THE ROLE OF OXIDATIVE STRESS IN THE DEVELOPMENT OF OBESITY AND OBESITY-RELATED METABOLIC DISORDERS

  • Emina Čolak Institute of Medical Biochemistry, Clinical Center of Serbia, Belgrade
  • Dragana Pap Students Health Protection Institute, Department of Laboratory Diagnostics, Novi Sad

Abstract


Obesity is a serious medical contition, defined as excessive accumulation of fat. Abdominal fat is recognized as the major risk for obesity related diseases such as: hypertension, dyslipidemia, type 2 diabetes mellitus, coronary heart disease, stroke, non-alcoholic fatty liver disease etc. Fat accumulation is also related to pro-oxidant and pro-inflammatory states. Recently published articles suggest that oxidative stress may be a link between obesity and related complications. Adiposity leads to increased oxidative stress via several multiple biochemical processes such as superoxide generation through the action of NADPH oxidase, glyceraldehyde auto-oxidation, oxidative phosphorylation, protein kinase C (PKC) activation, and polyol and hexosamine pathways. On the other hand, oxidative stress plays a causative role in the development of obesity, by stimulating the deposition of adipose tissue, including preadipocyte proliferation, adipocyte differentiation and growth. Exercise-induced weight loss can improve the redox state by modulating both oxidative stress and antioxidant promoters, which reduce endothelial dysfunction and inflammation.

References

Savini I, Catani MV, Evangelista D, Gasperi V, Avigliano L. Obesity-associated oxidative stres: Strategies finalized to improve redox state. Int J Mol Sci 2013,14:10497–538

Lobato NS, Filgueira FP, Akamine EH, Tostes RC, Carvalho MHC, Fortes ZB. Mechanisms of endothelial dysfunction in obesity-associated hypertension. Braz J Med Biol Res. 2012; 45(5): 392–400.

Pap D, Čolak E, Majkić.Singh N, Grubor-Lajšić G, Vicković S. Lipoproteins and other risk factors for cardiovascular disease in a student population J Med Biochem 2013; 32: 140–5

Čabarkapa V, Đerić M, Stojšić Z, Sakač V, Davidović S, Eremić N. Determining the relationship between homocysteinemia and biomarkers of inflammation, oxidative stress and functional kidney status in patients with diabetic nephropathy. J Med Biochem 2013;32:131–9

Fernandez-Sancez A, Madrigal-Santillan E, Bautista M, et al. Inflammation, oxidative stress and obesity. Int J Mol Sci 2011;12:3117–32

Colak E, Majkić-Singh N, Stanković S, et al. Parameters of antioxidative defense in type 2 diabetic patients with cardiovascular complications. Ann Med 2005;37(8):613-20.

Korita I, Bulo A, Langlois M, Blaton V. Inflammation markers in patients with cardiovascular disease and metabolic syndrome. J Med Biochem 2013;32: 214–9

Perez-Escamilla R, Obbagy JE, Altman JM, et al. Dietary energy density and body weight in adults and children: A systematic review. J Acad Nutr Diet 2012;112:671–84

Bego T, Čaušević A, Dujić T, Malenica M, Velija-Asimi Z, Prnjavorac B, et al. Association of FTO gene variant (RS8050136) with type 2 diabetes and markers of obesity, glycaemic control and inflammation. J Med Biochem 2019;38(2):153–63

Ates E, Set T, Caner Karahan S, Bicer C, Erel O. Thiol/disulphide homeostasis, ischemia modified albumin, and ferroxidase as oxidative stress markers in women with obesity with insulin resistance. J Med Biochem 2019;38 (4): 445 –51

Dandona P, Ghaim H, Chaudhuri A, Dhinndsa S, Kim SS. Micronutrient intake induces oxidative and inflammatory stress. Potential relevance to atherosclerosis and insulin resistance. EXP Mol Med 2010;42:245–53

Higuchi M, Dusting GJ, Peshavariva H, et al. Differentiation of human adipose-derived steam cells into fat involves reactive oxygen species and forkhead box o1 mediated upregulation of antioxidant enzymes. Stem cells Dev 2013;22:878–88

Marseglia L, Manti S, D`Angelo G, et al. Oxidative stress in obesity: a critical component in human disease. Int J Mol Sci 2014;16:378–400

Fonseca-Alaniz MH, Takada J, Alonso-Vale MI, Lima FB. Adipose tissue as an endocrine organ: From theory to practise. J Pediatr 2007;83:192–203

Wang B, Trayhurn P. Acute and prolonged effects of TNF-α on the expression and secretion of inflammation-related adipokines by human adipocites differentiated in culture. Pflug Arch 2006;452:418–27

Shoelson SE, Herrero L, Naaz A. Obesity, inflammation and insulin resistance. Gastroenterology 2007; 132:2169–80

Bedard K, Krause KH. The NOX family of ROS generating NADPH oxidases. Physilogy and pathophysiology. J Physiol Rev 2007;87:245–313

Stienstra R, Tack CJ, Kanneganti TD, Joosten LA, Netea MG. The inflammasome puts obesity in the danger zone. Cell metab 2012;15:10–8

Naugler WE, Karin M. The wolf in sheep`s cloting: the role of interleukin-6 in immunity, inflammation and cancer. Trends Mol Med 2008; 14:109-19

Dujić T, Bego T, Mlinar B, et al. Effects of the PPARG gene polymorphisms on markers of obesity and metabolic syndrome in bosnian subjects J Med Biochem 2014;33: 323–32

Stenlof K, Wernstedt I, Fjallman T, Wallenius V, Wallenius K, Jansson JO. Interleukin-6 levels in the central nervous system are negatively correlated with with fat mass in overweighr/obese subjects. J Endocrinol metab 2003;88:4379–83

Goossens GH. The role of adipose tissue dysfunction in the pathogenesis of obesity-related insulin resistance. Physiol Behav 2008;94:206–18

Colak E. New markers of oxidative damage to macromolecules. J Med Biochem 2008;27(1):1–16

Khan N, Naz L, Yasmeen G. Obesity: An independent risk factor of systemic oxidative stress. Pak J Pharm Sci 2006;19:62–9

Alberti KGMM, Zimmet P, Shaw J. The metabolic syndrome–A new worldwide definition. Lancet 2005;366;1059–62

Sabio G, Das M, Mora A, et al. A stress signalling pathway in adipose tissue regulates hepatic insulin resistance. Science 2008;322:1539–43

Maury E, Brichard SM. Adipokine dysregulation, adipose tissue inflammation and metabolic syndrome. Mol Cell Endocrinol. 2010;314:1–16

Lago F, Dieguez C, Gomez-Reino G, Gualillo O. Adipokines as emerging mediators of immune response and inflammation. Nat Clin Pract Rheumatol. 2007;3:716–24

Hopps E, Noto D, Caimi G, Averna MR. A novel comoponent of the metabolic syndrome: The oxidative stress. Nutr Metab Cardiovasc Dis. 2010;20:72–7

Dimitrijević-Srećković V, Čolak E, Djordjević P, et al. Prothrombogenic factors and reduced antioxidative defense in children and adolescents with pre-metabolic and metabolic syndrome. Clin Chem Lab Med. 2007;45(9):1140-4.

Hadi H, Carr C, Suwaidi J. Endothelial dysfunction: Cardiovascular risk factors, therapy, and outcome. Vasc Health Risk Manage. 2005;1:183–98

Couillard C, Ruel G, Archer WR, et al. Circulating levels of oxidative stress markers and endotelial adhesion molecules in men with abdominal obesity. J Clin Endocrinol Metab. 2005;90:6454–59

Galili O, Versari D, Sattler KJ, et al. Early experimental obesity is associated with endothelial dysfunction and oxidative stress. Am J Physiol Heart Circ Physiol. 2007;292:H904–H911

Poitout, V.; Robertson, R.P. Glucolipotoxicity: Fuel excess and β-cell dysfunction. Endocr. Rev. 2008; 29: 351–66

Čolak E, Majkić-Singh N. The effect of hyperglycemia and oxidative stress on the development and progress of vascular complications in type 2 diabetes. J Med Biochem 2009;28:63–71

Čolak E, Majkić-Singh N. Advanced glycosylated end products-new markers of oxidative stress and cell dysfunction. Acta Clinica 2010; 10 (2):72–97.

Kahn BB, Flier, JS. Obesity and insulin resistance. J. Clin. Investig. 2000;106:473–81

Jung UJ, Choi MS. Obesity and its metabolic complications: The role of adipokines and the relationship between obesity, inflammation, insulin resistance, dyslipidemia and nonalcoholic fatty liver disease. Int J Mol Sci 2014;15:6184–93

Schenk S, Saberi M, Olefsky JM. Insulin sensitivity: Modulation by nutrients and inflammation. J Clin Investig 2008; 118:2992–3002

Haus JM, Kashyap SR, Kasumov T, et al. Plasma ceramides are elevated in obese subjects with type 2 diabetes and correlate with the severity of insulin resistance. Diabetes 2009;58:337–43

Lim JM, Wollaston-Hayden E, Teo CF, Hausman D, Wells L. Quantitative secretome and glycome of primary human adipocytes during insulin resistance. Clinical Proteomics 2014;11:1–23

Teo CF, Wollaston-Hayden EE, Wells L. Hexosamine flux, the O-GlcNAc modification, and the development of insulin resistance in adipocytes. Mol Cell Endocrinol 2010;318:44–53

Yang X, Ongusaha PP, Miles PD, et al. Phosphoinositide signalling links O-GlcNAc transferase to insulin resistance. Nature 2008;451:964–9

Wahba IM, Mak RH. Obesity and obesity-initiated metabolic syndrome: Mechanistic links to chronic kidney disease. CJASN 2007; 2 (3): 550-62

Della Corte C, Ferrari F, Villani A, Nobili V. Epidemiology and natural history of NAFLD. J Med Biochem 2015;34:1317

Unger RH: Minireview: Weapons of lean body mass destruction—The role of ectopic lipids in the metabolic syndrome. Endocrinology 2003;144:5159–65

Unger RH, Orci L: Lipoapoptosis: Its mechanism and its diseases. Biochim Biophys Acta 2002;1585:202– 212

Horton JD, Shimomura I, Ikemoto S, Bashmakov Y, Hammer RE. Overexpression of sterol regulatory element-binding protein-1a in mouse adipose tissue produces adipocyte hypertrophy, increased fatty acid secretion, and fatty liver. J Biol Chem 2003;278 :36652–60

Nakamura MT, Cheon Y, Li Y, Nara TY. Mechanisms of regulation of gene expression by fatty acids. Lipids 2004; 39:1077– 83

Knight BL, Hebbachi A, Hauton D, et al. A role for PPAR alpha in the control of SREBP activity and lipid synthesis in the liver. Biochem 2005; J389:413– 21

Guan Y. Peroxisome proliferator-activated receptor family and its relationship to renal complications of the metabolic syndrome. J Am Soc Nephrol 2004;15:2801–15

Jia D, Yamamoto M, Otani M, Otsuki M. Bezafibrate on lipids and glucose metabolism in obese diabetic Otsuka Long-Evans Tokushima fatty rats. Metabolism 2004;53(4):405-13

Asma A, Azmi MN, Mazita A, Marina MB, Salina H, Norlaila M. A single blinded randomized comtrolled study of the effect of conventional oral hypoglycemic agents versus intensive short-term insulin therapy on pure tone audiometry in type II diabetes mellitus. Indian J Otolaryngol Head Neck Surg. 2011;63(2):114-8.

Ruan XZ, Moorhead JF, Fernando R, Wheeler DC, Powis SH, Varghese Z. Regulation of lipoprotein trafficking in the kidney: Role of inflammatory mediators and transcription factors. Biochem Soc Trans 2004;32 :88– 91

Jiang T, Wang Z, Proctor G, et al. Diet-induced obesity in C57BL/6J mice causes increased renal lipid accumulation and glomerulosclerosis via a sterol regulatory element-binding protein-1c-dependent pathway. J Biol Chem 2005;280 :32317–25

Sun L, Halaihel N, Zhang W, Rogers T, Levi M. Role of sterol regulatory element-binding protein 1 in regulation of renal lipid metabolism and glomerulosclerosis in diabetes mellitus. J Biol Chem 2002;277 :18919–27

Arici M, Chana R, Lewington A, Brown J, Brunskill NJ. Stimulation of proximal tubular cell apoptosis by albumin-bound fatty acids mediated by peroxisome proliferator activated receptor-gamma. J Am Soc Nephrol 2003;14 :17–27

Klop B, Jukema, JW, Rabelink TJ, Castro Cabezas M. A physician’s guide for the management of hypertriglyceridemia: The etiology of hypertriglyceridemia determines treatment strategy. Panminerva Med. 2012;54:91–103

Klop B, Elte JW, Cabezas M.C. Dyslipidemia in obesity: Mechanisms and potential targets. Nutrients 2013;5:1218–40

St-Pierre AC, Cantin B, Dagenais GR, et al. Low-density lipoprotein subfractions and the long-term risk of ischemic heart disease in men. Arterioscler. Thromb. Vasc. Biol. 2005;25:553–9

Sam S, Haffner S, Davidson MH, et al. Relationship of abdominal visceral and subcutaneous adipose tissue with lipoprotein particle number and size in type 2 diabetes. Diabetes 2008;57:2022–7

Cancello R, Tordjman J, Poitou C, et al. Increased infiltration of macrophages in omental adipose tissue is associated with marked hepatic lesions in morbid human obesity. Diabetes; 2006;55:1554–61

Huber J, Kiefer FW, Zeyda M, et al. CC chemokine and CC chemokine receptor profiles in visceral and subcutaneous adipose tissue are altered in human obesity. J Clin Endocrinol Metab. 2008;93:3215–21

Yang Y, Ju D, Zhang M, Yang G. Interleukin-6 stimulates lipolysis in porcine adipocytes. Endocrine 2008;33:261–9

Wells L, Vosseller K, Hart GW. A role for N-acetylglucosamine as a nutrient sensor and mediator of insulin resistance. Cell Mol Life Sci 2003;60:222–8

Tarantino G, Savastano S, Colao A. Hepatic steatosis, low-grade chronic inflammation and hormone/growth factor/adipokine imbalance. World J Gastroenterol 2010; 16:4773–83

Repič Lampret B, Murko S, Žerjav Tanšek M, et al. Selective screening for metabolic disorders in the Slovenian pediatric population. J Med Biochem 2015;34:58–63

Zdravković V, Sajić S, Mitrović J, et al. The diagnosis of prediabetes in adolescents. J Med Biochem 2015;34:38–45

Čolak E, Pap D, Majkić-Singh N, Obradović I. The association of obesity and liver enzymes activities in a student population at increased risk for cardiovascular disease. J Med Biochem 2013;32: 26–31

Bradbury MW, Berk PD. Lipid metabolism in hepatic steatosis. Clin Liver Dis 2004;8:639–71

Roden M. Mechanisms of disease: Hepatic steatosis in type 2 diabetes—Pathogenesis and clinical relevance. Nat Clin Pract Endocrinol Metab. 2006;2:335–48

Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS. TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Investig 2006;116:3015–25

Berg AH, Combs TP, Du X, Brownlee M, Scherer PE. The adipocyte-secreted protein Acrp30 enhances hepatic insulin action. Nat Med 2001;7:947–53

Yamauchi T, Kamon J, Minokoshi Y, et al. Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nat Med 2002;8:1288–95

Masaki T, Chiba S, Tatsukawa H, et al. Adiponectin protects LPS-induced liver injury through modulation of TNF-alpha in KK-Ay obese mice. Hepatology 2004;40:177–84.

Hui JM, Hodge A, Farrell GC, Kench JG, Kriketos A, George J. Beyond insulin resistance in NASH: TNF-alpha or adiponectin? Hepatology 2004;40:46–54

Kaser S, Moschen A, Cayon A, et al. Adiponectin and its receptors in non-alcoholic steatohepatitis. Gut 2005;54:117–21

Minokoshi Y, Kim YB, Peroni OD, et al. Leptin stimulates fatty-acid oxidation by activating AMP-activated protein kinase. Nature 2002;415:339–43.

Serin E, Ozer B, Gümürdülü Y, Kayaselçuk F, Kul K, Boyacioğlu S. Serum leptin level can be a negative marker of hepatocyte damage in nonalcoholic fatty liver. J Gastroenterol 2003;38:471–6

Poordad FF. The role of leptin in NAFLD contender or pretender? J Clin Gastroenterol 2004;38:841–3

Marra F. Leptin and liver fibrosis: A matter of fat. Gastroenterology 2002;122:1529–32

Cao Q, Mak KM., Ren C, Lieber CS. Leptin stimulates tissue inhibitor of metalloproteinase-1 in human hepatic stellate cells: Respective roles of the JAK/STAT and JAK-mediated H2O2-dependent MAPK pathways. J Biol Chem 2004;279:4292–304

Saxena NK, Titus MA, Ding X, et al. Leptin as a novel profibrogenic cytokine in hepatic stellate cells: Mitogenesis and inhibition of apoptosis mediated by extracellular regulated kinase (Erk) and Akt phosphorylation. FASEB J 2004;18:1612–4

Pagano C, Soardo G, Pilon C, et al. Increased serum resistin in nonalcoholic fatty liver disease is related to liver disease severity and not to insulin resistance. J Clin Endocrinol Metab 2006;91:1081–6

Manco M, Marcellini M, Giannone G, Nobili V. Correlation of serum TNF-alpha levels and histologic liver injury scores in pediatric nonalcoholic fatty liver disease. Am J Clin Pathol 2007;127:954–60

Gillett M, Royle P, Snaith A, et al. Non-pharmacological interventions to reduce the risk of diabetes in people with impaired glucose regulation: A systematic review and

Published
2020/02/17
Section
Review article