332~337
    [14] Lago F, Dieguez C, Gomez-Reino J, et al. The emerging role of adipokines asmediators of inflammation and immune responses. Cytokine Growth Factor Rev,2007,18:313~325
    [15] Minokoshi Y, Kim YB, Peroni OD, et al. Leptin stimulates fatty-acid oxidationby activating AMP-activated protein kinase. Nature,2002,415:339~343
    [16] Yamagishi SI, Edelstein D, Du XL, et al. Leptin induces mitochondrialsuperoxide production and monocyte chemoattractant protein-1expression inaortic endothelial cells by increasing fatty acid oxidation via protein kinase A. JBio Chem,2001,276:25096~25100
    [17] Otero M, Lago R, Lago F, et al. Leptin,from fat to inflammation: old questionsand new insights. Sell Immunol,2005,579(2):295~301
    [18] Sierra-Johnson J, Romero-Corral A, Lopez-Jimenez F, et al. Relation of increasedleptin concentrations to history of myocardial infarction and stroke in the UnitedStates population. Am J Cardiol,2007,100:234~239
    [19] Chen MM, Ashley EA, Deng DX, et al. Novel role for the potent endogenousinotrope apelin in human cardiac dysfunction. Circulation,2003,108:1432~1439
    [20] Ouchi N, Ohishi M, Kihara S, et al. Association of hypoadiponectinemia withimpaired vasoreactivity. Hypertension,2003,42:231~234
    [21] Ouedraogo R, Gong Y, Berzins B, et al. Adiponectin deficiency increasesleukocyte-endothelium interactions via upregulation of endothelial cell adhesionmolecules in vivo. J Clin Invest,2007,117:1718~1726
    [22] Cheng KK, Lam KS, Wang Y, et al. Adiponectin-induced endothelial nitric oxidesynthase activation and nitric oxide production aremediated by APPL1inendothelial cells. Diabetes,2007,56:1387~1394
    [23] Hopkins TA, Ouchi N, Shibata R, et al.Adiponectin actions in the cardiovascularsystem. Cardiovasc Res,2007,74:11~18
    [24] Hall JE. The kidney, hypertension, and obesity. Hypertension,2003,41:625~633
    [25] ChowWS, Cheung BM, Tso AW, et al. Hypoadiponectinemia as a predictor forthe development of hypertension: a5-year prospective study. Hypertension,2007,49:1455~1461
    [26] Kawanami D, Maemura K, Takeda N, et al. Direct reciprocal effects of resistinand adiponectin on vascular endothelial cells: a new insight intoadipocytokine–endothelial cell interactions. Biochem Biophys Res Commun,2004,314:415~419
    [27] RajalaMW, Qi Y, Patel HR, et al. Regulation of resistin expression andcirculating levels in obesity, diabetes, and fasting. Diabetes,2004,53:1671~1679
    [28] Norata GD, Ongari M, Garlaschelli K,et al. Plasma resistin levels correlate withdeterminants of the metabolic syndrome. Eur J Endocrinol,2007,156:279~284
    [29] Efstathiou SP, Tsiakou AG, Tsioulos DI, et al. Prognostic significance of plasmaresistin levels in patients with atherothrombotic ischemic stroke. Clinica ChimicaActa,2007,378:78~85
    [30] Lubos E,Messow CM, Schnabel R, et al. Resistin, acute coronary syndrome andprognosis results from the AtheroGene study. Atherosclerosis,2007,193:121~128
    [31] Dick GM, Katz PS, Farias M III, et al. Resistin impairs endothelium-dependentdilation to bradykinin, but not acetylcholine, in the coronary circulation. Am JPhysiol Heart Circ Physiol,2006,291: H2997~3002
    [32] Calabro P, Samudio I, Willerson JT, et al.Resistin promotes smooth muscle cellproliferation through activation of extracellular signal-regulated kinase1/2andphosphatidylinositol3-kinase pathways. Circulation,2004,110:3335~3340
    [33] Mu H, Ohashi R, Yan S, et al. Adipokine resistin promotes in vitro angiogenesisof human endothelial cells. Cardiovasc Res,2006,70:146~157
    [34] Smith J, Al-Amri M, Sniderman A, et al. Visfatin concentration in Asian Indiansis correlated with high density lipoprotein cholesterol and apolipoprotein A1. ClinEndocrinol(Oxf),2006,65:667~672
    [35] Yamawaki H, Hara N, OkadaM, et al. Visfatin causes endothelium dependentrelaxation in isolated blood vessels. Biochem Biophys Res Commun,2009,383:503~508
    [36] Lim SY, Davidson SM, Paramanathan AJ, et al. The novel adipocytokine visfatinexerts direct cardioprotective effects. J Cell and Mol Med,2008,12:1395~1403
    [37] Dahl TB, Yndestad A, Skjelland M, et al. Increased expression of visfatin inmacrophages of human unstable carotid and coronary atherosclerosis: possiblerole in inflammation and plaque destabilization. Circulation,2007,115:972~980
    [38] Moschen AR, Kaser A, Enrich B, et al. Visfatin, an adipocytokine withproinflammatory and immunomodulating properties. J Immunol,2007,178:1748~1758
    [39] Takebayashi K, Suetsugu M, Wakabayashi S, et al. Association between plasmavisfatin and vascular endothelial function in patients with type2diabetes mellitus.Metabolism,2007,56:451~458
    [40] Adya R, Tan BK, Punn A, et al. Visfatin induces human endothelial VEGF andMMP-2/9production via MAPK and PI3K/Akt signalling pathways: novelinsights into visfatin-induced angiogenesis. Cardiovasc Res,2008,78:356~365
    [41] Lee DK, Cheng R, Nguyen T, et al. Characterization of apelin, the ligand for theAPJ receptor. J Neurochem,2000,74:34~41
    [42] Tasci I, Erdem G, Ozgur G, et al. LDL-cholesterol lowering increases plasmaapelin in isolated hypercholesterolemia. Atherosclerosis,2009,204:222~228
    [43] Leeper NJ, Tedesco MM, Kojima Y, et al. Apelin prevents aortic aneurysmformation by inhibiting macrophage inflammation. Am J Physiol Heart CircPhysiol,2009,296: H1329~H1335
    [44] Tatemoto K, Takayama K, Zou MX, et al. The novel peptide apelin lowers bloodpressure via a nitric oxide-dependent mechanism. Regul Pept,2001,99:87~92
    [45] Ellinor PT, Low AF, Macrae CA. Reduced apelin levels in lone atrial fibrillation.Eur Heart J,2006,27:222~226
    [46] Chong KS, Gardner RS,Morton JJ, et al. Plasma concentrations of the novelpeptide apelin are decreased in patients with chronic heart failure. Eur J Heart Fail,2006,8:355~360
    [47] Iwanaga Y, Kihara Y, Takenaka H, et al. Down-regulation of cardiac apelinsystem in hypertrophied and failing hearts: possible role of angiotensinII-angiotensin type1receptor system. J Mol Cell Cardiol,2006,41:798~806
    [48] Saremi A, Asghari M, Ghorbani A. Effects of aerobic training on serumomentin-1and cardiometabolic risk factors in overweight and obese men. J SportsSci,2010,28:993~998
    [49] Yamawaki H, Tsubaki N, Mukohda M, et al. Omentin, a novel adipokine, inducesvasodilation in rat isolated blood vessels. Biochem Biophys Res Commun,2010,393:668~672
    [50] Yamawaki H, Kuramoto J, Kameshima S, et al. Omentin, a novel adipocytokineinhibits TNF-induced vascular inflammation in human endothelial cells. BiochemBiophys Res Commun,2011,408:339~343
    [51] Bozaoglu K, Bolton K, McMillan J, et al. Chemerin is a novel adipokineassociated with obesity and metabolic syndrome. Endocrinol,2007,148:4687~4694
    [52] Sell H, Divoux A, Poitou C, et al. Chemerin correlates with markers for fattyliver inmorbidly obese patients and strongly decreases after weight loss inducedby bariatric surgery. J Clin Endocrinol Metab,2010,95:2892~2896
    [53] Roh SG, Song SH, Choi KC, et al. Chemerin–a new adipokine that modulatesadipogenesis via its own receptor. Biochem Biophys Res Commun,2007,362:1013~1018
    [54] Lehrke M, Becker A, Greif M, et al. Chemerin is associated with markers ofinflammation and components of the metabolic syndrome but does not predictcoronary atherosclerosis. Eur J Endocrinol,2009,161:339~344
    [55] Yoo HJ, Choi HY, Yang SJ, et al. Circulating chemerin level is independentlycorrelated with arterial stiffness. J Atheroscler and Thromb,2012,19:59~66(discussion67-58)
    [56] Lobato NS, Neves KB, Filgueira FP, et al. The adipokine chemerin augmentsvascular reactivity to contractile stimuli via activation of the MEK-ERK1/2pathway. Life Sci,2012,91:600~606
    [57] Yang W, Lu J, Weng J, et al. Prevalence of diabetes among men and women inChina. N Engl J Med,2010,362(12):1090~1101
    [58] Dyck DJ,Heigenhauser GJ,Burce CR,et a1.The role of adipokinases regulators of skeletal muscle fatty acid metabolism and insuline sensitivity.AcatPhysiol(Oxf),2006,186(1):5~16
    [59] BenomarY,NaourN,AubourglA, et al.Insluin and leptin induce Glut4plsamamembrane translcoation and glucose uptake in a human neuronla cell line by aPI3-kinase dependent.Endcoirnol,2006,147(5):2550~2556
    [60] Scha lerA,ScholmeirchJ,BucbierC, et a1.Mechanisms of disease:adipocytokines and visceral adiopse tissue: emerging role in nonalcoholic fattyliver disease.Nat Clin Parct Gsatorenteorl Hepatol,2005,2(6):273~280
    [61] Kubota N,Teranchi Y,Yamanchi T, et al. Disruption of adiponectin causesinsulin resistance and neointimal formation. J Biol Chem,2002,277(29):25863~25866
    [62] Tomas E, Tsao TS, Saha AK, et al. Enhanced muscle fat oxidation and glucosetransport by ACRP30globular domain: acetyl-CoA carboxylase inhibition andAMP-activated protein kinase activation. PNAS,2002,99:16309~16313
    [63] Silha JV, Krsek M, Skrha JV, et al. Plasma resistin, adiponectin and leptin levelsin lean and obese subjects: correlations with insulin resistance. Eur J Endocrinol,2003,149:331~335
    [64] Steppan CM, Bailey ST, Bhat S, et al.The hormone resistin links obesity todiabetes.Nature,2001,409(6818):307~312
    [65] Graveleau C, Zaha VG, Mohajer A, et al. Mouse and human resistins impairglucose transport in primary mouse cardiomyocytes, and oligomerization isrequired for this biological action. J Bio Chem,2005,280:31679~31685
    [66] Nagaev I, Smith U. Insulin resistance and type2diabetes are not related toresistin expression in human fat cells or skeletal muscle.Biochem Biophys ResCommun.2001,285(2):561~564
    [67] Sethi JK, Vidal-Puig A. Visfatin: the missing link between intra-abdominalobesity and diabetes? Trends Mol Med,2005,11:344~347
    [68] Bailey SD, Loredo-Osti JC, Lepage P, et al. Common polymorphisms in thepromoter of the visfatin gene (PBEF1) influence plasma insulin levels in aFrench-Canadian population. Diabetes,2006,55:2896~2902
    [69] Brown JE, Onyango DJ, RamanjaneyaM, et al. Visfatin regulates insulinsecretion, insulin receptor signalling and mRNA expression of diabetes-relatedgenes in mouse pancreatic b-cells. J Mol Endocrinol,2010,44:171~178
    [70] Fukuhara A, Matsuda M, Nishizawa M, et al. Visfatin: a protein secreted byvisceral fat that mimics the effects of insulin. Science,2005,307(5708):426~430
    [71] Boucher J, Masri B, Daviaud D,et al. Apelin, a newly identified adipokineup-regulated by insulin and obesity. Endocrinol,2005,146(4):1764~1771
    [72] Yang RZ, Lee MJ, Hu H, et al. Identification of omentin as a novel depot-specificadipokine in human adipose tissue: possible role in modulating insulin action.American Journal of Physiology. Endocrinol Metab,2006,290: E1253~E1261
    [73] de Souza Batista CM, Yang RZ, Lee MJ, et al. Omentin plasma levels and geneexpression are decreased in obesity. Diabetes,2007,56:1655~1661
    [74] Zabel BA, Allen SJ, Kulig P, et al. Chemerin activation by serine proteases of thecoagulation, fibrinolytic, and inflammatory cascades. J Biol Chem,2005,280(41):34661~34666
    [75] Goralski KB, McCarthy TC, Hanniman EA, et al. Chemerin, a novel adipokinethat regulates adipogenesis and adipocyte metabolism. J Bio Chem,2007,282:28175~28188
    [76] Takahashi M, Takahashi Y, Takahashi K, et al. Chemerin enhances insulinsignaling and potentiates insulin-stimulated glucose uptake in3T3-L1adipocytes.FEBS Letters,2008,582:573~578
    [77] Sharma D, Saxena NK, Vertino PM, et al. Leptin promotes the proliferativeresponse and invasiveness in human endometrial cancer cells by activatingmultiple signal-transduction pathways. Endocr Relat Cancer,2006,13:629~640
    [78] Hu X, Juneja SC, Machle NJ, et al. Leptin a growth factor in normal andmaglignant breast cells and for normal mammary gland development. J NatlCancer Inst,2002,94(22):1704~1711
    [79] Hoda MR, Popken G. Mitogenic and anti-apoptotic actions of adipocyte-derivedhormone leptin in prostate cancer cells, BJU Int,2008,102:383~388
    [80] Petridou E, Mantzoros C, Dessypris N, et a1. Plasma adiponectin concentrationsin relation to endometrial cancer:a case control study in Greece. J Clin EndoerinolMetab,2003,88(3):993~997
    [81] Housa D, Housova J, Vernerova Z, et al. Adipocytokines and Cancer. J PhysiolRes,2006,55:233~244
    [82] Landskroner-Eiger S, Qian B, Muise ES, et al. Proangiogenic contribution ofadiponectin toward mammary tumor growth in vivo, Clin Cancer Res,2009,15:3265~3276
    [83] Poehls J, Wassel CL, Harris TB,et al. Association of adiponectin with mortalityin older adults: the Health, Aging, and Body Composition Study. Diabetologia,2009,52:591~595
    [84] Van Berendoncks AM, Garnier A, Beckers P, et al. Functional adiponectinresistance at the level of the skeletal muscle in mild to moderate chronic heartfailure. Circ Heart Fail,2010,3:185~194
    [85] Howard JK, Lord GM, Matarese G, et al. Leptin protects mice fromstarvation-induced lymphoid atrophy and increases thymic cellularity in ob/obmice. J Clin Invest,1999,104(8):1051~1059
    [86] Shamsuzzaman AS, Winnicki M, Wolk R, et al. Independent association betweenplasma leptin and C-reactive protein in healthy humans. Circulation,2004,109:2181~2185
    [87] Fantuzzi G. Adipose tissue, adipokines, and inflammation. J Allergy ClinImmunol,115:911~919, quiz920
    [88] Breyer MK, Rutten EP, Locantore NW, et al. Dysregulated adipokine metabolismin chronic obstructive pulmonary disease. Eur J Clin Invest,2012,42(9):983~991