Lifestyle Modifications and Nutraceutical Interventions in the Prevention and Management of Metabolic Syndrome
Abstract
Abdominal obesity, dyslipidemia, hypertension, and hyperglycemia are metabolic risk factors that are grouped together to define metabolic syndrome (MetS). It is now widely recognized that MetS is linked to a higher risk of type 2 diabetes (T2DM) and cardiovascular disease (CVD). Overall, the pathophysiology of MetS initiated by the imbalance of nutrition intake and physical activity. It involves a complex interplay of insulin resistance (IR), inflammation, dysregulated adipocyte function, and genetic susceptibility, all of which contribute to the metabolic dysfunction. Lifestyle modifications play a crucial role in managing and preventing MetS. Key strategies include adopting a balanced diet like Mediterranean diet, Dietary Approaches to Stop Hypertension (DASH), or caloric restriction (CR), engaging in regular physical activity, and maintaining a healthy weight. Nutraceuticals, including polyphenols and CR-mimetic agents, improve insulin sensitivity, reduce inflammation, lower blood pressure and cholesterol levels, reducing oxidative stress, and promoting autophagy. In addition to lifestyle changes, drug therapy may be necessary for some individuals to manage specific risk factors, such as diuretics, angiotensin-converting enzyme (ACE) inhibitors, angiotensin II receptor blockers (ARB), calcium channel blockers, and beta blockers for hypertension; biguanides, sulfonylureas, dipeptidyl peptidase-4 (DPP-4) inhibitors, glucagon-like peptide 1(GLP-1) receptor agonists, sodium-glucose co-transporter 2 (SGLT2) inhibitors, and thiazolidinediones for hyperglycemia; and statins for dyslipidemia. Early diagnosis, including waist circumference and blood pressure measurement, serum cholesterol and glucose testing, and intervention, is essential to effectively manage MetS and prevent the progression of associated diseases. In conclusion, understanding the risk factors and associated risks of MetS, along with the implementation of lifestyle modifications such as dietary and nutraceutical interventions including polyphenols and CR-mimetic agents, is vital for reducing the burden of this syndrome. Early diagnosis and proactive management are key to improving long-term health outcomes.
KEYWORDS: metabolic syndrome, abdominal obesity, insulin resistance, diet, nutraceuticals
Full Text:
PDFReferences
Gurka MJ, Guo Y, Filipp SL, DeBoer MD. Metabolic syndrome severity is significantly associated with future coronary heart disease in Type 2 diabetes. Cardiovasc Diabetol. 2018; 17(1): 17, CrossRef.
Ambarwati R, Santosa D, Pangarsa EA, Setiawan B, Tobing ML, Sofro MAU, et al. High leptin and low adiponectin levels in the metabolic syndrome patients with malignancy. Indones Biomed J. 2023; 15(5): 297-303, CrossRef.
Islam MS, Wei P, Suzauddula M, Nime I, Feroz F, Acharjee M, et al. The interplay of factors in metabolic syndrome: understanding its roots and complexity. Molecular Medicine. 2024; 30: 279, CrossRef.
Sargowo D, Handayani O. The association between cardiovascular risk and elevated triglycerides. Indones Biomed J. 2017; 9(1): 17-22, CrossRef.
Levesque J, Lamarche B. The metabolic syndrome: Definitions, prevalence and management. J Nutrigenet Nutrigenomics. 2008; 1(3): 100-8, CrossRef.
Meiliana A, Dewi NM, Wijaya A. Adipose tissue, inflammation (meta-inflammation) and obesity management. Indones Biomed J. 2015; 7(3): 129-46, CrossRef.
Kassi E, Pervanidou P, Kaltsas G, Chrousos G. Metabolic syndrome: Definitions and controversies. BMC Med. 2011; 9: 48, CrossRef.
National Heart, Lung, and Blood Institute [Internet]. Metabolic Syndrome - What Is Metabolic Syndrome? [updated 2022 May 18; cited 2025 Jan 21]. Available from: https://www.nhlbi.nih.gov.
Grundy SM, Cleeman JI, Daniels SR, Donato KA, Eckel RH, Franklin BA, et al. Diagnosis and management of the metabolic syndrome an American Heart Association/National Heart, Lung, and Blood Institute scientific statement underlying risk factors and metabolic syndrome. 2005; 112(17): 2735-52, CrossRef.
Zimmet P, Magliano D, Matsuzawa Y, Alberti G, Shaw J. The metabolic syndrome: A global public health problem and a new definition. J Atheroscler Thromb. 2005; 12(6): 295-300, CrossRef.
Alberti KGMM, Zimmet PZ. Definition, diagnosis and classification of diabetes mellitus and its complications. Part 1: diagnosis and classification of diabetes mellitus. Provisional report of a WHO Consultation. Diabet Med. 1998; 15(7): 539-53, CrossRef.
Balkau B, Charles MA. Comment on the provisional report from the WHO consultation. European Group for the Study of Insulin Resistance (EGIR). Diabet Med. 1999; 16(5): 442-3, CrossRef.
Lipsy RJ. The National Cholesterol Education Program Adult Treatment Panel III guidelines. J Manag Care Pharm. 2003; 9(Suppl 1): 2-5, CrossRef.
Lorenzo C, Serrano-Ríos M, Martínez-Larrad MT, González-Villalpando C, González-Sánchez JL, Martínez-Calatrava MJ, et al. Is waist circumference an essential component of the metabolic syndrome? Diabetes Care. 2007; 30(8): 2141-2, CrossRef.
Alberti KG, Zimmet P, Shaw J. Metabolic syndrome--A new world-wide definition. A Consensus Statement from the International Diabetes Federation. Diabet Med. 2006; 23(5): 469-80, CrossRef.
Watanabe S. Waist Circumference in the Diagnosis of Metabolic Syndrome Debate and Solution. Annals of Nutrition & Food Science. 2018; 2(3): 1022, article.
Desroches S, Lamarche B. The evolving definitions and increasing prevalence of the metabolic syndrome. Appl Physiol Nutr Metab. 2007; 32(1): 23-32, CrossRef.
Bays H, Ballantyne C. Adiposopathy: Why do adiposity and obesity cause metabolic disease? Future Lipidol. 2006; 1(4): 389-420, CrossRef.
Meiliana A, Dewi NM, Wijaya A. Current progress in adipose tissue biology: Implications in obesity and its comorbidities. Indones Biomed J. 2020; 12(2): 85-101, CrossRef.
Roberts CK, Hevener AL, Barnard RJ. Metabolic syndrome and insulin resistance: Underlying causes and modification by exercise training. Compr Physiol. 2013; 3(1): 1-58, CrossRef.
Rochlani Y, Pothineni NV, Kovelamudi S, Mehta JL. Metabolic syndrome: Pathophysiology, management, and modulation by natural compounds. Ther Adv Cardiovasc Dis. 2017; 11(8): 215-25, CrossRef.
Rizvi AA. Cytokine biomarkers, endothelial inflammation, and atherosclerosis in the metabolic syndrome: emerging concepts. Am J Med Sci. 2009; 338(4): 310-8, CrossRef.
Monda KL, North KE, Hunt SC, Rao DC, Province MA, Kraja AT. The genetics of obesity and the metabolic syndrome. Endocr Metab Immune Disord Drug Targets. 2010; 10(2): 86-108, CrossRef.
Matsuzawa Y, Funahashi T, Nakamura T. The concept of metabolic syndrome: contribution of visceral fat accumulation and its molecular mechanism. J Atheroscler Thromb. 2011; 18(8): 629-39, CrossRef.
Caprio S, Perry R, Kursawe R. Adolescent obesity and insulin resistance: Roles of ectopic fat accumulation and adipose inflammation. Gastroenterology. 2017; 152(7): 1638-46, CrossRef.
Darbre PD. Endocrine disruptors and obesity. Curr Obes Rep. 2017; 6(1): 18-27, CrossRef.
Saltiel AR, Olefsky JM. Inflammatory mechanisms linking obesity and metabolic disease. J Clin Invest. 2017; 127(1): 1-4, CrossRef.
Zmora N, Bashiardes S, Levy M, Elinav E. The role of the immune system in metabolic health and disease. Cell Metab. 2017; 25(3): 506-21, CrossRef.
Rodríguez A, Catalán V, Gómez-Ambrosi J, Frühbeck G. Visceral and subcutaneous adiposity: Are both potential therapeutic targets for tackling the metabolic syndrome? Curr Pharm Des. 2007; 13(21): 2169-75, CrossRef.
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. 2004; 114(12): 1752-61, CrossRef.
Wisse BE. The inflammatory syndrome: the role of adipose tissue cytokines in metabolic disorders linked to obesity. J Am Soc Nephrol. 2004; 15(11): 2792-800, CrossRef.
Leal Vde O, Mafra D. Adipokines in obesity. Clin Chim Acta. 2013; 419: 87-94, CrossRef.
Juhan-Vague I, Alessi MC, Mavri A, Morange PE. Plasminogen activator inhibitor-1, inflammation, obesity, insulin resistance and vascular risk. J Thromb Haemost. 2003; 1(7): 1575-9, CrossRef.
Pant S, Deshmukh A, Gurumurthy GS, Pothineni NV, Watts TE, Romeo F, et al. Inflammation and atherosclerosis--revisited. J Cardiovasc Pharmacol Ther. 2014; 19(2): 170-8, CrossRef.
Lackey DE, Olefsky JM. Regulation of metabolism by the innate immune system. Nat Rev Endocrinol. 2016; 12(1): 15-20, CrossRef.
Cancello R, Clément K. Is obesity an inflammatory illness? Role of low-grade inflammation and macrophage infiltration in human white adipose tissue. BJOG. 2006; 113(10): 1141-7, CrossRef.
Wu H, Ballantyne CM. Skeletal muscle inflammation and insulin resistance in obesity. J Clin Invest. 2017; 127(1): 43-54, CrossRef.
Yanai H, Tomono Y, Ito K, Furutani N, Yoshida H, Tada N. The underlying mechanisms for development of hypertension in the metabolic syndrome. Nutr J. 2008; 7: 10, CrossRef.
Mulè G. Metabolic syndrome in hypertensive patients: An unholy alliance. World J Cardiol. 2014; 6(9): 890, CrossRef.
Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annu Rev Immunol. 2011; 29: 415-45, CrossRef.
Aron-Wisnewsky J, Clément K. The gut microbiome, diet, and links to cardiometabolic and chronic disorders. Nat Rev Nephrol. 2016; 12(3): 169-81, CrossRef.
Rodríguez-Monforte M, Sánchez E, Barrio F, Costa B, Flores-Mateo G. Metabolic syndrome and dietary patterns: A systematic review and meta-analysis of observational studies. Eur J Nutr. 2017; 56(3): 925-47, CrossRef.
Ristic-Medic D, Vucic V. Dietary fats and metabolic syndrome. J Nutrit Health Food Sci. 2013; 1(1): 8, CrossRef.
Fan H, Wang Y, Ren Z, Liu X, Zhao J, Yuan Y, et al. Mediterranean diet lowers all-cause and cardiovascular mortality for patients with metabolic syndrome. Diabetol Metab Syndr. 2023; 15(1): 107, CrossRef.
Nikrad N, Hosseini B, Pakmehr A, Tousi AZ, Ardekani AM, Farhangi MA, Akhavan-Sigari R. Dietary carbohydrate quality index (CQI), cardio-metabolic risk factors and insulin resistance among adults with obesity. BMC Endocr Disord. 2023; 23(1): 171, CrossRef.
Muñoz-Cabrejas A, Laclaustra M, Guallar-Castillón P, Casasnovas JA, Marco-Benedí V, Calvo-Galiano N, Moreno-Franco B. Low-quality carbohydrate intake is associated with a higher prevalence of metabolic syndrome: The AWHS Study. J Clin Endocrinol Metab. 2024; 109(9): e1768-e1775, CrossRef.
Manoogian ENC, Wilkinson MJ, O'Neal M, Laing K, Nguyen J, Van D, et al. Time-restricted eating in adults with metabolic syndrome: A randomized controlled trial. Ann Intern Med. 2024; 177(11): 1462-70, CrossRef.
Engin AB. What is lipotoxicity? Adv Exp Med Biol. 2017; 960: 197-220, CrossRef.
Erion DM, Shulman GI. Diacylglycerol-mediated insulin resistance. Nat Med. 2010; 16(4): 400-2, CrossRef.
Meyer C, Dostou JM, Welle SL, Gerich JE. Role of human liver, kidney, and skeletal muscle in postprandial glucose homeostasis. Am J Physiol Endocrinol Metab. 2002; 282(2): E419-27, CrossRef.
Adams JM, Pratipanawatr T, Berria R, Wang E, DeFronzo RA, Sullards MC, et al. Ceramide content is increased in skeletal muscle from obese insulin-resistant humans. Diabetes. 2004; 53(1): 25-31, CrossRef.
Samuel VT, Shulman GI. Mechanisms for insulin resistance: Common threads and missing links. Cell. 2012; 148(5): 852-71, CrossRef.
Chait A, den Hartigh LJ. Adipose tissue distribution, inflammation and its metabolic consequences, including diabetes and cardiovascular disease. Front Cardiovasc Med. 2020; 7: 22, CrossRef.
Ota T. Chemokine systems link obesity to insulin resistance. Diabetes Metab J. 2013 37(3): 165-72, CrossRef.
Ahima RS, Lazar MA. "Portal" adipose tissue as a generator of risk factors for cardiovascular disease and diabetes. Arteriosclerosis. 1990; 10: 493-6, CrossRef.
Marchington JM, Mattacks CA, Pond CM. Adipose tissue in the mammalian heart and pericardium: structure, foetal development and biochemical properties. Comp Biochem Physiol B. 1989; 94(2): 225-32, CrossRef.
Chusyd DE, Wang D, Huffman DM, Nagy TR. Relationships between rodent white adipose fat pads and human white adipose fat depots. Front Nutr. 2016; 3: 10, CrossRef.
Mazurek T, Zhang LF, Zalewski A, Mannion JD, Diehl JT, Arafat H, et al. Human epicardial adipose tissue is a source of inflammatory mediators. Circulation. 2003; 108(20): 2460-6, CrossRef.
Szasz T, Webb RC. Perivascular adipose tissue: More than just structural support. Clin Sci. 2012; 122(1): 1-12, CrossRef.
Qi XY, Qu SL, Xiong WH, Rom O, Chang L, Jiang ZS. Perivascular adipose tissue (PVAT) in atherosclerosis: A double-edged sword. Cardiovasc Diabetol. 2018; 17(1): 134, CrossRef.
Freedland ES. Role of a critical visceral adipose tissue threshold (CVATT) in metabolic syndrome: Implications for controlling dietary carbohydrates: A review. Nutr Metab. 2004; 1(1): 12, CrossRef.
Kwok KHM, Lam KSL, Xu A. Heterogeneity of white adipose tissue: Molecular basis and clinical implications. Exp Mol Med. 2016; 48(3): e215, CrossRef.
Ho E, Shimada Y. Formation of the epicardium studied with the scanning electron microscope. Dev Biol. 1978; 66(2): 579-85, CrossRef.
Ahima RS, Lazar MA. Adipokines and the peripheral and neural control of energy balance. Mol Endocrinol. 2008; 22(5): 1023-31, CrossRef.
Goossens GH, Blaak EE. Adipose tissue dysfunction and impaired metabolic health in human obesity: A matter of oxygen? Front Endocrinol. 2015; 6: 55, CrossRef.
Smith GI, Mittendorfer B, Klein S. Metabolically healthy obesity: Facts and fantasies. J Clin Invest. 2019;129(10): 3978-89, CrossRef.
Echouffo-Tcheugui JB, Short MI, Xanthakis V, Field P, Sponholtz TR, Larson MG, Vasan RS. Natural history of obesity subphenotypes: Dynamic changes over two decades and prognosis in the Framingham Heart Study. J Clin Endocrinol Metab. 2019; 104(3): 738-52, CrossRef.
Yki-Järvinen H. Non-alcoholic fatty liver disease as a cause and a consequence of metabolic syndrome. Lancet Diabetes Endocrinol. 2014; 2(11): 901-10, CrossRef.
Fu J, Li Y, Esangbedo IC, Li G, Feng D, Li L, Xu L, Han L, Li M, Li C, et al. Circulating osteonectin and adipokine profiles in relation to metabolically healthy obesity in Chinese children: Findings from BCAMS. J Am Heart Assoc. 2018; 7(23): e009169, CrossRef.
Indulekha K, Surendar J, Anjana RM, Geetha L, Gokulakrishnan K, Pradeepa R, et al. Metabolic obesity, adipocytokines, and inflammatory markers in Asian Indians--CURES-124. Diabetes Technol Ther. 2015; 17(2): 134-41, CrossRef.
Goossens GH. The Metabolic phenotype in obesity: Fat mass, body fat distribution, and adipose tissue function. Obes Facts. 2017; 10(3): 207-15, CrossRef.
Bremer AA, Jialal I. Adipose tissue dysfunction in nascent metabolic syndrome. J Obes. 2013; 2013: 393192, CrossRef.
Barko PC, McMichael MA, Swanson KS, Williams DA. The gastrointestinal microbiome: A review. J Vet Intern Med. 2018; 32(1): 9-25, CrossRef.
O'Connor S, Chouinard-Castonguay S, Gagnon C, Rudkowska I. Prebiotics in the management of components of the metabolic syndrome. Maturitas. 2017; 104: 11-8, CrossRef.
Ramadhan AY, Rosdiana DS. The prospect of probiotics to treat metabolic syndrome. Mol Cell Biomed Sci. 2024; 8(2): 71-80, CrossRef.
Hur KY, Lee MS. Gut microbiota and metabolic disorders. Diabetes Metab J. 2015; 39(3): 198-203,
Meiliana A, Wijaya A. Gut microbiota, obesity and metabolic dysfunction. Indones Biomed J. 2011; 3(3): 150-67, CrossRef.
Gibson GR, Probert HM, Van Loo J, Rastall RA, Roberfroid MB. Dietary modulation of the human colonic microbiota: Updating the concept of prebiotics. Nutr Res Rev. 2004; 17(2): 259-75, CrossRef.
Asemi Z, Khorrami-Rad A, Alizadeh SA, Shakeri H, Esmaillzadeh A. Effects of synbiotic food consumption on metabolic status of diabetic patients: A double-blind randomized cross-over controlled clinical trial. Clin Nutr. 2014; 33(2): 198-203, CrossRef.
Sun J, Buys NJ. Glucose- and glycaemic factor-lowering effects of probiotics on diabetes: a meta-analysis of randomised placebo-controlled trials. Br J Nutr. 2016; 115(7): 1167-77, CrossRef.
Turnbaugh PJ, Ridaura VK, Faith JJ, Rey FE, Knight R, Gordon JI. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci Transl Med [Internet]. 2009; 1(6): 6ra14, CrossRef.
Vrieze A, Van Nood E, Holleman F, Salojärvi J, Kootte RS, Bartelsman JFWM, et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology. 2012; 143(4): 913-6.e7, CrossRef.
Burcelin R. Gut microbiota and immune crosstalk in metabolic disease. Mol Metab. 2016; 5(9): 771-81, CrossRef.
Abildinova GZ, Benberin V V., Vochshenkova TA, Afshar A, Mussin NM, Kaliyev AA, Zhussupova Z, Tamadon A. The gut-brain-metabolic axis: exploring the role of microbiota in insulin resistance and cognitive function. Front Microbiol. 2024; 15: 1463958, CrossRef.
Kellow NJ, Coughlan MT, Reid CM. Metabolic benefits of dietary prebiotics in human subjects: A systematic review of randomised controlled trials. Br J Nutr. 2014; 111(7): 1147-61, CrossRef.
Sariyanti M, Andita TA, Erlinawati ND, Yunita E, Nasution AA, Sari K, et al. Probiotic Lactobacillus acidophilus FNCC 0051 improves pancreatic histopathology in streptozotocin-induced type-1 diabetes mellitus rats. Indones Biomed J. 2022; 14(4): 410-5, CrossRef.
Kassaian N, Feizi A, Aminorroaya A, Amini M. Probiotic and synbiotic supplementation could improve metabolic syndrome in prediabetic adults: A randomized controlled trial. Diabetes Metab Syndr. 2019; 13(5): 2991-6, CrossRef.
Yassine F, Najm A, Bilen M. The role of probiotics, prebiotics, and synbiotics in the treatment of inflammatory bowel diseases: An overview of recent clinical trials. Fron Syst Biol. 2025; 5: 1561047, CrossRef.
Kaushik M, Madeswaraguptha P, Vanangamudi M, Surendran V, Subramaniyan NK, Kumar A. Targeting signaling pathway and mechanism of prebiotics, probiotics, and synbiotics to manage metabolic disorders. In: Synbiotics in Metabolic Disorders. Boca Raton: CRC Press; 2024. p. 10-38, CrossRef.
Nuotio ML, Pervjakova N, Joensuu A, Karhunen V, Hiekkalinna T, Milani L, et al. An epigenome-wide association study of metabolic syndrome and its components. Sci Rep. 2020; 10: 20567, CrossRef.
Chitrala KN, Hernandez DG, Nalls MA, Mode NA, Zonderman AB, Ezike N, et al. Race-specific alterations in DNA methylation among middle-aged African Americans and Whites with metabolic syndrome. Epigenetics. 2020; 15(5): 462-82, CrossRef.
Schlaepfer IR, Joshi M. CPT1A-mediated fat oxidation, mechanisms, and therapeutic potential. Endocrinology. 2020; 161(2): bqz046, CrossRef.
Akinyemiju T, Do AN, Patki A, Aslibekyan S, Zhi D, Hidalgo B, et al. Epigenome-wide association study of metabolic syndrome in African-American adults. Clin Epigenetics. 2018: 10: 49, CrossRef.
Akinyemiju T, Do AN, Patki A, Aslibekyan S, Zhi D, Hidalgo B, et al. Epigenome-wide association study of metabolic syndrome in African-American adults. Clin Epigenetics. 2018: 10: 49, CrossRef.
Castellano-Castillo D, Moreno-Indias I, Sanchez-Alcoholado L, Ramos-Molina B, Alcaide-Torres J, Morcillo S, et al. Altered adipose tissue DNA methylation status in metabolic syndrome: Relationships between global DNA methylation and specific methylation at adipogenic, lipid metabolism and inflammatory candidate genes and metabolic variables. J Clin Med. 2019; 8(1): 87, CrossRef.
Inagaki T. Histone demethylases regulate adipocyte thermogenesis. Diabetol Int. 2018; 9(4): 215-23, CrossRef.
Inagaki T, Tachibana M, Magoori K, Kudo H, Tanaka T, Okamura M, et al. Obesity and metabolic syndrome in histone demethylase JHDM2a-deficient mice. Genes Cells. 2009; 14(8): 991-1001, CrossRef.
Sathishkumar C, Prabu P, Balakumar M, Lenin R, Prabhu D, Anjana RM, et al. Augmentation of histone deacetylase 3 (HDAC3) epigenetic signature at the interface of proinflammation and insulin resistance in patients with type 2 diabetes. Clin Epigenetics. 2016; 8: 125, CrossRef.
Kotas ME, Gorecki MC, Gillum MP. Sirtuin-1 is a nutrient-dependent modulator of inflammation. Adipocyte. 2013; 2(2): 113-8, CrossRef.
Li F, Li H, Jin X, Zhang Y, Kang X, Zhang Z, et al. Adipose-specific knockdown of Sirt1 results in obesity and insulin resistance by promoting exosomes release. Cell Cycle. 2019; 18(17): 2067-82, CrossRef.
Stols-Gonçalves D, Tristão LS, Henneman P, Nieuwdorp M. Epigenetic markers and microbiota/metabolite-induced epigenetic modifications in the pathogenesis of obesity, metabolic syndrome, type 2 diabetes, and non-alcoholic fatty liver disease. Curr Diab Rep. 2019; 19(6): 31, CrossRef.
Zhou X, Wu R, Tang G, Shen T, Li W. The predictive function of miR-122-5p and its action mechanism by regulating PKM2 in metabolic syndrome. BMC Endocr Disord. 2025; 25: 54, CrossRef.
Liu S, Tang G, Duan F, Zeng C, Gong J, Chen Y, et al. MiR-17-5p inhibits TXNIP/NLRP3 inflammasome pathway and suppresses pancreatic β-cell pyroptosis in diabetic mice. Front Cardiovasc Med. 2021; 8: 768029, CrossRef.
Houshmand-Oeregaard A, Schrölkamp M, Kelstrup L, Hansen NS, Hjort L, Thuesen ACB, et al. Increased expression of microRNA-15a and microRNA-15b in skeletal muscle from adult offspring of women with diabetes in pregnancy. Hum Mol Genet. 2018; 27(10): 1763-71, CrossRef.
Vishnoi A, Rani S. MiRNA biogenesis and regulation of diseases: An overview. Methods Mol Biol. 2017: 1509: 1-10, CrossRef.
Ramos-Lopez O, Milagro FI, Riezu-Boj JI, Martinez JA. Epigenetic signatures underlying inflammation: An interplay of nutrition, physical activity, metabolic diseases, and environmental factors for personalized nutrition. Inflamm Res. 2021; 70(1): 29-49, CrossRef.
Sánchez I, Reynoso-Camacho R, Salgado LM. The diet-induced metabolic syndrome is accompanied by whole-genome epigenetic changes. Genes Nutr. 2015; 10(4): 471, CrossRef.
Chen P, Wang Y, Chen F, Zhou B. Epigenetics in obesity: Mechanisms and advances in therapies based on natural products. Pharmacol Res Perspect. 2024; 12(1): e1171, CrossRef.
Casanova E, Salvadó J, Crescenti A, Gibert-Ramos A. Epigallocatechin gallate modulates muscle homeostasis in type 2 diabetes and obesity by targeting energetic and redox pathways: A narrative review. Int J Mol Sci. 2019; 20(3): 532, CrossRef.
Číž M, Dvořáková A, Skočková V, Kubala L. The role of dietary phenolic compounds in epigenetic modulation involved in inflammatory processes. Antioxidants. 2020; 9(8): 691, CrossRef.
Milagro FI, Gómez-Abellán P, Campión J, Martínez JA, Ordovás JM, Garaulet M. CLOCK, PER2 and BMAL1 DNA methylation: association with obesity and metabolic syndrome characteristics and monounsaturated fat intake. Chronobiol Int. 2012; 29(9): 1180-94, CrossRef.
Yun JM, Jialal I, Devaraj S. Effects of epigallocatechin gallate on regulatory T cell number and function in obese v. lean volunteers. Br J Nutr. 2010; 103(12): 1771-7, CrossRef.
Bose M, Lambert JD, Ju J, Reuhl KR, Shapses SA, Yang CS. The major green tea polyphenol, (-)-epigallocatechin-3-gallate, inhibits obesity, metabolic syndrome, and fatty liver disease in high-fat-fed Mice. J Nutr. 2008; 138(9): 1677-83, CrossRef.
Yiannakopoulou EC. Targeting DNA methylation with green tea catechins. Pharmacology. 2015; 95(3-4): 111-6, CrossRef.
Carlos-Reyes Á, López-González JS, Meneses-Flores M, Gallardo-Rincón D, Ruíz-García E, Marchat LA, et al. Dietary compounds as epigenetic modulating agents in cancer. Front Genet. 2019; 10: 79, CrossRef.
Kaufman Szymczyk A, Majewski G, Lubecka Pietruszewska K, Fabianowska Majewska K. The role of sulforaphane in epigenetic mechanisms, including interdependence between histone modification and DNA methylation. Int J Mol Sci. 2015; 16(12): 29732-43, CrossRef.
Yun JM, Jialal I, Devaraj S. Epigenetic regulation of high glucose induced proinflammatory cytokine production in monocytes by curcumin. J Nutr Biochem. 2011; 22(5): 450-8, CrossRef.
Meiliana A, Dewi NM, Wijaya A. Resveratrol: The multifaceted roles and mechanisms of polyphenol to improve longevity, immunomodulation, and age-related diseases. Indones Biomed J. 2025; 17(2): 109-24, CrossRef.
Zhao H, Shu L, Huang W, Song G, Ma H. Resveratrol affects hepatic gluconeogenesis via histone deacetylase 4. Diabetes Metab Syndr Obes. 2019; 12: 401-11, CrossRef.
Asgary S, Karimi R, Momtaz S, Naseri R, Farzaei MH. Effect of resveratrol on metabolic syndrome components: A systematic review and meta analysis. Rev Endocr Metab Disord. 2019; 20(2): 173-86, CrossRef.
Moreno M, Moreno Navarrete JM, Serrano M, Ortega F, Delgado E, Sanchez Ragnarsson C, et al. Circulating irisin levels are positively associated with metabolic risk factors in sedentary subjects. PLoS One. 2015; 10(4): e0124100, CrossRef.
Daimiel L, Micó V, Valls RM, Pedret A, Motilva MJ, Rubió L, Fitó M, Farrás M, Covas MI, Solá R, et al. Impact of phenol enriched virgin olive oils on the postprandial levels of circulating microRNAs related to cardiovascular disease. Mol Nutr Food Res. 2020; 64(15): e2000049, CrossRef.
Ramzan F, D'Souza RF, Durainayagam BR, Milan AM, Roy NC, Kruger MC, et al. Inflexibility of the plasma miRNA response following a high-carbohydrate meal in overweight insulin-resistant women. Genes Nutr. 2020; 15: 2, CrossRef.
Marques-Rocha JL, Milagro FI, Mansego ML, Zulet MA, Bressan J, Martínez JA. Expression of inflammation-related miRNAs in white blood cells from subjects with metabolic syndrome after 8 wk of following a Mediterranean diet-based weight loss program. Nutrition. 2016; 32: 48-55, CrossRef.
Corrêa TA, Rogero MM. Polyphenols regulating microRNAs and inflammation biomarkers in obesity. Nutrition. 2019; 59: 150-7, CrossRef.
Otton R, Bolin AP, Ferreira LT, Marinovic MP, Rocha ALS, Mori MA. Polyphenol rich green tea extract improves adipose tissue metabolism by down-regulating miR 335 expression and mitigating insulin resistance and inflammation. J Nutr Biochem. 2018; 57: 170-9, CrossRef.
Tomé-Carneiro J, Larrosa M, Yáñez-Gascón MJ, Dávalos A, Gil-Zamorano J, Gonzálvez M, et al. One-year supplementation with a grape extract containing resveratrol modulates inflammatory-related microRNAs and cytokines expression in peripheral blood mononuclear cells of type 2 diabetes and hypertensive patients with coronary artery disease. Pharmacol Res. 2013; 72: 69-82, CrossRef.
Ramzan F, Vickers MH, Mithen RF. Epigenetics, microRNA and Metabolic Syndrome: A Comprehensive Review. Int J Mol Sci. 2021; 22(9): 5047, CrossRef.
Zhou J, Sun DQ, Targher G, Byrne CD, Lee B, Hamaguchi M, et al. Metabolic dysfunction associated fatty liver disease increases risk of chronic kidney disease: A systematic review and meta analysis. eGastroenterology. 2023; 1(1): e100005, CrossRef.
Loomba R, Sanyal AJ. The global NAFLD epidemic. Nat Rev Gastroenterol Hepatol. 2013; 10(12): 686-90, CrossRef.
Rinella ME, Sookoian S. From NAFLD to MASLD: updated naming and diagnosis criteria for fatty liver disease. J Lipid Res. 2024; 65(1): 100485, CrossRef.
Siddiqui MS, Harrison SA, Abdelmalek MF, Anstee QM, Bedossa P, Castera L, et al. Case definitions for inclusion and analysis of endpoints in clinical trials for nonalcoholic steatohepatitis through the lens of regulatory science. Hepatology. 2018; 67(5) :2001-12, CrossRef.
Singh S, Allen AM, Wang Z, Prokop LJ, Murad MH, Loomba R. Fibrosis progression in nonalcoholic fatty liver vs nonalcoholic steatohepatitis: A systematic review and meta analysis of paired biopsy studies. Clin Gastroenterol Hepatol. 2015; 13(4): 643-654.e1 9, CrossRef.
Lindenmeyer CC, McCullough AJ. The natural history of nonalcoholic fatty liver disease-an evolving view. Clin Liver Dis. 2018; 22(1): 11-21, CrossRef.
Rinella ME, Sanyal AJ. Management of NAFLD: A stage based approach. Nat Rev Gastroenterol Hepatol. 2016; 13(4): 196-205, CrossRef.
Caussy C, Soni M, Cui J, Bettencourt R, Schork N, Chen CH, et al. Nonalcoholic fatty liver disease with cirrhosis increases familial risk for advanced fibrosis. J Clin Invest. 2017; 127(7): 2697-04, CrossRef.
Loomba R, Schork N, Chen CH, Bettencourt R, Bhatt A, Ang B, et al. Heritability of hepatic fibrosis and steatosis based on a prospective twin study. Gastroenterology. 2015; 149(7): 1784-93, CrossRef.
Pouladi N, Bime C, Garcia JGN, Lussier YA, et al. Complex genetics of pulmonary diseases: lessons from genome wide association studies and next generation sequencing. Transl Res. 2016; 168: 22-39, CrossRef.
Huang PL. A comprehensive definition for metabolic syndrome. Dis Model Mech. 2009; 2(5-6): 231-7, CrossRef.
Käräjämäki AJ, Bloigu R, Kauma H, Kesäniemi YA, Koivurova OP, Perkiomäki J, et al. Non alcoholic fatty liver disease with and without metabolic syndrome: Different long term outcomes. Metabolism. 2017; 66: 55-63, CrossRef.
Allen AM, Therneau TM, Larson JJ, Coward A, Somers VK, Kamath PS. Nonalcoholic fatty liver disease incidence and impact on metabolic burden and death: A 20 year-community study. Hepatology. 2018; 67(5): 1726-36, CrossRef.
Bazick J, Donithan M, Neuschwander Tetri BA, Kleiner D, Brunt EM, Wilson L, et al. Clinical model for NASH and advanced fibrosis in adult patients with diabetes and NAFLD: guidelines for referral in NAFLD. Diabetes Care. 2015; 38(7): 1347-55, CrossRef.
Portillo Sanchez P, Bril F, Maximos M, Lomonaco R, Biernacki D, Orsak B, et al. High prevalence of nonalcoholic fatty liver disease in patients with type 2 diabetes mellitus and normal plasma aminotransferase levels. J Clin Endocrinol Metab. 2015; 100(5): 2231-8, CrossRef.
Kwok R, Choi KC, Wong GLH, Zhang Y, Chan HLY, Luk AOY, et al. Screening diabetic patients for non alcoholic fatty liver disease with controlled attenuation parameter and liver stiffness measurements: A prospective cohort study. Gut. 2016; 65(8): 1359-68, CrossRef.
Choudhury J, Sanyal AJ. Insulin resistance and the pathogenesis of nonalcoholic fatty liver disease. Clin Liver Dis. 2004; 8(3): 575-94, CrossRef.
Ballestri S, Zona S, Targher G, Romagnoli D, Baldelli E, Nascimbeni F, et al. Nonalcoholic fatty liver disease is associated with an almost twofold increased risk of incident type 2 diabetes and metabolic syndrome: evidence from a systematic review and meta-analysis. J Gastroenterol Hepatol. 2016; 31(5): 936-44, CrossRef.
Pal M, Febbraio MA, Lancaster GI. The roles of c Jun NH₂ terminal kinases (JNKs) in obesity and insulin resistance. J Physiol. 2016; 594(1): 267-79, CrossRef.
Han MS, Jung DY, Morel C, Lakhani SA, Kim JK, Flavell RA, et al. JNK expression by macrophages promotes obesity induced insulin resistance and inflammation. Science. 2013; 339(6116): 218-22, CrossRef.
Samuel VT, Shulman GI. The pathogenesis of insulin resistance: integrating signaling pathways and substrate flux. J Clin Invest. 2016; 126(1): 12-22, CrossRef.
Sabio G, Das M, Mora A, Zhang Z, Jun JY, Hwi JK, et al. A stress signaling pathway in adipose tissue regulates hepatic insulin resistance. Science. 2008; 322(5907): 1539-43, CrossRef.
Tilg H. The role of cytokines in non-alcoholic fatty liver disease. Dig Dis. 2010; 28(1): 179-85, CrossRef.
Ghorpade DS, Ozcan L, Zheng Z, Nicoloro SM, Shen Y, Chen E, et al. Hepatocyte-secreted DPP4 in obesity promotes adipose inflammation and insulin resistance. Nature. 2018; 555(7698): 673-7, CrossRef.
Gastaldelli A, Harrison SA, Belfort-Aguilar R, Hardies LJ, Balas B, Schenker S, et al. Importance of changes in adipose tissue insulin resistance to histological response during thiazolidinedione treatment of patients with nonalcoholic steatohepatitis. Hepatology. 2009; 50(4): 1087-93, CrossRef.
Sulaeman A, Amiruddin AR, Lawrence GS. Metabolic syndrome (MetS) and nonalcoholic steatohepatitis (NASH): Study of biochemical markers free fatty acid (FFA), total antioxidant status (TAOS), adiponectin, transforming growth factor (TGF-beta1), in occurence of NASH. Indones Biomed J. 2009; 1(1): 40-4, CrossRef.
Li Y, Yang P, Ye J, Xu Q, Wu J, Wang Y. Updated mechanisms of MASLD pathogenesis. Lipids Health Dis. 2024; 23(1): 117, CrossRef.
Takaki A, Kawai D, Yamamoto K. Multiple hits, including oxidative stress, as pathogenesis and treatment target in non-alcoholic steatohepatitis (NASH). Int J Mol Sci. 2013; 14(10): 20704-28, CrossRef.
Alonso C, Fernández-Ramos D, Varela-Rey M, Martínez-Arranz I, Navasa N, Van Liempd SM, et al. Metabolomic identification of subtypes of nonalcoholic steatohepatitis. Gastroenterology. 2017; 152(6): 1449-61.e7, CrossRef.
Donnelly KL, Smith CI, Schwarzenberg SJ, Jessurun J, Boldt MD, Parks EJ. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J Clin Invest. 2005; 115(5): 1343-51, CrossRef.
Jang C, Hui S, Lu W, Cowan AJ, Morscher RJ, Lee G, et al. The small intestine converts dietary fructose into glucose and organic acids. Cell Metab. 2018; 27(2): 351-61.e3, CrossRef.
Rao SSC, Attaluri A, Anderson L, Stumbo P. Ability of the normal human small intestine to absorb fructose: Evaluation by breath testing. Clin Gastroenterol Hepatol. 2007; 5(8): 959-63, CrossRef.
Yu S, Li C, Ji G, Zhang L. The contribution of dietary fructose to non-alcoholic fatty liver disease. Front Pharmacol. 2021; 12(1): 783393, CrossRef.
Abdelmalek MF, Lazo M, Horska A, Bonekamp S, Lipkin EW, Balasubramanyam A, et al. Higher dietary fructose is associated with impaired hepatic adenosine triphosphate homeostasis in obese individuals with type 2 diabetes. Hepatology. 2012; 56(3): 952-60, CrossRef.
Schwarz JM, Noworolski SM, Erkin-Cakmak A, Korn NJ, Wen MJ, Tai VW, et al. Effects of dietary fructose restriction on liver fat, de novo lipogenesis, and insulin kinetics in children with obesity. Gastroenterology. 2017; 153(3): 743-52, CrossRef.
Softic S, Cohen DE, Kahn CR. Role of dietary fructose and hepatic de novo lipogenesis in fatty liver disease. Dig Dis Sci. 2016; 61(5): 1282-93, CrossRef.
Luukkonen PK, Zhou Y, Sädevirta S, Leivonen M, Arola J, Orešič M, et al. Hepatic ceramides dissociate steatosis and insulin resistance in patients with non-alcoholic fatty liver disease. J Hepatol. 2016; 64(5): 1167-75, CrossRef.
Maurer S, Harms M, Boucher J. The colorful versatility of adipocytes: White‐to‐brown transdifferentiation and its therapeutic potential in humans. FEBS J. 2021; 288(11): 3628-46, CrossRef.
Myoung SH, Sun YP, Shinzawa K, Kim S, Kun WC, Lee JH, et al. Lysophosphatidylcholine as a death effector in the lipoapoptosis of hepatocytes. J Lipid Res. 2008; 49(1): 84-97, CrossRef.
Perry RJ, Samuel VT, Petersen KF, Shulman GI. The role of hepatic lipids in hepatic insulin resistance and type 2 diabetes. Nature. 2014; 510(7503): 84-91, CrossRef.
Ioannou GN. The role of cholesterol in the pathogenesis of NASH. Trends Endocrinol Metab. 2016; 27(2): 84-95, CrossRef.
Krishnan A, Abdullah TS, Mounajjed T, Hartono S, McConico A, White T, et al. A longitudinal study of whole body, tissue, and cellular physiology in a mouse model of fibrosing NASH with high fidelity to the human condition. Am J Physiol Gastrointest Liver Physiol. 2017; 312(6): G666-80, CrossRef.
Loomba R, Seguritan V, Li W, Long T, Klitgord N, Bhatt A, et al. Gut microbiome-based metagenomic signature for non-invasive detection of advanced fibrosis in human nonalcoholic fatty liver disease. Cell Metab. 2017; 25(5): 1054-62.e5, CrossRef.
Xie Z, Li Y, Cheng L, Huang Y, Rao W, Shi H, et al. Potential therapeutic strategies for MASH: from preclinical to clinical development. Life Metab. 2024; 3(5): loae029, CrossRef.
Pérez-Martínez P, Mikhailidis DP, Athyros VG, Bullo M, Couture P, Covas MI, et al. Lifestyle recommendations for the prevention and management of metabolic syndrome: an international panel recommendation. Nutr Rev. 2017; 75(5): 307-26, CrossRef.
Kapoor G, Chauhan P, Singh G, Malhotra N, Chahal A. Physical activity for health and fitness: Past, present and future. J Lifestyle Med. 2022; 12(1): 9-14, CrossRef.
Xie Y, Wu Z, Sun L, Zhou L, Wang G, Xiao L, Wang H. The effects and mechanisms of exercise on the treatment of depression. Front Psychiatry. 2021; 12: 705559, CrossRef.
Pucci G, Alcidi R, Tap L, Battista F, Mattace Raso F, Schillaci G. Sex- and gender-related prevalence, cardiovascular risk and therapeutic approach in metabolic syndrome: A review of the literature. Pharmacol Res. 2017; 120: 34-42, CrossRef.
Bull F, Goenka S, Lambert V, Pratt M. Physical activity for the prevention of cardiometabolic disease. In: Prabhakaran D, Anand S, Gaziano TA, Mbanya JC, Wu Y, Nugent R, editors. Disease Control Priorities. 3rd Edition. Washington (DC): The International Bank for Reconstruction and Development/The World Bank; 2017. p.79-99, CrossRef.
Geidenstam N, Al Majdoub M, Ekman M, Spégel P, Ridderstråle M. Metabolite profiling of obese individuals before and after a one year weight loss program. Int J Obes. 2017; 41(9): 1369-78, CrossRef.
Geidenstam N, Hsu YHH, Astley CM, Mercader JM, Ridderstråle M, Gonzalez ME, et al. Using metabolite profiling to construct and validate a metabolite risk score for predicting future weight gain. PLoS One. 2019; 14(9): e0222445, CrossRef.
Pigsborg K, Magkos F. Metabotyping for precision nutrition and weight management: hype or hope? Curr Nutr Rep. 2022; 11(2): 117-23, CrossRef.
Hillesheim E, Brennan L. Metabotyping: a tool for identifying subgroups for tailored nutrition advice. Proc Nutr Soc. 2023; 82(2): 130-41, CrossRef.
Myers J, Kokkinos P, Nyelin E. Physical activity, cardiorespiratory fitness, and the metabolic syndrome. Nutrients. 2019. 11(7): 1652, CrossRef.
Knežević SK, Filippi Arriaga F, Belančić A, Božina T, Mršić J, Pelčić M P, et al. Metabolic syndrome drug therapy: the potential interplay of pharmacogenetics and pharmacokinetic interactions in clinical practice: a narrative review. Diabetology. 2024; 5(4): 406-29, CrossRef.
Kim MY, Kim EJ, Kim YN, Choi C, Lee BH. Comparison of the chemical compositions and nutritive values of various pumpkin (Cucurbitaceae) species and parts. Nutr Res Pract. 2012; 6(1): 21-7, CrossRef.
Abdelkader C, Cherif FZH, Elius EAE, Lucchesi D, Pucci L, Yahia DA. Pumpkin seed proteins (Cucurbita pepo L.) protect against diet-induced metabolic syndrome by improving insulin resistance and markers of oxidative stress and inflammation in rats. Biologia. 2022; 77(10): 2677-87, CrossRef.
Fu Z, Lv J, Gao X, Zheng H, Shi S, Xu X, et al. Effects of garlic supplementation on components of metabolic syndrome: a systematic review, meta-analysis, and meta-regression of randomized controlled trials. BMC Complement Med Ther. 2023; 23(1): 260, CrossRef.
Ambroselli D, Masciulli F, Romano E, Catanzaro G, Besharat ZM, Massari MC, et al. New advances in metabolic syndrome, from prevention to treatment: The role of diet and food. Nutrients. 2023; 15(3): 640, CrossRef.
Maphosa Y, Jideani VA. The role of legumes in human nutrition. In: Pacheco Aguilar R, Vitor Diaz Rodríguez M, editors. Functional Food - Improve Health through Adequate Food. London: IntechOpen; 2017, CrossRef.
Azadbakht L, Kimiagar M, Mehrabi Y, Esmaillzadeh A, Padyab M, Hu FB, et al. Soy inclusion in the diet improves features of the metabolic syndrome: A randomized crossover study in postmenopausal women. Am J Clin Nutr. 2007; 85(3): 735-41, CrossRef.
Ruscica M, Pavanello C, Gandini S, Gomaraschi M, Vitali C, Macchi C, et al. Effect of soy on metabolic syndrome and cardiovascular risk factors: A randomized controlled trial. Eur J Nutr. 2018; 57(2): 499-511, CrossRef.
Guo H, Ding J, Liang J, Zhang Y. Associations of whole grain and refined grain consumption with metabolic syndrome: a meta-analysis of observational studies. Front Nutr. 2021; 8: 695620, CrossRef.
Li N, Jia M, Deng Q, Wang Z, Huang F, Hou H, et al. Effect of low ratio n 6/n 3 PUFA on blood lipid level: A meta-analysis. Hormones. 2021; 20(5): 697-706, CrossRef.
Ambroselli D, Masciulli F, Romano E, Catanzaro G, Besharat ZM, Massari MC, et al. New advances in metabolic syndrome, from prevention to treatment: the role of diet and food. Nutrients. 2023; 15(3): 640, CrossRef.
Carresi C, Gliozzi M, Musolino V, Scicchitano M, Scarano F, Bosco F, et al. The effect of natural antioxidants in the development of metabolic syndrome: Focus on bergamot polyphenolic fraction. Nutrients. 2020; 12(5): 1504, CrossRef.
Salerno R, Casale F, Calandruccio C, Procopio A Characterization of flavonoids in Citrus bergamia (bergamot) polyphenolic fraction by liquid chromatography-high resolution mass spectrometry (LC/HRMS). Pharma Nutrition. 2016; 4(Suppl 1): S1-S7, CrossRef.
Mollace V, Sacco I, Janda E, Malara C, Ventrice D, Colica C, et al. Hypolipemic and hypoglycaemic activity of bergamot polyphenols: from animal models to human studies. Fitoterapia. 2011; 82(3): 309-16, CrossRef.
Bahrami A, Khalesi S, Makiabadi E, Alibeyk S, Hajigholam Saryazdi M, Hejazi E. Adherence to the Mediterranean diet and the risk of lung cancer: A systematic review and dose-response meta-analysis of observational studies. Nutr Rev. 2022; 80(11): 1118-28, CrossRef.
Gorzynik Debicka M, Przychodzen P, Cappello F, Kuban Jankowska A, Marino Gammazza A, Knap N, et al .Potential health benefits of olive oil and plant polyphenols. Int J Mol Sci. 2018; 19(3): 686, CrossRef.
Scaglione S, Di Chiara T, Daidone M, Tuttolomondo A. Effects of the Mediterranean diet on the components of metabolic syndrome concerning the cardiometabolic risk. Nutrients. 2025; 17(2): 358, CrossRef.
Azadbakht L, Mirmiran P, Esmaillzadeh A, Azizi T, Azizi F. Beneficial effects of a dietary approaches to stop hypertension eating plan on features of the metabolic syndrome. Diabetes Care. 2005; 28(12): 2823-31, CrossRef.
Wang Y, Wang Y, Shehzad Q, Su Y, Xu L, Yu L, et al. Does omega 3 PUFAs supplementation improve metabolic syndrome and related cardiovascular diseases? A systematic review and meta analysis of randomized controlled trials. Crit Rev Food Sci Nutr. 2024; 64(26): 9455-82, CrossRef.
Al Shammaa A, Al Thani A, Al Kaabi M, Al Saeed K, Alanazi M, Shi Z. Serum magnesium is inversely associated with body composition and metabolic syndrome. Diabetes Metab Syndr Obes. 2023; 16: 95-104, CrossRef.
Qi KJ, Zhao ZT, Zhang W, Yang F. The impacts of vitamin D supplementation in adults with metabolic syndrome: A systematic review and meta analysis of randomized controlled trials. Front Pharmacol. 2022; 13: 1033026, CrossRef.
Gholami Z, Paknahad Z. Effect of psyllium consumption on metabolic syndrome indices: Systematic review and dose-response meta analysis of randomized controlled trials. J Funct Foods. 2023; 107: 105685, CrossRef.
Wastyk HC, Perelman D, Topf M, Fragiadakis GK, Robinson JL, Sonnenburg JL, et al. Randomized controlled trial demonstrates response to a probiotic intervention for metabolic syndrome that may correspond to diet. Gut Microbes. 2023; 15(1): 2178794, CrossRef.
Zhang P, Chen K, He T, Guo H, Chen X. Coenzyme Q10 supplementation improves adipokine profile in dyslipidemic individuals: A randomized controlled trial. Nutr Metab. 2022;19(1): 13, CrossRef.
Pandey KB, Rizvi SI. Plant polyphenols as dietary antioxidants in human health and disease. Oxid Med Cell Longev. 2009; 2(5): 270-78, CrossRef.
Shahidi F, Ambigaipalan P. Phenolics and polyphenolics in foods, beverages and spices: Antioxidant activity and health effects - Areview. J Funct Foods. 2015; 18: 820-97, CrossRef.
Singla RK, Dubey AK, Garg A, Sharma RK, Fiorino M, Ameen SM, et al. Natural polyphenols: chemical classification, definition of classes, subcategories, and structures. J AOAC Int. 2019; 102(5): 1397-400, CrossRef.
Ciupei D, Colişar A, Leopold L, Stănilă A, Diaconeasa ZM. Polyphenols: from classification to therapeutic potential and bioavailability. Foods. 2024; 13(24): 4131, CrossRef.
Parhiz H, Roohbakhsh A, Soltani F, Rezaee R, Iranshahi M. Antioxidant and anti-inflammatory properties of the citrus flavonoids hesperidin and hesperetin: An updated review of their molecular mechanisms and experimental models. Phytother Res. 2015; 29(3): 323-31, CrossRef.
Barth SW, Koch TCL, Watzl B, Dietrich H, Will F, Bub A. Moderate effects of apple juice consumption on obesity-related markers in obese men: impact of diet-gene interaction on body fat content. Eur J Nutr. 2012; 51(7): 841-50, CrossRef.
Medina-Remón A, Tresserra-Rimbau A, Pons A, Tur JA, Martorell M, Ros E, et al. Effects of total dietary polyphenols on plasma nitric oxide and blood pressure in a high cardiovascular risk cohort: the PREDIMED randomized trial. Nutr Metab Cardiovasc Dis. 2015; 25(1): 60-7, CrossRef.
Macready AL, George TW, Chong MF, Alimbetov DS, Jin Y, Vidal A, et al. Flavonoid-rich fruit and vegetables improve microvascular reactivity and inflammatory status in men at risk of cardiovascular disease-FLAVURS: A randomized controlled trial. Am J Clin Nutr. 2014; 99(3): 479-89, CrossRef.
Qin W, Ren B, Wang S, Liang S, He B, Shi X, et al. Apigenin and naringenin ameliorate PKCβII-associated endothelial dysfunction via regulating ROS/caspase-3 and NO pathway in endothelial cells exposed to high glucose. Vasc Pharmacol. 2016; 85(1): 39-49, CrossRef.
Duarte S, Arango D, Parihar A, Hamel P, Yasmeen R, Doseff AI. Apigenin protects endothelial cells from lipopolysaccharide (LPS)-induced inflammation by decreasing caspase-3 activation and modulating mitochondrial function. Int J Mol Sci. 2013; 14(9): 17664-79, CrossRef.
Huang K, Liang X, Zhong Y, He W, Wang Z. 5-Caffeoylquinic acid decreases diet-induced obesity in rats by modulating PPARα and LXRα transcription. J Sci Food Agric. 2015; 95(9): 1903-10, CrossRef.
Peng BJ, Zhu Q, Zhong YL, Xu SH, Wang Z. Chlorogenic acid maintains glucose homeostasis through modulating the expression of SGLT-1, GLUT-2, and PLG in different intestinal segments of Sprague-Dawley rats fed a high-fat diet. Biomed Environ Sci. 2015; 28(12): 894-903, CrossRef.
Jin S, Chang C, Zhang L, Liu Y, Huang X, Chen Z. Chlorogenic acid improves late diabetes through adiponectin receptor signaling pathways in db/db mice. PLoS One. 2015; 10(4): e0120842, CrossRef.
Bondonno CP, Mubarak A, Liu AH, Considine MJ, Mas E, Croft KD, et al. Acute effects of chlorogenic acid on nitric oxide status, endothelial function and blood pressure in healthy volunteers: a randomised trial. Free Radic Biol Med. 2012; 53(Suppl 1): S191-2, CrossRef.
Ma Y, Gao M, Liu D. Chlorogenic acid improves high fat diet-induced hepatic steatosis and insulin resistance in mice. Pharm Res. 2015; 32(4): 1200-9, CrossRef.
Batista-Jorge GC, Barcala-Jorge AS, Lelis DF, Santos DE, Jorge AH, Monteiro-Junior RS, et al. Resveratrol effects on metabolic syndrome features: A systematic review and meta-analysis. Endocrines. 2024; 5(2): 225-43, CrossRef.
Dhanya R, Arya AD, Nisha P, Jayamurthy P. Quercetin, a lead compound against type 2 diabetes ameliorates glucose uptake via AMPK pathway in skeletal muscle cell line. Front Pharmacol. 2017; 8: 336, CrossRef.
Nurcahyanti ADR, Cokro F, Wulanjati MP, Mahmoud MF, Wink M, Sobeh M. Curcuminoids for metabolic syndrome: meta-analysis evidences toward personalized prevention and treatment management. Front Nutr. 2022; 9: 891339, CrossRef.
Fisher NDL, Hughes M, Gerhard-Herman M, Hollenberg NK. Flavanol-rich cocoa induces nitric-oxide-dependent vasodilation in healthy humans. J Hypertens. 2003; 21(12): 2281-6, CrossRef.
Zelicha H, Kloting N, Kaplan A, Yaskolka Meir A, Rinott E, Tsaban G, et al. The effect of high-polyphenol Mediterranean diet on visceral adiposity: The DIRECT PLUS randomized controlled trial. BMC Med. 2022; 20(1): 327, CrossRef.
Barona J, Aristizabal JC, Blesso CN, Volek JS, Fernandez ML. Grape polyphenols reduce blood pressure and increase flow-mediated vasodilation in men with metabolic syndrome. J Nutr. 2012; 142(9): 1626-32, CrossRef.
Edmands WMB, Ferrari P, Rothwell JA, Rinaldi S, Slimani N, Barupal DK, et al. Polyphenol metabolome in human urine and its association with intake of polyphenol-rich foods across European countries. Am J Clin Nutr. 2015; 102(4): 905-13, CrossRef.
Chiva-Blanch G, Badimon L, Estruch R. Latest evidence of the effects of the Mediterranean diet in prevention of cardiovascular disease. Curr Atheroscler Rep. 2014; 16(11): 446, CrossRef.
Walther G, Obert P, Dutheil F, Chapier R, Lesourd B, Naughton G, et al. Metabolic syndrome individuals with and without type 2 diabetes mellitus present generalized vascular dysfunction: cross-sectional study. Arterioscler Thromb Vasc Biol. 2015; 35(4): 1022-9, CrossRef.
Amiot MJ, Riva C, Vinet A. Effects of dietary polyphenols on metabolic syndrome features in humans: a systematic review. Obes Rev. 2016; 17(7): 573-86, CrossRef.
Duda-Chodak A, Tarko T. Possible side effects of polyphenols and their interactions with medicines. Molecules. 2023; 28(6): 2536, CrossRef.
DOI: https://doi.org/10.18585/inabj.v17i3.3412
Copyright (c) 2025 The Prodia Education and Research Institute

This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.
Indexed by:
The Prodia Education and Research Institute