Korean Circ J.  2018 Nov;48(11):951-963. 10.4070/kcj.2018.0307.

Functional Implications of HMG-CoA Reductase Inhibition on Glucose Metabolism

Affiliations
  • 1Department of Internal Medicine, College of Medicine Ulsan University, Asan Medical Center, Seoul, Korea. steadyhan@amc.seoul.kr

Abstract

HMG-CoA reductase inhibitors, i.e. statins, are effective in reducing cardiovascular disease events but also in cardiac-related and overall mortality. Statins are in general well-tolerated, but currently the concerns are raised if statins may increase the risk of new-onset diabetes mellitus (NOD). In this review, the possible effects of statins on organs/tissues being involved in glucose metabolism, i.e. liver, pancreas, adipose tissue, and muscles, had been discussed. The net outcome seems to be inconsistent and often contradictory, which may be largely affected by in vitro experimental settings or/and in vivo animal conditions. The majority of studies point out statin-induced changes of regulations of isoprenoid metabolites and cell-associated cholesterol contents as predisposing factors related to the statin-induced NOD. On the other hand, it should be considered that dysfunctions of isoprenoid pathway and mitochondrial ATP production and the cholesterol homeostasis are already developed under (pre)diabetic and hypercholesterolemic conditions. In order to connect the basic findings with the clinical manifestation more clearly, further research efforts are needed.

Keyword

HMG-CoA reductase; Statins; Diabetes; Insulin sensitivity; Glucogenesis

MeSH Terms

Adenosine Triphosphate
Adipose Tissue
Animals
Cardiovascular Diseases
Causality
Cholesterol
Diabetes Mellitus
Glucose*
Hand
Homeostasis
Hydroxymethylglutaryl-CoA Reductase Inhibitors
In Vitro Techniques
Insulin Resistance
Liver
Metabolism*
Mortality
Muscles
Oxidoreductases*
Pancreas
Social Control, Formal
Adenosine Triphosphate
Cholesterol
Glucose
Hydroxymethylglutaryl-CoA Reductase Inhibitors
Oxidoreductases

Reference

1. Endo A, Hasumi K. Biochemical aspect of HMG CoA reductase inhibitors. Adv Enzyme Regul. 1989; 28:53–64.
Article
2. Stone NJ, Robinson JG, Lichtenstein AH, et al. 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2014; 63:2889–2934.
3. Augustin R. The protein family of glucose transport facilitators: it's not only about glucose after all. IUBMB Life. 2010; 62:315–333.
Article
4. Nordlie RC, Foster JD, Lange AJ. Regulation of glucose production by the liver. Annu Rev Nutr. 1999; 19:379–406.
Article
5. Keembiyehetty C, Augustin R, Carayannopoulos MO, et al. Mouse glucose transporter 9 splice variants are expressed in adult liver and kidney and are up-regulated in diabetes. Mol Endocrinol. 2006; 20:686–697.
Article
6. McVie-Wylie AJ, Lamson DR, Chen YT. Molecular cloning of a novel member of the GLUT family of transporters, SLC2a10 (GLUT10), localized on chromosome 20q13.1: a candidate gene for NIDDM susceptibility. Genomics. 2001; 72:113–117.
Article
7. Elliott KR, Bate AJ, Craik JD. Specificity of the rat hepatocyte monosaccharide transporter. Int J Biochem. 1984; 16:1251–1253.
Article
8. Leturque A, Brot-Laroche E, Le Gall M. GLUT2 mutations, translocation, and receptor function in diet sugar managing. Am J Physiol Endocrinol Metab. 2009; 296:E985–92.
Article
9. Fraulob JC, Souza-Mello V, Aguila MB, Mandarim-de-Lacerda CA. Beneficial effects of rosuvastatin on insulin resistance, adiposity, inflammatory markers and non-alcoholic fatty liver disease in mice fed on a high-fat diet. Clin Sci (Lond). 2012; 123:259–270.
Article
10. Tappy L, Dussoix P, Iynedjian P, et al. Abnormal regulation of hepatic glucose output in maturity-onset diabetes of the young caused by a specific mutation of the glucokinase gene. Diabetes. 1997; 46:204–208.
Article
11. Steele AM, Wensley KJ, Ellard S, et al. Use of HbA1c in the identification of patients with hyperglycaemia caused by a glucokinase mutation: observational case control studies. PLoS One. 2013; 8:e65326.
Article
12. Iynedjian PB, Pilot PR, Nouspikel T, et al. Differential expression and regulation of the glucokinase gene in liver and islets of Langerhans. Proc Natl Acad Sci U S A. 1989; 86:7838–7842.
Article
13. Hegarty BD, Bobard A, Hainault I, Ferré P, Bossard P, Foufelle F. Distinct roles of insulin and liver X receptor in the induction and cleavage of sterol regulatory element-binding protein-1c. Proc Natl Acad Sci U S A. 2005; 102:791–796.
14. Kim TH, Kim H, Park JM, et al. Interrelationship between liver X receptor alpha, sterol regulatory element-binding protein-1c, peroxisome proliferator-activated receptor gamma, and small heterodimer partner in the transcriptional regulation of glucokinase gene expression in liver. J Biol Chem. 2009; 284:15071–15083.
15. Pramfalk C, Parini P, Gustafsson U, Sahlin S, Eriksson M. Effects of high-dose statin on the human hepatic expression of genes involved in carbohydrate and triglyceride metabolism. J Intern Med. 2011; 269:333–339.
Article
16. O'Doherty RM, Lehman DL, Télémaque-Potts S, Newgard CB. Metabolic impact of glucokinase overexpression in liver: lowering of blood glucose in fed rats is accompanied by hyperlipidemia. Diabetes. 1999; 48:2022–2027.
17. Chakera AJ, Steele AM, Gloyn AL, et al. Recognition and management of individuals with hyperglycemia because of a heterozygous glucokinase mutation. Diabetes Care. 2015; 38:1383–1392.
Article
18. Schonewille M, de Boer JF, Mele L, et al. Statins increase hepatic cholesterol synthesis and stimulate fecal cholesterol elimination in mice. J Lipid Res. 2016; 57:1455–1464.
Article
19. Tsai AC, Dyer IA. Influence of dietary cholesterol and cholic acid on liver carbohydrate metabolism enzymes in rats. J Nutr. 1973; 103:93–101.
Article
20. Miao J, Haas JT, Manthena P, et al. Hepatic insulin receptor deficiency impairs the SREBP-2 response to feeding and statins. J Lipid Res. 2014; 55:659–667.
Article
21. Naples M, Federico LM, Xu E, Nelken J, Adeli K. Effect of rosuvastatin on insulin sensitivity in an animal model of insulin resistance: evidence for statin-induced hepatic insulin sensitization. Atherosclerosis. 2008; 198:94–103.
Article
22. Lalli CA, Pauli JR, Prada PO, et al. Statin modulates insulin signaling and insulin resistance in liver and muscle of rats fed a high-fat diet. Metabolism. 2008; 57:57–65.
Article
23. Gotoh S, Negishi M. Statin-activated nuclear receptor PXR promotes SGK2 dephosphorylation by scaffolding PP2C to induce hepatic gluconeogenesis. Sci Rep. 2015; 5:14076.
Article
24. Black RN, Ennis CN, Young IS, Hunter SJ, Atkinson AB, Bell PM. The peroxisome proliferator-activated receptor alpha agonist fenofibrate has no effect on insulin sensitivity compared to atorvastatin in type 2 diabetes mellitus; a randomised, double-blind controlled trial. J Diabetes Complications. 2014; 28:323–327.
Article
25. Szendroedi J, Anderwald C, Krssak M, et al. Effects of high-dose simvastatin therapy on glucose metabolism and ectopic lipid deposition in nonobese type 2 diabetic patients. Diabetes Care. 2009; 32:209–214.
Article
26. Zhao W, Zhao SP. Different effects of statins on induction of diabetes mellitus: an experimental study. Drug Des Devel Ther. 2015; 9:6211–6223.
27. Coppieters KT, Wiberg A, Amirian N, Kay TW, von Herrath MG. Persistent glucose transporter expression on pancreatic beta cells from longstanding type 1 diabetic individuals. Diabetes Metab Res Rev. 2011; 27:746–754.
Article
28. Liang Y, Cushman SM, Whitesell RR, Matschinsky FM. GLUT1 is adequate for glucose uptake in GLUT2-deficient insulin-releasing β-cells. Horm Metab Res. 1997; 29:255–260.
Article
29. Fex M, Nicholas LM, Vishnu N, et al. The pathogenetic role of β-cell mitochondria in type 2 diabetes. J Endocrinol. 2018; 236:R145–59.
Article
30. Zhang Y, Feng F, Chen T, Li Z, Shen QW. Antidiabetic and antihyperlipidemic activities of Forsythia suspensa (Thunb.) Vahl (fruit) in streptozotocin-induced diabetes mice. J Ethnopharmacol. 2016; 192:256–263.
Article
31. Hao M, Head WS, Gunawardana SC, Hasty AH, Piston DW. Direct effect of cholesterol on insulin secretion: a novel mechanism for pancreatic beta-cell dysfunction. Diabetes. 2007; 56:2328–2338.
32. The Human Protein Atlas. Gene information: LDLR [Internet]. place unknown: The Human Protein Atlas;cited 2018 Aug. Available from https://www.proteinatlas.org/ENSG00000130164-LDLR/tissue.
33. Ivarsson R, Quintens R, Dejonghe S, et al. Redox control of exocytosis: regulatory role of NADPH, thioredoxin, and glutaredoxin. Diabetes. 2005; 54:2132–2142.
Article
34. Henquin JC. Triggering and amplifying pathways of regulation of insulin secretion by glucose. Diabetes. 2000; 49:1751–1760.
Article
35. MacDonald MJ, Longacre MJ, Langberg EC, et al. Decreased levels of metabolic enzymes in pancreatic islets of patients with type 2 diabetes. Diabetologia. 2009; 52:1087–1091.
Article
36. Lupi R, Marselli L, Dionisi S, et al. Improved insulin secretory function and reduced chemotactic properties after tissue culture of islets from type 1 diabetic patients. Diabetes Metab Res Rev. 2004; 20:246–251.
Article
37. Anello M, Lupi R, Spampinato D, et al. Functional and morphological alterations of mitochondria in pancreatic beta cells from type 2 diabetic patients. Diabetologia. 2005; 48:282–289.
Article
38. Brand MD, Parker N, Affourtit C, Mookerjee SA, Azzu V. Mitochondrial uncoupling protein 2 in pancreatic β-cells. Diabetes Obes Metab. 2010; 12:Suppl 2. 134–140.
Article
39. Yang C, Zhao D, Liu G, et al. Atorvastatin attenuates metabolic remodeling in ischemic myocardium through the downregulation of UCP2 expression. Int J Med Sci. 2018; 15:517–527.
Article
40. Wang L, Lin R, Guo L, Hong M. Rosuvastatin relieves myocardial ischemia/reperfusion injury by upregulating PPAR-γ and UCP2. Mol Med Rep. 2018; 18:789–798.
Article
41. Urbano F, Bugliani M, Filippello A, et al. Atorvastatin but not pravastatin impairs mitochondrial function in human pancreatic islets and rat β-cells. Direct effect of oxidative stress. Sci Rep. 2017; 7:11863.
Article
42. Yaluri N, Modi S, López Rodríguez M, et al. Simvastatin impairs insulin secretion by multiple mechanisms in MIN6 cells. PLoS One. 2015; 10:e0142902.
Article
43. Salunkhe VA, Elvstam O, Eliasson L, Wendt A. Rosuvastatin treatment affects both basal and glucose-induced insulin secretion in INS-1 832/13 cells. PLoS One. 2016; 11:e0151592.
Article
44. Zaki NM. Strategies for oral delivery and mitochondrial targeting of CoQ10. Drug Deliv. 2016; 23:1868–1881.
Article
45. Chew GT, Watts GF. Coenzyme Q10 and diabetic endotheliopathy: oxidative stress and the ‘recoupling hypothesis’. QJM. 2004; 97:537–548.
Article
46. Tomita T. Apoptosis in pancreatic β-islet cells in Type 2 diabetes. Bosn J Basic Med Sci. 2016; 16:162–179.
Article
47. Sadighara M, Amirsheardost Z, Minaiyan M, et al. Toxicity of atorvastatin on pancreas mitochondria: a justification for increased risk of diabetes mellitus. Basic Clin Pharmacol Toxicol. 2017; 120:131–137.
Article
48. Chen ZY, Liu SN, Li CN, et al. Atorvastatin helps preserve pancreatic β cell function in obese C57BL/6 J mice and the effect is related to increased pancreas proliferation and amelioration of endoplasmic-reticulum stress. Lipids Health Dis. 2014; 13:98.
49. Otani M, Yamamoto M, Harada M, Otsuki M. Effect of long- and short-term treatments with pravastatin on diabetes mellitus and pancreatic fibrosis in the Otsuka-Long-Evans-Tokushima fatty rat. Br J Pharmacol. 2010; 159:462–473.
Article
50. Lorza-Gil E, Salerno AG, Wanschel AC, et al. Chronic use of pravastatin reduces insulin exocytosis and increases β-cell death in hypercholesterolemic mice. Toxicology. 2016; 344-346:42–52.
Article
51. Contreras JL, Smyth CA, Bilbao G, Young CJ, Thompson JA, Eckhoff DE. Simvastatin induces activation of the serine-threonine protein kinase AKT and increases survival of isolated human pancreatic islets. Transplantation. 2002; 74:1063–1069.
Article
52. Mohammed A, Qian L, Janakiram NB, Lightfoot S, Steele VE, Rao CV. Atorvastatin delays progression of pancreatic lesions to carcinoma by regulating PI3/AKT signaling in p48Cre/+ LSL-KrasG12D/+ mice. Int J Cancer. 2012; 131:1951–1962.
53. Favaro E, Miceli I, Bussolati B, et al. Hyperglycemia induces apoptosis of human pancreatic islet endothelial cells: effects of pravastatin on the Akt survival pathway. Am J Pathol. 2008; 173:442–450.
54. De Meyts P. The insulin receptor and its signal transduction network. In : De Groot LJ, Chrousos G, Dungan K, editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.;2000-. updated 2016 Apr 27. cited 2018 Aug. Available from https://www.ncbi.nlm.nih.gov/books/NBK378978/.
55. Jiang Z, Yu B, Li Y. Effect of three statins on glucose uptake of cardiomyocytes and its mechanism. Med Sci Monit. 2016; 22:2825–2830.
Article
56. Li W, Liang X, Zeng Z, et al. Simvastatin inhibits glucose uptake activity and GLUT4 translocation through suppression of the IR/IRS-1/Akt signaling in C2C12 myotubes. Biomed Pharmacother. 2016; 83:194–200.
Article
57. Yaluri N, Modi S, Kokkola T. Simvastatin induces insulin resistance in L6 skeletal muscle myotubes by suppressing insulin signaling, GLUT4 expression and GSK-3β phosphorylation. Biochem Biophys Res Commun. 2016; 480:194–200.
Article
58. Chamberlain LH. Inhibition of isoprenoid biosynthesis causes insulin resistance in 3T3-L1 adipocytes. FEBS Lett. 2001; 507:357–361.
Article
59. Takaguri A, Satoh K, Itagaki M, Tokumitsu Y, Ichihara K. Effects of atorvastatin and pravastatin on signal transduction related to glucose uptake in 3T3L1 adipocytes. J Pharmacol Sci. 2008; 107:80–89.
Article
60. Khan T, Hamilton MP, Mundy DI, Chua SC, Scherer PE. Impact of simvastatin on adipose tissue: pleiotropic effects in vivo. Endocrinology. 2009; 150:5262–5272.
Article
61. Sahebkar A, Simental-Mendía LE, Pedone C, et al. Statin therapy and plasma free fatty acids: a systematic review and meta-analysis of controlled clinical trials. Br J Clin Pharmacol. 2016; 81:807–818.
Article
62. Berk-Planken II, Hoogerbrugge N, Stolk RP, Bootsma AH, Jansen H. DALI Study Group. Atorvastatin dose-dependently decreases hepatic lipase activity in type 2 diabetes: effect of sex and the LIPC promoter variant. Diabetes Care. 2003; 26:427–432.
Article
63. Bey L, Maigret P, Laouenan H, Hamilton MT. Induction of lipoprotein lipase gene expression in 3T3-L1 preadipocytes by atorvastatin, a cholesterol- and triglyceride-lowering drug. Pharmacology. 2002; 66:51–56.
Article
64. Ohira M, Endo K, Saiki A, et al. Atorvastatin and pitavastatin enhance lipoprotein lipase production in L6 skeletal muscle cells through activation of adenosine monophosphate-activated protein kinase. Metabolism. 2012; 61:1452–1460.
Article
65. Roden M. How free fatty acids inhibit glucose utilization in human skeletal muscle. News Physiol Sci. 2004; 19:92–96.
Article
66. Williams ML, Menon GK, Hanley KP. HMG-CoA reductase inhibitors perturb fatty acid metabolism and induce peroxisomes in keratinocytes. J Lipid Res. 1992; 33:193–208.
Article
67. Murthy S, Tong H, Hohl RJ. Regulation of fatty acid synthesis by farnesyl pyrophosphate. J Biol Chem. 2005; 280:41793–41804.
Article
68. Chruściel P, Sahebkar A, Rembek-Wieliczko M, et al. Impact of statin therapy on plasma adiponectin concentrations: a systematic review and meta-analysis of 43 randomized controlled trial arms. Atherosclerosis. 2016; 253:194–208.
Article
69. Sahebkar A, Giua R, Pedone C. Impact of statin therapy on plasma leptin concentrations: a systematic review and meta-analysis of randomized placebo-controlled trials. Br J Clin Pharmacol. 2016; 82:1674–1684.
Article
70. Sahebkar A, Giorgini P, Ludovici V, et al. Impact of statin therapy on plasma resistin and visfatin concentrations: a systematic review and meta-analysis of controlled clinical trials. Pharmacol Res. 2016; 111:827–837.
Article
71. Singh P, Zhang Y, Sharma P, et al. Statins decrease leptin expression in human white adipocytes. Physiol Rep. 2018; 6:e13566.
Article
72. Kralova Lesna I, Petras M, Cejkova S, et al. Cardiovascular disease predictors and adipose tissue macrophage polarization: is there a link? Eur J Prev Cardiol. 2018; 25:328–334.
Article
73. Abe M, Matsuda M, Kobayashi H, et al. Effects of statins on adipose tissue inflammation: their inhibitory effect on MyD88-independent IRF3/IFN-β pathway in macrophages. Arterioscler Thromb Vasc Biol. 2008; 28:871–877.
74. Henriksbo BD, Schertzer JD. Is immunity a mechanism contributing to statin-induced diabetes? Adipocyte. 2015; 4:232–238.
Article
75. Henriksbo BD, Lau TC, Cavallari JF, et al. Fluvastatin causes NLRP3 inflammasome-mediated adipose insulin resistance. Diabetes. 2014; 63:3742–3747.
Article
76. Singh LP. The NLRP3 inflammasome and diabetic cardiomyopathy: editorial to: “Rosuvastatin alleviates diabetic cardiomyopathy by inhibiting NLRP3 inflammasome and MAPK pathways in a type 2 diabetes rat model” by Beibei Luo et al. Cardiovasc Drugs Ther. 2014; 28:5–6.
77. Luo B, Li B, Wang W, et al. Rosuvastatin alleviates diabetic cardiomyopathy by inhibiting NLRP3 inflammasome and MAPK pathways in a type 2 diabetes rat model. Cardiovasc Drugs Ther. 2014; 28:33–43.
Article
78. Altaf A, Qu P, Zhao Y, Wang H, Lou D, Niu N. NLRP3 inflammasome in peripheral blood monocytes of acute coronary syndrome patients and its relationship with statins. Coron Artery Dis. 2015; 26:409–421.
Article
79. Kong F, Ye B, Lin L, Cai X, Huang W, Huang Z. Atorvastatin suppresses NLRP3 inflammasome activation via TLR4/MyD88/NF-κB signaling in PMA-stimulated THP-1 monocytes. Biomed Pharmacother. 2016; 82:167–172.
Article
80. Yu SY, Tang L, Zhao GJ, Zhou SH. Statin protects the heart against ischemia-reperfusion injury via inhibition of the NLRP3 inflammasome. Int J Cardiol. 2017; 229:23–24.
Article
81. Wang S, Xie X, Lei T, et al. Statins attenuate activation of the NLRP3 inflammasome by oxidized LDL or TNFα in vascular endothelial cells through a PXR-dependent mechanism. Mol Pharmacol. 2017; 92:256–264.
Article
82. Gazzerro P, Proto MC, Gangemi G, et al. Pharmacological actions of statins: a critical appraisal in the management of cancer. Pharmacol Rev. 2012; 64:102–146.
Article
Full Text Links
  • KCJ
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