J Lipid Atheroscler.  2020 Jan;9(1):92-109. 10.12997/jla.2020.9.1.92.

Positioning Metabolism as a Central Player in the Diabetic Heart

Affiliations
  • 1Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK. lisa.heather@dpag.ox.ac.uk

Abstract

In type 2 diabetes (T2D), the leading cause of death is cardiovascular complications. One mechanism contributing to cardiac pathogenesis is alterations in metabolism, with the diabetic heart exhibiting increased fatty acid oxidation and reduced glucose utilisation. The processes classically thought to underlie this metabolic shift include the Randle cycle and changes to gene expression. More recently, alternative mechanisms have been proposed, most notably, changes in post-translational modification of mitochondrial proteins in the heart. This increased understanding of how metabolism is altered in the diabetic heart has highlighted new therapeutic targets, with an aim to improve cardiac function in T2D. This review focuses on metabolism in the healthy heart and how this is modified in T2D, providing evidence for the mechanisms underlying this shift. There will be emphasis on the current treatments for the heart in diabetes, alongside efforts for metabocentric pharmacological therapies.

Keyword

Type 2 diabetes; Cardiac metabolism; Cardiovascular complications; Mitochondrial acetylation

MeSH Terms

Cause of Death
Gene Expression
Glucose
Heart*
Metabolism*
Mitochondrial Proteins
Protein Processing, Post-Translational
Glucose
Mitochondrial Proteins

Figure

  • Fig. 1 Schematic representing glucose and fatty acid metabolism in the healthy heart. Glucose uptake into cardiomyocytes occurs via GLUTs, namely GLUT1 and GLUT4. Inside the cell, glucose is phosphorylated by HK to G-6-P, which is a central intermediate of metabolism and can enter many pathways. One such pathway is glycolysis, whereby glucose is broken down to pyruvate and a small amount of ATP is generated under anaerobic conditions. Pyruvate can then enter the mitochondria for oxidation or be reduced to lactate in the cytoplasm. Mitochondrial PDH catalyses the oxidative decarboxylation of pyruvate to acetyl-CoA, which can then enter the Krebs cycle to generate hydrogen carriers. In the case of FA, uptake across the sarcolemma occurs primarily by the transporter fatty acid translocase (FAT/CD36). Once within the cardiomyocyte, FA are esterified to LCFA-CoA, which enters mitochondria via CPT1 for β-oxidation, or is incorporated into the myocardial TAG pool. The Krebs cycle yields hydrogen carriers for ATP production at the electron transport chain. GLUT, glucose transporter; HK, hexokinase; G-6-P, glucose-6-phosphate; ATP, adenosine triphosphate; PDH, pyruvate dehydrogenase; FA, fatty acids; LCFA-CoA, long chain fatty acyl coenzyme A; CPT1, carnitine palmitoyl transferase 1; TAG, triglyceride.

  • Fig. 2 Cellular mechanisms that favour FA use within the diabetic cardiomyocyte. Intermediates from FA breakdown inhibit components of glucose metabolism. LCFA-CoA can inhibit HK, the primary enzyme involved in glucose breakdown. Acetyl-CoA from increased FA oxidation can also activate PDK, the inhibitor of PDH. This subsequently reduces PDH activity, reducing pyruvate metabolism. Citrate from the Krebs cycle generated by increased FA metabolism can also inhibit PFK in glycolysis. Overall, this reduces glycolytic flux in the cardiomyocyte. Increased LCFA-CoA leads to increased DAG accumulation, which contributes to altered signalling and increased storage of fats as triglycerides. DAG can activate PKCθ, which has been suggested as the enzyme driving lipid-induced insulin resistance. It has been proposed that PKCθ can phosphorylate serine residues on the insulin receptor and its adaptor protein, IRS1/2. This prevents tyrosine phosphorylation, which is necessary for signalling, reducing translocation of vesicles containing GLUT4 to the membrane, reducing insulin-stimulated glucose uptake. FA, fatty acids; LCFA-CoA, long chain fatty acyl coenzyme A; HK, hexokinase; PDK, pyruvate dehydrogenase kinase; PDH, pyruvate dehydrogenase; PFK, phosphofructokinase; DAG, diacylglycerol; PKCθ, protein kinase C theta; IRS1/2, insulin receptor substrates 1/2; GLUT, glucose transporter; G-6-P, glucose-6-phosphate.

  • Fig. 3 Schematic representing changes mediated by PPARα, which is upregulated in the diabetic heart. Upon FA binding, PPARα becomes activated and dimerises with the retinoic acid receptor. This heterodimer can then bind to the PPAR response element and activate a plethora of genes. This includes genes involved in FA uptake, mitochondrial FA uptake and β-oxidation, including fatty acid translocase (FAT/CD36) and CPT1. Furthermore, PPARα also promotes upregulation of PDK, inhibiting PDH and reducing glycolytic flux. PPAR, peroxisome proliferator-activated receptor; FA, fatty acids; CPT1, carnitine palmitoyl transferase 1; PDK, pyruvate dehydrogenase kinase; PDH, pyruvate dehydrogenase; HK, hexokinase; G-6-P, glucose-6-phosphate; ATP, adenosine triphosphate; LCFA-CoA, long chain fatty acyl coenzyme A.

  • Fig. 4 Schematic representing acetylation and deacetylation of mitochondrial proteins. Acetyltransferases mediate the transfer of an acetyl moiety from acetyl CoA onto lysine (K) residues of proteins in the mitochondria. SIRTs mediate deacetylation, which requires NAD+ as a cofactor, and removes the acetyl group from lysine residues. SIRT3 displays the most robust deacetylating capacity of the mitochondrial SIRTs. CoA, coenzyme A; SIRTs, sirtuins; Ac, acetylation.


Reference

1. Diabetes UK. Us, diabetes and a lot of facts and stats [Internet]. London Diabetes UK;2019. cited 3 October 2019. Available from: https://www.diabetes.org.uk/resources-s3/2019-02/1362B_Facts%20and%20stats%20Update%20Jan%202019_LOW%20RES_EXTERNAL.pdf.
2. DeFronzo RA, Ferrannini E, Groop L, Henry RR, Herman WH, Holst JJ, et al. Type 2 diabetes mellitus. Nat Rev Dis Primers. 2015; 1:15019.
Article
3. Shah AD, Langenberg C, Rapsomaniki E, Denaxas S, Pujades-Rodriguez M, Gale CP, et al. Type 2 diabetes and incidence of cardiovascular diseases: a cohort study in 1·9 million people. Lancet Diabetes Endocrinol. 2015; 3:105–113.
Article
4. Chong CR, Clarke K, Levelt E. Metabolic remodeling in diabetic cardiomyopathy. Cardiovasc Res. 2017; 113:422–430.
5. Kodde IF, van der Stok J, Smolenski RT, de Jong JW. Metabolic and genetic regulation of cardiac energy substrate preference. Comp Biochem Physiol A Mol Integr Physiol. 2007; 146:26–39.
Article
6. Heather LC, Clarke K. Metabolism, hypoxia and the diabetic heart. J Mol Cell Cardiol. 2011; 50:598–605.
Article
7. McGill JB, Peterson LR, Herrero P, Saeed IM, Recklein C, Coggan AR, et al. Potentiation of abnormalities in myocardial metabolism with the development of diabetes in women with obesity and insulin resistance. J Nucl Cardiol. 2011; 18:421–429.
Article
8. Mansor LS, Mehta K, Aksentijevic D, Carr CA, Lund T, Cole MA, et al. Increased oxidative metabolism following hypoxia in the type 2 diabetic heart, despite normal hypoxia signalling and metabolic adaptation. J Physiol. 2016; 594:307–320.
Article
9. Mather KJ, Hutchins GD, Perry K, Territo W, Chisholm R, Acton A, et al. Assessment of myocardial metabolic flexibility and work efficiency in human type 2 diabetes using 16-[18F]fluoro-4-thiapalmitate, a novel PET fatty acid tracer. Am J Physiol Endocrinol Metab. 2016; 310:E452–E460.
10. Levelt E, Mahmod M, Piechnik SK, Ariga R, Francis JM, Rodgers CT, et al. Relationship between left ventricular structural and metabolic remodeling in type 2 diabetes. Diabetes. 2016; 65:44–52.
Article
11. Rijzewijk LJ, van der Meer RW, Smit JW, Diamant M, Bax JJ, Hammer S, et al. Myocardial steatosis is an independent predictor of diastolic dysfunction in type 2 diabetes mellitus. J Am Coll Cardiol. 2008; 52:1793–1799.
Article
12. Jagasia D, Whiting JM, Concato J, Pfau S, McNulty PH. Effect of non-insulin-dependent diabetes mellitus on myocardial insulin responsiveness in patients with ischemic heart disease. Circulation. 2001; 103:1734–1739.
Article
13. Desrois M, Sidell RJ, Gauguier D, King LM, Radda GK, Clarke K. Initial steps of insulin signaling and glucose transport are defective in the type 2 diabetic rat heart. Cardiovasc Res. 2004; 61:288–296.
Article
14. Randle PJ, Garland PB, Hales CN, Newsholme EA. The glucose fatty-acid cycle. Its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus. Lancet. 1963; 1:785–789.
Article
15. Thompson AL, Cooney GJ. Acyl-CoA inhibition of hexokinase in rat and human skeletal muscle is a potential mechanism of lipid-induced insulin resistance. Diabetes. 2000; 49:1761–1765.
Article
16. Berg JM, Tymoczko JL, Stryer L. Biochemistry. 5th ed. New York (NY): W H Freeman;2002.
17. Yu C, Chen Y, Cline GW, Zhang D, Zong H, Wang Y, et al. Mechanism by which fatty acids inhibit insulin activation of insulin receptor substrate-1 (IRS-1)-associated phosphatidylinositol 3-kinase activity in muscle. J Biol Chem. 2002; 277:50230–50236.
Article
18. Szendroedi J, Yoshimura T, Phielix E, Koliaki C, Marcucci M, Zhang D, et al. Role of diacylglycerol activation of PKCθ in lipid-induced muscle insulin resistance in humans. Proc Natl Acad Sci U S A. 2014; 111:9597–9602.
Article
19. Samuel VT, Petersen KF, Shulman GI. Lipid-induced insulin resistance: unravelling the mechanism. Lancet. 2010; 375:2267–2277.
Article
20. Pégorier JP, Le May C, Girard J. Control of gene expression by fatty acids. J Nutr. 2004; 134:2444S–2449S.
21. Bookout AL, Jeong Y, Downes M, Yu RT, Evans RM, Mangelsdorf DJ. Anatomical profiling of nuclear receptor expression reveals a hierarchical transcriptional network. Cell. 2006; 126:789–799.
Article
22. Cannon MV, van Gilst WH, de Boer RA. Emerging role of liver X receptors in cardiac pathophysiology and heart failure. Basic Res Cardiol. 2016; 111:3.
Article
23. Cozzone D, Debard C, Dif N, Ricard N, Disse E, Vouillarmet J, et al. Activation of liver X receptors promotes lipid accumulation but does not alter insulin action in human skeletal muscle cells. Diabetologia. 2006; 49:990–999.
Article
24. Osborne TF. Sterol regulatory element-binding proteins (SREBPs): key regulators of nutritional homeostasis and insulin action. J Biol Chem. 2000; 275:32379–32382.
Article
25. Foufelle F, Ferré P. New perspectives in the regulation of hepatic glycolytic and lipogenic genes by insulin and glucose: a role for the transcription factor sterol regulatory element binding protein-1c. Biochem J. 2002; 366:377–391.
Article
26. Varga T, Czimmerer Z, Nagy L. PPARs are a unique set of fatty acid regulated transcription factors controlling both lipid metabolism and inflammation. Biochim Biophys Acta. 2011; 1812:1007–1022.
Article
27. Young ME, McNulty P, Taegtmeyer H. Adaptation and maladaptation of the heart in diabetes. Part II: potential mechanisms. Circulation. 2002; 105:1861–1870.
Article
28. Campbell FM, Kozak R, Wagner A, Altarejos JY, Dyck JR, Belke DD, et al. A role for peroxisome proliferator-activated receptor α (PPARα) in the control of cardiac malonyl-CoA levels: reduced fatty acid oxidation rates and increased glucose oxidation rates in the hearts of mice lacking PPARα are associated with higher concentrations of malonyl-CoA and reduced expression of malonyl-CoA decarboxylase. J Biol Chem. 2002; 277:4098–4103.
Article
29. Finck BN, Han X, Courtois M, Aimond F, Nerbonne JM, Kovacs A, et al. A critical role for PPARα-mediated lipotoxicity in the pathogenesis of diabetic cardiomyopathy: modulation by dietary fat content. Proc Natl Acad Sci U S A. 2003; 100:1226–1231.
Article
30. Parodi-Rullán RM, Chapa-Dubocq XR, Javadov S. Acetylation of mitochondrial proteins in the heart: the role of SIRT3. Front Physiol. 2018; 9:1094.
Article
31. Glozak MA, Sengupta N, Zhang X, Seto E. Acetylation and deacetylation of non-histone proteins. Gene. 2005; 363:15–23.
Article
32. Kim SC, Sprung R, Chen Y, Xu Y, Ball H, Pei J, et al. Substrate and functional diversity of lysine acetylation revealed by a proteomics survey. Mol Cell. 2006; 23:607–618.
Article
33. Venkat S, Gregory C, Sturges J, Gan Q, Fan C. Studying the lysine acetylation of malate dehydrogenase. J Mol Biol. 2017; 429:1396–1405.
Article
34. Still AJ, Floyd BJ, Hebert AS, Bingman CA, Carson JJ, Gunderson DR, et al. Quantification of mitochondrial acetylation dynamics highlights prominent sites of metabolic regulation. J Biol Chem. 2013; 288:26209–26219.
Article
35. Kelley DE, He J, Menshikova EV, Ritov VB. Dysfunction of mitochondria in human skeletal muscle in type 2 diabetes. Diabetes. 2002; 51:2944–2950.
Article
36. Alrob OA, Sankaralingam S, Ma C, Wagg CS, Fillmore N, Jaswal JS, et al. Obesity-induced lysine acetylation increases cardiac fatty acid oxidation and impairs insulin signalling. Cardiovasc Res. 2014; 103:485–497.
Article
37. Fan J, Shan C, Kang HB, Elf S, Xie J, Tucker M, et al. Tyr phosphorylation of PDP1 toggles recruitment between ACAT1 and SIRT3 to regulate the pyruvate dehydrogenase complex. Mol Cell. 2014; 53:534–548.
Article
38. Baeza J, Smallegan MJ, Denu JM. Site-specific reactivity of nonenzymatic lysine acetylation. ACS Chem Biol. 2015; 10:122–128.
Article
39. Jing E, Emanuelli B, Hirschey MD, Boucher J, Lee KY, Lombard D, et al. Sirtuin-3 (Sirt3) regulates skeletal muscle metabolism and insulin signaling via altered mitochondrial oxidation and reactive oxygen species production. Proc Natl Acad Sci U S A. 2011; 108:14608–14613.
Article
40. Pham T, Loiselle D, Power A, Hickey AJ. Mitochondrial inefficiencies and anoxic ATP hydrolysis capacities in diabetic rat heart. Am J Physiol Cell Physiol. 2014; 307:C499–C507.
Article
41. Kuo TH, Moore KH, Giacomelli F, Wiener J. Defective oxidative metabolism of heart mitochondria from genetically diabetic mice. Diabetes. 1983; 32:781–787.
Article
42. Dabkowski ER, Baseler WA, Williamson CL, Powell M, Razunguzwa TT, Frisbee JC, et al. Mitochondrial dysfunction in the type 2 diabetic heart is associated with alterations in spatially distinct mitochondrial proteomes. Am J Physiol Heart Circ Physiol. 2010; 299:H529–H540.
Article
43. Duncan JG. Mitochondrial dysfunction in diabetic cardiomyopathy. Biochim Biophys Acta. 2011; 1813:1351–1359.
Article
44. Sloan RC, Moukdar F, Frasier CR, Patel HD, Bostian PA, Lust RM, et al. Mitochondrial permeability transition in the diabetic heart: contributions of thiol redox state and mitochondrial calcium to augmented reperfusion injury. J Mol Cell Cardiol. 2012; 52:1009–1018.
Article
45. Tsutsui H, Kinugawa S, Matsushima S. Oxidative stress and mitochondrial DNA damage in heart failure. Circ J. 2008; 72 Suppl A:A31–A37.
Article
46. Boudina S, Sena S, Theobald H, Sheng X, Wright JJ, Hu XX, et al. Mitochondrial energetics in the heart in obesity-related diabetes: direct evidence for increased uncoupled respiration and activation of uncoupling proteins. Diabetes. 2007; 56:2457–2466.
Article
47. Rousset S, Alves-Guerra MC, Mozo J, Miroux B, Cassard-Doulcier AM, Bouillaud F, et al. The biology of mitochondrial uncoupling proteins. Diabetes. 2004; 53 Suppl 1:S130–S135.
Article
48. Cole MA, Murray AJ, Cochlin LE, Heather LC, McAleese S, Knight NS, et al. A high fat diet increases mitochondrial fatty acid oxidation and uncoupling to decrease efficiency in rat heart. Basic Res Cardiol. 2011; 106:447–457.
Article
49. Bielawska AE, Shapiro JP, Jiang L, Melkonyan HS, Piot C, Wolfe CL, et al. Ceramide is involved in triggering of cardiomyocyte apoptosis induced by ischemia and reperfusion. Am J Pathol. 1997; 151:1257–1263.
50. Finck BN, Lehman JJ, Leone TC, Welch MJ, Bennett MJ, Kovacs A, et al. The cardiac phenotype induced by PPARα overexpression mimics that caused by diabetes mellitus. J Clin Invest. 2002; 109:121–130.
Article
51. Boardman N, Hafstad AD, Larsen TS, Severson DL, Aasum E. Increased O2 cost of basal metabolism and excitation-contraction coupling in hearts from type 2 diabetic mice. Am J Physiol Heart Circ Physiol. 2009; 296:H1373–H1379.
Article
52. Neubauer S, Horn M, Cramer M, Harre K, Newell JB, Peters W, et al. Myocardial phosphocreatine-to-ATP ratio is a predictor of mortality in patients with dilated cardiomyopathy. Circulation. 1997; 96:2190–2196.
Article
53. Diamant M, Lamb HJ, Groeneveld Y, Endert EL, Smit JW, Bax JJ, et al. Diastolic dysfunction is associated with altered myocardial metabolism in asymptomatic normotensive patients with well-controlled type 2 diabetes mellitus. J Am Coll Cardiol. 2003; 42:328–335.
Article
54. Control Group. Turnbull FM, Abraira C, Anderson RJ, Byington RP, Chalmers JP, et al. Intensive glucose control and macrovascular outcomes in type 2 diabetes. Diabetologia. 2009; 52:2288–2298.
Article
55. Sarma S, Mentz RJ, Kwasny MJ, Fought AJ, Huffman M, Subacius H, et al. Association between diabetes mellitus and post-discharge outcomes in patients hospitalized with heart failure: findings from the EVEREST trial. Eur J Heart Fail. 2013; 15:194–202.
Article
56. Rena G, Hardie DG, Pearson ER. The mechanisms of action of metformin. Diabetologia. 2017; 60:1577–1585.
Article
57. UK Prospective Diabetes Study (UKPDS) Group. Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). Lancet. 1998; 352:854–865.
58. Evans JM, Ogston SA, Emslie-Smith A, Morris AD. Risk of mortality and adverse cardiovascular outcomes in type 2 diabetes: a comparison of patients treated with sulfonylureas and metformin. Diabetologia. 2006; 49:930–936.
59. Eurich DT, Majumdar SR, McAlister FA, Tsuyuki RT, Johnson JA. Improved clinical outcomes associated with metformin in patients with diabetes and heart failure. Diabetes Care. 2005; 28:2345–2351.
Article
60. Andersson C, Søgaard P, Hoffmann S, Hansen PR, Vaag A, Major-Pedersen A, et al. Metformin is associated with improved left ventricular diastolic function measured by tissue Doppler imaging in patients with diabetes. Eur J Endocrinol. 2010; 163:593–599.
Article
61. Bertrand L, Ginion A, Beauloye C, Hebert AD, Guigas B, Hue L, et al. AMPK activation restores the stimulation of glucose uptake in an in vitro model of insulin-resistant cardiomyocytes via the activation of protein kinase B. Am J Physiol Heart Circ Physiol. 2006; 291:H239–H250.
62. Yang J, Holman GD. Long-term metformin treatment stimulates cardiomyocyte glucose transport through an AMP-activated protein kinase-dependent reduction in GLUT4 endocytosis. Endocrinology. 2006; 147:2728–2736.
Article
63. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015; 373:2117–2128.
Article
64. Neal B, Perkovic V, Matthews DR. Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med. 2017; 377:2099.
Article
65. Wiviott SD, Raz I, Sabatine MS. Dapagliflozin and cardiovascular outcomes in type 2 diabetes. Reply. N Engl J Med. 2019; 380:1881–1882.
66. Santos-Gallego CG, Ibanez JA, San Antonio R, Ishikawa K, Watanabe S, Botija MB, et al. Empagliflozin induces a myocardial metabolic shift from glucose consumption to ketone metabolism that mitigates adverse cardiac remodeling and improves myocardial contractility. J Am Coll Cardiol. 2018; 71:A674.
Article
67. Santos-Gallego CG, Requena-Ibanez JA, San Antonio R, Ishikawa K, Picatoste B, Garcia-Ropero J, et al. Infusion of the ketone body β-hydroxybutyrate improves left ventricular systolic function in an animal model of heart failure with reduced ejection fraction. Circulation. 2018; 138:A17367.
Article
68. Garcia-Ropero A, Santos-Gallego CG, Zafar MU, Badimon JJ. Metabolism of the failing heart and the impact of SGLT2 inhibitors. Expert Opin Drug Metab Toxicol. 2019; 15:275–285.
Article
69. Lepore JJ, Olson E, Demopoulos L, Haws T, Fang Z, Barbour AM, et al. Effects of the novel long-acting GLP-1 agonist, albiglutide, on cardiac function, cardiac metabolism, and exercise capacity in patients with chronic heart failure and reduced ejection fraction. JACC Heart Fail. 2016; 4:559–566.
Article
70. Picatoste B, Ramírez E, Caro-Vadillo A, Iborra C, Ares-Carrasco S, Egido J, et al. Sitagliptin reduces cardiac apoptosis, hypertrophy and fibrosis primarily by insulin-dependent mechanisms in experimental type-II diabetes. Potential roles of GLP-1 isoforms. PLoS One. 2013; 8:e78330.
Article
71. Rizzo M, Rizvi AA, Patti AM, Nikolic D, Giglio RV, Castellino G, et al. Liraglutide improves metabolic parameters and carotid intima-media thickness in diabetic patients with the metabolic syndrome: an 18-month prospective study. Cardiovasc Diabetol. 2016; 15:162.
Article
72. Nikolaidis LA, Elahi D, Hentosz T, Doverspike A, Huerbin R, Zourelias L, et al. Recombinant glucagon-like peptide-1 increases myocardial glucose uptake and improves left ventricular performance in conscious dogs with pacing-induced dilated cardiomyopathy. Circulation. 2004; 110:955–961.
Article
73. Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016; 375:311–322.
Article
74. Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jódar E, Leiter LA, et al. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 2016; 375:1834–1844.
Article
75. Holman RR, Bethel MA, Mentz RJ, Thompson VP, Lokhnygina Y, Buse JB, et al. Effects of once-weekly exenatide on cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2017; 377:1228–1239.
Article
76. Margulies KB, Hernandez AF, Redfield MM, Givertz MM, Oliveira GH, Cole R, et al. Effects of liraglutide on clinical stability among patients with advanced heart failure and reduced ejection fraction: a randomized clinical trial. JAMA. 2016; 316:500–508.
Article
77. Tanaka A, Node K. Clinical application of glucagon-like peptide-1 receptor agonists in cardiovascular disease: lessons from recent clinical cardiovascular outcomes trials. Cardiovasc Diabetol. 2018; 17:85.
Article
78. Eldor R, DeFronzo RA, Abdul-Ghani M. In vivo actions of peroxisome proliferator-activated receptors: glycemic control, insulin sensitivity, and insulin secretion. Diabetes Care. 2013; 36 Suppl 2:S162–S174.
79. Yki-Järvinen H. Thiazolidinediones. N Engl J Med. 2004; 351:1106–1118.
Article
80. Dormandy JA, Charbonnel B, Eckland DJ, Erdmann E, Massi-Benedetti M, Moules IK, et al. Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial In macroVascular Events): a randomised controlled trial. Lancet. 2005; 366:1279–1289.
Article
81. Liao HW, Saver JL, Wu YL, Chen TH, Lee M, Ovbiagele B. Pioglitazone and cardiovascular outcomes in patients with insulin resistance, pre-diabetes and type 2 diabetes: a systematic review and meta-analysis. BMJ Open. 2017; 7:e013927.
Article
82. Roumie CL, Hung AM, Greevy RA, Grijalva CG, Liu X, Murff HJ, et al. Comparative effectiveness of sulfonylurea and metformin monotherapy on cardiovascular events in type 2 diabetes mellitus: a cohort study. Ann Intern Med. 2012; 157:601–610.
Article
83. Azoulay L, Suissa S. Sulfonylureas and the risks of cardiovascular events and death: a methodological meta-regression analysis of the observational studies. Diabetes Care. 2017; 40:706–714.
Article
84. Douros A, Dell'Aniello S, Yu OH, Filion KB, Azoulay L, Suissa S. Sulfonylureas as second line drugs in type 2 diabetes and the risk of cardiovascular and hypoglycaemic events: population based cohort study. BMJ. 2018; 362:k2693.
Article
85. Scirica BM, Bhatt DL, Braunwald E, Steg PG, Davidson J, Hirshberg B, et al. Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus. N Engl J Med. 2013; 369:1317–1326.
Article
86. White WB, Cannon CP, Heller SR, Nissen SE, Bergenstal RM, Bakris GL, et al. Alogliptin after acute coronary syndrome in patients with type 2 diabetes. N Engl J Med. 2013; 369:1327–1335.
Article
87. Nicholl TA, Lopaschuk GD, McNeill JH. Effects of free fatty acids and dichloroacetate on isolated working diabetic rat heart. Am J Physiol. 1991; 261:H1053–H1059.
Article
88. Le Page LM, Rider OJ, Lewis AJ, Ball V, Clarke K, Johansson E, et al. Increasing pyruvate dehydrogenase flux as a treatment for diabetic cardiomyopathy: a combined 13C hyperpolarized magnetic resonance and echocardiography study. Diabetes. 2015; 64:2735–2743.
Article
89. Kuda O, Pietka TA, Demianova Z, Kudova E, Cvacka J, Kopecky J, et al. Sulfo-N-succinimidyl oleate (SSO) inhibits fatty acid uptake and signaling for intracellular calcium via binding CD36 lysine 164: SSO also inhibits oxidized low density lipoprotein uptake by macrophages. J Biol Chem. 2013; 288:15547–15555.
Article
90. Mansor LS, Sousa Fialho MD, Yea G, Coumans WA, West JA, Kerr M, et al. Inhibition of sarcolemmal FAT/CD36 by sulfo-N-succinimidyl oleate rapidly corrects metabolism and restores function in the diabetic heart following hypoxia/reoxygenation. Cardiovasc Res. 2017; 113:737–748.
Article
91. Keung W, Ussher JR, Jaswal JS, Raubenheimer M, Lam VH, Wagg CS, et al. Inhibition of carnitine palmitoyltransferase-1 activity alleviates insulin resistance in diet-induced obese mice. Diabetes. 2013; 62:711–720.
Article
92. Haynie KR, Vandanmagsar B, Wicks SE, Zhang J, Mynatt RL. Inhibition of carnitine palymitoyltransferase1b induces cardiac hypertrophy and mortality in mice. Diabetes Obes Metab. 2014; 16:757–760.
Article
93. Mourouzis I, Mantzouratou P, Galanopoulos G, Kostakou E, Dhalla AK, Belardinelli L, et al. The beneficial effects of ranolazine on cardiac function after myocardial infarction are greater in diabetic than in nondiabetic rats. J Cardiovasc Pharmacol Ther. 2014; 19:457–469.
Article
94. Morrow DA, Scirica BM, Chaitman BR, McGuire DK, Murphy SA, Karwatowska-Prokopczuk E, et al. Evaluation of the glycometabolic effects of ranolazine in patients with and without diabetes mellitus in the MERLIN-TIMI 36 randomized controlled trial. Circulation. 2009; 119:2032–2039.
Article
95. Sankaralingam S, Abo Alrob O, Zhang L, Jaswal JS, Wagg CS, Fukushima A, et al. Lowering body weight in obese mice with diastolic heart failure improves cardiac insulin sensitivity and function: implications for the obesity paradox. Diabetes. 2015; 64:1643–1657.
Article
96. Koentges C, Bode C, Bugger H. SIRT3 in cardiac physiology and disease. Front Cardiovasc Med. 2016; 3:38.
Article
97. Pillai VB, Samant S, Sundaresan NR, Raghuraman H, Kim G, Bonner MY, et al. Honokiol blocks and reverses cardiac hypertrophy in mice by activating mitochondrial Sirt3 . Nat Commun. 2015; 6:6656.
Article
98. Pillai VB, Kanwal A, Fang YH, Sharp WW, Samant S, Arbiser J, et al. Honokiol, an activator of sirtuin-3 (SIRT3) preserves mitochondria and protects the heart from doxorubicin-induced cardiomyopathy in mice. Oncotarget. 2017; 8:34082–34098.
Article
Full Text Links
  • JLA
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr