Oxidative stress and obesity

  • Maja Malenica University of Sarajevo - Faculty of Pharmacy, Department of Pharmaceutical Biochemistry and Laboratory Diagnostics
  • Neven Meseldžić University of Sarajevo - Faculty of Pharmacy, Department of Pharmaceutical Biochemistry and Laboratory Diagnostics
Keywords: oxidative stress, insulin resistance, inflammation, biomarkers

Abstract


Obesity is a disease of excessive accumulation of adipose tissue due to an increased energy intake which is disproportionate to the energy expenditure in the body. The visceral adipose tissue in the obese accumulated in that way increases the risk of developing a number of metabolic and cardiovascular diseases. Disorders such as diabetes, dyslipidemia, inflammation, endothelial dysfunction and mitochondria can contribute to the development of oxidative stress, which is especially pronounced in the abdominal type of obesity. Obesity can induce systemic oxidative stress through a variety of biochemical mechanisms. Although ROS is generated in a large number of cells, mitochondria play a significant role in their intracellular production through the process of oxidative phosphorylation of the respiratory chain, and in fatty acid oxidation reactions. Oxidative stress is a unique link between the various molecular disorders present in the development of insulin resistance that plays a key role in the pathogenesis and progression of chronic metabolic, proinflammatory diseases. The progression of insulin resistance is also affected by inflammation. Both of these can be the cause and the consequence of obesity. The synthesis of the inflammatory mediators is induced by oxidative stress, thus bringing the inflammation and the oxidative stress into a very significant relation. This review aims to highlight recent findings on the role of oxidative stress in the pathogenesis of obesity, with special reference to the mechanisms that explain its occurrence.

 

References

A-Mansia Biotech [Internet]. Mont-Saint-Guibert, Belgium, Obesity and Diabetes in the world; 2021 [cited 2020 Oct 10]. Available from: https://www.a-mansia.com/obesity-and-diabetes-in-the-world/.

Eurostat Statistics Explained [Internet]. Overweight and obesity – BMI statistics; 2021 [cited 2020 Oct 15]. Available from: https://ec.europa.eu/eurostat/web/products-eurostat-news/-/ddn-20210721-2.

Blüher M. Obesity: global epidemiology and pathogenesis. Nat Rev Endocrinol. 2019;15(5):288-298.

Lee EY, Yoon KH. Epidemic obesity in children and adolescents: risk factors and prevention. Front Med. 2018;12(6):658-666.

Marseglia L, Manti S, D’Angelo G, Nicotera A, Parisi E, Di Rosa G, et al. Oxidative Stress in Obesity: A Critical Component in Human Diseases. Int J Mol Sci. 2015;16(1):378-400.

Manna P, Jain SK. Obesity, Oxidative Stress, Adipose Tissue Dysfunction, and the Associated Health Risks: Causes and Therapeutic Strategies. Metab Syndr Relat Disord. 2015;13(10):423-444.

Fernández-Sánchez A, Madrigal-Santillán E, Bautista M, Esquivel-Soto J, Morales-González A, Esquivel-Chirino C, et al. Inflammation, oxidative stress, and obesity. Int J Mol Sci. 2011;12(5):3117-32.

Alfadda AA, Sallam RM. Reactive oxygen species in health and disease. J Biomed Biotechnol. 2012;2012:936486.

Burhans MS, Hagman DK, Kuzma JN, Schmidt KA, Kratz M. Contribution of Adipose Tissue Inflammation to the Development of Type 2 Diabetes Mellitus. Compr Physiol. 2018;9(1):1-58.

Chawla A, Nguyen KD, Goh YP. Macrophage-mediated inflammation in metabolic disease. Nat Rev Immunol. 2011;11(11):738-49.

Zatterale F, Longo M, Naderi J, Raciti GA, Desiderio A, Miele C, et al. Chronic Adipose Tissue Inflammation Linking Obesity to Insulin Resistance and Type 2 Diabetes. Front Physiol. 2020;10:1607.

Newsholme P, Keane KN, Carlessi R, Cruzat V. Oxidative stress pathways in pancreatic β-cells and insulin-sensitive cells and tissues: importance to cell metabolism, function, and dysfunction. Am J Physiol Cell Physiol. 2019;317:C420–C433.

Jankovic A, Korac A, Buzadzic B, Otasevic V, Stanci A, Daiber A, et al. Redox implications in adipose tissue (dys)function—A new look at old acquaintances. Redox Biology. 2015;6:19-32.

Skovsø S. Modeling type 2 diabetes in rats using high fat diet and streptozotocin. J Diabetes Investig. 2014;5(4):349-358.

Schröder K, Wandzioch K, Helmcke I, Brandes RP. Nox4 acts as a switch between differentiation and proliferation in preadipocytes. Arterioscler Thromb Vasc Biol. 2009;29 (2):239-245.

Tormos KV, Anso E, Hamanaka RB, Eisenbart J, Joseph J, Kalyanaraman B, et.al. Mitochondrial complex III ROS regulate adipocyte differentiation. Cell Metab. 2011;14(4):537-44.

Han CY. Roles of Reactive Oxygen Species on Insulin Resistance in Adipose Tissue. Diabetes Metab J. 2016;40(4):272-279.

Schulz E, Wenzel P, Münzel T, Daiber A. Mitochondrial redox signaling: Interaction of mitochondrial reactive oxygen species with other sources of oxidative stress. Antioxid Redox Signal. 2014;20(2):308-324.

Takac I, Schröder K, Zhang L, Lardy B, Anilkumar N, Lambeth JD, et al. The E-loop is involved in hydrogen peroxide formation by the NADPH oxidase Nox4. J Biol Chem. 2011;286(15):13304-13.

Fujishiro M, Gotoh Y, Katagiri H, Sakoda H, Ogihara T, Anai M, et al. Three mitogen-activated protein kinases inhibit insulin signaling by different mechanisms in 3T3-L1 adipocytes. Mol Endocrinol. 2003;17(3):487-97.

Boucher J, Kleinridders A, Kahn CR. Insulin receptor signaling in normal and insulin-resistant states. Cold Spring Harb Perspect Biol. 2014;6(1):a009191.

Czech MP, Tencerova M, Pedersen DJ, Aouadi M. Insulin signalling mechanisms for triacylglycerol storage. Diabetologia. 2013;56(5):949-64.

Den Hartigh LJ, Omer M, Goodspeed L, Wang S, Wietecha T, O’Brien KD, et al. Adipocyte-Specific Deficiency of NADPH Oxidase 4 Delays the Onset of Insulin Resistance and Attenuates Adipose Tissue Inflammation in Obesity. Arterioscler Thromb Vasc Biol. 2017;37(3):466–475.

Patergnani S, Bouhamida E, Leo S, Pinton P, Rimessi A. Mitochondrial Oxidative Stress and “Mito-Inflammation”: Actors in the Diseases. Biomedicines. 2021;9(2):216.

Yeop Han C, Kargi AY, Omer M, Chan CK, Wabitsch M, O'Brien KD, et al. Differential effect of saturated and unsaturated free fatty acids on the generation of monocyte adhesion and chemotactic factors by adipocytes: dissociation of adipocyte hypertrophy from inflammation. Diabetes. 2010;59(2):386-96.

Wang L, Hu J, Zhou H. Macrophage and Adipocyte Mitochondrial Dysfunction in Obesity-Induced Metabolic Diseases. World J Mens Health. 2021;39(4):606-614.

Cusi K. The role of adipose tissue and lipotoxicity in the pathogenesis of type 2 diabetes. Curr Diab Rep. 2010;10(4):306–315.

Lee HY, Lee JS, Alves T, et al. Mitochondrial-Targeted Catalase Protects Against High-Fat Diet-Induced Muscle Insulin Resistance by Decreasing Intramuscular Lipid Accumulation. Diabetes. 2017;66(8):2072-2081.

Sivitz WI. Mitochondrial Dysfunction in Obesity and Diabetes. US Endocrinology, 2010;6(1):20-27.

Mullins CA, Gannaban RB, Khan MS, Shah H, Siddik MAB, Hegde VK, et al. Neural Underpinnings of Obesity: The Role of Oxidative Stress and Inflammation in the Brain. Antioxidants (Basel). 2020;9(10):1018.

Gealekman O, Guseva N, Hartigan C, Apotheker S, Gorgoglione M, Gurav K, et al. Depot-specific differences and insufficient subcutaneous adipose tissue angiogenesis in human obesity. Circulation. 2011;123(2):186-94.

Sun K, Kusminski CM, Scherer PE. Adipose tissue remodeling and obesity. J Clin Invest. 2011;121(6):2094-2101.

Neels JG. A role for 5-lipoxygenase products in obesity-associated inflammation and insulin resistance. Adipocyte. 2013;2(4):262-5.

Zhang H, Park Y, Wu J, Chen XP, Lee S, Yang J, et al. Role of TNF-alpha in vascular dysfunction. Clin Sci (Lond). 2009;116(3):219-30.

Zumbach MS, Boehme MW, Wahl P, Stremmel W, Ziegler R, Nawroth PP. Tumor necrosis factor increases serum leptin levels in humans. J Clin Endocrinol Metab. 1997;82(12):4080-2.

Yan S, Zhang X, Zheng H, Hu D, Zhang Y, Guan Q, et al. Clematichinenoside inhibits VCAM-1 and ICAM-1 expression in TNF-α-treated endothelial cells via NADPH oxidase-dependent IκB kinase/NF-κB pathway. Free Radic Biol Med. 2015;78:190-201.

Thomas D, Apovian CM. Macrophage functions in lean and obese adipose tissue. Metabolism. 2017 July;72:120–143.

Sánchez E, Baena-Fustegueras JA, de la Fuente MC, Gutiérrez L, Bueno M, Ros S, et al. Advanced glycation end-products in morbid obesity and after bariatric surgery: When glycemic memory starts to fail. Endocrinol Diabetes Nutr. 2017;64(1):4-10.

Uribarri J, Cai W, Woodward M, Tripp E, Goldberg L, Pyzik R, et al. Elevated Serum Advanced Glycation Endproducts in Obese Indicate Risk for the Metabolic Syndrome: A Link Between Healthy and Unhealthy Obesity? J Clin Endocrinol Metab. 2015;100(5):1957–1966.

Pérez-Matute P, Zulet MA, Martínez JA. Reactive species and diabetes: counteracting oxidative stress to improve health. Curr Opin Pharmacol. 2009;9(6):771-9.

Sankhla M, Sharma TK, Mathur K, Rathor JS, Butolia V, Gadhok AK, et al. Relationship of oxidative stress with obesity and its role in obesity induced metabolic syndrome. Clin Lab. 2012;58(5-6):385-92.

Furukawa S, Fujita T, Shimabukuro M, Iwaki M, Yamada Y, Nakajima Y, et al. Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest. 2002; 114:1752–1761.

Kumawat M, Sharma TK, Singh I, Singh N, Ghalaut VS, Vardey SK, et al. Antioxidant enzymes and lipid peroxidation in Type 2 diabetes mellitus patients with and without nephropathy. North Am J Med Sci. 2013;5(3):213-219.

Marantos C, Mukaro V, Ferrante J, Hii C, Ferrant A. Inhibition of the Lipopolysaccharide-Induced Stimulation of the Members of the MAPK Family in Human Monocytes/Macrophages by 4-Hydroxynonenal, a Product of Oxidized Omega-6 Fatty Acids. Am J Pathol. 2009;173(4):1057-66.

Zhang X, Wang Z, Li J, Gu D, Li S, Shen C, et al. Increased 4-Hydroxynonenal Formation Contributes to Obesity-Related Lipolytic Activation in Adipocytes. PloS ONE. 2013;8(8):e70663.

Guo L, Zhang XM, Zhang YB, Huang X, Chi MH. Association of 4-hydroxynonenal with classical adipokines and insulin resistance in a Chinese non-diabetic obese population. Nutr Hosp. 2017;34:363-368.

Il'yasova D, Wong BJ, Waterstone A, Kinev A, Okosun IS. Systemic F2-Isoprostane Levels in Predisposition to Obesity and Type 2 Diabetes: Emphasis on Racial Differences. Divers Equal Health Care. 2017;14(2):91-101.

Masschelin PM, Cox AR, Chernis N, Hartig SM. The impact of oxidative stress on adipose tissue energy balance. Front Physiol. 2019;10:1638.

Milne GL, Dai Q, Roberts LJ 2nd. The isoprostanes-25 years later. Biochim Biophys Acta. 2015;1851(4):433-45.

Gonzalez-Alvarez C, Ramos-Ibanez N, Azprioz-Leehan J, Ortiz-Hernandez L. Intra-abdominal and subcutaneous abdominal fat as predictors of cardiometabolic risk in a sample of Mexican children. Eur J Clin Nutr. 2017;71(9):1068–73.

Liu JB, Li WJ, Fu FM, Zhang XL, Jiao L, Cao LJ, et al. Inverse correlation between serum adiponectin and 8-iso-prostaglandin F2α in newly diagnosed type 2 diabetes patients. Int J Clin Exp Med. 2015;8(4):6085-90.

Hou N, Luo JD. Leptin and cardiovascular diseases. Clin Exp Pharmacol Physiol. 201;38(12):905-13.

Broedbaek K, Siersma V, Henriksen T, Weimann A, Petersen M, Andersen JT, et al. Association between urinary markers of nucleic acid oxidation and mortality in type 2 diabetes: a population-based cohort study. Diabetes Care. 2013;36(3):669-76.

Carlsson ER, Fenger M, Henriksen T, Kjaer LK, Worm D, Hansen DL, et al. Reduction of oxidative stress on DNA and RNA in obese patients after Roux-en-Y gastric bypass surgery—An observational cohort study of changes in urinary markers. PLoS ONE. 2020;15(12):e0243918.

Cejvanovic V, Asferg C, Kjær LK, Andersen UB, Linneberg A, Frystyk J, et al. Markers of oxidative stress in obese men with and without hypertension. Scand J Clin Lab Invest. 2016;76(8):620–5.

Sies H, Berndt C, Jones DP. Oxidative Stress. Annu Rev Biochem. 2017;86(1):715–48.

Go YM, Jones DP. Thiol/disulfide redox states in signaling and sensing. Crit Rev Biochem Mol Biol. 2013;48(2):173-81.

Jones DP, Sies H. The Redox Code. Antioxid Redox Signal. 2015; 23(9):734-746.

Alkazemi D, Rahman A, Habra B. Alterations in glutathione redox homeostasis among adolescents with obesity and anemia. Sci Rep. 2011;11:3034.

Langhardt J, Flehmig G, Klöting N, Lehmann S, Ebert T, Kern M, et al. Effects of Weight Loss on Glutathione Peroxidase 3 Serum Concentrations and Adipose Tissue Expression in Human Obesity. Obes Facts. 2018;11:475-490.

Ambad RS, Butola LK, Bankar N, Dhok A. Clinical Correlation Of Oxidative Stress Andantioxidant In Obese Individuals. Eur J Mol Clin Me. 2021;8(1):349-355.

Wang J, Wang H. Oxidative Stress in Pancreatic Beta Cell Regeneration. Oxid Med Cell Longev. 2017;2017:1930261. doi: 10.1155/2017/1930261

Sharifi-Rad M, Anil Kumar NV, Zucca P, Varoni EM, Dini L, Panzarini E, et al. Lifestyle, Oxidative Stress, and Antioxidants: Back and Forth in the Pathophysiology of Chronic Diseases. Front Physiol. 2020;11:694.

Gunawardenaa HP, Silvaa KDRR, Sivakanesanb R, Katulandac P. Increased lipid peroxidation and erythrocyte glutathione peroxidase activity of patients with type 2 diabetes mellitus: Implications for obesity and central obesity. Obesity Med. 2019;15:1-7.

Koncsos P, Seres I, Harangi M, Illyés I, Józsa L, Gönczi F, et al. Human Paraoxonase-1 Activity in Childhood Obesity and Its Relation to Leptin and Adiponectin Levels. Pediatr Res. 2010;67:309–313.

Zhou C, Cao J, Shang L, Tong C, Hu H, Wang H. Reduced Paraoxonase 1 Activity as a Marker for Severe Coronary Artery Disease. Dis Markers. 2013;35(2):97-103.

Durrington PN, Mackness B, Mackness MI. Paraoxonase and atherosclerosis. Arterioscler Thromb Vasc Biol. 2001;21:473.

Mackness B, Durrington P, McElduff P, Yarnell J, Azam N, Watt M, et al. Low paraoxonase activity predicts coronary events in the Caerphilly prospective study. Circulation. 2003;107:2775–9.

Paragh G, Seres I, Balogh Z, Varga Z, Karpati I, Matyus J, et al. The serum paraoxonase activity in patients with chronic renal failure and hyperlipidemia. Nephron. 1998;80:166–70.

Boemi M, Leviev I, Sirolla C, Pieri C, Marra M, James RW. Serum paraoxonase is reduced in Type 1 diabetic patients compared to non-diabetic, first-degree relatives: influence on the ability of HDL to protect LDL from oxidation. Atherosclerosis. 2001;155:229–235.

Mastorikou M, Mackness B, Liu Y, Mackness M. Glycation of paraoxonase-1 inhibits its activity and impairs the ability of high-density lipoprotein to metabolize membrane lipid hydroperoxides. Diabet Med. 2008;25(9):1049-1055.

Sampson MJ, Braschi S, Willis G, Astley SB. Paraoxonase-1 (PON-1) genotype and activity and in vivo oxidized plasma low-density lipoprotein in Type II diabetes. Clin Sci (Lond). 2005; 109(2):189-97.

Beer S, Moren X, Ruiz J, James RW: Postprandial modulation of serum paraoxonase activity and concentration in diabetic and non-diabetic subjects. Nutr Metab Cardiovasc Dis. 2006;16:457-465.

Kopprasch S, Pietzsch J, Kuhlisch E, Graessler J. Lack of association between paraoxonase 1 activities and increased oxidized low-density lipoprotein levels in impaired glucose tolerance and newly diagnosed diabetes mellitus. J Clin Endocrinol Metab. 2003;88:1711-1716.

Bajnok L, Csongradi E, Seres I, Varga Z, Jeges S, Peti A, et al. Relationship of adiponectin to serum paraoxonase 1. Atherosclerosis. 2008;197:363-367.

Garin MC, Kalix B, Morabia A, James RW. Small, dense lipoprotein particles and reduced paraoxonase-1 in patients with the metabolic syndrome. J Clin Endocrinol Metab. 2005;90(4):2264-9.

Tabur S, Torun AN, Sabuncu T, Turan MN, Celik H, Ocak AR, et al. Non-diabetic metabolic syndrome and obesity do not affect serum paraoxonase and arylesterase activities but do affect oxidative stress and inflammation. Eur J Endocrinol. 2010;162(3):535-41.

Liang KW, Lee WJ, Lee IT, Lee WL, Lin SY, Hsu SL, et al. Persistent elevation of paraoxonase-1 specific enzyme activity after weight reduction in obese non-diabetic men with metabolic syndrome. Clin Chim Acta. 2011;412(19-20):1835-41.

Nakamura T, Nampei M, Murase T, Satoh E, Akari S, Katoh N, et al. Influence of xanthine oxidoreductase inhibitor, topiroxostat, on body weight of diabetic obese mice. Nutr Diabetes. 2021;11:12.

Tam HK, Kelly AS, Fox CK, Nathan BM, Johnson LA. Weight Loss Mediated Reduction in Xanthine Oxidase Activity and Uric Acid Clearance in Adolescents with Severe Obesity. Child Obes. 2016;12(4):286-91.

Li X, Meng X, Gao X, Pang X, Wang Y, Wu X, et.al. Elevated Serum Xanthine Oxidase Activity Is Associated With the Development of Type 2 Diabetes: A Prospective Cohort Study. Diabetes Care. 2018;41(4):884-890.

Tam HK, Kelly AS, Fox CK, Nathan BM, Johnson LA. Weight Loss Mediated Reduction in Xanthine Oxidase Activity and Uric Acid Clearance in Adolescents with Severe Obesity. Child Obes. 2016;12(4):286-91.

Battelli MG, Bortolotti M, Polito L, Bolognesi A. The role of xanthine oxidoreductase and uric acid in metabolic syndrome, Biochim Biophys Acta (BBA)- Molecular Basis of Disease. 2018;1864(8):2557-2565.

Sodhi K, Hilgefort J, Banks G, Gilliam C, Stevens S, Ansinelli HA, et al. Uric Acid-Induced Adipocyte Dysfunction Is Attenuated by HO-1 Upregulation: Potential Role of Antioxidant Therapy to Target Obesity. Stem Cells Int. 2016;2016:8197325.

Garcia-Diaz DF, Lopez-Legarrea P, Quintero P, Martinez JA. Vitamin C in the treatment and/or prevention of obesity. J Nutr Sci Vitaminol. 2014;60(6):367-79.

Alcalá M, Sánchez-Vera I, Sevillano J, Herrero L, Serra D, Ramos MP, et al. Vitamin E reduces adipose tissue fibrosis, inflammation, and oxidative stress and improves metabolic profile in obesity. Obesity. 2015;23(8):1598-606.

Published
2022/04/28
Section
Review articles