Best exercise for metabolic syndrome

Exercise is known to prevent or treat metabolic syndrome, which is a group of health conditions that raise the risk of heart problems and type 2 diabetes. Now, a new study suggests that less than 1 hour per week of resistance training, even without aerobic exercise, can be of benefit.

Share on PinterestNew research has shown that a person’s risk of developing metabolic syndrome can be reduced with modest resistance training.

Lead author Esmée Bakker, of Radboud University Medical Center in Nijmegen in the Netherlands, and colleagues report their findings in the journal Mayo Clinic Proceedings.

Metabolic syndrome is a cluster of health risk factors that make it more likely that a person will develop type 2 diabetes and cardiovascular diseases such as heart disease and stroke.

To be diagnosed with metabolic syndrome, a person must have at least three of the risk factors, which include: a large waistline, high levels of triglycerides (a type of blood fat), low levels of high-density lipoprotein (“good” cholesterol), high blood pressure, and high blood sugar.

Today, metabolic syndrome affects more than 1 in 3 adults in the United States, up from 1 in 4 in the 1990s. It is thought that this rate is likely to continue rising as the U.S. population ages.

In their study paper, Bakker and colleagues explain that increasing exercise is a “cornerstone for preventing and treating” metabolic syndrome, and they cite several studies that show the benefits of aerobic exercise – such as running and cycling – in reducing metabolic risk factors.

Some studies have also found that higher levels of resistance exercise are tied to lower risk of type 2 diabetes in men and women.

However, the authors note that while there is evidence that higher levels of resistance exercise or weight training are linked to lower incidence of metabolic syndrome, this has largely come from cross-sectional studies and not from following large groups over a period of time to see how exercise habits link to the emergence of a condition.

Therefore, they decided to explore the relationship between resistance training – separate from, and together with, aerobic exercise – and the development of metabolic syndrome.

For their analysis, the team used data from the Aerobics Center Longitudinal Study on more than 7,000 adults. The average age of the participants was 46 years and 19 percent were women.

The participants underwent extensive medical exams between 1987 and 2006 and completed questionnaires about frequency and intensity of resistance and aerobic exercise. All were free of metabolic syndrome when they enrolled.

Of the 7,418 participants that the researchers included in the analysis, 1,147 (15 percent) of them developed metabolic syndrome during their follow-up. While the maximum follow-up was 19 years, the median follow-up was 4 years.

The researchers found that any amount of resistance training that met the 2008 U.S. guidelines for physical activity was linked to a 17 percent lower risk of developing metabolic syndrome, compared with not doing any at all.

The U.S. guidelines suggest that adults, “Do muscle-strengthening activities (such as lifting weights or using resistance bands) that are moderate or high intensity and involve all major muscle groups on 2 or more days a week.”

The analysis also showed that doing up to an hour each week of resistance training was linked to a 29 percent lower risk of developing metabolic syndrome, even after adjusting for other potential influencers such as smoking status and endurance training.

More intensive resistance training appeared to show no additional benefits in relation to metabolic risk. Also, whether participants did their resistance exercise in one or two sessions at the weekend or spread it over the week appeared to make no difference to the results.

However, the greatest benefits – in terms of preventing metabolic syndrome – appeared to come from combining resistance training with aerobic exercise, note the researchers.

Our results indicate that a modest amount of resistance exercise, such as two 30-minute sessions per week, has the most beneficial effect. These findings should be included in the standard medical recommendations for preventing metabolic syndrome and future cardiovascular disease.”

Esmée Bakker

Learn how lack of sleep can raise death risk in people with metabolic syndrome.

1. Fujita T. Insulin resistance and salt-sensitive hypertension in metabolic syndrome. Nephrology Dialysis Transplantation. 2007;22(11):3102–3107. [PubMed] [Google Scholar]

2. Fujita T. Aldosterone in salt-sensitive hypertension and metabolic syndrome. Journal of Molecular Medicine. 2008;86(6):729–734. [PubMed] [Google Scholar]

3. Eckel RH, Grundy SM, Zimmet PZ. The metabolic syndrome. The Lancet. 2005;365(9468):1415–1428. [PubMed] [Google Scholar]

4. Park YW, Zhu S, Palaniappan L, Heshka S, Carnethon MR, Heymsfield SB. The metabolic syndrome: prevalence and associated risk factor findings in the US population from the Third National Health and Nutrition Examination Survey, 1988–1994. Archives of Internal Medicine. 2003;163(4):427–436. [PMC free article] [PubMed] [Google Scholar]

5. Caterson ID, Hubbard V, Bray GA, et al. Prevention conference VII: obesity, a worldwide epidemic related to heart disease and stroke: group III: worldwide comorbidities of obesity. Circulation. 2004;110(18):e476–e483. [PubMed] [Google Scholar]

6. Eckel RH, Grundy SM, Zimmet PZ. The metabolic syndrome. The Lancet. 2005;365(9468):1415–1428. [PubMed] [Google Scholar]

7. Galassi A, Reynolds K, He J. Metabolic syndrome and risk of cardiovascular disease: a meta-analysis. American Journal of Medicine. 2006;119(10):812–819. [PubMed] [Google Scholar]

8. Reaven GM. Role of insulin resistance in human disease. Diabetes. 1988;37(12):1595–1607. [PubMed] [Google Scholar]

9. Reaven GM. Insulin resistance: the link between obesity and cardiovascular disease. Medical Clinics of North America. 2011;95:875–892. [PubMed] [Google Scholar]

10. Uretsky S, Messerli FH, Bangalore S, et al. Obesity paradox in patients with hpertension and coronary artery disease. American Journal of Medicine. 2007;120(10):863–870. [PubMed] [Google Scholar]

11. Aguilar-Salinas CA, García E, Robles L, et al. High adiponectin concentrations are associated with the metabolically healthy obese phenotype. Journal of Clinical Endocrinology and Metabolism. 2008;93(10):4075–4079. [PubMed] [Google Scholar]

12. Wildman RP, Muntner P, Reynolds K, et al. The obese without cardiometabolic risk factor clustering and the normal weight with cardiometabolic risk factor clustering: prevalence and correlates of 2 phenotypes among the US population (NHANES 1999–2004) Archives of Internal Medicine. 2008;168(15):1617–1624. [PubMed] [Google Scholar]

13. Stefan N, Kantartzis K, Machann J, et al. Identification and characterization of metabolically benign obesity in humans. Archives of Internal Medicine. 2008;168(15):1609–1616. [PubMed] [Google Scholar]

14. Wildman RP. Healthy obesity. Current Opinion in Clinical Nutrition & Metabolic Care. 2009;12:438–443. [PubMed] [Google Scholar]

15. Van Gaal LF, Mertens IL, De Block CE. Mechanisms linking obesity with cardiovascular disease. Nature. 2006;444(7121):875–880. [PubMed] [Google Scholar]

16. Després JP, Lemieux I. Abdominal obesity and metabolic syndrome. Nature. 2006;444(7121):881–887. [PubMed] [Google Scholar]

17. Abate N, Garg A, Peshock RM, Stray-Gundersen J, Grundy SM. Relationships of generalized and regional adiposity to insulin sensitivity in men. Journal of Clinical Investigation. 1995;96(1):88–98. [PMC free article] [PubMed] [Google Scholar]

18. Fernández-Real JM, Ricart W. Insulin resistance and chronic cardiovascular inflammatory syndrome. Endocrine Reviews. 2003;24(3):278–301. [PubMed] [Google Scholar]

19. Welsh P, Polisecki E, Robertson M, et al. Unraveling the directional link between adiposity and inflammation: a bidirectional mendelian randomization approach. Journal of Clinical Endocrinology and Metabolism. 2010;95(1):93–99. [PMC free article] [PubMed] [Google Scholar]

20. Wellen KE, Hotamisligil GS. Inflammation, stress, and diabetes. Journal of Clinical Investigation. 2005;115(5):1111–1119. [PMC free article] [PubMed] [Google Scholar]

21. Stocker R, Keaney JF. Role of oxidative modifications in atherosclerosis. Physiological Reviews. 2004;84(4):1381–1478. [PubMed] [Google Scholar]

22. Furukawa S, Fujita T, Shimabukuro M, et al. Increased oxidative stress in obesity and its impact on metabolic syndrome. Journal of Clinical Investigation. 2004;114(12):1752–1761. [PMC free article] [PubMed] [Google Scholar]

23. Keaney JF, Jr., Larson MG, Vasan RS, et al. Obesity and systemic oxidative stress: clinical correlates of oxidative stress in the Framingham study. Arteriosclerosis, Thrombosis, and Vascular Biology. 2003;23(3):434–439. [PubMed] [Google Scholar]

24. Pinzani M, Marra F, Carloni V. Signal transduction in hepatic stellate cells. Liver. 1998;18(1):2–13. [PubMed] [Google Scholar]

25. Gastaldelli A, Basta G. Ectopic fat and cardiovascular disease: what is the link? Nutrition, Metabolism and Cardiovascular Diseases. 2010;20(7):481–490. [PubMed] [Google Scholar]

26. Sironi AM, Gastaldelli A, Mari A, et al. Visceral fat in hypertension: influence on insulin resistance and β-cell function. Hypertension. 2004;44(2):127–133. [PubMed] [Google Scholar]

27. Iacobellis G, Ribaudo MC, Assael F, et al. Echocardiographic epicardial adipose tissue is related to anthropometric and clinical parameters of metabolic syndrome: a new indicator of cardiovascular risk. Journal of Clinical Endocrinology and Metabolism. 2003;88(11):5163–5168. [PubMed] [Google Scholar]

28. Mazurek T, Zhang L, Zalewski A, et al. Human epicardial adipose tissue is a source of inflammatory mediators. Circulation. 2003;108(20):2460–2466. [PubMed] [Google Scholar]

29. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-α: direct role in obesity-linked insulin resistance. Science. 1993;259(5091):87–91. [PubMed] [Google Scholar]

30. Kern PA, Saghizadeh M, Ong JM, Bosch RJ, Deem R, Simsolo RB. The expression of tumor necrosis factor in human adipose tissue. Regulation by obesity, weight loss, and relationship to lipoprotein lipase. Journal of Clinical Investigation. 1995;95(5):2111–2119. [PMC free article] [PubMed] [Google Scholar]

31. Diehl AM. Tumor necrosis factor and its potential role in insulin resistance and nonalcoholic fatty liver disease. Clinics in Liver Disease. 2004;8(3):619–638. [PubMed] [Google Scholar]

32. Uysal KT, Wiesbrock SM, Marino MW, Hotamisligil GS. Protection from obesity-induced insulin resistance in mice lacking TNF- α function. Nature. 1997;389(6651):610–614. [PubMed] [Google Scholar]

33. Uysal KT, Wiesbrock SM, Hotamisligil GS. Functional analysis of tumor necrosis factor (TNF) receptors in TNF-α- mediated insulin resistance in genetic obesity. Endocrinology. 1998;139(12):4832–4838. [PubMed] [Google Scholar]

34. Katsuki A, Sumida Y, Urakawa H, et al. Increased visceral fat and serum levels of triglyceride are associated with insulin resistance in Japanese metabolically obese, normal weight subjects with normal glucose tolerance. Diabetes Care. 2003;26(8):2341–2344. [PubMed] [Google Scholar]

35. Gabriely I, Barzilai N. Surgical removal of visceral adipose tissue: effects on insulin action. Current Diabetes Reports. 2003;3(3):201–206. [PubMed] [Google Scholar]

36. Chaldakov GN, Stankulov IS, Hristova M, Ghenev PI. Adipobiology of disease: adipokines and adipokine-targeted pharmacology. Current Pharmaceutical Design. 2003;9(12):1023–1031. [PubMed] [Google Scholar]

37. Berk PD, Zhou SL, Bradbury M, Stump D, Kiang CL, Isola LM. Regulated membrane transport of free fatty acids in adipocytes: role in obesity and non-insulin dependent diabetes mellitus. Transactions of the American Clinical and Climatological Association. 1996;108:26–43. [PMC free article] [PubMed] [Google Scholar]

38. Robertson G, Leclercq I, Farrell GC. Nonalcoholic steatosis and steatohepatitis. II. Cytochrome P-450 enzymes and oxidative stress. American Journal of Physiology. 2001;281(5):G1135–G1139. [PubMed] [Google Scholar]

39. Hashimoto T, Fujita T, Usuda N, et al. Peroxisomal and mitochondrial fatty acid β-oxidation in mice nullizygous for both peroxisome proliferator-activated receptor and peroxisomal fatty acyl-CoA oxidase: genotype correlation with fatty liver phenotype. Journal of Biological Chemistry. 1999;274(27):19228–19236. [PubMed] [Google Scholar]

40. Schulze-Osthoff K, Bakker AC, Vanhaesebroeck B, Beyaert R, Jacob WA, Fiers W. Cytotoxic activity of tumor necrosis factor is mediated by early damage of mitochondrial functions. Evidence for the involvement of mitochondrial radical generation. Journal of Biological Chemistry. 1992;267(8):5317–5323. [PubMed] [Google Scholar]

41. Bournat JC, Brown CW. Mitochondrial dysfunction in obesity. Current Opinion in Endocrinology, Diabetes and Obesity. 2010;17(5):446–452. [PMC free article] [PubMed] [Google Scholar]

42. Lemasters JJ, Qian T, Bradham CA, et al. Mitochondrial dysfunction in the pathogenesis of necrotic and apoptotic cell death. Journal of Bioenergetics and Biomembranes. 1999;31(4):305–319. [PubMed] [Google Scholar]

43. Joshi-Barve S, Barve SS, Butt W, Klein J, McClain CJ. Inhibition of proteasome function leads to NF-κB-independent IL-8 expression in human hepatocytes. Hepatology. 2003;38(5):1178–1187. [PubMed] [Google Scholar]

44. Monteiro R, Azevedo I. Chronic inflammation in obesity and the metabolic syndrome. Mediators of Inflammation. 2010;2010:10 pages.289645 [PMC free article] [PubMed] [Google Scholar]

45. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444(7121):860–867. [PubMed] [Google Scholar]

46. Ruan H, Lodish HF. Insulin resistance in adipose tissue: direct and indirect effects of tumor necrosis factor-α Cytokine and Growth Factor Reviews. 2003;14(5):447–455. [PubMed] [Google Scholar]

47. Frayn KN, Karpe F, Fielding BA, Macdonald IA, Coppack SW. Integrative physiology of human adipose tissue. International Journal of Obesity. 2003;27(8):875–888. [PubMed] [Google Scholar]

48. Ronti T, Lupattelli G, Mannarino E. The endocrine function of adipose tissue: an update. Clinical Endocrinology. 2006;64(4):355–365. [PubMed] [Google Scholar]

49. Yamauchi T, Kamon J, Ito Y, et al. Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature. 2003;423(6941):762–769. [PubMed] [Google Scholar]

50. Yamauchi T, Kamon J, Minokoshi Y, et al. Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nature Medicine. 2002;8(11):1288–1295. [PubMed] [Google Scholar]

51. Grattagliano I, Palmieri VO, Portincasa P, Moschetta A, Palasciano G. Oxidative stress-induced risk factors associated with the metabolic syndrome: a unifying hypothesis. Journal of Nutritional Biochemistry. 2008;19(8):491–504. [PubMed] [Google Scholar]

52. Roberts CK, Barnard RJ, Sindhu RK, Jurczak M, Ehdaie A, Vaziri ND. A high-fat, refined-carbohydrate diet induces endothelial dysfunction and oxidant/antioxidant imbalance and depresses NOS protein expression. Journal of Applied Physiology. 2005;98(1):203–210. [PubMed] [Google Scholar]

53. Kim HC, Choi SH, Shin HW, et al. Severity of ultrasonographic liver steatosis and metabolic syndrome in Korean men and women. World Journal of Gastroenterology. 2005;11(34):5314–5321. [PMC free article] [PubMed] [Google Scholar]

54. Diehl AM. Fatty liver, hypertension, and the metabolic syndrome. Gut. 2004;53(7):923–924. [PMC free article] [PubMed] [Google Scholar]

55. Vega-López S, Devaraj S, Jialal I. Oxidative stress and antioxidant-supplementation in the management of diabetic cardiovascular disease. Journal of Investigative Medicine. 2004;52(1):24–32. [PubMed] [Google Scholar]

56. Ceriello A, Mercuri F, Quagliaro L, et al. Detection of nitrotyrosine in the diabetic plasma: evidence of oxidative stress. Diabetologia. 2001;44(7):834–838. [PubMed] [Google Scholar]

57. Turko IV, Marcondes S, Murad F. Diabetes-associated nitration of tyrosine and inactivation of succinyl-CoA:3-oxoacid CoA-transferase. American Journal of Physiology. 2001;281(6):H2289–H2294. [PubMed] [Google Scholar]

58. Guzik TJ, West NE, Black E, et al. Vascular superoxide production by NAD(P)H oxidase: association with endothelial dysfunction and clinical risk factors. Circulation Research. 2000;86(9):E85–E90. [PubMed] [Google Scholar]

59. Guzik TJ, Mussa S, Gastaldi D, et al. Mechanisms of increased vascular superoxide production in human diabetes mellitus: role of NAD(P)H oxidase and endothelial nitric oxide synthase. Circulation. 2002;105(14):1656–1662. [PubMed] [Google Scholar]

60. Aliciguzel Y, Ozen I, Aslan M, Karayalcin U. Activities of xanthine oxidoreductose and antioxidant enzymes in different tissues of diabetic rats. Journal of Laboratory and Clinical Medicine. 2003;142(3):172–177. [PubMed] [Google Scholar]

61. Maritim AC, Sanders RA, Watkins JB. Diabetes, oxidative stress, and antioxidants: a review. Journal of Biochemical and Molecular Toxicology. 2003;17(1):24–38. [PubMed] [Google Scholar]

62. Kitada M, Koya D, Sugimoto T, et al. Translocation of glomerular p47phox and p67phox by protein kinase C-β activation is required for oxidative stress in diabetic nephropathy. Diabetes. 2003;52(10):2603–2614. [PubMed] [Google Scholar]

63. Etoh T, Inoguchi T, Kakimoto M, et al. Increased expression of NAD(P)H oxidase subunits, NOX4 and p22phox, in the kidney of streptozotocin-induced diabetic rats and its reversibity by interventive insulin treatment. Diabetologia. 2003;46(10):1428–1437. [PubMed] [Google Scholar]

64. Green K, Brand MD, Murphy MP. Prevention of mitochondrial oxidative damage as a therapeutic strategy in diabetes. Diabetes. 2004;53(supplement 1):S110–S118. [PubMed] [Google Scholar]

65. Nishikawa T, Edelstein D, Du XL, et al. Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature. 2000;404(6779):787–790. [PubMed] [Google Scholar]

66. Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature. 2001;414(6865):813–820. [PubMed] [Google Scholar]

67. Taniyama Y, Griendling KK. Reactive oxygen species in the vasculature: molecular and cellular mechanisms. Hypertension. 2003;42(6):1075–1081. [PubMed] [Google Scholar]

68. Liu Y, Gutterman DD. The coronary circulation in diabetes: influence of reactive oxygen species on K+ channel-mediated vasodilation. Vascular Pharmacology. 2002;38(1):43–49. [PubMed] [Google Scholar]

69. Liu Y, Terata K, Chai Q, Li H, Kleinman LH, Gutterman DD. Peroxynitrite inhibits Ca2+-activated K+ channel activity in smooth muscle of human coronary arterioles. Circulation Research. 2002;91(11):1070–1076. [PubMed] [Google Scholar]

70. Soriano FG, Virág L, Szabó C. Diabetic endothelial dysfunction: role of reactive oxygen and nitrogen species production and poly(ADP-ribose) polymerase activation. Journal of Molecular Medicine. 2001;79(8):437–448. [PubMed] [Google Scholar]

71. Griendling KK, FitzGerald GA. Oxidative stress and cardiovascular injury part I: basic mechanisms and in vivo monitoring of ROS. Circulation. 2003;108(16):1912–1916. [PubMed] [Google Scholar]

72. Lastra G, Whaley-Connell A, Manrique C, et al. Low-dose spironolactone reduces reactive oxygen species generation and improves insulin-stimulated glucose transport in skeletal muscle in the TG(mRen2)27 rat. American Journal of Physiology. 2008;295(1):E110–E116. [PMC free article] [PubMed] [Google Scholar]

73. Hayden MR, Whaley-Connell A, Sowers JR. Renal redox stress and remodeling in metabolic syndrome, type 2 diabetes mellitus, and diabetic nephropathy: paying homage to the podocyte. American Journal of Nephrology. 2005;25(6):553–569. [PubMed] [Google Scholar]

74. Sowers JR, Whaley-Connell A, Epstein M. The emerging clinical implications of the role of aldosterone in the metabolic syndrome and resistant hypertension. Annals of Internal Medicine. 2009;150(11):776–783. [PMC free article] [PubMed] [Google Scholar]

75. Caprio M, Fève B, Claës A, Viengchareun S, Lombès M, Zennaro MC. Pivotal role of the mineralocorticoid receptor in corticosteroid-induced adipogenesis. The FASEB Journal. 2007;21(9):2185–2194. [PubMed] [Google Scholar]

76. Rondinone CM, Rodbard D, Baker ME. Aldosterone stimulates differentiation of mouse 3T3-L1 cells into adipocytes. Endocrinology. 1993;132(6):2421–2426. [PubMed] [Google Scholar]

77. Catena C, Lapenna R, Baroselli S, et al. Insulin sensitivity in patients with primary aldosteronism: a follow-up study. Journal of Clinical Endocrinology and Metabolism. 2006;91(9):3457–3463. [PubMed] [Google Scholar]

78. Ehrhart-Bornstein M, Arakelyan K, Krug AW, Scherbaum WA, Bornstein SR. Fat cells may be the obesity-hypertension link: human adipogenic factors stimulate aldosterone secretion from adrenocortical cells. Endocrine Research. 2004;30(4):865–870. [PubMed] [Google Scholar]

79. Guo C, Ricchiuti V, Lian BQ, et al. Mineralocorticoid receptor blockade reverses obesity-related changes in expression of adiponectin, peroxisome proliferator-activated receptor-γ, and proinflammatory adipokines. Circulation. 2008;117(17):2253–2261. [PMC free article] [PubMed] [Google Scholar]

80. Goodfriend TL, Ball DL, Egan BM, Campbell WB, Nithipatikom K. Epoxy-keto derivative of linoleic acid stimulates aldosterone secretion. Hypertension. 2004;43(2):358–363. [PubMed] [Google Scholar]

81. Tuck ML, Sowers J, Dornfeld L. The effect of weight reduction on blood pressure, plasma renin activity, and plasma aldosterone levels in obese patients. The New England Journal of Medicine. 1981;304(16):930–933. [PubMed] [Google Scholar]

82. Dall’Asta C, Vedani P, Manunta P, et al. Effect of weight loss through laparoscopic gastric banding on blood pressure, plasma renin activity and aldosterone levels in morbid obesity. Nutrition, Metabolism and Cardiovascular Diseases. 2009;19(2):110–114. [PubMed] [Google Scholar]

83. Gilbert KC, Brown NJ. Aldosterone and inflammation. Current Opinion in Clinical Nutrition & Metabolic Care. 2010;17:199–204. [PMC free article] [PubMed] [Google Scholar]

84. Virdis A, Neves MF, Amiri F, Viel E, Touyz RM, Schiffrin EL. Spironolactone improves angiotensin-induced vascular changes and oxidative stress. Hypertension. 2002;40(4):504–510. [PubMed] [Google Scholar]

85. Johar S, Cave AC, Narayanapanicker A, Grieve DJ, Shah AM. Aldosterone mediates angiotensin II-induced interstitial cardiac fibrosis via a Nox2-containing NADPH oxidase. The FASEB Journal. 2006;20(9):1546–1548. [PubMed] [Google Scholar]

86. Stas S, Whaley-Connell A, Habibi J, et al. Mineralocorticoid receptor blockade attenuates chronic overexpression of the renin-angiotensin-aldosterone system stimulation of reduced nicotinamide adenine dinucleotide phosphate oxidase and cardiac remodeling. Endocrinology. 2007;148(8):3773–3780. [PubMed] [Google Scholar]

87. Cooper SA, Whaley-Connell A, Habibi J, et al. Renin-angiotensin-aldosterone system and oxidative stress in cardiovascular insulin resistance. American Journal of Physiology. 2007;293(4):H2009–H2023. [PubMed] [Google Scholar]

88. Callera GE, Touyz RM, Tostes RC, et al. Aldosterone activates vascular p38MAP kinase and NADPH oxidase via c-Src. Hypertension. 2005;45(4):773–779. [PubMed] [Google Scholar]

89. Blasi ER, Rocha R, Rudolph AE, Blomme EAG, Polly ML, McMahon EG. Aldosterone/salt induces renal inflammation and fibrosis in hypertensive rats. Kidney International. 2003;63(5):1791–1800. [PubMed] [Google Scholar]

90. Benetos A, Lacolley P, Safar ME. Prevention of aortic fibrosis by spironolactone in spontaneously hypertensive rats. Arteriosclerosis, Thrombosis, and Vascular Biology. 1997;17(6):1152–1156. [PubMed] [Google Scholar]

91. Lacolley P, Labat C, Pujol A, Delcayre C, Benetos A, Safar M. Increased carotid wall elastic modulus and fibronectin in aldosterone-salt-treated rats: effects of eplerenone. Circulation. 2002;106(22):2848–2853. [PubMed] [Google Scholar]

92. Neves MF, Amiri F, Virdis A, Diep QN, Schiffrin EL. Role of aldosterone in angiotensin II-induced cardiac and aortic inflammation, fibrosis, and hypertrophy. Canadian Journal of Physiology and Pharmacology. 2005;83(11):999–1006. [PubMed] [Google Scholar]

93. Suzuki J, Iwai M, Mogi M, et al. Eplerenone with valsartan effectively reduces atherosclerotic lesion by attenuation of oxidative stress and inflammation. Arteriosclerosis, Thrombosis, and Vascular Biology. 2006;26(4):917–921. [PubMed] [Google Scholar]

94. Mosso LM, Carvajal CA, Maiz A, et al. A possible association between primary aldosteronism and a lower β-cell function. Journal of Hypertension. 2007;25(10):2125–2130. [PubMed] [Google Scholar]

95. Hayden MR, Sowers JR. Pancreatic renin-angiotensin-aldosterone system in the cardiometabolic syndrome and type 2 diabetes mellitus. Journal of the Cardiometabolic Syndrome. 2008;3(3):129–131. [PubMed] [Google Scholar]

96. Olsen RH, Krogh-Madsen R, Thomsen C, Booth FW, Pedersen BK. Metabolic responses to reduced daily steps in healthy nonexercising men. Journal of the American Medical Association. 2008;299(11):1261–1263. [PubMed] [Google Scholar]

97. Burstein R, Polychronakos C, Toews CJ. Acute reversal of the enhanced insulin action in trained athletes. Association with insulin receptor changes. Diabetes. 1985;34(8):756–760. [PubMed] [Google Scholar]

98. Nunn AV, Guy GW, Brodie JS, Bell JD. Inflammatory modulation of exercise salience: using hormesis to return to a healthy lifestyle. Nutrition and Metabolism. 2010;7, article 87 [PMC free article] [PubMed] [Google Scholar]

99. Dantzer R, O’Connor JC, Freund GG, Johnson RW, Kelley KW. From inflammation to sickness and depression: when the immune system subjugates the brain. Nature Reviews Neuroscience. 2008;9(1):46–56. [PMC free article] [PubMed] [Google Scholar]

100. Miller AH, Maletic V, Raison CL. Inflammation and its discontents: the role of cytokines in the pathophysiology of major depression. Biological Psychiatry. 2009;65(9):732–741. [PMC free article] [PubMed] [Google Scholar]

101. Calabrese EJ, Baldwin LA. Hormesis: a generalizable and unifying hypothesis. Critical Reviews in Toxicology. 2001;31(4-5):353–424. [PubMed] [Google Scholar]

102. Gustat J, Srinivasan SR, Elkasabany A, Berenson GS. Relation of self-rated measures of physical activity to multiple risk factors of insulin resistance syndrome in young adults: the Bogalusa Heart Study. Journal of Clinical Epidemiology. 2002;55(10):997–1006. [PubMed] [Google Scholar]

103. Lakka TA, Laaksonen DE, Lakka HM, et al. Sedentary lifestyle, poor cardiorespiratory fitness, and the metabolic syndrome. Medicine and Science in Sports and Exercise. 2003;35(8):1279–1286. [PubMed] [Google Scholar]

104. Rennie KL, McCarthy N, Yazdgerdi S, Marmot M, Brunner E. Association of the metabolic syndrome with both vigorous and moderate physical activity. International Journal of Epidemiology. 2003;32(4):600–606. [PubMed] [Google Scholar]

105. Marks BL, Ward A, Morris DH, Castellani J, Rippe JM. Fat-free mass is maintained in women following a moderate diet and exercise program. Medicine and Science in Sports and Exercise. 1995;27(9):1243–1251. [PubMed] [Google Scholar]

106. McMurray RG, Andersen LB. The influence of exercise on metabolic syndrome in youth: a review. American Journal of Lifestyle Medicine. 2010;4(2):176–186. [Google Scholar]

107. Erdei N, Bagi Z, Édes I, Kaley G, Koller A. H2O2 increases production of constrictor prostaglandins in smooth muscle leading to enhanced arteriolar tone in Type 2 diabetic mice. American Journal of Physiology. 2007;292(1):H649–H656. [PubMed] [Google Scholar]

108. Nieman DC, Davis JM, Henson DA, et al. Carbohydrate ingestion influences skeletal muscle cytokine mRNA and plasma cytokine levels after a 3-h run. Journal of Applied Physiology. 2003;94(5):1917–1925. [PubMed] [Google Scholar]

109. Keller C, Steensberg A, Pilegaard H, et al. Transcriptional activation of the IL-6 gene in human contracting skeletal muscle: influence of muscle glycogen content. The FASEB Journal. 2001;15(14):2748–2750. [PubMed] [Google Scholar]

110. Febbraio MA, Pedersen BK. Contraction-induced myokine production and release: is skeletal muscle an endocrine organ? Exercise and Sport Sciences Reviews. 2005;33(3):114–119. [PubMed] [Google Scholar]

111. Pedersen BK, Febbraio MA, Mooney RA. Interleukin-6 does/does not have a beneficial role in insulin sensitivity and glucose homeostasis. Journal of Applied Physiology. 2007;102(2):814–819. [PubMed] [Google Scholar]

112. Festa A, D’Agostino R, Howard G, Mykkänen L, Tracy RP, Haffner SM. Chronic subclinical inflammation as part of the insulin resistance syndrome: the insulin resistance atherosclerosis study (IRAS) Circulation. 2000;102(1):42–47. [PubMed] [Google Scholar]

113. Starkie R, Ostrowski SR, Jauffred S, Febbraio M, Pedersen BK. Exercise and IL-6 infusion inhibit endotoxin-induced TNF-alpha production in humans. The FASEB Journal. 2003;17(8):884–886. [PubMed] [Google Scholar]

114. Keller C, Keller P, Giralt M, Hidalgo J, Pedersen BK. Exercise normalises overexpression of TNF-α in knockout mice. Biochemical and Biophysical Research Communications. 2004;321(1):179–182. [PubMed] [Google Scholar]

115. van der Poll T, Coyle SM, Barbosa K, Braxton CC, Lowry SF. Epinephrine inhibits tumor necrosis factor-α and potentiates interleukin 10 production during human endotoxemia. Journal of Clinical Investigation. 1996;97(3):713–719. [PMC free article] [PubMed] [Google Scholar]

116. Petersen EW, Carey AL, Sacchetti M, et al. Acute IL-6 treatment increases fatty acid turnover in elderly humans in vivo and in tissue culture in vitro. American Journal of Physiology. 2005;288(1):E155–E162. [PubMed] [Google Scholar]

117. Wallenius V, Wallenius K, Ahrén B, et al. Interleukin-6-deficient mice develop mature-onset obesity. Nature Medicine. 2002;8(1):75–79. [PubMed] [Google Scholar]

118. van Hall G, Steensberg A, Sacchetti M, et al. Interleukin-6 stimulates lipolysis and fat oxidation in humans. Journal of Clinical Endocrinology and Metabolism. 2003;88(7):3005–3010. [PubMed] [Google Scholar]

119. Brandt C, Pedersen BK. The role of exercise-induced myokines in muscle homeostasis and the defense against chronic diseases. Journal of Biomedicine and Biotechnology. 2010;2010:6 pages.520258 [PMC free article] [PubMed] [Google Scholar]

120. Hopps E, Canino B, Caimi G. Effects of exercise on inflammation markers in type 2 diabetic subjects. Acta Diabetologica. 2011;48(3):183–189. [PubMed] [Google Scholar]

121. Hayashi T, Wojtaszewski JFP, Goodyear LJ. Exercise regulation of glucose transport in skeletal muscle. American Journal of Physiology. 1997;273(6):E1039–E1051. [PubMed] [Google Scholar]

122. Lund S, Holman GD, Schmitz O, Pedersen O. Contraction stimulates translocation of glucose transporter GLUT4 in skeletal muscle through a mechanism distinct from that of insulin. Proceedings of the National Academy of Sciences of the United States of America. 1995;92(13):5817–5821. [PMC free article] [PubMed] [Google Scholar]

123. Zierath JR, Tsao TS, Stenbit AE, Ryder JW, Galuska D, Charron MJ. Restoration of hypoxia-stimulated glucose uptake in GLUT4-deficient muscles by muscle-specific GLUT4 transgenic complementation. Journal of Biological Chemistry. 1998;273(33):20910–20915. [PubMed] [Google Scholar]

124. Cheatham B, Vlahos CJ, Cheatham L, Wang L, Blenis J, Kahn CR. Phosphatidylinositol 3-kinase activation is required for insulin stimulation of pp70 S6 kinase, DNA synthesis, and glucose transporter translocation. Molecular and Cellular Biology. 1994;14(7):4902–4911. [PMC free article] [PubMed] [Google Scholar]

125. Goodyear LJ. AMP-activated protein kinase: a critical signaling intermediary for exercise-stimulated glucose transport? Exercise and Sport Sciences Reviews. 2000;28(3):113–116. [PubMed] [Google Scholar]

126. Krook A, Björnholm M, Galuska D, et al. Characterization of signal transduction and glucose transport in skeletal muscle from type 2 diabetic patients. Diabetes. 2000;49(2):284–292. [PubMed] [Google Scholar]

127. Houmard JA, Shaw CD, Hickey MS, Tanner CJ. Effect of short-term exercise training on insulin-stimulated PI 3-kinase activity in human skeletal muscle. American Journal of Physiology. 1999;277(6):E1055–E1060. [PubMed] [Google Scholar]

128. King DS, Dalsky GP, Staten MA, Clutter WE, Van Houten DR, Holloszy JO. Insulin action and secretion in endurance-trained and untrained humans. Journal of Applied Physiology. 1987;63(6):2247–2252. [PubMed] [Google Scholar]

129. Goodpaster BH, Katsiaras A, Kelley DE. Enhanced fat oxidation through physical activity is associated with improvements in insulin sensitivity in obesity. Diabetes. 2003;52(9):2191–2197. [PubMed] [Google Scholar]

130. Hughes VA, Fiatarone MA, Fielding RA, et al. Exercise increases muscle GLUT-4 levels and insulin action in subjects with impaired glucose tolerance. American Journal of Physiology. 1993;264(6):E855–E862. [PubMed] [Google Scholar]

131. Bruce CR, Kriketos AD, Cooney GJ, Hawley JA. Disassociation of muscle triglyceride content and insulin sensitivity after exercise training in patients with Type 2 diabetes. Diabetologia. 2004;47(1):23–30. [PubMed] [Google Scholar]

132. Poirier P, Tremblay A, Broderick T, Catellier C, Tancrède G, Nadeau A. Impact of moderate aerobic exercise training on insulin sensitivity in type 2 diabetic men treated with oral hypoglycemic agents: is insulin sensitivity enhanced only in nonobese subjects? Medical Science Monitor. 2002;8(2):CR59–CR65. [PubMed] [Google Scholar]

133. Dohm GL. Invited review: regulation of skeletal muscle GLUT-4 expression by exercise. Journal of Applied Physiology. 2002;93(2):782–787. [PubMed] [Google Scholar]

134. Zierath JR. Invited review: exercise training-induced changes in insulin signaling in skeletal muscle. Journal of Applied Physiology. 2002;93(2):773–781. [PubMed] [Google Scholar]

135. MacLean PS, Zheng D, Dohm GL. Muscle glucose transporter (GLUT 4) gene expression during exercise. Exercise and Sport Sciences Reviews. 2000;28(4):148–152. [PubMed] [Google Scholar]

136. Irrcher I, Adhihetty PJ, Joseph AM, Ljubicic V, Hood DA. Regulation of mitochondrial biogenesis in muscle by endurance exercise. Sports Medicine. 2003;33(11):783–793. [PubMed] [Google Scholar]

137. Lehman JJ, Barger PM, Kovacs A, Saffitz JE, Medeiros DM, Kelly DP. Peroxisome proliferator-activated receptor γ coactivator-1 promotes cardiac mitochondrial biogenesis. Journal of Clinical Investigation. 2000;106(7):847–856. [PMC free article] [PubMed] [Google Scholar]

138. Liang H, Ward WF. PGC-1α: a key regulator of energy metabolism. American Journal of Physiology. 2006;30(4):145–151. [PubMed] [Google Scholar]

139. Baar K, Wende AR, Jones TE, et al. Adaptations of skeletal muscle to exercise: rapid increase in the transcriptional coactivator PGC-1. The FASEB Journal. 2002;16(14):1879–1886. [PubMed] [Google Scholar]

140. Garvey WT, Maianu L, Hancock JA, Golichowski AM, Baron A. Gene expression of GLUT4 in skeletal muscle from insulin-resistant patients with obesity, IGT, GDM, and NIDDM. Diabetes. 1992;41(4):465–475. [PubMed] [Google Scholar]

141. Pedersen O, Bak JF, Andersen PH, et al. Evidence against altered expression of GLUT1 or GLUT4 in skeletal muscle of patients with obesity for NIDDM. Diabetes. 1990;39(7):865–870. [PubMed] [Google Scholar]

142. Dela F, Ploug T, Handberg A, et al. Physical training increases muscle GLUT4 protein and mRNA in patients with NIDDM. Diabetes. 1994;43(7):862–865. [PubMed] [Google Scholar]

143. Zierath JR, Wallberg-Henriksson H. From receptor to effector: insulin signal transduction in skeletal muscle from type II diabetic patients. Annals of the New York Academy of Sciences. 2002;967:120–134. [PubMed] [Google Scholar]

144. Kirwan JP, Jing M. Modulation of insulin signaling in human skeletal muscle in response to exercise. Exercise and Sport Sciences Reviews. 2002;30(2):85–90. [PubMed] [Google Scholar]

145. Ishiki M, Klip A. Minireview: recent developments in the regulation of glucose transporter-4 traffic: new signals, locations, and partners. Endocrinology. 2005;146(12):5071–5078. [PubMed] [Google Scholar]

146. Farese RV, Sajan MP, Standaert ML. Atypical protein kinase C in insulin action and insulin resistance. Biochemical Society Transactions. 2005;33(2):350–353. [PubMed] [Google Scholar]

147. Frøsig C, Richter EA. Improved insulin sensitivity after exercise: focus on insulin signaling. Obesity. 2009;17(3):S15–S20. [PubMed] [Google Scholar]

148. Dugani CB, Klip A. Glucose transporter 4: cycling, compartments and controversies. EMBO Reports. 2005;6(12):1137–1142. [PMC free article] [PubMed] [Google Scholar]

149. Le Roith D, Zick Y. Recent advances in our understanding of insulin action and insulin resistance. Diabetes Care. 2001;24(3):588–597. [PubMed] [Google Scholar]

150. Turcotte LP, Richter EA, Kiens B. Increased plasma FFA uptake and oxidation during prolonged exercise in trained vs. untrained humans. American Journal of Physiology. 1992;262(6):E791–E799. [PubMed] [Google Scholar]

151. Tunstall RJ, Mehan KA, Wadley GD, et al. Exercise training increases lipid metabolism gene expression in human skeletal muscle. American Journal of Physiology. 2002;283(1):E66–E72. [PubMed] [Google Scholar]

152. Keizer HA, Schaart G, Tandon NN, Glatz JFC, Luiken JJFP. Subcellular immunolocalisation of fatty acid translocase (FAT)/CD36 in human type-1 and type-2 skeletal muscle fibres. Histochemistry and Cell Biology. 2004;121(2):101–107. [PubMed] [Google Scholar]

153. Kiens B, Roepstorff C, Glatz JFC, et al. Lipid-binding proteins and lipoprotein lipase activity in human skeletal muscle: influence of physical activity and gender. Journal of Applied Physiology. 2004;97(4):1209–1218. [PubMed] [Google Scholar]

154. Kiens B, Kristiansen S, Jensen P, Richter EA, Turcotte LP. Membrane associated fatty acid binding protein (FABPpm) in human skeletal muscle is increased by endurance training. Biochemical and Biophysical Research Communications. 1997;231(2):463–465. [PubMed] [Google Scholar]

155. Kola B, Boscaro M, Rutter GA, Grossman AB, Korbonits M. Expanding role of AMPK in endocrinology. Trends in Endocrinology and Metabolism. 2006;17(5):205–215. [PubMed] [Google Scholar]

156. Winder WW. Energy-sensing and signaling by AMP-activated protein kinase in skeletal muscle. Journal of Applied Physiology. 2001;91(3):1017–1028. [PubMed] [Google Scholar]

157. Viollet B, Lantier L, Devin-Leclerc J, et al. Targeting the AMPK pathway for the treatment of type 2 diabetes. Frontiers in Bioscience. 2009;14(9):3380–3400. [PMC free article] [PubMed] [Google Scholar]

158. Evans JL, Goldfine ID, Maddux BA, Grodsky GM. Oxidative stress and stress-activated signaling pathways: a unifying hypothesis of type 2 diabetes. Endocrine Reviews. 2002;23(5):599–622. [PubMed] [Google Scholar]

159. Winder WW, Hardie DG. Inactivation of acetyl-CoA carboxylase and activation of AMP-activated protein kinase in muscle during exercise. American Journal of Physiology. 1996;270(2):E299–E304. [PubMed] [Google Scholar]

160. Ruderman N, Flier JS. Chewing the fat–ACC and energy balance. Science. 2001;291(5513):2558–2559. [PubMed] [Google Scholar]

161. Bergeron R, Ren JM, Cadman KS, et al. Chronic activation of AMP kinase results in NRF-1 activation and mitochondrial biogenesis. American Journal of Physiology. 2001;281(6):E1340–E1346. [PubMed] [Google Scholar]

162. Mackenzie R. New NICE guidelines for hypertension. British Medical Journal. 2011;343d5644 [Google Scholar]

163. Eriksson J, Taimela S, Koivisto VA. Exercise and the metabolic syndrome. Diabetologia. 1997;40(2):125–135. [PubMed] [Google Scholar]

164. Miller ER, III, Erlinger TP, Young DR, et al. Results of the diet, exercise, and weight loss intervention trial (DEW-IT) Hypertension. 2002;40(5):612–618. [PubMed] [Google Scholar]

165. Hermansen K. Diet, blood pressure and hypertension. British Journal of Nutrition. 2000;83(1):S113–S119. [PubMed] [Google Scholar]

166. Francischetti EA, Genelhu VA. Obesity-hypertension: an ongoing pandemic. International Journal of Clinical Practice. 2007;61(2):269–280. [PubMed] [Google Scholar]

167. Rahmouni K, Haynes WG, Morgan DA, Mark AL. Selective resistance to central neural administration of leptin in agouti obese mice. Hypertension. 2002;39(2):486–490. [PubMed] [Google Scholar]

168. Eikelis N, Schlaich M, Aggarwal A, Kaye D, Esler M. Interactions between leptin and the human sympathetic nervous system. Hypertension. 2003;41(5):1072–1079. [PubMed] [Google Scholar]

169. Shek EW, Brands MW, Hall JE. Chronic leptin infusion increases arterial pressure. Hypertension. 1998;31(1):409–414. [PubMed] [Google Scholar]

170. Dunbar JC, Hu Y, Lu H. Intracerebroventricular leptin increases lumbar and renal sympathetic nerve activity and blood pressure in normal rats. Diabetes. 1997;46(12):2040–2043. [PubMed] [Google Scholar]

171. Quehenberger P, Exner M, Sunder-Plassmann R, et al. Leptin induces endothelin-1 in endothelial cells in vitro. Circulation Research. 2002;90(6):711–718. [PubMed] [Google Scholar]

172. Bouloumié A, Marumo T, Lafontan M, Busse R. Leptin induces oxidative stress in human endothelial cells. The FASEB Journal. 1999;13(10):1231–1238. [PubMed] [Google Scholar]

173. Fleming I, Busse R. Molecular mechanisms involved in the regulation of the endothelial nitric oxide synthase. American Journal of Physiology. 2003;284(1):R1–R12. [PubMed] [Google Scholar]

174. Fukai T, Siegfried MR, Ushio-Fukai M, Cheng Y, Kojda G, Harrison DG. Regulation of the vascular extracellular superoxide dismutase by nitric oxide and exercise training. Journal of Clinical Investigation. 2000;105(11):631–1639. [PMC free article] [PubMed] [Google Scholar]

175. Kojda G, Cheng YC, Burchfield J, Harrison DG. Dysfunctional regulation of endothelial nitric oxide synthase (eNOS) expression in response to exercise in mice lacking one eNOS gene. Circulation. 2001;103(23):2839–2844. [PubMed] [Google Scholar]

176. Hambrecht R, Adams V, Erbs S, et al. Regular physical activity improves endothelial function in patients with coronary artery disease by increasing phosphorylation of endothelial nitric oxide synthase. Circulation. 2003;107(25):3152–3158. [PubMed] [Google Scholar]

177. Hambrecht R, Wolf A, Gielen S, et al. Effect of exercise on coronary endothelial function in patients with coronary artery disease. The New England Journal of Medicine. 2000;342(7):454–460. [PubMed] [Google Scholar]

178. Hambrecht R, Gielen S, Linke A, et al. Effects of exercise training on left ventricular function and peripheral resistance in patients with chronic heart failure: a randomized trial. Journal of the American Medical Association. 2000;283(23):3095–3101. [PubMed] [Google Scholar]

179. Gielen S, Schuler G, Adams V. Cardiovascular effects of exercise training: molecular mechanisms. Circulation. 2010;122(12):1221–1238. [PubMed] [Google Scholar]

180. Laurindo FRM, Pedro MDA, Barbeiro HV, et al. Vascular free radical release: ex vivo and in vivo evidence for a flow- dependent endothelial mechanism. Circulation Research. 1994;74(4):700–709. [PubMed] [Google Scholar]

181. De Keulenaer GW, Chappell DC, Ishizaka N, Nerem RM, Wayne Alexander R, Griendling KK. Oscillatory and steady laminar shear stress differentially affect human endothelial redox state: role of a superoxide-producing NADH oxidase. Circulation Research. 1998;82(10):1094–1101. [PubMed] [Google Scholar]

182. Drummond GR, Cai H, Davis ME, Ramasamy S, Harrison DG. Transcriptional and posttranscriptional regulation of endothelial nitric oxide synthase expression by hydrogen peroxide. Circulation Research. 2000;86(3):347–354. [PubMed] [Google Scholar]

183. Cai H, Davis ME, Drummond GR, Harrison DG. Induction of endothelial NO synthase by hydrogen peroxide via a Ca2+/calmodulin-dependent protein kinase II/janus kinase 2-dependent pathway. Arteriosclerosis, Thrombosis, and Vascular Biology. 2001;21(10):1571–1576. [PubMed] [Google Scholar]

184. Rush JWE, Turk JR, Laughlin MH. Exercise training regulates SOD-1 and oxidative stress in porcine aortic endothelium. American Journal of Physiology. 2003;284(4):H1378–H1387. [PubMed] [Google Scholar]

185. Maeda S, Sugawara J, Yoshizawa M, et al. Involvement of endothelin-1 in habitual exercise-induced increase in arterial compliance. Acta Physiologica. 2009;196(2):223–229. [PMC free article] [PubMed] [Google Scholar]

186. Leung FP, Yung LM, Laher I, Yao X, Chen ZY, Huang Y. Exercise, vascular wall and cardiovascular diseases: an update (part 1) Sports Medicine. 2008;38(12):1009–1024. [PubMed] [Google Scholar]

Page 2

Comparison of definitions of the metabolic syndrome.

WHOEGIRNCEPIDF
Presence of one of the following:Insulin resistance AND two or more of the following:Presence of three of the following (2001):Central obesity(13) AND any two of the following:
 (i) DM (i) Central obesity(4) (i) Central obesity(6) (i) Raised TG(14)
 (ii) IGT (ii) Dyslipidemia(5) (ii) Dislipidemia(7) (ii) ↓ HDL(15)
 (iii) IFG (iii) BP ≥ 140/90 (iii) BP ≥ 130/85 (iii) ↑ BP(16)
 (iv) Insulin resistance (iv) FBG ≥ 6.1 mmol/L (110 mg/dL) (iv) FPG ≥ 6.1 mmol/L (110 mg/dL) (iv) ↑ FBG(17)
AND two of the following:Update (2004):
 (i) BP ≥ 140/90 (i) Elevated waist circumferences(8)
 (ii) Dyslipidemia(1) (ii) Elevated TG(9)
 (iii) Central obesity(2) (iii) Reduced HDL(10)
 (iv) Microalbuminuria(3) (iv) Elevated BP(11)
 (v) Elevated fasting glucose(12)

Toplist

Latest post

TAGs