AccScience Publishing / GTM / Volume 1 / Issue 2 / DOI: 10.36922/gtm.v1i2.85
ORIGINAL RESEARCH ARTICLE

Effect of leptin on aortic dissection

Ling Chen1,2 Yang Xi1,2 Fan Xu1,2 Liangwan Chen1,2*
Show Less
1 Department of Cardiac Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian 350001, People’s Republic of China
2 Fujian Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fuzhou, Fujian 350001, People’s Republic of China
Global Translational Medicine 2022, 1(2), 85 https://doi.org/10.36922/gtm.v1i2.85
Submitted: 6 May 2022 | Accepted: 14 November 2022 | Published: 9 December 2022
© 2022 by the Author(s). This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution 4.0 International License ( https://creativecommons.org/licenses/by/4.0/ )
Abstract

The most important clinical features of aortic dissection (AD) are its acute onset, rapid progress, and high fatality rate. The exact pathogenesis of AD is unclear, and the focus of current research on the mechanism of AD has been primarily on hypertension and changes in metalloproteinases, among which leptin plays an important role. The purpose of this study is to evaluate the effect of leptin on AD. We conducted a computerized literature search on animal studies related to leptin and dissecting aortic aneurysm in PubMed, EMBASE, Cochrane Library, MEDLINE, and other databases from their inception to present. Meta-analysis was conducted to compare the changes in aortic diameter, aortic dilatation, and the incidence of AD in mice under the local intervention of leptin or leptin antagonist (LepA). A total of four studies were included, involving five batches of animal experiments. According to the results of the meta-analysis, the increase in local leptin content led to the enlargement of aortic diameter (relative risk [RR] = 0.18; 95% confidence interval [CI]: 0.09 – 0.27; P < 0.0001) and increased aortic dilatation (RR = 0.11; 95% CI: 0.01 – 0.22; P < 0.0001). This meta-analysis showed that local leptin administration increased the aortic diameter and aortic dilatation. However, due to high heterogeneity between the results, it is difficult to draw a clear conclusion on the effect of leptin on AD.

Keywords
Leptin
Aortic dissection
Leptin antagonist
Funding
Fujian Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University)
References
[1]

Bossone E, LaBounty TM, Eagle KA. 2018, Acute aortic syndromes: diagnosis and management, an update. Eur Heart J, 39: 739–749d. https://doi.org/10.1093/eurheartj/ehx319 

[2]

Zindovic I, Gudbjartsson T, Ahlsson A, et al., 2019, Malperfusion in acute type A aortic dissection: An update from the Nordic Consortium for Acute Type A Aortic Dissection. J Thorac Cardiovasc Surg, 157: 1324–1333.e6. https://doi.org/10.1016/j.jtcvs.2018.10.134 

[3]

Criado FJ. 2011, Aortic dissection: A 250-year perspective. Tex Heart Inst J, 38: 694–700.

[4]

Ma LY, Wang Z, Fan J, et al., 2022, China Cardiovascular Health and Disease Report 2021 on the prevalence and prevention of hypertension in China. Chin J General Pract, 25: 6. 

[5]

Chen LW, Wu XJ, Dai XF, et al., 2016, Repair of acute type A aortic dissection with ascending aorta replacement combined with open fenestrated stent graft placement. Ann Thorac Surg, 101: 644–649. https://doi.org/10.1016/j.athoracsur.2015.07.060

[6]

Di S, Wang Y, Han L, et al., 2019, The intervention effect of traditional Chinese medicine on the intestinal flora and its metabolites in glycolipid metabolic disorders. Evid Based Complement Altern Med, 2019: 2958920. https://doi.org/10.1155/2019/2958920

[7]

Saxena NK, Vertino PM, Anania FA, et al., 2007, Leptin-induced growth stimulation of breast cancer cells involves recruitment of histone acetyltransferases and mediator complex to CYCLIN D1 promoter via activation of Stat3. J Biol Chem, 282: 13316–13325. https://doi.org/10.1074/jbc.M609798200

[8]

Otero M, Lago R, Lago F, et al., 2005, Signalling pathway involved in nitric oxide synthase Type II activation in chondrocytes: Synergistic effect of leptin with interleukin-1. Arthritis Res Ther, 7: R581–R591. https://doi.org/10.1186/ar1708

[9]

Maguire EM, Pearce SW, Xiao R, et al., 2019, Matrix metalloproteinase in abdominal aortic aneurysm and aortic dissection. Pharmaceuticals (Basel), 12: 371–394. https://doi.org/10.3390/ph12030118 

[10]

Mimler T, Nebert C, Eichmair E, et al., 2019, Extracellular matrix in ascending aortic aneurysms and dissections-what we learn from decellularization and scanning electron microscopy. PLoS One, 14: e0213794. https://doi.org/10.1371/journal.pone.0213794

[11]

Zhang L, Wang C, Xi Z, et al., 2018, Mercaptoethanol protects the aorta from dissection by inhibiting oxidative stress, inflammation, and extracellular matrix degeneration in a mouse model. Med Sci Monit, 24: 1802–1812. https://doi.org/10.12659/msm.905151

[12]

Vuolteenaho K, Koskinen A, Kukkonen M, et al., 2009, Leptin enhances synthesis of proinflammatory mediators in human osteoarthritic cartilage--mediator role of NO in leptin-induced PGE2, IL-6, and IL-8 production. Mediators Inflamm, 2009: 345838. https:/doi.org/10.1155/2009/345838

[13]

Mechanick JI, Zhao S, Garvey WT. 2018, Leptin, an adipokine with central importance in the global obesity problem. Global Heart, 13: 113–127. https://doi.org/10.1016/j.gheart.2017.10.003

[14]

Park HY, Kwon HM, Lim HJ, et al., 2001, Potential role of leptin in angiogenesis: Leptin induces endothelial cell proliferation and expression of matrix metalloproteinases in vivo and in vitro. Exp Mol Med, 33: 95–102. https://doi.org/10.1038/emm.2001.17

[15]

Cronin O, Walker PJ, Golledge J. 2013, The association of obesity with abdominal aortic aneurysm presence and growth. Atherosclerosis, 226: 321–327. https://doi.org/10.1016/j.atherosclerosis.2012.10.041

[16]

Higgins JPT, Green S, editors. Cochrane Handbook for Systematic Reviews of Interventions 4.2.6. In: The Cochrane Library, Issue 3, 2005. Chichester, UK: John Wiley & Sons, Ltd. 

[17]

Zhan Y, Yuan H, Bu P, et al., 2018, Recombinant leptin attenuates abdominal aortic aneurysm formation in angiotensin II-infused apolipoprotein E-deficient mice. Biochem Biophys Res Commun, 503: 1450–1456. https://doi.org/10.1016/j.bbrc.2018.07.062

[18]

Wu Z, Wang Z, Wu H, et al., 2020, Obesity is a risk factor for preoperative hypoxemia in Stanford A acute aortic dissection. Medicine (Baltimore), 99: e19186. https://doi.org/10.1097/MD.0000000000019186

[19]

Ellmark SH, Dusting GJ, Mark NT, et al., 2005, The contribution of Nox4 to NADPH oxidase activity in mouse vascular smooth muscle. Cardiovasc Res, 65: 495–504. https://doi.org/10.1016/j.cardiores.2004.10.026

[20]

Zheng XL, Yuan SG, Peng DQ. 2007, Phenotype-specific inhibition of the vascular smooth muscle cell cycle by high glucose treatment. Diabetologia, 50(4): 881–890. https://doi.org/10.1007/s00125-006-0543-6 

[21]

Werle M, Kreuzer J, Höfele J, et al., 2005, Metabolic control analysis of the Warburg-effect in proliferating vascular smooth muscle cells. J Biomed Sci, 12: 827–834. https://doi.org/10.1007/s11373-005-9010-5 

[22]

Zheng H, Qiu Z, Chai T, et al., 2022, Insulin resistance promotes the formation of aortic dissection by inducing the phenotypic switch of vascular smooth muscle cells. Front Cardiovasc Med, 8: 732122. https://doi.org/10.3389/fcvm.2021.732122

[23]

Yang L, Gao L, Nickel T, et al., 2017, Lactate promotes synthetic phenotype in vascular smooth muscle cells. Circ Res, 121: 1251–1262. https://doi.org/10.1161/CIRCRESAHA.117.311819 

[24]

Dusserre E, Bourdillon MC, Ciavatti M, et al., 1993, Lipid biosynthesis in cultured arterial smooth muscle cells is related to their phenotype. Lipids, 28: 589–592. https://doi.org/10.1007/BF02536051

[25]

Salabei JK, Hill BG. 2013, Mitochondrial fission induced by platelet-derived growth factor regulates vascular smooth muscle cell bioenergetics and cell proliferation. Redox Biol, 1: 542–551. https://doi.org/10.1016/j.redox.2013.10.011

[26]

Wu X, Ye J, Cai W, et al., 2022, LDHA mediated degradation of extracellular matrix is a potential target for the treatment of aortic dissection. Pharmacol Res, 176: 106051. https://doi.org/10.1016/j.phrs.2021.106051

[27]

Wang Y, Zhao ZM, Zhang GX, et al., 2016, Dynamic autophagic activity affected the development of thoracic aortic dissection by regulating functional properties of smooth muscle cells. Biochem Biophys Res Commun, 479: 358–364. https://doi.org/10.1016/j.bbrc.2016.09.080

[28]

Landsverk ML, Li S, Hutagalung AH, et al., 2007, The UNC- 45 chaperone mediates sarcomere assembly through myosin degradation in Caenorhabditis elegans. J Cell Biol, 177: 205–210. https://doi.org/10.1083/jcb.200607084

[29]

Bhatnagar S, Mittal A, Gupta SK, et al., 2011, TWEAK causes myotube atrophy through coordinated activation of ubiquitin-proteasome system, autophagy, and caspases. J Cell Physiol, 227: 1042–1051. https://doi.org/10.1002/jcp.22821

[30]

Karsli-Uzunbas G, Guo JY, Price S, et al., 2014, Autophagy is required for glucose homeostasis and lung tumor maintenance. Cancer Discov, 4: 914–927. https://doi.org/10.1158/2159-8290.CD-14-0363 

[31]

Zhang S, Ye J, Dong G. 2010, Neuroprotective effect of baicalein on hydrogen peroxide-mediated oxidative stress and mitochondrial dysfunction in PC12 cells. J Mol Neurosci, 40: 311–320. https://doi.org/10.1007/s12031-009-9285-5

[32]

Aliberti G, Proietta M, Tabacco F, et al., 2010, Inflammation and immune response in acute aortic dissection. Ann Med, 42: 622–629. https://doi.org/10.3109/07853890.2010.518156

[33]

Bersi MR, Khosravi R, Wujciak AJ, et al., 2017, Differential cell-matrix mechanoadaptations and inflammation drive regional propensities to aortic fibrosis, aneurysm or dissection in hypertension. J R Soc Interface, 14: 20170327. https:/doi.org/10.1098/rsif.2017.0327

[34]

Csongrádi É, Káplár M, Nagy B Jr., et al., 2017, Adipokines as atherothrombotic risk factors in obese subjects: Associations with haemostatic markers and common carotid wall thickness. Nutr Metab Cardiovasc Dis, 27: 571–580. https://doi.org/10.1016/j.numecd.2017.02.007

[35]

Ciccone M, Vettor R, Pannacciulli N, et al., 2001, Plasma leptin is independently associated with the intima-media thickness of the common carotid artery. Int J Obes Relat Metab Disord, 25: 805–810. https://doi.org/10.1038/sj.ijo.0801623

[36]

Stroescu R, Bizerea T, Doro G, et al., 2017, Correlation between adipokines and carotid intima media thickness in a group of obese Romanian children: Is small for gestational age status an independent factor for cardiovascular risk? Arch Endocrinol Metab, 61: 14–20. https://doi.org/10.1590/2359-3997000000201

[37]

Robati RM, Partovi-Kia M, Haghighatkhah HR, et al., 2014, Increased serum leptin and resistin levels and increased carotid intima-media wall thickness in patients with psoriasis: Is psoriasis associated with atherosclerosis? J Am Acad Dermatol, 71: 642–648. https://doi.org/10.1016/j.jaad.2014.06.006

[38]

Asha K, Sharma SB, Singal A, et al., 2014, Association of carotid intima-media thickness with leptin and apoliprotein B/apoliprotein A-I ratio reveals imminent predictors of subclinical atherosclerosis in psoriasis patients. Acta Med (Hradec Kralove), 57: 21–27. https://doi.org/10.14712/18059694.2014.4

[39]

McMahon M, Skaggs BJ, Grossman JM, et al., 2014, A panel of biomarkers is associated with increased risk of the presence and progression of atherosclerosis in women with systemic lupus erythematosus. Arthritis Rheumatol, 66: 130–139. https://doi.org/10.1002/art.38204

[40]

Gasbarrino K, Mantzoros C, Gorgui J, et al., 2016, Circulating chemerin is associated with carotid plaque instabaltty, whereas resistin is related to cerebrovascular symptomatology. Arterioscler Thromb Vasc Biol, 36: 1670–1678. https://doi.org/10.1161/ATVBAHA.115.306741

[41]

Schneiderman J, Simon AJ, Schroeter MR, et al., 2008, Leptin receptor is elevated in carotid plaques from neurologically symptomatic patients and positively correlated with augmented macrophage density. J Vasc Surg, 48: 1146–1155. https://doi.org/10.1016/j.jvs.2008.06.054

[42]

Bountouris I, Paraskevas Kl, Koutouzis M, et al., 2009, Serum leptin levels in patients undergoing carotid endarterectomy: A pilot study. Angilogy, 60: 698–704. https://doi.org/10.1177/0003319709350133

[43]

Tsai YC, Leu SY, Peng YJ, et al., 2017, Genistein suppresses leptin-induced proliferation and migration of vascular smooth muscle cells and neointima formation. J Cell Mol Med, 21: 422–431. https://doi.org/10.1111/jcmm.12986 

[44]

Andrade-Oliveira V, Camara NO, Moraes-Vieira PM. 2015, Adipokines as drug targets in diabetes and underlying disturbances. J Diabetes Res, 2015: 681612. https://doi.org/10.1155/2015/681612 

[45]

Kurajoh M, Koyama H, Kadoya M, et al., 2015, Plasma leptin level is associated with cardiac autonomic dysfunction in patients with Type 2 diabetes: HSCAA study. Cardiovasc Diabetol, 14: 117. https://doi.org/10.1186/s12933-015-0280-6

[46]

Vavruch C, Lanne T, Fredrikson M, et al., 2015, Serum leptin levels are independently related to the incidence of ischemic heart disease in a prospective study of patients with Type 2 diabetes. Cardiovasc Diabetol, 14: 62. https://doi.org/10.1186/s12933-015-0208-1

[47]

Rodriguez AJ, Nunes VD, Mastronardi CA, et al., 2016, Association between circulating adipocytokine concentrations and microvascular complications in patients with Type 2 diabetes mellitus: A systematic review and meta-analysis of controlled cross-sectional studies. J Diabetes Complications, 30: 357–367. https://doi.org/10.1016/j.jdiacomp.2015.11.004

[48]

Katsiki N, Yovos JG, Gotzamani-Psarrakou A, Karamitsos DT. Adipokines and vascular risk in Type 2 diabetes mellitus. Angiology, 62: 601–604. https://doi.org/10.1177/0003319711409201

[49]

Guzel S, Seven A, Kocaoglu A, et al., 2013, Osteoprotegerin, leptin and IL6: Association with silent myocardial ischemia in Type 2 diabetes mellitus. Diab Vasc Dis Res, 10: 25–31. https://doi.org/10.1177/1479164112440815 

[50]

Yamazaki Y, Emoto M, Morioka T, et al., 2013, Clinical impact of the leptin to soluble leptin receptor ratio on subclinical carotid atherosclerosis in patients with Type 2 diabetes. J Atheroscler Thromb, 20: 186–194. https://doi.org/10.5551/jat.14662

[51]

Liu X, Li X, Li C, et al., 2019, Study on regulation of adipokines on body fat distribution and its correlation with metabolic syndrome in Type 2 diabetes mellitus. Minerva Endocrinol, 44: 259–263. https://doi.org/10.23736/s0391-1977.17.02773-0

[52]

Asakawa H, Tokunaga K, Kawakami F. 2001, Relationship of leptin level with metabolic disorders and hypertension in Japanese Type 2 diabetes mellitus patients. J Diabetes Complications, 15: 57–62. https://doi.org/10.1016/s1056-8727(00)00145-8

[53]

Morioka T, Emoto M, Yamazaki Y, et al., 2014, Leptin is associated with vascular endothelial function in overweight patients with Type 2 diabetes. Cardiovasc Diabetol, 13: 10. https://doi.org/10.1186/1475-2840-13-10

[54]

Iraklianou S, Melidonis A, Tournis S, et al., 2001, Postprandial leptin responses after an oral fat tolerance test: Differences in Type 2 diabetes. Diabetes Care, 24: 1299–1301. https://doi.org/10.2337/diacare.24.7.1299-a

[55]

Zhang L, Qin Y, Liang D, et al., 2018, Association of polymorphisms in LEPR with Type 2 diabetes and related metabolic traits in a Chinese population. Lipids Health Dis, 17: 2. https://doi.org/10.1186/s12944-017-0644-x

[56]

Motawi T, Salman T, Shaker O, et al., 2015, Association of polymorphism in adiponectin (+45 T/G) and leptin (-2548 G/A) genes with Type 2 diabetes mellitus in male Egyptians. Arch Med Sci, 11: 937–944. https://doi.org/10.5114/aoms.2015.54848 

[57]

Su S, Zhang C, Zhang F, et al., 2016, The association between leptin receptor gene polymorphisms and Type 2 diabetes mellitus: A systematic review and meta-analysis. Diabetes Res Clin Pract, 121: 49–58. https://doi.org/10.1016/j.diabres.2016.08.008

[58]

Yang MM, Wang J, Fan JJ, et al., 2016, Variations in the obesity gene “LEPR” contribute to risk of Type 2 diabetes mellitus: Evidence from a meta-analysis. J Diabetes Res, 2016: 5412084. https://doi.org/10.1155/2016/5412084

[59]

Perry RJ, Petersen KF, Shulman GI. 2016, Pleotropic effects of leptin to reverse insulin resistance and diabetic ketoacidosis. Diabetologia, 59: 933–937. https://doi.org/10.1007/s00125-016-3909-4 

[60]

Feijoo-Bandin S, Portoles M, Rosello-Lleti E, et al., 2015, 20 years of leptin: Role of leptin in cardiomyocyte physiology and physiopathology. Life Sci, 140: 10–18. https://doi.org/10.1016/j.lfs.2015.02.016 

[61]

Gruzdeva OV, Akbasheva OE, Dyleva YA, et al., 2017, Adipokine and cytokine profiles of epicardial and subcutaneous adipose tissue in patients with coronary heart disease. Bull Exp Biol Med, 163: 608–611. https://doi.org/10.1007/s10517-017-3860-5

[62]

Basati G, Razavi AE, Abdi S, et al., Association between adipokine and myeloperoxidase levels in patients with coronary artery disease. Acta Med Iran, 53: 25–29.

[63]

Lodh M, Goswami B, Parida A, et al., 2012, Assessment of serum leptin, pregnancy-associated plasma protein A and CRP levels as indicators of plaque vulnerability in patients with acute coronary syndrome. Cardiovasc J Afr, 23: 330–335. https://doi.org/10.5830/CVJA-2012-008 

[64]

Karatela RA, Sainani GS. 2010, Interrelationships of factor VII activity and plasma leptin with insulin resistance in coronary heart disease. Atherosclerosis, 209: 235–240. https://doi.org/10.1016/j.atherosclerosis.2009.08.043

[65]

Gormez S, Demirkan A, Atalar F, et al., 2011, Adipose tissue gene expression of adiponectin, tumor necrosis factor-α and leptin in metabolic syndrome patients with coronary artery disease. Intern Med, 50: 805–810. https://doi.org/10.2169/internalmedicine.50.4753

[66]

Bigalke B, Stellos K, Geisler T, et al., 2010, High plasma levels of adipocytokines are associated with platelet activation in patients with coronary artery disease. Platelets, 21: 11–19. https://doi.org/10.3109/09537100903377584

[67]

Parhami F, Tintut Y, Ballard A, et al., 2001, Leptin enhances the calcification of vascular cells: Artery wall as a target of leptin. Circ Res, 88: 954–960. https://doi.org/10.1161/hh0901.090975

[68]

Cirillo P, Angri V, De Rosa S, et al., 2010, Proatherothrombotic effects of leptin in human coronary endothelial cells. Thromb Haemost, 103: 1065–1075. https://doi.org/10.1160/TH09-06-0392 

[69]

Ikmal SI, Huri H, Vethakkan SR, et al., 2013, Potential biomarkers of insulin resistance and atherosclerosis in Type 2 diabetes mellitus patients with coronary artery disease. Int J Endocrinol, 2013: 698567. https://doi.org/10.1155/2013/698567

[70]

Sainani GS, Karatela RA. 2009, Plasma leptin in insulin-resistant and insulin-nonresistant coronary artery disease and its association with cardio-metabolic risk factors among Asian Indians. Metab Syndr Relat Disord, 7: 335–340. https://doi.org/10.1089/met.2008.0097

[71]

Takahashi Y, Satoh M, Tabuchi T, et al., 2012, Prospective, randomized, single-blind comparison of effects of 6 months’ treatment with atorvastatin versus pravastatin on leptin and angiogenic factors in patients with coronary artery disease. Heart Vessels, 27: 337–343. https://doi.org/10.1007/s00380-011-0156-y

[72]

Sun YM, Li J, Luan Y, et al., 2010, Effect of statin therapy on leptin levels in patients with coronary heart disease. Peptides, 31: 1205–1207. https://doi.org/10.1016/j.peptides.2010.03.023

[73]

Katsiki N, Mikhailidis DP, Gotzamani-Psarrakou A, et al., 2011, Effect of various treatments on leptin, adiponectin, ghrelin and neuropeptide Y in patients with Type 2 diabetes mellitus. Expert Opin Ther Targets, 15: 401–420. https://doi.org/10.1517/14728222.2011.553609

[74]

Katsiki N, Mikhailidis DP, Gotzamani-Psarrakou A, et al., 2011, Effects of improving glycemic control with insulin on leptin, adiponectin, ghrelin and neuropeptide Y levels in patients with Type 2 diabetes mellitus: A pilot study. Open Cardiovasc Med J, 5: 136–147. https://doi.org/10.2174/1874192401105010136 

[75]

Pérez-Pérez A, Vilariño-García T, Fernández-Riejos P, et al., 2017, Role of leptin as a link between metabolism and the immune system. Cytokine Growth Factor Rev, 35: 71–84. https://doi.org/10.1016/j.cytogfr.2017.03.001 

[76]

Liberale L, Bonaventura A, Vecchiè A, et al., 2017, The role of adipocytokines in coronary atherosclerosis. Curr Atheroscler Rep, 19: 10. https://doi.org/10.1007/s11883-017-0644-3

[77]

Beltowski J. 2006, Leptin and atherosclerosis. Atherosclerosis, 189: 47–60. https://doi.org/10.1016/j.atherosclerosis.2006.03.003 

[78]

Tahergorabi Z, Khazaei M. 2015, Leptin and its cardiovascular effects: Focus on angiogenesis. Adv Biomed Res, 4: 79. https://doi.org/10.4103/2277-9175.156526 

[79]

Shäfer K, Halle M, Goeschen C, et al., 2004, Leptin promotes vascular remodeling and neointimal growth in mice. Arterioscler Thromb Vasc Biol, 24: 112–117. https://doi.org/10.1161/01.ATV.0000105904.02142.e7

[80]

Bodary PF, Gu S, Shen Y, et al., 2005, Recombinant leptin promotes atherosclerosis and thrombosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol, 25: e119–e122. https://doi.org/10.1161/01.ATV.0000173306.47722.ec

[81]

Reilly MP, Iqbal N, Schutta M, et al., 2004, Plasma leptin levels are associated with coronary atherosclerosis in Type 2 diabetes. J Clin Endocrinol Metab, 89: 3872–3878. 

[82]

Wallace AM, McMahon AD, Packard CJ, et al., 2001, Plasma leptin and the risk of cardiovascular disease in the West of Scotland Coronary Prevention Study (WOSCOPS). Circulation, 104: 3052–3056. https://doi.org/10.1161/hc5001.101061

[83]

Konstantinides S, Schafer K, Koschnick S, et al., 2001, Leptin-dependent platelet aggregation and arterial thrombosis suggests a mechanism for atherothrombotic disease in obesity. J Clin Invest, 108: 1533–1540. https://doi.org/10.1172/JCI13143

[84]

Loffreda S, Yang SQ, Lin HZ, et al., 1998, Leptin regulates proinflammatory responses. FASEB J, 12: 57–65.

[85]

Yamagishi SI, Edelstein D, Du XL, et al., 2001, Leptin induces mitochondrial superoxide production and monocyte chemoattractant protein-1 expression in aortic endothelial cells by increasing fatty acid oxidation via protein kinase A. J Biol Chem, 276: 25096–25100. https://doi.org/10.1074/jbc.M007383200

[86]

Oda A, Taniguchi T, Yokoyama M. 2001, Leptin stimulates rat aortic smooth muscle cell proliferation and migration. Kobe J Med Sci, 47: 141–150.

[87]

Cassano S, Pucino V, La Rocca C, et al., 2014, Leptin modulates autophagy in human CD4+CD25-conventional T cells. Metabolism, 63: 1272–1279. https://doi.org/10.1016/j.metabol.2014.06.010

[88]

Golledge J, Clancy P, Jamrozik K, et al., 2007, Obesity, adipokines, and abdominal aortic aneurysm: Health in men study. Circulation, 116: 2275–2279. https://doi.org/10.1161/CIRCULATIONAHA.107.717926

Conflict of interest
The paper has no financial interest in any individual or organization; it does not infringe on the intellectual property rights of others. The manuscript and images are original and have not been published before. All authors have no conflicts of interest or financial ties to disclose.
Share
Back to top
Global Translational Medicine, Electronic ISSN: 2811-0021 Published by AccScience Publishing