Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front Med    2013, Vol. 7 Issue (3) : 301-305    https://doi.org/10.1007/s11684-013-0283-1
REVIEW
Adiponectin: mechanisms and new therapeutic approaches for restoring diabetic heart sensitivity to ischemic post-conditioning
Tingting Wang1,2(), Shanglong Yao1, Zhengyuan Xia2(), Michael G. Irwin2
1. Department of Anesthesiology and Critical Care, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; 2. Department of Anesthesiology, The University of Hong Kong, Hong Kong SAR, China
 Download: PDF(165 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Systemic inflammatory response following myocardial ischemia-reperfusion injury (IRI) to a specific organ may cause injuries. Ischemic post-conditioning (IPostC) has emerged as a promising method for myocardial protection against IRI both in experimental and in clinical settings. Enhancement of endogenous nitric oxide (NO) is one of the major mechanisms by which IPostC confers cardioprotection. However, the sensitivity of the diabetic heart to IPostC is impaired and the underlying mechanism is unknown. Adiponectin (APN) is an adipocyte-derived plasma protein with anti-diabetic and anti-inflammatory properties. Plasma levels of APN are decreased in obese subjects and in patients with type 2 diabetes. APN supplementation has been shown to increase NO production and attenuate myocardial IRI in normal (non-diabetic) animals. However, the effect of APN on myocardial injury in diabetic subjects, especially its potential in restoring the sensitivity of the diabetic heart to IPostC has not been investigated. In the current paper, we discussed the possible reasons why the myocardium of diabetic subjects loses sensitivity to IPostC and also highlighted the potential effectiveness and mechanism of APN in restoring IPostC cardioprotection in diabetes. This review proposes to conduct studies that may facilitate the development of novel and optimal therapies to enhance cardioprotection in patients with severe diseases such as diabetes.

Keywords adiponectin      ischemic post-conditioning      ischemia reperfusion injury      diabetes     
Corresponding Author(s): Wang Tingting,Email:wangtingting721@yahoo.com; Xia Zhengyuan,Email:zyxia@hku.hk   
Issue Date: 05 September 2013
 Cite this article:   
Tingting Wang,Shanglong Yao,Zhengyuan Xia, et al. Adiponectin: mechanisms and new therapeutic approaches for restoring diabetic heart sensitivity to ischemic post-conditioning[J]. Front Med, 2013, 7(3): 301-305.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-013-0283-1
https://academic.hep.com.cn/fmd/EN/Y2013/V7/I3/301
Fig.1  Schematic representation of the proposed mechanism for restoration of diabetic heart sensitivity to ischemic post-conditioning by adiponectin supplementation. Tumor necrosis factor-alpha (TNF-α) at low concentrations binds to TNF-α receptor-2 which activates in sequence JAK and STAT-3, confering cardioprotection via the SAFE pathway. Although cross-talk may exist between the RISK and SAFE pathways, ischemic post-conditioning can trigger SAFE pathway independent of its ability to activate the classic RISK pathway. Adiponectin may activate the SAFE pathway by stimulating endothelial NO production and subsequent TNF production. Adiponectin can also activate AMPK which may trigger JAK/STAT-3 signaling. On the other hand, AMPK can directly activate eNOS and inhibit iNOS. The resulting increase in nitric oxide bioavailability then confers cardioprotection.
1 Roffi M, Eberli FR. Diabetes and acute coronary syndromes. Best Pract Res Clin Endocrinol Metab 2009; 23(3): 305-316
doi: 10.1016/j.beem.2009.01.003 pmid:19520305
2 Andersen HR, Nielsen TT, Rasmussen K, Thuesen L, Kelbaek H, Thayssen P, Abildgaard U, Pedersen F, Madsen JK, Grande P, Villadsen AB, Krusell LR, Haghfelt T, Lomholt P, Husted SE, Vigholt E, Kjaergard HK, Mortensen LS ; DANAMI-2 Investigators. A comparison of coronary angioplasty with fibrinolytic therapy in acute myocardial infarction. N Engl J Med 2003; 349(8): 733-742
doi: 10.1056/NEJMoa025142 pmid:12930925
3 Murry CE, Jennings RB, Reimer KA. Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium. Circulation 1986; 74(5): 1124-1136
doi: 10.1161/01.CIR.74.5.1124 pmid:3769170
4 Zhao ZQ, Corvera JS, Halkos ME, Kerendi F, Wang NP, Guyton RA, Vinten-Johansen J. Inhibition of myocardial injury by ischemic postconditioning during reperfusion: comparison with ischemic preconditioning. Am J Physiol Heart Circ Physiol 2003; 285(2): H579-H588
pmid:12860564
5 Ouchi N, Walsh K. Adiponectin as an anti-inflammatory factor. Clin Chim Acta 2007; 380(1-2): 24-30
doi: 10.1016/j.cca.2007.01.026 pmid:17343838
6 Pajvani UB, Scherer PE. Adiponectin: systemic contributor to insulin sensitivity. Curr Diab Rep 2003; 3(3): 207-213
doi: 10.1007/s11892-003-0065-2 pmid:12762967
7 Hotta K, Funahashi T, Arita Y, Takahashi M, Matsuda M, Okamoto Y, Iwahashi H, Kuriyama H, Ouchi N, Maeda K, Nishida M, Kihara S, Sakai N, Nakajima T, Hasegawa K, Muraguchi M, Ohmoto Y, Nakamura T, Yamashita S, Hanafusa T, Matsuzawa Y. Plasma concentrations of a novel, adipose-specific protein, adiponectin, in type 2 diabetic patients. Arterioscler Thromb Vasc Biol 2000; 20(6): 1595-1599
doi: 10.1161/01.ATV.20.6.1595 pmid:10845877
8 Wang T, Qiao S, Lei S, Liu Y, Ng KF, Xu A, Lam KS, Irwin MG, Xia Z. N-acetylcysteine and allopurinol synergistically enhance cardiac adiponectin content and reduce myocardial reperfusion injury in diabetic rats. PLoS ONE 2011; 6(8): e23967
doi: 10.1371/journal.pone.0023967 pmid:21912612
9 Shibata R, Sato K, Pimentel DR, Takemura Y, Kihara S, Ohashi K, Funahashi T, Ouchi N, Walsh K. Adiponectin protects against myocardial ischemia-reperfusion injury through AMPK- and COX-2-dependent mechanisms. Nat Med 2005; 11(10): 1096-1103
doi: 10.1038/nm1295 pmid:16155579
10 Tao L, Gao E, Jiao X, Yuan Y, Li S, Christopher TA, Lopez BL, Koch W, Chan L, Goldstein BJ, Ma XL. Adiponectin cardioprotection after myocardial ischemia/reperfusion involves the reduction of oxidative/nitrative stress. Circulation 2007; 115(11): 1408-1416
doi: 10.1161/CIRCULATIONAHA.106.666941 pmid:17339545
11 Liu X, Chen H, Zhan B, Xing B, Zhou J, Zhu H, Chen Z. Attenuation of reperfusion injury by renal ischemic postconditioning: the role of NO. Biochem Biophys Res Commun 2007; 359(3): 628-634
doi: 10.1016/j.bbrc.2007.05.129 pmid:17548062
12 Amour J, Brzezinska AK, Weihrauch D, Billstrom AR, Zielonka J, Krolikowski JG, Bienengraeber MW, Warltier DC, Pratt PFJ Jr, Kersten JR. Role of heat shock protein 90 and endothelial nitric oxide synthase during early anesthetic and ischemic preconditioning. Anesthesiology 2009; 110(2): 317-325
pmid:19194158
13 Lecour S. Activation of the protective survivor activating factor enhancement (SAFE) pathway against reperfusion injury: does it go beyond the RISK pathway? J Mol Cell Cardiol 2009; 47(1): 32-40
doi: 10.1016/j.yjmcc.2009.03.019 pmid:19344728
14 Suleman N, Somers S, Smith R, Opie LH, Lecour SC. Dual activation of STAT-3 and Akt is required during the trigger phase of ischaemic preconditioning. Cardiovasc Res 2008; 79(1): 127-133
doi: 10.1093/cvr/cvn067 pmid:18339648
15 Gross ER, Hsu AK, Gross GJ. The JAK/STAT pathway is essential for opioid-induced cardioprotection: JAK2 as a mediator of STAT3, Akt, and GSK-3 beta. Am J Physiol Heart Circ Physiol 2006; 291(2): H827-H834
doi: 10.1152/ajpheart.00003.2006 pmid:16517948
16 Oshima Y, Fujio Y, Nakanishi T, Itoh N, Yamamoto Y, Negoro S, Tanaka K, Kishimoto T, Kawase I, Azuma J. STAT3 mediates cardioprotection against ischemia/reperfusion injury through metallothionein induction in the heart. Cardiovasc Res 2005; 65(2): 428-435
doi: 10.1016/j.cardiores.2004.10.021 pmid:15639482
17 Xuan YT, Guo Y, Han H, Zhu Y, Bolli R. An essential role of the JAK-STAT pathway in ischemic preconditioning. Proc Natl Acad Sci USA 2001; 98(16): 9050-9055
doi: 10.1073/pnas.161283798 pmid:11481471
18 Skyschally A, van Caster P, Boengler K, Gres P, Musiolik J, Schilawa D, Schulz R, Heusch G. Ischemic postconditioning in pigs: no causal role for RISK activation. Circ Res 2009; 104(1): 15-18
doi: 10.1161/CIRCRESAHA.108.186429 pmid:19038864
19 Boengler K, Buechert A, Heinen Y, Roeskes C, Hilfiker-Kleiner D, Heusch G, Schulz R. Cardioprotection by ischemic postconditioning is lost in aged and STAT3-deficient mice. Circ Res 2008; 102(1): 131-135
doi: 10.1161/CIRCRESAHA.107.164699 pmid:17967780
20 Yin X, Zheng Y, Zhai X, Zhao X, Cai L.Diabetic inhibition of preconditioning- and postconditioning-mediated myocardial protection against ischemia/reperfusion injury. Exp Diabetes Res 2012 ; 2012: 198048
doi: 10.1155/2012/198048
21 Takimoto E, Kass DA. Role of oxidative stress in cardiac hypertrophy and remodeling. Hypertension 2007; 49(2): 241-248
doi: 10.1161/01.HYP.0000254415.31362.a7 pmid:17190878
23 Nagareddy PR, Xia Z, McNeill JH, MacLeod KM. Increased expression of iNOS is associated with endothelial dysfunction and impaired pressor responsiveness in streptozotocin-induced diabetes. Am J Physiol Heart Circ Physiol 2005; 289(5): H2144-H2152
doi: 10.1152/ajpheart.00591.2005 pmid:16006542
24 Xia Z, Nagareddy PR, Guo Z, Zhang W, McNeill JH. Antioxidant N-acetylcysteine restores systemic nitric oxide availability and corrects depressions in arterial blood pressure and heart rate in diabetic rats. Free Radic Res 2006; 40(2): 175-184
doi: 10.1080/10715760500484336 pmid:16390827
25 Nagareddy PR, Xia Z, MacLeod KM, McNeill JH. N-acetylcysteine prevents nitrosative stress-associated depression of blood pressure and heart rate in streptozotocin diabetic rats. J Cardiovasc Pharmacol 2006; 47(4): 513-520
doi: 10.1097/01.fjc.0000211744.93701.25 pmid:16680064
26 Zhang P, Xu X, Hu X, van Deel ED, Zhu G, Chen Y. Inducible nitric oxide synthase deficiency protects the heart from systolic overload-induced ventricular hypertrophy and congestive heart failure. Circ Res 2007; 100(7): 1089-1098
doi: 10.1161/01.RES.0000264081.78659.45 pmid:17363700
27 Lacerda L, Somers S, Opie LH, Lecour S. Ischaemic postconditioning protects against reperfusion injury via the SAFE pathway. Cardiovasc Res 2009; 84(2): 201-208
doi: 10.1093/cvr/cvp274 pmid:19666677
28 Gross ER, Hsu AK, Gross GJ. Diabetes abolishes morphine-induced cardioprotection via multiple pathways upstream of glycogen synthase kinase-3beta. Diabetes 2007; 56(1): 127-136
doi: 10.2337/db06-0907 pmid:17192474
29 Siasos G, Tousoulis D, Kollia C, Oikonomou E, Siasou Z, Stefanadis C, Papavassiliou AG. Adiponectin and cardiovascular disease: mechanisms and new therapeutic approaches. Curr Med Chem 2012; 19(8): 1193-1209
doi: 10.2174/092986712799320583 pmid:22257055
30 Lindsay RS, Funahashi T, Hanson RL, Matsuzawa Y, Tanaka S, Tataranni PA, Knowler WC, Krakoff J. Adiponectin and development of type 2 diabetes in the Pima Indian population. Lancet 2002; 360(9326): 57-58
doi: 10.1016/S0140-6736(02)09335-2 pmid:12114044
31 Cheng KK, Lam KS, Wang Y, Huang Y, Carling D, Wu D, Wong C, Xu A. Adiponectin-induced endothelial nitric oxide synthase activation and nitric oxide production are mediated by APPL1 in endothelial cells. Diabetes 2007; 56(5): 1387-1394
doi: 10.2337/db06-1580 pmid:17287464
32 Chang J, Li Y, Huang Y, Lam KS, Hoo RL, Wong WT, Cheng KK, Wang Y, Vanhoutte PM, Xu A. Adiponectin prevents diabetic premature senescence of endothelial progenitor cells and promotes endothelial repair by suppressing the p38 MAP kinase/p16INK4A signaling pathway. Diabetes 2010; 59(11): 2949-2959
doi: 10.2337/db10-0582 pmid:20802255
33 Cai XJ, Chen L, Li L, Feng M, Li X, Zhang K, Rong YY, Hu XB, Zhang MX, Zhang Y, Zhang M. Adiponectin inhibits lipopolysaccharide-induced adventitial fibroblast migration and transition to myofibroblasts via AdipoR1-AMPK-iNOS pathway. Mol Endocrinol 2010; 24(1): 218-228
doi: 10.1210/me.2009-0128 pmid:19889816
34 Liao W, Yu C, Wen J, Jia W, Li G, Ke Y, Zhao S, Campell W. Adiponectin induces interleukin-6 production and activates STAT3 in adult mouse cardiac fibroblasts. Biol Cell 2009; 101(5): 263-272
doi: 10.1042/BC20080117 pmid:18795895
35 Morrow VA, Foufelle F, Connell JM, Petrie JR, Gould GW, Salt IP. Direct activation of AMP-activated protein kinase stimulates nitric-oxide synthesis in human aortic endothelial cells. J Biol Chem 2003; 278(34): 31629-31639
doi: 10.1074/jbc.M212831200 pmid:12791703
36 Shin MJ, Lee YP, Kim DW, An JJ, Jang SH, Cho SM, Sheen SH, Lee HR, Kweon HY, Kang SW, Lee KG, Park J, Eum WS, Cho YJ, Choi SY. Transduced PEP-1-AMPK inhibits the LPS-induced expression of COX-2 and iNOS in Raw264.7 cells. BMB Rep 2010; 43(1): 40-45
doi: 10.5483/BMBRep.2010.43.1.040 pmid:20132734
37 Pilon G, Dallaire P, Marette A. Inhibition of inducible nitric-oxide synthase by activators of AMP-activated protein kinase: a new mechanism of action of insulin-sensitizing drugs. J Biol Chem 2004; 279(20): 20767-20774
doi: 10.1074/jbc.M401390200 pmid:14985344
38 Hwang JT, Kwon DY, Park OJ, Kim MS. Resveratrol protects ROS-induced cell death by activating AMPK in H9c2 cardiac muscle cells. Genes Nutr 2008; 2(4): 323-326
doi: 10.1007/s12263-007-0069-7 pmid:18850225
39 Bouhidel O, Pons S, Souktani R, Zini R, Berdeaux A, Ghaleh B. Myocardial ischemic postconditioning against ischemia-reperfusion is impaired in ob/ob mice. Am J Physiol Heart Circ Physiol 2008; 295(4): H1580-H1586
doi: 10.1152/ajpheart.00379.2008 pmid:18689499
40 Shimano M, Ouchi N, Shibata R, Ohashi K, Pimentel DR, Murohara T, Walsh K. Adiponectin deficiency exacerbates cardiac dysfunction following pressure overload through disruption of an AMPK-dependent angiogenic response. J Mol Cell Cardiol 2010; 49(2): 210-220
doi: 10.1016/j.yjmcc.2010.02.021 pmid:20206634
41 Szmitko PE, Teoh H, Stewart DJ, Verma S. Adiponectin and cardiovascular disease: state of the art? Am J Physiol Heart Circ Physiol 2007; 292(4): H1655-H1663
doi: 10.1152/ajpheart.01072.2006 pmid:17142348
42 Guo Z, Xia Z, Jiang J, McNeill JH. Downregulation of NADPH oxidase, antioxidant enzymes, and inflammatory markers in the heart of streptozotocin-induced diabetic rats by N-acetyl-L-cysteine. Am J Physiol Heart Circ Physiol 2007; 292(4): H1728-H1736
doi: 10.1152/ajpheart.01328.2005 pmid:17122189
43 Peng T, Lu X, Lei M, Feng Q. Endothelial nitric-oxide synthase enhances lipopolysaccharide-stimulated tumor necrosis factor-alpha expression via cAMP-mediated p38 MAPK pathway in cardiomyocytes. J Biol Chem 2003; 278(10): 8099-8105
doi: 10.1074/jbc.M207288200 pmid:12506117
44 Heusch G, Boengler K, Schulz R. Cardioprotection: nitric oxide, protein kinases, and mitochondria. Circulation 2008; 118(19): 1915-1919
doi: 10.1161/CIRCULATIONAHA.108.805242 pmid:18981312
45 Thielmann M, D?rge H, Martin C, Belosjorow S, Schwanke U, van De Sand A, Konietzka I, Büchert A, Krüger A, Schulz R, Heusch G. Myocardial dysfunction with coronary microembolization: signal transduction through a sequence of nitric oxide, tumor necrosis factor-alpha, and sphingosine. Circ Res 2002; 90(7): 807-813
doi: 10.1161/01.RES.0000014451.75415.36 pmid:11964374
46 L’Abbate A, Neglia D, Vecoli C, Novelli M, Ottaviano V, Baldi S, Barsacchi R, Paolicchi A, Masiello P, Drummond GS, McClung JA, Abraham NG. Beneficial effect of heme oxygenase-1 expression on myocardial ischemia-reperfusion involves an increase in adiponectin in mildly diabetic rats. Am J Physiol Heart Circ Physiol 2007; 293(6): H3532-H3541
doi: 10.1152/ajpheart.00826.2007 pmid:17906103
47 Xia Z, Vanhoutte PM. Nitric oxide and protection against cardiac ischemia. Curr Pharm Des 2011; 17(18): 1774-1782
doi: 10.2174/138161211796391047 pmid:21631419
[1] Hui Wang, Yang Zhang, Zhujun Shen, Ligang Fang, Zhenyu Liu, Shuyang Zhang. Prognostic value of fasting glucose on the risk of heart failure and left ventricular systolic dysfunction in non-diabetic patients with ST-segment elevation myocardial infarction[J]. Front. Med., 2021, 15(1): 70-78.
[2] Di Cheng, Chunyan Hu, Rui Du, Hongyan Qi, Lin Lin, Xueyan Wu, Lina Ma, Kui Peng, Mian Li, Min Xu, Yu Xu, Yufang Bi, Weiqing Wang, Yuhong Chen, Jieli Lu. Serum uric acid and risk of incident diabetes in middle-aged and elderly Chinese adults: prospective cohort study[J]. Front. Med., 2020, 14(6): 802-810.
[3] Huiwen Ren, Can Wu, Ying Shao, Shuang Liu, Yang Zhou, Qiuyue Wang. Correlation between serum miR-154-5p and urinary albumin excretion rates in patients with type 2 diabetes mellitus: a cross-sectional cohort study[J]. Front. Med., 2020, 14(5): 642-650.
[4] Pengju Zhang, Tao Li, Xingyun Wu, Edouard C. Nice, Canhua Huang, Yuanyuan Zhang. Oxidative stress and diabetes: antioxidative strategies[J]. Front. Med., 2020, 14(5): 583-600.
[5] Ning Jiang, Yao Li, Ting Shu, Jing Wang. Cytokines and inflammation in adipogenesis: an updated review[J]. Front. Med., 2019, 13(3): 314-329.
[6] Xiaoqing Li, Xinxin Li, Genbei Wang, Yan Xu, Yuanyuan Wang, Ruijia Hao, Xiaohui Ma. Xiao Ke Qing improves glycometabolism and ameliorates insulin resistance by regulating the PI3K/Akt pathway in KKAy mice[J]. Front. Med., 2018, 12(6): 688-696.
[7] Liping Xuan, Zhiyun Zhao, Xu Jia, Yanan Hou, Tiange Wang, Mian Li, Jieli Lu, Yu Xu, Yuhong Chen, Lu Qi, Weiqing Wang, Yufang Bi, Min Xu. Type 2 diabetes is causally associated with depression: a Mendelian randomization analysis[J]. Front. Med., 2018, 12(6): 678-687.
[8] Jiemin Pan, Weiping Jia. Early-onset diabetes: an epidemic in China[J]. Front. Med., 2018, 12(6): 624-633.
[9] Cynthia Rajani, Wei Jia. Bile acids and their effects on diabetes[J]. Front. Med., 2018, 12(6): 608-623.
[10] Meng Dong, Jun Lin, Wonchung Lim, Wanzhu Jin, Hyuek Jong Lee. Role of brown adipose tissue in metabolic syndrome, aging, and cancer cachexia[J]. Front. Med., 2018, 12(2): 130-138.
[11] Chao Chen, Chang Wang, Chun Hu, Yachun Han, Li Zhao, Xuejing Zhu, Li Xiao, Lin Sun. Normoalbuminuric diabetic kidney disease[J]. Front. Med., 2017, 11(3): 310-318.
[12] Palka Kaur Khanuja,Satish Chander Narula,Rajesh Rajput,Rajinder Kumar Sharma,Shikha Tewari. Association of periodontal disease with glycemic control in patients with type 2 diabetes in Indian population[J]. Front. Med., 2017, 11(1): 110-119.
[13] Huiqin Zhong,Ya Shao,Ling Fan,Tangshen Zhong,Lu Ren,Yan Wang. Perceived resource support for chronic illnesses among diabetics in north-western China[J]. Front. Med., 2016, 10(2): 219-227.
[14] Juan Zheng,Shih-Lung Woo,Xiang Hu,Rachel Botchlett,Lulu Chen,Yuqing Huo,Chaodong Wu. Metformin and metabolic diseases: a focus on hepatic aspects[J]. Front. Med., 2015, 9(2): 173-186.
[15] Jie Zheng,Yuzhen Gao,Yuejuan Jing,Xiaoshuang Zhou,Yuanyuan Shi,Yanhong Li,Lihua Wang,Ruiying Wang,Maolian Li,Chuanshi Xiao,Yafeng Li,Rongshan Li. Gender differences in the relationship between plasma lipids and fasting plasma glucose in non-diabetic urban Chinese population: a cross-section study[J]. Front. Med., 2014, 8(4): 477-483.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed