Global Impacts of Western Diet and Its Effects on Metabolism and Health: A Narrative Review
Abstract
:1. Introduction to the Western Diet
1.1. Health Consequences, Costs, and Impact
1.2. Scientific Evidence
2. Methods
3. Nutritional Characteristic of Western Diet
3.1. Western Diet Concept
3.2. Western Diet-Related Diseases
3.3. Western Diet’s Impact on Genetics
4. Western Diet and Lifestyle
5. Western Diet and Antioxidant Status
6. Western Diet and Inflammation
7. The Effect of Nutrition and the Western Diet on the Intestinal Microbiota
8. Western Diet and Mitochondrial Fitness
9. The Effect of Western Diet on Cardiovascular Health
10. The Effect of Western Diet on Mental Health
11. The Effect of Western Diet on Metabolism
12. The Effect of Western Diet on Cancer
13. Sanitary Costs of Western Diet
14. Practical Applications
- Increase intake of fruits and vegetables: A diet rich in fruits and vegetables provides a range of vitamins, minerals, and antioxidants that can promote health and reduce the risk of chronic diseases. Encouraging people to eat a variety of colorful fruits and vegetables and including them in meals and snacks can help increase their intake.
- Reduce intake of processed and fast food: Processed and fast foods tend to be high in calories, unhealthy fats, sugar, and sodium and low in nutrients. Encouraging people to cook meals at home, emphasizing the importance of reading food labels, and promoting healthier fast food options can help reduce intake.
- Encourage physical activity: Regular physical activity is essential for maintaining a healthy weight and reducing the risk of chronic diseases. Encouraging people to find enjoyable ways to be active, such as walking, biking, or dancing, and emphasizing the importance of incorporating physical activity into daily routines can help increase physical activity levels.
- Limit sedentary behavior: Sedentary behavior, such as sitting for long periods of time, can increase the risk of chronic diseases. Encouraging people to take breaks from sitting, such as standing or walking, and promoting workplace wellness programs can help reduce sedentary behavior.
- Promote mindful eating: Mindful eating involves paying attention to the present moment and being aware of food choices, hunger, and fullness cues. Encouraging people to eat slowly, savor their food, and listen to their body’s cues can help promote mindful eating.
- Emphasize the importance of sleep: Getting enough sleep is important for overall health and well-being. Encouraging people to prioritize sleep and establish healthy sleep habits, such as going to bed and waking up at consistent times, can help improve sleep quality.
- By incorporating and enhancing psychometric evaluation questionnaires specifically designed for assessing one’s lifestyle choices, health professionals will be empowered to provide more personalized and targeted interventions.
15. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- González Olmo, B.M.; Butler, M.J.; Barrientos, R.M. Evolution of the Human Diet and Its Impact on Gut Microbiota, Immune Responses, and Brain Health. Nutrients 2021, 13, 196. [Google Scholar] [CrossRef] [PubMed]
- Shively, C.A.; Appt, S.E.; Vitolins, M.Z.; Uberseder, B.; Michalson, K.T.; Silverstein-Metzler, M.G.; Register, T.C. Mediterranean versus Western Diet Effects on Caloric Intake, Obesity, Metabolism, and Hepatosteatosis in Nonhuman Primates. Obesity (Silver Spring Md.) 2019, 27, 777–784. [Google Scholar] [CrossRef] [PubMed]
- Kennedy, G.; Nantel, G.; Shetty, P.; Food and Agriculture Organization of the United Nations. Globalization of food systems in developing countries: Impact on food security and nutrition. FAO Food Nutr. Pap. 2004, 83, 1–300. [Google Scholar]
- Bustamante-Sanchez, A.; Villegas-Mora, B.E.; Martínez-Guardado, I.; Tornero-Aguilera, J.F.; Ardigò, L.P.; Nobari, H.; Clemente-Suárez, V.J. Physical Activity and Nutritional Pattern Related to Maturation and Development. Sustainability 2022, 14, 16958. [Google Scholar] [CrossRef]
- Kopp, W. How Western Diet and Lifestyle Drive The Pandemic Of Obesity And Civilization Diseases. Diabetes Metab. Syndr. Obes. Targets Ther. 2019, 12, 2221–2236. [Google Scholar] [CrossRef] [Green Version]
- Zinöcker, M.K.; Lindseth, I.A. The Western Diet-Microbiome-Host Interaction and Its Role in Metabolic Disease. Nutrients 2018, 10, 365. [Google Scholar] [CrossRef] [Green Version]
- Rakhra, V.; Galappaththy, S.L.; Bulchandani, S.; Cabandugama, P.K. Obesity and the Western Diet: How We Got Here. Mo. Med. 2020, 117, 536–538. [Google Scholar]
- Rice Bradley, B.H. Dietary Fat and Risk for Type 2 Diabetes: A Review of Recent Research. Curr. Nutr. Rep. 2018, 7, 214–226. [Google Scholar] [CrossRef] [Green Version]
- Al Ghorani, H.; Götzinger, F.; Böhm, M.; Mahfoud, F. Arterial hypertension—Clinical trials update 2021. Nutr. Metab. Cardiovasc. Dis. 2022, 32, 21–31. [Google Scholar] [CrossRef]
- Carrera-Bastos, P.; Fontes-Villalba, M.; O’Keefe, J.H.; Lindeberg, S.; Cordain, L. The western diet and lifestyle and diseases of civilization. Res. Rep. Clin. Cardiol. 2011, 2, 15–35. [Google Scholar] [CrossRef] [Green Version]
- Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2019. CA Cancer J. Clin. 2019, 69, 7–34. [Google Scholar] [CrossRef] [Green Version]
- Cordain, L.; Eaton, S.B.; Sebastian, A.; Mann, N.; Lindeberg, S.; Watkins, B.A.; O’Keefe, J.H.; Brand-Miller, J. Origins and evolution of the Western diet: Health implications for the 21st century. Am. J. Clin. Nutr. 2005, 81, 341–354. [Google Scholar] [CrossRef] [Green Version]
- Naude, C.E.; Schoonees, A.; A Nguyen, K.; Senekal, M.; Young, T.; Garner, P.; Chaplin, M.; Volmink, J.; Richardson, M. Low-carbohydrate versus balanced-carbohydrate diets for reducing weight and cardiovascular risk. Cochrane Database Syst. Rev. 2022, 1, CD013334. [Google Scholar] [CrossRef] [PubMed]
- Hariharan, D.; Vellanki, K.; Kramer, H. The Western Diet and Chronic Kidney Disease. Curr. Hypertens. Rep. 2015, 17, 16. [Google Scholar] [CrossRef] [PubMed]
- Arsene, M.M.; Jorelle, A.B.; Sarra, S.; Viktorovna, P.I.; Davares, A.K.; Ingrid, N.K.; Steve, A.A.; Andreevna, S.L.; Vyacheslavovna, Y.N.; Carime, B.Z. Short review on the potential alternatives to antibiotics in the era of antibiotic resistance. J. Appl. Pharm. Sci. 2021, 12, 29–40. [Google Scholar]
- Salas-Salvadó, J.; Becerra-Tomás, N.; García-Gavilán, J.F.; Bulló, M.; Barrubés, L. Mediterranean Diet and Cardiovascular Disease Prevention: What Do We Know? Prog. Cardiovasc. Dis. 2018, 61, 62–67. [Google Scholar] [CrossRef] [PubMed]
- Mariotti, F. Animal and Plant Protein Sources and Cardiometabolic Health. Adv. Nutr. 2019, 10, S351–S366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chun, Y.J.; Sohn, S.-K.; Song, H.K.; Lee, S.M.; Youn, Y.H.; Lee, S.; Park, H. Associations of colorectal cancer incidence with nutrient and food group intakes in korean adults: A case-control study. Clin. Nutr. Res. 2015, 4, 110–123. [Google Scholar] [CrossRef] [Green Version]
- Malik, V.S.; Li, Y.; Tobias, D.K.; Pan, A.; Hu, F.B. Dietary Protein Intake and Risk of Type 2 Diabetes in US Men and Women. Am. J. Epidemiol. 2016, 183, 715–728. [Google Scholar] [CrossRef] [Green Version]
- Hunter, J.E.; Zhang, J.; Kris-Etherton, P.M. Cardiovascular disease risk of dietary stearic acid compared with trans, other saturated, and unsaturated fatty acids: A systematic review. Am. J. Clin. Nutr. 2010, 91, 46–63. [Google Scholar] [CrossRef] [Green Version]
- Wolever, T. Carbohydrates and health—The FAO/WHO consultation. Aust. J. Nutr. Diet. 2001, 58, S3. [Google Scholar]
- Mann, J.; Cummings, J.H.; Englyst, H.N.; Key, T.; Liu, S.; Riccardi, G.; Summerbell, C.; Uauy, R.; van Dam, R.M.; Venn, B.; et al. FAO/WHO Scientific Update on carbohydrates in human nutrition: Conclusions. Eur. J. Clin. Nutr. 2007, 61, S132–S137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- The State of Food Security and Nutrition in the World 2018|Agrifood Economics|Food and Agriculture Organization of the United Nations. Available online: https://www.fao.org/agrifood-economics/publications/detail/en/c/1153252/ (accessed on 20 April 2023).
- Wu, H.; Flint, A.J.; Qi, Q.; Van Dam, R.M.; Sampson, L.A.; Rimm, E.B.; Holmes, M.D.; Willett, W.C.; Hu, F.B.; Sun, Q. Association between dietary whole grain intake and risk of mortality: Two large prospective studies in US men and women. JAMA Intern. Med. 2015, 175, 373–384. [Google Scholar] [CrossRef] [PubMed]
- Ogden, C.L.; Fryar, C.D.; Martin, C.B.; Freedman, D.S.; Carroll, M.D.; Gu, Q.; Hales, C.M. Trends in Obesity Prevalence by Race and Hispanic Origin-1999-2000 to 2017-2018. JAMA 2020, 324, 1208–1210. [Google Scholar] [CrossRef]
- de Souza, R.J.; Mente, A.; Maroleanu, A.; Cozma, A.I.; Ha, V.; Kishibe, T.; Uleryk, E.; Budylowski, P.; Schünemann, H.; Beyene, J.; et al. Intake of saturated and trans unsaturated fatty acids and risk of all cause mortality, cardiovascular disease, and type 2 diabetes: Systematic review and meta-analysis of observational studies. BMJ 2015, 351, h3978. [Google Scholar] [CrossRef] [Green Version]
- Park, Y.; Lee, J.; Oh, J.H.; Shin, A.; Kim, J. Dietary patterns and colorectal cancer risk in a Korean population: A case-control study. Medicine 2016, 95, e3759. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.H.; Duster, M.; Roberts, T.; Devinsky, O. United States Dietary Trends Since 1800: Lack of Association Between Saturated Fatty Acid Consumption and Non-communicable Diseases. Front. Nutr. 2022, 8, 1267. [Google Scholar] [CrossRef] [PubMed]
- Cerdá, B.; Pérez, M.; Pérez-Santiago, J.D.; Tornero-Aguilera, J.F.; González-Soltero, R.; Larrosa, M. Gut Microbiota Modification: Another Piece in the Puzzle of the Benefits of Physical Exercise in Health? Front. Physiol. 2016, 7, 51. [Google Scholar] [CrossRef] [Green Version]
- Clemente-Suárez, V.J.; Mielgo-Ayuso, J.; Martín-Rodríguez, A.; Ramos-Campo, D.J.; Redondo-Flórez, L.; Tornero-Aguilera, J.F. The Burden of Carbohydrates in Health and Disease. Nutrients 2022, 14, 3809. [Google Scholar] [CrossRef]
- López-Taboada, I.; González-Pardo, H.; Conejo, N.M. Western Diet: Implications for Brain Function and Behavior. Front. Psychol. 2020, 11, 564413. [Google Scholar] [CrossRef]
- Cinquina, V.; Calvigioni, D.; Farlik, M.; Halbritter, F.; Fife-Gernedl, V.; Shirran, S.L.; Fuszard, M.A.; Botting, C.H.; Poullet, P.; Piscitelli, F.; et al. Life-long epigenetic programming of cortical architecture by maternal ‘Western’ diet during pregnancy. Mol. Psychiatry 2020, 25, 22–36. [Google Scholar] [CrossRef]
- Johnson, C.S.; Shively, C.A.; Michalson, K.T.; Lea, A.J.; DeBo, R.J.; Howard, T.D.; Hawkins, G.A.; Appt, S.E.; Liu, Y.; McCall, C.E.; et al. Contrasting effects of Western vs Mediterranean diets on monocyte inflammatory gene expression and social behavior in a primate model. eLife 2021, 10, e68293. [Google Scholar] [CrossRef] [PubMed]
- Cordero, M.D.; De Miguel, M.; Moreno Fernández, A.M.; Carmona López, I.M.; Garrido Maraver, J.; Cotán, D.; Gómez Izquierdo, L.; Bonal, P.; Campa, F.; Bullon, P.; et al. Mitochondrial dysfunction and mitophagy activation in blood mononuclear cells of fibromyalgia patients: Implications in the pathogenesis of the disease. Arthritis Res. Ther. 2010, 12, R17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Turnbaugh, P.J.; Ridaura, V.K.; Faith, J.J.; Rey, F.E.; Knight, R.; Gordon, J.I. The effect of diet on the human gut microbiome: A metagenomic analysis in humanized gnotobiotic mice. Sci. Transl. Med. 2009, 1, 6ra14. [Google Scholar] [CrossRef] [Green Version]
- Roberts, M.D.; Mobley, C.B.; Toedebush, R.G.; Heese, A.J.; Zhu, C.; Krieger, A.E.; Cruthirds, C.L.; Lockwood, C.M.; Hofheins, J.C.; Wiedmeyer, C.E.; et al. Western diet-induced hepatic steatosis and alterations in the liver transcriptome in adult Brown-Norway rats. BMC Gastroenterol. 2015, 15, 151. [Google Scholar] [CrossRef] [Green Version]
- Arendt, J.; Aulinas, A. Physiology of the Pineal Gland and Melatonin. In Endotext [Internet]; Feingold, K.R., Anawalt, B., Blackman, M.R., Boyce, A., Chrousos, G., Corpas, E., de Herder, W.W., Dhatariya, K., Dungan, K., Hofland, J., et al., Eds.; MDText.com, Inc.: South Dartmouth, MA, USA, 2000. [Google Scholar]
- Burrows, T.L.; Whatnall, M.C.; Patterson, A.J.; Hutchesson, M.J. Associations between Dietary Intake and Academic Achievement in College Students: A Systematic Review. Healthcare 2017, 5, 60. [Google Scholar] [CrossRef] [Green Version]
- Yuan, M.; Chen, W.; Teng, B.; Fang, Y. Occupational Disparities in the Association between Self-Reported Salt-Eating Habit and Hypertension in Older Adults in Xiamen, China. Int. J. Environ. Res. Public Health 2016, 13, 148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hu, G.; Eriksson, J.; Barengo, N.C.; Lakka, T.A.; Valle, T.T.; Nissinen, A.; Jousilahti, P.; Tuomilehto, J. Occupational, commuting, and leisure-time physical activity in relation to total and cardiovascular mortality among Finnish subjects with type 2 diabetes. Circulation 2004, 110, 666–673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, M.; Veeranki, S.P.; Li, S.; Steffen, L.M.; Xi, B. Beneficial associations of low and large doses of leisure time physical activity with all-cause, cardiovascular disease and cancer mortality: A national cohort study of 88,140 US adults. Br. J. Sport. Med. 2019, 53, 1405–1411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salam, N.; Jiao, J.-Y.; Zhang, X.-T.; Li, W.-J. Update on the classification of higher ranks in the phylum Actinobacteria. Int. J. Syst. Evol. Microbiol. 2020, 70, 1331–1355. [Google Scholar] [CrossRef]
- Lally, P.; Bartle, N.; Wardle, J. Social norms and diet in adolescents. Appetite 2011, 57, 623–627. [Google Scholar] [CrossRef] [PubMed]
- Cooksey Stowers, K.; Jiang, Q.; Atoloye, A.; Lucan, S.; Gans, K. Racial Differences in Perceived Food Swamp and Food Desert Exposure and Disparities in Self-Reported Dietary Habits. Int. J. Environ. Res. Public Health 2020, 17, 7143. [Google Scholar] [CrossRef]
- Caspi, C.E.; Sorensen, G.; Subramanian, S.V.; Kawachi, I. The local food environment and diet: A systematic review. Health Place 2012, 18, 1172–1187. [Google Scholar] [CrossRef] [Green Version]
- Ballal, K.; Wilson, C.R.; Harmancey, R.; Taegtmeyer, H. Obesogenic high fat western diet induces oxidative stress and apoptosis in rat heart. Mol. Cell. Biochem. 2010, 344, 221–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheeseman, K.H.; Slater, T.F. An introduction to free radical biochemistry. Br. Med. Bull. 1993, 49, 481–493. [Google Scholar] [CrossRef] [PubMed]
- McCord, J.M. The evolution of free radicals and oxidative stress. Am. J. Med. 2000, 108, 652–659. [Google Scholar] [CrossRef]
- Sies, H. Hydrogen peroxide as a central redox signaling molecule in physiological oxidative stress: Oxidative eustress. Redox Biol. 2017, 11, 613–619. [Google Scholar] [CrossRef]
- Halliwell, B. How to characterize an antioxidant: An update. Biochem. Soc. Symp. 1995, 61, 73–101. [Google Scholar]
- Shi, H.; Noguchi, N.; Niki, E. Comparative study on dynamics of antioxidative action of alpha-tocopheryl hydroquinone, ubiquinol, and alpha-tocopherol against lipid peroxidation. Free Radic. Biol. Med. 1999, 27, 334–346. [Google Scholar] [CrossRef]
- Kunwar, A.; Priyadarsini, K.I. Free radicals, oxidative stress and importance of antioxidants in human health. J. Med. Allied Sci. 2011, 1, 53–60. [Google Scholar]
- Galli, C.; Calder, P.C. Effects of fat and fatty acid intake on inflammatory and immune responses: A critical review. Ann. Nutr. Metab. 2009, 55, 123–139. [Google Scholar] [CrossRef]
- Wu, G.; Fang, Y.-Z.; Yang, S.; Lupton, J.R.; Turner, N.D. Glutathione metabolism and its implications for health. J. Nutr. 2004, 134, 489–492. [Google Scholar] [CrossRef] [Green Version]
- Nani, A.; Murtaza, B.; Sayed Khan, A.; Khan, N.A.; Hichami, A. Antioxidant and Anti-Inflammatory Potential of Polyphenols Contained in Mediterranean Diet in Obesity: Molecular Mechanisms. Molecules 2021, 26, 985. [Google Scholar] [CrossRef]
- Lu, W.; Shi, Y.; Wang, R.; Su, D.; Tang, M.; Liu, Y.; Li, Z. Antioxidant Activity and Healthy Benefits of Natural Pigments in Fruits: A Review. Int. J. Mol. Sci. 2021, 22, 4945. [Google Scholar] [CrossRef]
- Manzel, A.; Muller, D.N.; Hafler, D.A.; Erdman, S.E.; Linker, R.A.; Kleinewietfeld, M. Role of ‘Western diet’ in inflammatory autoimmune diseases. Curr. Allergy Asthma. Rep. 2014, 14, 404. [Google Scholar] [CrossRef] [Green Version]
- Blaner, W.S.; Shmarakov, I.O.; Traber, M.G. Vitamin A and Vitamin E: Will the Real Antioxidant Please Stand Up? Annu. Rev. Nutr. 2021, 41, 105–131. [Google Scholar] [CrossRef]
- Lloyd-Jones, D.M.; Hong, Y.; Labarthe, D.; Mozaffarian, D.; Appel, L.J.; Van Horn, L.; Greenlund, K.; Daniels, S.; Nichol, G.; Tomaselli, G.F.; et al. Defining and setting national goals for cardiovascular health promotion and disease reduction: The American Heart Association’s strategic Impact Goal through 2020 and beyond. Circulation 2010, 121, 586–613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hodge, A.M.; Simpson, J.A.; Gibson, R.A.; Sinclair, A.J.; Makrides, M.; O’Dea, K.; English, D.R.; Giles, G.G. Plasma phospholipid fatty acid composition as a biomarker of habitual dietary fat intake in an ethnically diverse cohort. Nutr. Metab. Cardiovasc. Dis. NMCD 2007, 17, 415–426. [Google Scholar] [CrossRef] [PubMed]
- Packer, J.E.; Slater, T.F.; Willson, R.L. Direct observation of a free radical interaction between vitamin E and vitamin C. Nature 1979, 278, 737–738. [Google Scholar] [CrossRef]
- Slavin, J.L.; Lloyd, B. Health benefits of fruits and vegetables. Adv. Nutr. 2012, 3, 506–516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lobo, V.; Patil, A.; Phatak, A.; Chandra, N. Free radicals, antioxidants and functional foods: Impact on human health. Pharmacogn. Rev. 2010, 4, 118–126. [Google Scholar] [CrossRef] [Green Version]
- Aune, D.; Keum, N.; Giovannucci, E.; Fadnes, L.T.; Boffetta, P.; Greenwood, D.C.; Tonstad, S.; Vatten, L.J.; Riboli, E.; Norat, T. Whole grain consumption and risk of cardiovascular disease, cancer, and all cause and cause specific mortality: Systematic review and dose-response meta-analysis of prospective studies. BMJ 2016, 353, i2716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mozaffarian, D.; Appel, L.J.; Van Horn, L. Components of a cardioprotective diet: New insights. Circulation 2011, 123, 2870–2891. [Google Scholar] [CrossRef] [PubMed]
- Charles-Messance, H.; Mitchelson, K.A.; Castro, E.D.M.; Sheedy, F.J.; Roche, H.M. Regulating metabolic inflammation by nutritional modulation. J. Allergy Clin. Immunol. 2020, 146, 706–720. [Google Scholar] [CrossRef] [PubMed]
- Gunnerud, U.J.; Heinzle, C.; Holst, J.J.; Östman, E.M.; Björck IM, E. Effects of pre-meal drinks with protein and amino acids on glycemic and metabolic responses at a subsequent composite meal. PLoS ONE 2012, 7, e44731. [Google Scholar] [CrossRef]
- De la Fuente, M. Effects of antioxidants on immune system ageing. Eur. J. Clin. Nutr. 2002, 56 (Suppl. 3), S5–S8. [Google Scholar] [CrossRef]
- Carr, A.C.; Maggini, S. Vitamin C and Immune Function. Nutrients 2017, 9, 1211. [Google Scholar] [CrossRef] [Green Version]
- Bertoia, M.L.; Mukamal, K.J.; Cahill, L.E.; Hou, T.; Ludwig, D.S.; Mozaffarian, D.; Willett, W.C.; Hu, F.B.; Rimm, E.B. Changes in Intake of Fruits and Vegetables and Weight Change in United States Men and Women Followed for Up to 24 Years: Analysis from Three Prospective Cohort Studies. PLoS Med. 2015, 12, e1001878. [Google Scholar] [CrossRef]
- Sesso, H.D.; Buring, J.E.; Christen, W.G.; Kurth, T.; Belanger, C.; MacFadyen, J.; Bubes, V.; Manson, J.E.; Glynn, R.J.; Gaziano, J.M. Vitamins E and C in the prevention of cardiovascular disease in men: The Physicians’ Health Study II randomized controlled trial. JAMA 2008, 300, 2123–2133. [Google Scholar] [CrossRef] [Green Version]
- Aguilar TA, F.; Navarro BC, H.; Pérez, J.A.M. Endogenous antioxidants: A review of their role in oxidative stress. In A Master Regulator of Oxidative Stress-the Transcription Factor nrf2; BoD–Books on Demand: Norderstedt, Germany, 2016; pp. 3–20. [Google Scholar]
- Fernández-Sánchez, A.; Madrigal-Santillán, E.; Bautista, M.; Esquivel-Soto, J.; Morales-González, Á.; Esquivel-Chirino, C.; Durante-Montiel, I.; Sánchez-Rivera, G.; Valadez-Vega, C.; Morales-González, J.A. Inflammation, oxidative stress, and obesity. Int. J. Mol. Sci. 2011, 12, 3117–3132. [Google Scholar] [CrossRef] [Green Version]
- Zorov, D.B.; Juhaszova, M.; Sollott, S.J. Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol. Rev. 2014, 94, 909–950. [Google Scholar] [CrossRef] [Green Version]
- Dato, S.; Crocco, P.; D’Aquila, P.; De Rango, F.; Bellizzi, D.; Rose, G.; Passarino, G. Exploring the role of genetic variability and lifestyle in oxidative stress response for healthy aging and longevity. Int. J. Mol. Sci. 2013, 14, 16443–16472. [Google Scholar] [CrossRef] [Green Version]
- Valko, M.; Rhodes, C.J.; Moncol, J.; Izakovic, M.; Mazur, M. Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chem.-Biol. Interact. 2006, 160, 1–40. [Google Scholar] [CrossRef]
- Christ, A.; Lauterbach, M.; Latz, E. Western Diet and the Immune System: An Inflammatory Connection. Immunity 2019, 51, 794–811. [Google Scholar] [CrossRef]
- Macho-González, A.; Garcimartín, A.; López-Oliva, M.E.; Bastida, S.; Benedí, J.; Ros, G.; Nieto, G.; Sánchez-Muniz, F.J. Can Meat and Meat-Products Induce Oxidative Stress? Antioxidants 2020, 9, 638. [Google Scholar] [CrossRef] [PubMed]
- Aune, D.; Ursin, G.; Veierød, M.B. Meat consumption and the risk of type 2 diabetes: A systematic review and meta-analysis of cohort studies. Diabetologia 2009, 52, 2277–2287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kaczmarczyk, M.M.; Miller, M.J.; Freund, G.G. The health benefits of dietary fiber: Beyond the usual suspects of type 2 diabetes mellitus, cardiovascular disease and colon cancer. Metab. Clin. Exp. 2012, 61, 1058–1066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Slavin, J. Why whole grains are protective: Biological mechanisms. Proc. Nutr. Soc. 2003, 62, 129–134. [Google Scholar] [CrossRef]
- Alzoubi, K.H.; Khabour, O.F.; Salah, H.A.; Abu Rashid, B.E. The combined effect of sleep deprivation and Western diet on spatial learning and memory: Role of BDNF and oxidative stress. J. Mol. Neurosci. MN 2013, 50, 124–133. [Google Scholar] [CrossRef]
- Halliwell, B. Free radicals and other reactive species in disease. e LS 2001. [Google Scholar] [CrossRef]
- Radak, Z.; Zhao, Z.; Koltai, E.; Ohno, H.; Atalay, M. Oxygen consumption and usage during physical exercise: The balance between oxidative stress and ROS-dependent adaptive signaling. Antioxid. Redox Signal. 2013, 18, 1208–1246. [Google Scholar] [CrossRef] [Green Version]
- Gomez-Cabrera, M.C.; Domenech, E.; Romagnoli, M.; Arduini, A.; Borras, C.; Pallardo, F.V.; Sastre, J.; Vina, J. Oral administration of vitamin C decreases muscle mitochondrial biogenesis and hampers training-induced adaptations in endurance performance. Am. J. Clin. Nutr. 2008, 87, 142–149. [Google Scholar] [CrossRef] [Green Version]
- Hecht, S.S. Cigarette smoking and lung cancer: Chemical mechanisms and approaches to prevention. Lancet Oncol. 2002, 3, 461–469. [Google Scholar] [CrossRef] [PubMed]
- Ravn-Haren, G.; Olsen, A.; Tjønneland, A.; Dragsted, L.O.; Nexø, B.A.; Wallin, H.; Overvad, K.; Raaschou-Nielsen, O.; Vogel, U. Associations between GPX1 Pro198Leu polymorphism, erythrocyte GPX activity, alcohol consumption and breast cancer risk in a prospective cohort study. Carcinogenesis 2006, 27, 820–825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barton, G.M. A calculated response: Control of inflammation by the innate immune system. J. Clin. Investig. 2008, 118, 413–420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hwang, J.S.; Wu, T.L.; Chou, S.C.; Ho, C.; Chang, P.Y.; Tsao, K.C.; Huang, J.Y.; Sun, C.F.; Wu, J.T. Development of multiple complications in type 2 diabetes is associated with the increase of multiple markers of chronic inflammation. J. Clin. Lab. Anal. 2008, 22, 6–13. [Google Scholar] [CrossRef]
- Multhoff, G.; Molls, M.; Radons, J. Chronic inflammation in cancer development. Front. Immunol. 2011, 2, 98. [Google Scholar] [CrossRef] [Green Version]
- Esmaillzadeh, A.; Kimiagar, M.; Mehrabi, Y.; Azadbakht, L.; Hu, F.B.; Willett, W.C. Dietary patterns, insulin resistance, and prevalence of the metabolic syndrome in women. Am. J. Clin. Nutr. 2007, 85, 910–918. [Google Scholar] [CrossRef] [Green Version]
- Calder, P.C.; Ahluwalia, N.; Brouns, F.; Buetler, T.; Clement, K.; Cunningham, K.; Esposito, K.; Jönsson, L.S.; Kolb, H.; Lansink, M.; et al. Dietary factors and low-grade inflammation in relation to overweight and obesity. Br. J. Nutr. 2011, 106 (Suppl. 3), S5–S78. [Google Scholar] [CrossRef]
- Mittal, M.; Siddiqui, M.R.; Tran, K.; Reddy, S.P.; Malik, A.B. Reactive oxygen species in inflammation and tissue injury. Antioxid. Redox Signal. 2014, 20, 1126–1167. [Google Scholar] [CrossRef] [Green Version]
- Chen, J.; Ding, X.; Wu, R.; Tong, B.; Zhao, L.; Lv, H.; Meng, X.; Liu, Y.; Ren, B.; Li, J.; et al. Novel Sesquiterpene Glycoside from Loquat Leaf Alleviates Type 2 Diabetes Mellitus Combined with Nonalcoholic Fatty Liver Disease by Improving Insulin Resistance, Oxidative Stress, Inflammation, and Gut Microbiota Composition. J. Agric. Food Chem. 2021, 69, 14176–14191. [Google Scholar] [CrossRef]
- Sies, H. Oxidative stress: A concept in redox biology and medicine. Redox Biol. 2015, 4, 180–183. [Google Scholar] [CrossRef] [Green Version]
- Kim, M.-S.; Chun, S.-S.; Choi, J.-H. Effects of turmeric (Curcuma longa L.) on antioxidative systems and oxidative damage in rats fed a high fat and cholesterol diet. J. Korean Soc. Food Sci. Nutr. 2013, 42, 570–576. [Google Scholar] [CrossRef] [Green Version]
- Niki, E.; Yoshida, Y.; Saito, Y.; Noguchi, N. Lipid peroxidation: Mechanisms, inhibition, and biological effects. Biochem. Biophys. Res. Commun. 2005, 338, 668–676. [Google Scholar] [CrossRef]
- Micha, R.; Mozaffarian, D. Saturated fat and cardiometabolic risk factors, coronary heart disease, stroke, and diabetes: A fresh look at the evidence. Lipids 2010, 45, 893–905. [Google Scholar] [CrossRef] [Green Version]
- Djuricic, I.; Calder, P.C. Beneficial Outcomes of Omega-6 and Omega-3 Polyunsaturated Fatty Acids on Human Health: An Update for 2021. Nutrients 2021, 13, 2421. [Google Scholar] [CrossRef] [PubMed]
- Calder, P.C. Nutrition, immunity and COVID-19. BMJ Nutr. Prev. Health 2020, 3, 74–92. [Google Scholar] [CrossRef] [PubMed]
- Ghanim, H.; Abuaysheh, S.; Sia, C.L.; Korzeniewski, K.; Chaudhuri, A.; Fernandez-Real, J.M.; Dandona, P. Increase in plasma endotoxin concentrations and the expression of Toll-like receptors and suppressor of cytokine signaling-3 in mononuclear cells after a high-fat, high-carbohydrate meal: Implications for insulin resistance. Diabetes Care 2009, 32, 2281–2287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Takeda, K.; Akira, S. TLR signaling pathways. Semin. Immunol. 2004, 16, 3–9. [Google Scholar] [CrossRef] [PubMed]
- Vlassara, H.; Uribarri, J. Advanced glycation end products (AGE) and diabetes: Cause, effect, or both? Curr. Diabetes Rep. 2014, 14, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Birlouez-Aragon, I.; Saavedra, G.; Tessier, F.J.; Galinier, A.; Ait-Ameur, L.; Lacoste, F.; Niamba, C.N.; Alt, N.; Somoza, V.; Lecerf, J.M. A diet based on high-heat-treated foods promotes risk factors for diabetes mellitus and cardiovascular diseases. Am. J. Clin. Nutr. 2010, 91, 1220–1226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Perrone, A.; Giovino, A.; Benny, J.; Martinelli, F. Advanced Glycation End Products (AGEs): Biochemistry, Signaling, Analytical Methods, and Epigenetic Effects. Oxid. Med. Cell Longev. 2020, 2020, 3818196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khalid, M.; Petroianu, G.; Adem, A. Advanced Glycation End Products and Diabetes Mellitus: Mechanisms and Perspectives. Biomolecules 2022, 12, 542. [Google Scholar] [CrossRef] [PubMed]
- Stadler, R.H.; Theurillat, V. Heat-generated toxicants in foods (acrylamide, MCPD esters, glycidyl esters, furan, and related compounds). In Chemical Contaminants and Residues in Food; Elsevier: Amsterdam, The Netherlands, 2017; pp. 171–195. [Google Scholar]
- Uribarri, J.; Woodruff, S.; Goodman, S.; Cai, W.; Chen, X.; Pyzik, R.; Yong, A.; Striker, G.E.; Vlassara, H. Advanced glycation end products in foods and a practical guide to their reduction in the diet. J. Am. Diet. Assoc. 2010, 110, 911–916.e12. [Google Scholar] [CrossRef] [Green Version]
- Hills, R.D., Jr.; Pontefract, B.A.; Mishcon, H.R.; Black, C.A.; Sutton, S.C.; Theberge, C.R. Gut Microbiome: Profound Implications for Diet and Disease. Nutrients 2019, 11, 1613. [Google Scholar] [CrossRef] [Green Version]
- Makki, K.; Deehan, E.C.; Walter, J.; Bäckhed, F. The Impact of Dietary Fiber on Gut Microbiota in Host Health and Disease. Cell Host Microbe 2018, 23, 705–715. [Google Scholar] [CrossRef] [Green Version]
- Singh, R.K.; Chang, H.-W.; Yan, D.; Lee, K.M.; Ucmak, D.; Wong, K.; Abrouk, M.; Farahnik, B.; Nakamura, M.; Zhu, T.H.; et al. Influence of diet on the gut microbiome and implications for human health. J. Transl. Med. 2017, 15, 73. [Google Scholar] [CrossRef] [Green Version]
- Cani, P.D.; Knauf, C. How gut microbes talk to organs: The role of endocrine and nervous routes. Mol. Metab. 2016, 5, 743–752. [Google Scholar] [CrossRef]
- Świątecka, D.; Narbad, A.; Ridgway, K.P.; Kostyra, H. The study on the impact of glycated pea proteins on human intestinal bacteria. Int. J. Food Microbiol. 2011, 145, 267–272. [Google Scholar]
- De Bandt, J.-P.; Waligora-Dupriet, A.-J.; Butel, M.-J. Intestinal microbiota in inflammation and insulin resistance: Relevance to humans. Curr. Opin. Clin. Nutr. Metab. Care 2011, 14, 334–340. [Google Scholar] [CrossRef]
- Kasai, C.; Sugimoto, K.; Moritani, I.; Tanaka, J.; Oya, Y.; Inoue, H.; Tameda, M.; Shiraki, K.; Ito, M.; Takei, Y.; et al. Comparison of the gut microbiota composition between obese and non-obese individuals in a Japanese population, as analyzed by terminal restriction fragment length polymorphism and next-generation sequencing. BMC Gastroenterol. 2015, 15, 100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Finucane, M.M.; Sharpton, T.J.; Laurent, T.J.; Pollard, K.S. A taxonomic signature of obesity in the microbiome? Getting to the guts of the matter. PLoS ONE 2014, 9, e84689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stojanov, S.; Berlec, A.; Štrukelj, B. The Influence of Probiotics on the Firmicutes/Bacteroidetes Ratio in the Treatment of Obesity and Inflammatory Bowel disease. Microorganisms 2020, 8, 1715. [Google Scholar] [CrossRef]
- Beam, A.; Clinger, E.; Hao, L. Effect of Diet and Dietary Components on the Composition of the Gut Microbiota. Nutrients 2021, 13, 2795. [Google Scholar] [CrossRef]
- Magne, F.; Gotteland, M.; Gauthier, L.; Zazueta, A.; Pesoa, S.; Navarrete, P.; Balamurugan, R. The Firmicutes/Bacteroidetes Ratio: A Relevant Marker of Gut Dysbiosis in Obese Patients? Nutrients 2020, 12, 1474. [Google Scholar] [CrossRef]
- Statovci, D.; Aguilera, M.; MacSharry, J.; Melgar, S. The Impact of Western Diet and Nutrients on the Microbiota and Immune Response at Mucosal Interfaces. Front. Immunol. 2017, 8, 838. [Google Scholar] [CrossRef] [Green Version]
- Bolte, L.A.; Vich Vila, A.; Imhann, F.; Collij, V.; Gacesa, R.; Peters, V.; Wijmenga, C.; Kurilshikov, A.; Campmans-Kuijpers, M.J.E.; Fu, J.; et al. Long-term dietary patterns are associated with pro-inflammatory and anti-inflammatory features of the gut microbiome. Gut 2021, 70, 1287–1298. [Google Scholar] [CrossRef]
- Astudillo-López, C.C.; Castro-Alarcón, N.; Ariza, A.C.; Muñoz-Valle, J.F.; de la Cruz-Mosso, U.; Flores-Alfaro, E.; del Moral-Hernández, O.; Moreno-Godínez, M.E.; Ramírez-Vargas, M.A.; Matia-Garcia, I.; et al. Influence of Diet and Levels of Zonulin, Lipopolysaccharide and C-Reactive Protein on Cardiometabolic Risk Factors in Young Subjects. Nutrients 2021, 13, 4472. [Google Scholar] [CrossRef]
- Celiberto, L.; Graef, F.; Healey, G.; Bosman, E.S.; Jacobson, K.; Sly, L.M.; Vallance, B. Inflammatory bowel disease and immunonutrition: Novel therapeutic approaches through modulation of diet and the gut microbiome. Immunology 2018, 155, 36–52. [Google Scholar] [CrossRef]
- Tsigalou, C.; Konstantinidis, T.; Paraschaki, A.; Stavropoulou, E.; Voidarou, C.; Bezirtzoglou, E. Mediterranean Diet as a Tool to Combat Inflammation and Chronic Diseases. An Overview. Biomedicines 2020, 8, 201. [Google Scholar] [CrossRef]
- Illescas, O.; Rodríguez-Sosa, M.; Gariboldi, M. Mediterranean Diet to Prevent the Development of Colon Diseases: A Meta-Analysis of Gut Microbiota Studies. Nutrients 2021, 13, 2234. [Google Scholar] [CrossRef]
- Bottero, V.; Potashkin, J.A. A Comparison of Gene Expression Changes in the Blood of Individuals Consuming Diets Supplemented with Olives, Nuts or Long-Chain Omega-3 Fatty Acids. Nutrients 2020, 12, 3765. [Google Scholar] [CrossRef]
- Clemente-Suárez, V.J.; Ramos-Campo, D.J.; Mielgo-Ayuso, J.; Dalamitros, A.A.; Nikolaidis, P.A.; Hormeño-Holgado, A.; Tornero-Aguilera, J.F. Nutrition in the Actual COVID-19 Pandemic. A Narrative Review. Nutrients 2021, 13, 1924. [Google Scholar] [CrossRef] [PubMed]
- David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hullar MA, J.; Fu, B.C. Diet, the gut microbiome, and epigenetics. Cancer J. 2014, 20, 170–175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wilson, A.S.; Koller, K.R.; Ramaboli, M.C.; Nesengani, L.T.; Ocvirk, S.; Chen, C.; Flanagan, C.A.; Sapp, F.R.; Merritt, Z.T.; Bhatti, F.; et al. Diet and the Human Gut Microbiome: An International Review. Dig. Dis. Sci. 2020, 65, 723–740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, E.Y.; Devkota, S.; Moscoso, D.; Chang, E.B.; Leone, V.A. The role of diet in triggering human inflammatory disorders in the modern age. Microbes Infect. 2013, 15, 765–774. [Google Scholar] [CrossRef]
- Shi, Z. Gut Microbiota: An Important Link between Western Diet and Chronic Diseases. Nutrients 2019, 11, 2287. [Google Scholar] [CrossRef] [Green Version]
- Srour, B.; Kordahi, M.C.; Bonazzi, E.; Deschasaux-Tanguy, M.; Touvier, M.; Chassaing, B. Ultra-processed foods and human health: From epidemiological evidence to mechanistic insights. Lancet Gastroenterol. Hepatol. 2022, 7, 1128–1140. [Google Scholar] [CrossRef] [PubMed]
- Monteiro, C.A.; Cannon, G.; Levy, R.B.; Moubarac, J.-C.; Louzada, M.L.C.; Rauber, F.; Khandpur, N.; Cediel, G.; Neri, D.; Martinez-Steele, E.; et al. Ultra-processed foods: What they are and how to identify them. Public Health Nutr. 2019, 22, 936–941. [Google Scholar] [CrossRef]
- Schnabel, L.; Kesse-Guyot, E.; Allès, B.; Touvier, M.; Srour, B.; Hercberg, S.; Buscail, C.; Julia, C. Association Between Ultraprocessed Food Consumption and Risk of Mortality Among Middle-aged Adults in France. JAMA Intern. Med. 2019, 179, 490–498. [Google Scholar] [CrossRef] [PubMed]
- Machado, P.P.; Steele, E.M.; Levy, R.B.; Sui, Z.; Rangan, A.; Woods, J.; Gill, T.; Scrinis, G.; Monteiro, C.A. Ultra-processed foods and recommended intake levels of nutrients linked to non-communicable diseases in Australia: Evidence from a nationally representative cross-sectional study. BMJ Open 2019, 9, e029544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, F.; Liu, X.; Wang, W.; Zhang, D. Consumption of vegetables and fruit and the risk of inflammatory bowel disease: A meta-analysis. Eur. J. Gastroenterol. Hepatol. 2015, 27, 623–630. [Google Scholar] [CrossRef] [PubMed]
- Lane, M.M.; Gamage, E.; Travica, N.; Dissanayaka, T.; Ashtree, D.N.; Gauci, S.; Lotfaliany, M.; O’Neil, A.; Jacka, F.N.; Marx, W. Ultra-Processed Food Consumption and Mental Health: A Systematic Review and Meta-Analysis of Observational Studies. Nutrients 2022, 14, 2568. [Google Scholar] [CrossRef]
- Rinninella, E.; Cintoni, M.; Raoul, P.; Lopetuso, L.R.; Scaldaferri, F.; Pulcini, G.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. Food Components and Dietary Habits: Keys for a Healthy Gut Microbiota Composition. Nutrients 2019, 11, 2393. [Google Scholar] [CrossRef] [Green Version]
- Frankenfeld, C.L.; Sikaroodi, M.; Lamb, E.; Shoemaker, S.; Gillevet, P.M. High-intensity sweetener consumption and gut microbiome content and predicted gene function in a cross-sectional study of adults in the United States. Ann. Epidemiol. 2015, 25, 736–742.e4. [Google Scholar] [CrossRef]
- Ketnawa, S.; Reginio, F.C., Jr.; Thuengtung, S.; Ogawa, Y. Changes in bioactive compounds and antioxidant activity of plant-based foods by gastrointestinal digestion: A review. Crit. Rev. Food Sci. Nutr. 2022, 62, 4684–4705. [Google Scholar] [CrossRef]
- Redondo-Useros, N.; Nova, E.; González-Zancada, N.; Díaz, L.E.; Gómez-Martínez, S.; Marcos, A. Microbiota and Lifestyle: A Special Focus on Diet. Nutrients 2020, 12, 1776. [Google Scholar] [CrossRef]
- Ray, S.K.; Mukherjee, S. Evolving Interplay Between Dietary Polyphenols and Gut Microbiota—An Emerging Importance in Healthcare. Front. Nutr. 2021, 8, 634944. [Google Scholar] [CrossRef]
- Wu, T.; Grootaert, C.; Pitart, J.; Vidovic, N.K.; Kamiloglu, S.; Possemiers, S.; Glibetic, M.; Smagghe, G.; Raes, K.; Van de Wiele, T.; et al. Aronia (Aronia melanocarpa) Polyphenols Modulate the Microbial Community in a Simulator of the Human Intestinal Microbial Ecosystem (SHIME) and Decrease Secretion of Proinflammatory Markers in a Caco-2/endothelial Cell Coculture Model. Mol. Nutr. Food Res. 2018, 62, e1800607. [Google Scholar] [CrossRef]
- Cory, H.; Passarelli, S.; Szeto, J.; Tamez, M.; Mattei, J. The Role of Polyphenols in Human Health and Food Systems: A Mini-Review. Front Nutr 2018, 5, 87. [Google Scholar] [CrossRef] [Green Version]
- Neufeld, K.M.; Kang, N.; Bienenstock, J.; Foster, J.A. Reduced anxiety-like behavior and central neurochemical change in germ-free mice. Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc. 2011, 23, 255–264.e119. [Google Scholar] [CrossRef] [PubMed]
- Martín-Rodríguez, A.; Bustamante-Sánchez, Á; Martínez-Guardado, I.; Navarro-Jiménez, E.; Plata-SanJuan, E.; Tornero-Aguilera, J.F.; Clemente-Suárez, V.J. Infancy Dietary Patterns, Development, and Health: An Extensive Narrative Review. Children 2022, 9, 1072. [Google Scholar] [CrossRef] [PubMed]
- Cena, H.; Calder, P.C. Defining a Healthy Diet: Evidence for The Role of Contemporary Dietary Patterns in Health and Disease. Nutrients 2020, 12, 334. [Google Scholar] [CrossRef] [Green Version]
- García-García, F.J.; Monistrol-Mula, A.; Cardellach, F.; Garrabou, G. Nutrition, Bioenergetics, and Metabolic Syndrome. Nutrients 2020, 12, 2785. [Google Scholar] [CrossRef] [PubMed]
- Burtscher, J.; Burtscher, M.; Millet, G.P. The central role of mitochondrial fitness on antiviral defenses: An advocacy for physical activity during the COVID-19 pandemic. Redox Biol. 2021, 43, 101976. [Google Scholar] [CrossRef]
- Bach, D.; Pich, S.; Soriano, F.X.; Vega, N.; Baumgartner, B.; Oriola, J.; Daugaard, J.R.; Lloberas, J.; Camps, M.; Zierath, J.R.; et al. Mitofusin-2 determines mitochondrial network architecture and mitochondrial metabolism. A novel regulatory mechanism altered in obesity. J. Biol. Chem. 2003, 278, 17190–17197. [Google Scholar] [CrossRef] [Green Version]
- Yang, A.; Mottillo, E.P. Adipocyte lipolysis: From molecular mechanisms of regulation to disease and therapeutics. Biochem. J. 2020, 477, 985–1008. [Google Scholar] [CrossRef]
- Chen, Z.; Tao, S.; Li, X.; Yao, Q. Resistin destroys mitochondrial biogenesis by inhibiting the PGC-1α/NRF1/TFAM signaling pathway. Biochem. Biophys. Res. Commun. 2018, 504, 13–18. [Google Scholar] [CrossRef]
- Yamauchi, T.; Kadowaki, T. Physiological and pathophysiological roles of adiponectin and adiponectin receptors in the integrated regulation of metabolic and cardiovascular diseases. Int. J. Obes. 2008, 32 (Suppl. 7), S13–S18. [Google Scholar] [CrossRef] [Green Version]
- Ouchi, N.; Parker, J.L.; Lugus, J.J.; Walsh, K. Adipokines in inflammation and metabolic disease. Nature reviews. Immunology 2011, 11, 85–97. [Google Scholar] [CrossRef]
- Sparks, L.M.; Xie, H.; Koza, R.A.; Mynatt, R.; Hulver, M.W.; Bray, G.A.; Smith, S.R. A high-fat diet coordinately downregulates genes required for mitochondrial oxidative phosphorylation in skeletal muscle. Diabetes 2005, 54, 1926–1933. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, D.; Li, X.; Zhang, L.; Zhu, M.; Gao, L. A high-fat diet impairs mitochondrial biogenesis, mitochondrial dynamics, and the respiratory chain complex in rat myocardial tissues. J. Cell. Biochem. 2018, 119, 9602. [Google Scholar] [CrossRef] [Green Version]
- Hancock, C.R.; Han, D.H.; Chen, M.; Terada, S.; Yasuda, T.; Wright, D.C.; Holloszy, J.O. High-fat diets cause insulin resistance despite an increase in muscle mitochondria. Proc. Natl. Acad. Sci. USA 2008, 105, 7815–7820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lionetti, L.; Mollica, M.P.; Donizzetti, I.; Gifuni, G.; Sica, R.; Pignalosa, A.; Cavaliere, G.; Gaita, M.; De Filippo, C.; Zorzano, A.; et al. High-lard and high-fish-oil diets differ in their effects on function and dynamic behaviour of rat hepatic mitochondria. PLoS ONE 2014, 9, e92753. [Google Scholar] [CrossRef] [Green Version]
- Li, N.; Li, H.P.; Zhang, B.Y.; Zhang, L.; Shen, J.M.; Li, Q.Y. Effect of high-fat diet on respiratory function and diaphragm fibers in mice and its mitochondrial mechanism. Zhonghua Yi Xue Za Zhi 2021, 101, 2893–2899. [Google Scholar] [PubMed]
- Jheng, H.-F.; Tsai, P.-J.; Guo, S.-M.; Kuo, L.-H.; Chang, C.-S.; Su, I.-J.; Chang, C.-R.; Tsai, Y.-S. Mitochondrial fission contributes to mitochondrial dysfunction and insulin resistance in skeletal muscle. Mol. Cell. Biol. 2012, 32, 309–319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, T.; Robotham, J.L.; Yoon, Y. Increased production of reactive oxygen species in hyperglycemic conditions requires dynamic change of mitochondrial morphology. Proc. Natl. Acad. Sci. USA 2006, 103, 2653–2658. [Google Scholar] [CrossRef] [Green Version]
- Tarpey, M.D.; Davy, K.P.; McMillan, R.P.; Bowser, S.M.; Halliday, T.M.; Boutagy, N.E.; Davy, B.M.; Frisard, M.I.; Hulver, M.W. Skeletal muscle autophagy and mitophagy in endurance-trained runners before and after a high-fat meal. Mol. Metab. 2017, 6, 1597–1609. [Google Scholar] [CrossRef]
- Tong, M.; Saito, T.; Zhai, P.; Oka, S.; Mizushima, W.; Nakamura, M.; Sadoshima, J. Mitophagy Is Essential for Maintaining Cardiac Function During High Fat Diet-Induced Diabetic Cardiomyopathy. Circ. Res. 2019, 124, 1360–1371. [Google Scholar] [CrossRef]
- Shao, D.; Kolwicz, S.C., Jr.; Wang, P.; Roe, N.D.; Villet, O.; Nishi, K.; Hsu, Y.-W.A.; Flint, G.V.; Caudal, A.; Wang, W.; et al. Increasing Fatty Acid Oxidation Prevents High-Fat Diet-Induced Cardiomyopathy through Regulating Parkin-Mediated Mitophagy. Circulation 2020, 142, 983–997. [Google Scholar] [CrossRef] [PubMed]
- Psaltopoulou, T.; Hatzis, G.; Papageorgiou, N.; Androulakis, E.; Briasoulis, A.; Tousoulis, D. Socioeconomic status and risk factors for cardiovascular disease: Impact of dietary mediators. Hell. J. Cardiol. HJC Hell. Kardiol. Ep. 2017, 58, 32–42. [Google Scholar] [CrossRef]
- Méjean, C.; Droomers, M.; van der Schouw, Y.T.; Sluijs, I.; Czernichow, S.; Grobbee, D.E.; Bueno-De-Mesquita, H.B.; Beulens, J.W. The contribution of diet and lifestyle to socioeconomic inequalities in cardiovascular morbidity and mortality. Int. J. Cardiol. 2013, 168, 5190–5195. [Google Scholar] [CrossRef] [PubMed]
- Abdelhamid, A.S.; Brown, T.J.; Brainard, J.S.; Biswas, P.; Thorpe, G.C.; Moore, H.J.; Deane, K.H.; AlAbdulghafoor, F.K.; Summerbell, C.D.; Worthington, H.V.; et al. Omega-3 fatty acids for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst. Rev. 2018, 7, CD003177. [Google Scholar] [PubMed]
- Cho, S.S.; Qi, L.; Fahey, G.C.; Klurfeld, D.M. Consumption of cereal fiber, mixtures of whole grains and bran, and whole grains and risk reduction in type 2 diabetes, obesity, and cardiovascular disease. Am. J. Clin. Nutr. 2013, 98, 594–619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hartley, L.; Igbinedion, E.; Holmes, J.; Flowers, N.; Thorogood, M.; Clarke, A.; Stranges, S.; Hooper, L.; Rees, K. Increased consumption of fruit and vegetables for the primary prevention of cardiovascular diseases. Cochrane Database Syst. Rev. 2013, 2013, CD009874. [Google Scholar] [PubMed] [Green Version]
- Wallace, T.C.; Murray, R.; Zelman, K.M. The Nutritional Value and Health Benefits of Chickpeas and Hummus. Nutrients 2016, 8, 766. [Google Scholar] [CrossRef] [Green Version]
- Salehin, S.; Rasmussen, P.; Mai, S.; Mushtaq, M.; Agarwal, M.; Hasan, S.M.; Salehin, S.; Raja, M.; Gilani, S.; Khalife, W.I. Plant Based Diet and Its Effect on Cardiovascular Disease. Int. J. Environ. Res. Public Health 2023, 20, 3337. [Google Scholar] [CrossRef]
- Ghosh, J.C.; Perego, M.; Agarwal, E.; Bertolini, I.; Wang, Y.; Goldman, A.R.; Tang, H.-Y.; Kossenkov, A.V.; Landis, C.J.; Languino, L.R.; et al. Ghost mitochondria drive metastasis through adaptive GCN2/Akt therapeutic vulnerability. Proc. Natl. Acad. Sci. USA 2022, 119, e2115624119. [Google Scholar] [CrossRef]
- Benjamin, E.J.; Virani, S.S.; Callaway, C.W.; Chamberlain, A.M.; Chang, A.R.; Cheng, S.; Chiuve, S.E.; Cushman, M.; Delling, F.N.; Deo, R.; et al. Heart Disease and Stroke Statistics-2018 Update: A Report From the American Heart Association. Circulation 2018, 137, e67–e492. [Google Scholar] [CrossRef]
- Artinian, N.T.; Fletcher, G.F.; Mozaffarian, D.; Kris-Etherton, P.; Van Horn, L.; Lichtenstein, A.H.; Kumanyika, S.; Kraus, W.E.; Fleg, J.L.; Nancy, S.; et al. Redeker Interventions to promote physical activity and dietary lifestyle changes for cardiovascular risk factor reduction in adults: A scientific statement from the American Heart Association. Circulation 2010, 122, 406–441. [Google Scholar] [CrossRef] [Green Version]
- Anand, S.S.; Hawkes, C.; De Souza, R.J.; Mente, A.; Dehghan, M.; Nugent, R.; Zulyniak, M.A.; Weis, T.; Bernstein, A.M.; Krauss, R.M.; et al. Food Consumption and its Impact on Cardiovascular Disease: Importance of Solutions Focused on the Globalized Food System: A Report From the Workshop Convened by the World Heart Federation. J. Am. Coll. Cardiol. 2015, 66, 1590–1614. [Google Scholar] [CrossRef] [Green Version]
- Mozaffarian, D.; Ludwig, D.S. Dietary guidelines in the 21st century--a time for food. JAMA 2010, 304, 681–682. [Google Scholar] [CrossRef] [PubMed]
- Bowen, K.J.; Sullivan, V.K.; Kris-Etherton, P.M.; Petersen, K.S. Nutrition and cardiovascular disease—an update. Curr. Atheroscler. Rep. 2018, 20, 8. [Google Scholar] [CrossRef] [PubMed]
- Libby, P. Interleukin-1 Beta as a Target for Atherosclerosis Therapy: Biological Basis of CANTOS and Beyond. J. Am. Coll. Cardiol. 2017, 70, 2278–2289. [Google Scholar] [CrossRef] [PubMed]
- Ley, K.; Gerdes, N.; Winkels, H. ATVB Distinguished Scientist Award: How Costimulatory and Coinhibitory Pathways Shape Atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 764–777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Virmani, R.; Joner, M.; Sakakura, K. Recent highlights of ATVB: Calcification. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 1329–1332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Low Wang, C.C.; Hess, C.N.; Hiatt, W.R.; Goldfine, A.B. Clinical Update: Cardiovascular Disease in Diabetes Mellitus: Atherosclerotic Cardiovascular Disease and Heart Failure in Type 2 Diabetes Mellitus—Mechanisms, Management, and Clinical Considerations. Circulation 2016, 133, 2459–2502. [Google Scholar] [CrossRef]
- Yubero-Serrano, E.M.; Fernandez-Gandara, C.; Garcia-Rios, A.; Rangel-Zuñiga, O.A.; Gutierrez-Mariscal, F.M.; Torres-Peña, J.D.; Marin, C.; Lopez-Moreno, J.; Castaño, J.P.; Delgado-Lista, J.; et al. Mediterranean diet and endothelial function in patients with coronary heart disease: An analysis of the CORDIOPREV randomized controlled trial. PLoS Med. 2020, 17, e1003282. [Google Scholar] [CrossRef]
- Usui, F.; Kimura, H.; Ohshiro, T.; Tatsumi, K.; Kawashima, A.; Nishiyama, A.; Iwakura, Y.-I.; Ishibashi, S.; Takahashi, M. Interleukin-17 deficiency reduced vascular inflammation and development of atherosclerosis in Western diet-induced apoE-deficient mice. Biochem. Biophys. Res. Commun. 2012, 420, 72–77. [Google Scholar] [CrossRef]
- Denova-Gutiérrez, E.; Tucker, K.L.; Flores, M.; Barquera, S.; Salmerón, J. Dietary Patterns Are Associated with Predicted Cardiovascular Disease Risk in an Urban Mexican Adult Population. J. Nutr. 2016, 146, 90–97. [Google Scholar] [CrossRef] [Green Version]
- Atkins, J.L.; Whincup, P.H.; Morris, R.W.; Lennon, L.T.; Papacosta, O.; Wannamethee, S.G. Dietary patterns and the risk of CVD and all-cause mortality in older British men. Br. J. Nutr. 2016, 116, 1246–1255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oikonomou, E.; Psaltopoulou, T.; Georgiopoulos, G.; Siasos, G.; Kokkou, E.; Antonopoulos, A.; Vogiatzi, G.; Tsalamandris, S.; Gennimata, V.; Papanikolaou, A.; et al. Western Dietary Pattern Is Associated with Severe Coronary Artery Disease. Angiology 2018, 69, 339–346. [Google Scholar] [CrossRef] [PubMed]
- Rees, K.; Takeda, A.; Martin, N.; Ellis, L.; Wijesekara, D.; Vepa, A.; Das, A.; Hartley, L.; Stranges, S. Mediterranean-style diet for the primary and secondary prevention of cardiovascular disease. Cochrane Database Syst. Rev. 2019, 3, CD009825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Viscogliosi, G.; Cipriani, E.; Liguori, M.L.; Marigliano, B.; Saliola, M.; Ettorre, E.; Andreozzi, P.; Georgousopoulou, E.N.; D’Cunha, N.M.; Mellor, D.D.; et al. Mediterranean dietary pattern adherence: Associations with prediabetes, metabolic syndrome, and related microinflammation. Metab. Syndr. Relat. Disord. 2013, 11, 210–216. [Google Scholar] [CrossRef]
- Martín-Peláez, S.; Fito, M.; Castaner, O. Mediterranean Diet Effects on Type 2 Diabetes Prevention, Disease Progression, and Related Mechanisms. A Review. Nutrients 2020, 12, 2236. [Google Scholar] [CrossRef]
- Badimon, L.; Chagas, P.; Chiva-Blanch, G. Diet and Cardiovascular Disease: Effects of Foods and Nutrients in Classical and Emerging Cardiovascular Risk Factors. Curr. Med. Chem. 2019, 26, 3639–3651. [Google Scholar] [CrossRef]
- Casas, R.; Castro-Barquero, S.; Estruch, R.; Sacanella, E. Nutrition and Cardiovascular Health. Int. J. Mol. Sci. 2018, 19, 3988. [Google Scholar] [CrossRef] [Green Version]
- Gao, M.; Jebb, S.A.; Aveyard, P.; Ambrosini, G.L.; Perez-Cornago, A.; Carter, J.; Sun, X.; Piernas, C. Associations between dietary patterns and the incidence of total and fatal cardiovascular disease and all-cause mortality in 116,806 individuals from the UK Biobank: A prospective cohort study. BMC Med. 2021, 19, 83. [Google Scholar] [CrossRef]
- Paglia, L. The sweet danger of added sugars. Eur. J. Paediatr. Dent. 2019, 20, 89. [Google Scholar]
- Khan, T.A.; Tayyiba, M.; Agarwal, A.; Mejia, S.B.; de Souza, R.J.; Wolever, T.M.; Leiter, L.A.; Kendall, C.W.C.; Jenkins, D.J.A.; Sievenpiper, J.L. Relation of Total Sugars, Sucrose, Fructose, and Added Sugars with the Risk of Cardiovascular Disease: A Systematic Review and Dose-Response Meta-analysis of Prospective Cohort Studies. Mayo Clin. Proc. 2019, 94, 2399–2414. [Google Scholar] [CrossRef] [Green Version]
- Batista, E.S.; da Silva Rios, T.; Muñoz, V.R.; Jesus, J.S.; Vasconcelos, M.M.; da Cunha, D.T.; Marques-Rocha, J.L.; Nakandakari, S.C.B.R.; Lara, R.; da Silva, A.S.R.; et al. Omega-3 mechanism of action in inflammation and endoplasmic reticulum stress in mononuclear cells from overweight non-alcoholic fatty liver disease participants: Study protocol for the ‘Brazilian Omega Study’ (BROS)-a randomized controlled trial. Trials 2021, 22, 927. [Google Scholar] [CrossRef]
- Mozaffarian, D. Dietary and Policy Priorities for Cardiovascular Disease, Diabetes, and Obesity: A Comprehensive Review. Circulation 2016, 133, 187–225. [Google Scholar] [CrossRef]
- Silveira BK, S.; Oliveira TM, S.; Andrade, P.A.; Hermsdorff HH, M.; Rosa CD, O.B.; Franceschini SD, C.C. Dietary Pattern and Macronutrients Profile on the Variation of Inflammatory Biomarkers: Scientific Update. Cardiol. Res. Pract. 2018, 2018, 4762575. [Google Scholar] [PubMed] [Green Version]
- Dinu, M.; Abbate, R.; Gensini, G.F.; Casini, A.; Sofi, F. Vegetarian, vegan diets and multiple health outcomes: A systematic review with meta-analysis of observational studies. Crit. Rev. Food Sci. Nutr. 2017, 57, 3640–3649. [Google Scholar] [CrossRef]
- Zampelas, A.; Magriplis, E. Dietary patterns and risk of cardiovascular diseases: A review of the evidence. Proc. Nutr. Soc. 2020, 79, 68–75. [Google Scholar] [CrossRef]
- Zhong, V.W.; Van Horn, L.; Cornelis, M.C.; Wilkins, J.T.; Ning, H.; Carnethon, M.R.; Greenland, P.; Mentz, R.J.; Tucker, K.L.; Zhao, L.; et al. Associations of Dietary Cholesterol or Egg Consumption with Incident Cardiovascular Disease and Mortality. JAMA 2019, 321, 1081–1095. [Google Scholar] [CrossRef] [PubMed]
- Shah, B.; Newman, J.D.; Woolf, K. Anti-Inflammatory Effects of a Vegan Diet Versus the American Heart Association-Recommended Diet in Coronary Artery Disease Trial. J. Am. Heart Assoc. 2018, 7, e011367. [Google Scholar] [CrossRef] [Green Version]
- Djekic, D.; Shi, L.; Brolin, H.; Carlsson, F.; Särnqvist, C.; Savolainen, O.; Cao, Y.; Bäckhed, F.; Tremaroli, V.; Landberg, R.; et al. Effects of a Vegetarian Diet on Cardiometabolic Risk Factors, Gut Microbiota, and Plasma Metabolome in Subjects with Ischemic Heart Disease: A Randomized, Crossover Study. J. Am. Heart Assoc. 2020, 9, e016518. [Google Scholar] [CrossRef] [PubMed]
- Djekic, D.; Shi, L.; Calais, F.; Carlsson, F.; Landberg, R.; Hyötyläinen, T.; Frøbert, O. Effects of a Lacto-Ovo-Vegetarian Diet on the Plasma Lipidome and Its Association with Atherosclerotic Burden in Patients with Coronary Artery Disease-A Randomized, Open-Label, Cross-over Study. Nutrients 2020, 12, 3586. [Google Scholar] [CrossRef]
- Mishra, S.; Xu, J.; Agarwal, U.; Gonzales, J.; Levin, S.; Barnard, N.D. A multicenter randomized controlled trial of a plant-based nutrition program to reduce body weight and cardiovascular risk in the corporate setting: The GEICO study. Eur. J. Clin. Nutr. 2013, 67, 718–724. [Google Scholar] [CrossRef] [Green Version]
- Turner-McGrievy, G.M.; Wilcox, S.; Frongillo, E.A.; Murphy, E.A.; Hutto, B.; Wilson, M.; Davey, M.; Bernhart, J.A.; Okpara, N.; Bailey, S.; et al. Effect of a Plant-Based vs Omnivorous Soul Food Diet on Weight and Lipid Levels Among African American Adults. JAMA Netw. Open 2023, 6, e2250626. [Google Scholar] [CrossRef]
- Wright, N.; Wilson, L.; Smith, M.; Duncan, B.; McHugh, P. The BROAD study: A randomised controlled trial using a whole food plant-based diet in the community for obesity, ischaemic heart disease or diabetes. Nutr. Diabetes 2017, 7, e256. [Google Scholar] [CrossRef] [Green Version]
- Jenkins, D.J.A.; Wong, J.M.W.; Kendall, C.W.C.; Esfahani, A.; Ng, V.W.Y.; Leong, T.C.K.; A Faulkner, D.; Vidgen, E.; Paul, G.; Mukherjea, R.; et al. Effect of a 6-month vegan low-carbohydrate (‘Eco-Atkins’) diet on cardiovascular risk factors and body weight in hyperlipidaemic adults: A randomised controlled trial. BMJ Open 2014, 4, e003505. [Google Scholar] [CrossRef]
- Carson, J.A.S.; Lichtenstein, A.H.; Anderson, C.A.; Appel, L.J.; Kris-Etherton, P.M.; Meyer, K.A.; Petersen, K.; Polonsky, T.; Van Horn, L.; American Heart Association Nutrition Committee of the Council on Lifestyle and Cardiometabolic Health; et al. Dietary cholesterol and cardiovascular risk: A science advisory from the American Heart Association. Circulation 2020, 141, e39–e53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kuwahara, A.; Matsuda, K.; Kuwahara, Y.; Asano, S.; Inui, T.; Marunaka, Y. Microbiota-gut-brain axis: Enteroendocrine cells and the enteric nervous system form an interface between the microbiota and the central nervous system. Biomed. Res. 2020, 41, 199–216. [Google Scholar] [CrossRef] [PubMed]
- Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef]
- Bienenstock, J.; Kunze, W.; Forsythe, P. Microbiota and the gut-brain axis. Nutr. Rev. 2015, 73 (Suppl. 1), 28–31. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.-X.; Wang, Y.-P. Gut Microbiota-brain Axis. Chin. Med. J. 2016, 129, 2373–2380. [Google Scholar] [CrossRef] [PubMed]
- Dowling, L.R.; Strazzari, M.R.; Keely, S.; Kaiko, G.E. Enteric nervous system and intestinal epithelial regulation of the gut-brain axis. J. Allergy Clin. Immunol. 2022, 150, 513–522. [Google Scholar] [CrossRef]
- Bonaz, B.; Bazin, T.; Pellissier, S. The Vagus Nerve at the Interface of the Microbiota-Gut-Brain Axis. Front. Neurosci. 2018, 12, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cussotto, S.; Sandhu, K.V.; Dinan, T.G.; Cryan, J.F. The Neuroendocrinology of the Microbiota-Gut-Brain Axis: A Behavioural Perspective. Front. Neuroendocrinol. 2018, 51, 80–101. [Google Scholar] [CrossRef] [PubMed]
- Barber, T.M.; Valsamakis, G.; Mastorakos, G.; Hanson, P.; Kyrou, I.; Randeva, H.S.; Weickert, M.O. Dietary Influences on the Microbiota–Gut–Brain Axis. Int. J. Mol. Sci. 2021, 22, 3502. [Google Scholar] [CrossRef] [PubMed]
- Durgan, D.J.; Lee, J.; McCullough, L.D.; Bryan RM, J. Examining the Role of the Microbiota-Gut-Brain Axis in Stroke. Stroke 2019, 50, 2270–2277. [Google Scholar] [CrossRef] [PubMed]
- Rönnbäck, C.; Hansson, E. The Importance and Control of Low-Grade Inflammation Due to Damage of Cellular Barrier Systems That May Lead to Systemic Inflammation. Front. Neurol. 2019, 10, 533. [Google Scholar] [CrossRef] [Green Version]
- Minihane, A.M.; Vinoy, S.; Russell, W.R.; Baka, A.; Roche, H.M.; Tuohy, K.M.; Teeling, J.L.; Blaak, E.E.; Fenech, M.; Vauzour, D.; et al. Low-grade inflammation, diet composition and health: Current research evidence and its translation. Br. J. Nutr. 2015, 114, 999–1012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martinez-Medina, M.; Denizot, J.; Dreux, N.; Robin, F.; Billard, E.; Bonnet, R.; Darfeuille-Michaud, A.; Barnich, N. Western diet induces dysbiosis with increased E coli in CEABAC10 mice, alters host barrier function favouring AIEC colonisation. Gut 2014, 63, 116–124. [Google Scholar] [CrossRef]
- Rohleder, N. Stimulation of systemic low-grade inflammation by psychosocial stress. Psychosom. Med. 2014, 76, 181–189. [Google Scholar] [CrossRef] [PubMed]
- Walker, W.A. Dysbiosis. In The Microbiota in Gastrointestinal Pathophysiology; Elsevier: Amsterdam, The Netherlands, 2017; pp. 227–232. [Google Scholar]
- Noble, E.E.; Hsu, T.M.; Kanoski, S.E. Gut to Brain Dysbiosis: Mechanisms Linking Western Diet Consumption, the Microbiome, and Cognitive Impairment. Front. Behav. Neurosci. 2017, 11, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stopińska, K.; Radziwoń-Zaleska, M.; Domitrz, I. The Microbiota-Gut-Brain Axis as a Key to Neuropsychiatric Disorders: A Mini Review. J. Clin. Med. 2021, 10, 4640. [Google Scholar] [CrossRef]
- Shahda, M.; El-Sayed, A. Study of the prevalence of metabolic syndrome among psychiatric patients and its correlation with diagnosis and medications. Egypt. J. Psychiatry 2010, 31, n2. [Google Scholar]
- Mousa, F.A.; Dessoki, H.H.; El Kateb, S.M.; Ezzat, A.A.; Soltan, M.R. Metabolic syndrome in psychiatric patients (comparative study). Egypt. J. Psychiatry 2017, 38, 179. [Google Scholar]
- Penninx, B.W.J.H.; Lange, S.M.M. Metabolic syndrome in psychiatric patients: Overview, mechanisms, and implications. Dialogues Clin. Neurosci. 2018, 20, 63–73. [Google Scholar] [CrossRef]
- De Leon, J. Metabolic Syndrome and Psychiatric Illness: Interactions, Pathophysiology, Assessment, and Treatment. Am. J. Psychiatry 2008, 165, 1056–1057. [Google Scholar] [CrossRef]
- Kim, S.-H.; Kim, K.; Kwak, M.H.; Kim, H.J.; Kim, H.-S.; Han, K.H. The contribution of abdominal obesity and dyslipidemia to metabolic syndrome in psychiatric patients. Korean J. Intern. Med. 2010, 25, 168–173. [Google Scholar] [CrossRef]
- Fabbri, C.; Corponi, F.; Albani, D.; Raimondi, I.; Forloni, G.; Schruers, K.; Kasper, S.; Kautzky, A.; Zohar, J.; Souery, D.; et al. Pleiotropic genes in psychiatry: Calcium channels and the stress-related FKBP5 gene in antidepressant resistance. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2018, 81, 203–210. [Google Scholar] [CrossRef]
- Lee, P.H.; Feng, Y.-C.A.; Smoller, J.W. Pleiotropy and Cross-Disorder Genetics Among Psychiatric Disorders. Biol. Psychiatry 2021, 89, 20–31. [Google Scholar] [CrossRef]
- Gorwood, P. Generalized anxiety disorder and major depressive disorder comorbidity: An example of genetic pleiotropy? Eur. Psychiatry J. Assoc. Eur. Psychiatr. 2004, 19, 27–33. [Google Scholar] [CrossRef]
- Zheutlin, A.B.; Dennis, J.; Linnér, R.K.; Moscati, A.; Restrepo, N.; Straub, P.; Ruderfer, D.; Castro, V.M.; Chen, C.-Y.; Ge, T.; et al. Penetrance and Pleiotropy of Polygenic Risk Scores for Schizophrenia in 106,160 Patients Across Four Health Care Systems. Am. J. Psychiatry 2019, 176, 846–855. [Google Scholar] [CrossRef]
- Torrico, B.; Shaw, A.D.; Mosca, R.; Vivó-Luque, N.; Hervás, A.; Fernàndez-Castillo, N.; Aloy, P.; Bayés, M.; Fullerton, J.M.; Cormand, B.; et al. Truncating variant burden in high-functioning autism and pleiotropic effects of LRP1 across psychiatric phenotypes. J. Psychiatry Neurosci. JPN 2019, 44, 350–359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, F.; Tu, H.; Chen, T. The Microbiota-Gut-Brain Axis in Depression: The Potential Pathophysiological Mechanisms and Microbiota Combined Antidepression Effect. Nutrients 2022, 14, 2081. [Google Scholar] [CrossRef] [PubMed]
- Hamamah, S.; Aghazarian, A.; Nazaryan, A.; Hajnal, A.; Covasa, M. Role of Microbiota-Gut-Brain Axis in Regulating Dopaminergic Signaling. Biomedicines 2022, 10, 436. [Google Scholar] [CrossRef] [PubMed]
- Mayer, E.A.; Nance, K.; Chen, S. The Gut-Brain Axis. Annu. Rev. Med. 2022, 73, 439–453. [Google Scholar] [CrossRef] [PubMed]
- Holzer, P.; Farzi, A. Neuropeptides and the microbiota-gut-brain axis. Adv. Exp. Med. Biol. 2014, 817, 195–219. [Google Scholar]
- Xie, Y.; Zhou, G.; Wang, C.; Xu, X.; Li, C. Specific Microbiota Dynamically Regulate the Bidirectional Gut-Brain Axis Communications in Mice Fed Meat Protein Diets. J. Agric. Food Chem. 2019, 67, 1003–1017. [Google Scholar] [CrossRef]
- Walker, W.A.; Duffy, L.C. Diet and bacterial colonization: Role of probiotics and prebiotics. J. Nutr. Biochem. 1998, 9, 668–675. [Google Scholar] [CrossRef]
- Douglas, L.C.; Sanders, M.E. Probiotics and prebiotics in dietetics practice. J. Am. Diet. Assoc. 2008, 108, 510–521. [Google Scholar] [CrossRef]
- Green, M.; Arora, K.; Prakash, S. Microbial Medicine: Prebiotic and Probiotic Functional Foods to Target Obesity and Metabolic Syndrome. Int. J. Mol. Sci. 2020, 21, 2890. [Google Scholar] [CrossRef] [Green Version]
- Muir, J.G.; Lu, Z.X.; Young, G.P.; Cameron-Smith, D.; Collier, G.R.; O’dea, K. Resistant starch in the diet increases breath hydrogen and serum acetate in human subjects. Am. J. Clin. Nutr. 1995, 61, 792–799. [Google Scholar] [CrossRef]
- Popova, M.; Martin, C.; Eugène, M.; Mialon, M.; Doreau, M.; Morgavi, D. Effect of fibre-and starch-rich finishing diets on methanogenic Archaea diversity and activity in the rumen of feedlot bulls. Anim. Feed. Sci. Technol. 2011, 166, 113–121. [Google Scholar] [CrossRef] [Green Version]
- Giacco, R.; Clemente, G.; Luongo, D.; Lasorella, G.; Fiume, I.; Brouns, F.; Bornet, F.; Patti, L.; Cipriano, P.; Rivellese, A.A.; et al. Effects of short-chain fructo-oligosaccharides on glucose and lipid metabolism in mild hypercholesterolaemic individuals. Clin. Nutr. 2004, 23, 331–340. [Google Scholar] [CrossRef]
- Nanno, M.; Matsumoto, S.; Shida, K. Lactobacillus casei strain Shirota: Benefits based on a long history of usage. In ECAB Health Impact of Probiotics: Vision & Opportunities-E-Book; Elsevier: Bengaluru, India, 2014; p. 85. [Google Scholar]
- Weichselbaum, E. Potential benefits of probiotics--main findings of an in-depth review. Br. J. Community Nurs. 2010, 15, 110–114. [Google Scholar] [CrossRef] [Green Version]
- Pimentel, T.C.; Brandão, L.R.; de Oliveira, M.P.; da Costa WK, A.; Magnani, M. Health benefits and technological effects of Lacticaseibacillus casei-01: An overview of the scientific literature. Trends Food Sci. Technol. 2021, 114, 722–737. [Google Scholar] [CrossRef]
- Foster, L.M.; Tompkins, T.A.; Dahl, W.J. A comprehensive post-market review of studies on a probiotic product containing Lactobacillus helveticus R0052 and Lactobacillus rhamnosus R0011. Benef. Microbes 2011, 2, 319–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Taverniti, V.; Guglielmetti, S. Health-Promoting Properties of Lactobacillus helveticus. Front. Microbiol. 2012, 3, 392. [Google Scholar] [CrossRef] [Green Version]
- Benno, Y.; Mitsuoka, T. Impact of Bifidobacterium longum on human fecal microflora. Microbiol. Immunol. 1992, 36, 683–694. [Google Scholar] [CrossRef] [PubMed]
- Underwood, M.A.; German, J.B.; Lebrilla, C.B.; Mills, D.A. Bifidobacterium longum subspecies infantis: Champion colonizer of the infant gut. Pediatr. Res. 2015, 77, 229–235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abhari, K.; Hosseini, H. Psychobiotics: Next Generation Treatment for Mental Disorders. J. Clin. Nutr. Diet 2018, 4, 1–2. [Google Scholar] [CrossRef]
- Malesza, I.J.; Malesza, M.; Walkowiak, J.; Mussin, N.; Walkowiak, D.; Aringazina, R.; Bartkowiak-Wieczorek, J.; Mądry, E. High-Fat, Western-Style Diet, Systemic Inflammation, and Gut Microbiota: A Narrative Review. Cells 2021, 10, 3164. [Google Scholar] [CrossRef]
- Halton, T.L.; Willett, W.C.; Liu, S.; E Manson, J.; Stampfer, M.J.; Hu, F.B. Potato and french fry consumption and risk of type 2 diabetes in women. Am. J. Clin. Nutr. 2006, 83, 284–290. [Google Scholar] [CrossRef] [Green Version]
- Shan, Z.; Rehm, C.D.; Rogers, G. Trends in Dietary Carbohydrate, Protein, and Fat Intake and Diet Quality Among US Adults, 1999–2016. JAMA 2019, 322, 1178–1187. [Google Scholar] [CrossRef] [Green Version]
- Jain, A.P.; Aggarwal, K.K.; Zhang, P.-Y. Omega-3 fatty acids and cardiovascular disease. Eur. Rev. Med. Pharmacol. Sci. 2015, 19, 441–445. [Google Scholar]
- Shahidi, F.; Ambigaipalan, P. Omega-3 Polyunsaturated Fatty Acids and Their Health Benefits. Annu. Rev. Food Sci. Technol. 2018, 9, 345–381. [Google Scholar] [CrossRef]
- Nicholls, S.J.; Lincoff, A.M.; Garcia, M.; Bash, D.; Ballantyne, C.M.; Barter, P.J.; Davidson, M.H.; Kastelein, J.J.P.; Koenig, W.; McGuire, D.K.; et al. Effect of High-Dose Omega-3 Fatty Acids vs Corn Oil on Major Adverse Cardiovascular Events in Patients at High Cardiovascular Risk: The STRENGTH Randomized Clinical Trial. JAMA 2020, 324, 2268–2280. [Google Scholar] [CrossRef]
- Dreher, M.L. Whole Fruits and Fruit Fiber Emerging Health Effects. Nutrients 2018, 10, 1833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lonnie, M.; Hooker, E.; Brunstrom, J.M.; Corfe, B.M.; Green, M.A.; Watson, A.W.; Williams, E.A.; Stevenson, E.J.; Penson, S.; Johnstone, A.M. Protein for Life: Review of Optimal Protein Intake, Sustainable Dietary Sources and the Effect on Appetite in Ageing Adults. Nutrients 2018, 10, 360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, X.-T.; Liao, W.; Yu, H.-J.; Liu, M.-W.; Yuan, S.; Tang, B.-W.; Yang, X.-H.; Song, Y.; Huang, Y.; Cheng, S.-L.; et al. Combined effects of fruit and vegetables intake and physical activity on the risk of metabolic syndrome among Chinese adults. PLoS ONE 2017, 12, e0188533. [Google Scholar] [CrossRef] [Green Version]
- Rochlani, Y.; Pothineni, N.V.; Kovelamudi, S.; Mehta, J.L. Metabolic syndrome: Pathophysiology, management, and modulation by natural compounds. Ther. Adv. Cardiovasc. Dis. 2017, 11, 215–225. [Google Scholar] [CrossRef] [Green Version]
- Black, P.H. The inflammatory response is an integral part of the stress response: Implications for atherosclerosis, insulin resistance, type II diabetes and metabolic syndrome X. Brain Behav. Immun. 2003, 17, 350–364. [Google Scholar] [CrossRef]
- Drews, G.; Krippeit-Drews, P.; Düfer, M. Oxidative stress and beta-cell dysfunction. Pflug. Arch. Eur. J. Physiol. 2010, 460, 703–718. [Google Scholar] [CrossRef]
- Park, K.-H.; Park, W.J. Endothelial Dysfunction: Clinical Implications in Cardiovascular Disease and Therapeutic Approaches. J. Korean Med. Sci. 2015, 30, 1213–1225. [Google Scholar] [CrossRef] [Green Version]
- Cohen, D.H.; LeRoith, D. Obesity, type 2 diabetes, and cancer: The insulin and IGF connection. Endocr.-Relat. Cancer 2012, 19, F27–F45. [Google Scholar] [CrossRef]
- Margioris, A.N. Fatty acids and postprandial inflammation. Curr. Opin. Clin. Nutr. Metab. Care 2009, 12, 129–137. [Google Scholar] [CrossRef]
- Soeters, M.R.; Soeters, P.B.; Schooneman, M.G.; Houten, S.M.; Romijn, J.A. Adaptive reciprocity of lipid and glucose metabolism in human short-term starvation. Am. J. Physiology. Endocrinol. Metab. 2012, 303, E1397–E1407. [Google Scholar] [CrossRef] [Green Version]
- Ohashi, K.; Shibata, R.; Murohara, T.; Ouchi, N. Role of anti-inflammatory adipokines in obesity-related diseases. Trends Endocrinol. Metab. TEM 2014, 25, 348–355. [Google Scholar] [CrossRef] [PubMed]
- Shi, H.; Kokoeva, M.V.; Inouye, K.; Tzameli, I.; Yin, H.; Flier, J.S. TLR4 links innate immunity and fatty acid-induced insulin resistance. J. Clin. Investig. 2006, 116, 3015–3025. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luu, N.-T.; Madden, J.; Calder, P.C.; Grimble, R.F.; Shearman, C.P.; Chan, T.; Dastur, N.; Howell, W.M.; Rainger, G.E.; Nash, G.B. Dietary supplementation with fish oil modifies the ability of human monocytes to induce an inflammatory response. J. Nutr. 2007, 137, 2769–2774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Calder, P.C. Polyunsaturated fatty acids and inflammation. Biochem. Soc. Trans. 2005, 33, 423–427. [Google Scholar] [CrossRef] [Green Version]
- Bogani, P.; Galli, C.; Villa, M.; Visioli, F. Postprandial anti-inflammatory and antioxidant effects of extra virgin olive oil. Atherosclerosis 2007, 190, 181–186. [Google Scholar] [CrossRef]
- Pacheco, Y.M.; López, S.; Bermúdez, B.; Abia, R.; Villar, J.; Muriana, F.J.G. A meal rich in oleic acid beneficially modulates postprandial sICAM-1 and sVCAM-1 in normotensive and hypertensive hypertriglyceridemic subjects. J. Nutr. Biochem. 2008, 19, 200–205. [Google Scholar] [CrossRef]
- Calder, P.C. Immunomodulation by omega-3 fatty acids. Prostaglandins Leukot. Essent. Fat. Acids 2007, 77, 327–335. [Google Scholar] [CrossRef]
- Calder, P.C.; Deckelbaum, R.J. Omega-3 fatty acids: Time to get the messages right! Curr. Opin. Clin. Nutr. Metab. Care 2008, 11, 91–93. [Google Scholar] [CrossRef] [PubMed]
- Bozzetto, L.; Annuzzi, G.; Ragucci, M.; Di Donato, O.; Della Pepa, G.; Della Corte, G.; Griffo, E.; Anniballi, G.; Giacco, A.; Mancini, M.; et al. Insulin resistance, postprandial GLP-1 and adaptive immunity are the main predictors of NAFLD in a homogeneous population at high cardiovascular risk. Nutr. Metab. Cardiovasc. Dis. NMCD 2016, 26, 623–629. [Google Scholar] [CrossRef] [PubMed]
- Tang, M.W.; Koopman, F.A.; Visscher, J.P.; de Hair, M.J.; Gerlag, D.M.; Tak, P.P. Hormone, metabolic peptide, and nutrient levels in the earliest phases of rheumatoid arthritis-contribution of free fatty acids to an increased cardiovascular risk during very early disease. Clin. Rheumatol. 2017, 36, 269–278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zilversmit, D.B. Atherogenesis: A postprandial phenomenon. Circulation 1979, 60, 473–485. [Google Scholar] [CrossRef] [Green Version]
- Miles, E.A.; Calder, P.C. Effects of Citrus Fruit Juices and Their Bioactive Components on Inflammation and Immunity: A Narrative Review. Front. Immunol. 2021, 12, 712608. [Google Scholar] [CrossRef]
- Gantenbein, K.V.; Kanaka-Gantenbein, C. Mediterranean Diet as an Antioxidant: The Impact on Metabolic Health and Overall Wellbeing. Nutrients 2021, 13, 1951. [Google Scholar] [CrossRef]
- Anderson, J.W.; Baird, P.; Davis, R.H., Jr.; Ferreri, S.; Knudtson, M.; Koraym, A.; Waters, V.; Williams, C.L. Health benefits of dietary fiber. Nutr. Rev. 2009, 67, 188–205. [Google Scholar] [CrossRef]
- De Filippo, C.; Di Paola, M.; Ramazzotti, M.; Albanese, D.; Pieraccini, G.; Banci, E.; Miglietta, F.; Cavalieri, D.; Lionetti, P. Diet, Environments, and Gut Microbiota. A Preliminary Investigation in Children Living in Rural and Urban Burkina Faso and Italy. Front. Microbiol. 2017, 8, 1979. [Google Scholar] [CrossRef] [Green Version]
- Calabrese, C.M.; Valentini, A.; Calabrese, G. Gut Microbiota and Type 1 Diabetes Mellitus: The Effect of Mediterranean Diet. Front. Nutr. 2020, 7, 612773. [Google Scholar] [CrossRef]
- Tremblay, M.S.; Aubert, S.; Barnes, J.D.; Saunders, T.J.; Carson, V.; Latimer-Cheung, A.E.; Chastin, S.F.M.; Altenburg, T.M.; Chinapaw, M.J.M. SBRN Terminology Consensus Project Participants. Sedentary Behavior Research Network (SBRN)—Terminology Consensus Project process and outcome. Int. J. Behav. Nutr. Phys. Act. 2017, 14, 75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alkhatib, A.; Tsang, C.; Tiss, A.; Bahorun, T.; Arefanian, H.; Barake, R.; Khadir, A.; Tuomilehto, J. Functional Foods and Lifestyle Approaches for Diabetes Prevention and Management. Nutrients 2017, 9, 1310. [Google Scholar] [CrossRef] [Green Version]
- Gopinath, K.; Sudhandiran, G. Naringin modulates oxidative stress and inflammation in 3-nitropropionic acid-induced neurodegeneration through the activation of nuclear factor-erythroid 2-related factor-2 signalling pathway. Neuroscience 2012, 227, 134–143. [Google Scholar] [CrossRef] [PubMed]
- Alam, M.A.; Subhan, N.; Rahman, M.M.; Uddin, S.J.; Reza, H.M.; Sarker, S.D. Effect of citrus flavonoids, naringin and naringenin, on metabolic syndrome and their mechanisms of action. Adv. Nutr. 2014, 5, 404–417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Y.; Sun, Y.; Brenna, J.T.; Shen, Y.; Ye, K. Higher ratio of plasma omega-6/omega-3 fatty acids is associated with greater risk of all-cause, cancer, and cardiovascular mortality: A population-based cohort study in UK Biobank. medRxiv Prepr. Serv. Health Sci. 2023; in press. [Google Scholar] [CrossRef]
- Calder, P.C. Omega-3: The good oil. Nutr. Bull. 2017, 42, 132–140. [Google Scholar] [CrossRef] [Green Version]
- Calder, P.C. Very long-chain n-3 fatty acids and human health: Fact, fiction and the future. Proc. Nutr. Soc. 2018, 77, 52–72. [Google Scholar] [CrossRef] [Green Version]
- Baker, E.J.; Miles, E.A.; Burdge, G.C.; Yaqoob, P.; Calder, P.C. Metabolism and functional effects of plant-derived omega-3 fatty acids in humans. Prog. Lipid Res. 2016, 64, 30–56. [Google Scholar] [CrossRef]
- Widmer, R.J.; Flammer, A.J.; Lerman, L.O.; Lerman, A. The Mediterranean diet, its components, and cardiovascular disease. Am. J. Med. 2015, 128, 229–238. [Google Scholar] [CrossRef] [Green Version]
- Charbonneau, B.; O’Connor, H.M.; Wang, A.H.; Liebow, M.; Thompson, C.A.; Fredericksen, Z.S.; Macon, W.R.; Slager, S.L.; Call, T.G.; Habermann, T.M.; et al. Trans fatty acid intake is associated with increased risk and n3 fatty acid intake with reduced risk of non-hodgkin lymphoma. J. Nutr. 2013, 143, 672–681. [Google Scholar] [CrossRef] [Green Version]
- Kolb, H.; Martin, S. Environmental/lifestyle factors in the pathogenesis and prevention of type 2 diabetes. BMC Med. 2017, 15, 131. [Google Scholar] [CrossRef] [Green Version]
- Koene, R.J.; Prizment, A.E.; Blaes, A.; Konety, S.H. Shared Risk Factors in Cardiovascular Disease and Cancer. Circulation 2016, 133, 1104–1114. [Google Scholar] [CrossRef] [Green Version]
- Minelli, A.; Bellezza, I.; Conte, C.; Culig, Z. Oxidative stress-related aging: A role for prostate cancer? Biochim. Biophys. Acta 2009, 1795, 83–91. [Google Scholar] [CrossRef]
- Stieg, D.C.; Wang, Y.; Liu, L.-Z.; Jiang, B.-H. ROS and miRNA Dysregulation in Ovarian Cancer Development, Angiogenesis and Therapeutic Resistance. Int. J. Mol. Sci. 2022, 23, 6702. [Google Scholar] [CrossRef]
- Poyton, R.O.; Ball, K.A.; Castello, P.R. Mitochondrial generation of free radicals and hypoxic signaling. Trends Endocrinol. Metab. TEM 2009, 20, 332–340. [Google Scholar] [CrossRef]
- Walens, A.; DiMarco, A.V.; Lupo, R.; Kroger, B.R.; Damrauer, J.S.; Alvarez, J.V. CCL5 promotes breast cancer recurrence through macrophage recruitment in residual tumors. eLife 2019, 8, e43653. [Google Scholar] [CrossRef]
- Chao, T.; Furth, E.E.; Vonderheide, R.H. CXCR2-Dependent Accumulation of Tumor-Associated Neutrophils Regulates T-cell Immunity in Pancreatic Ductal Adenocarcinoma. Cancer Immunol. Res. 2016, 4, 968–982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ames, B.N. Low micronutrient intake may accelerate the degenerative diseases of aging through allocation of scarce micronutrients by triage. Proc. Natl. Acad. Sci. USA 2006, 103, 17589–17594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aune, D. Plant Foods, Antioxidant Biomarkers, and the Risk of Cardiovascular Disease, Cancer, and Mortality: A Review of the Evidence. Adv. Nutr. 2019, 10, S404–S421. [Google Scholar] [CrossRef] [Green Version]
- Harris, W.S.; Tintle, N.L.; Imamura, F.; Qian, F.; Korat, A.V.A.; Marklund, M.; Djoussé, L.; Bassett, J.K.; Carmichael, P.-H.; Chen, Y.-Y.; et al. Blood n-3 fatty acid levels and total and cause-specific mortality from 17 prospective studies. Nat. Commun. 2021, 12, 2329. [Google Scholar] [CrossRef]
- Nguyen, S.; Li, H.; Yu, D.; Cai, H.; Gao, J.; Gao, Y.; Luu, H.N.; Tran, H.; Xiang, Y.B.; Zheng, W.; et al. Dietary fatty acids and colorectal cancer risk in men: A report from the Shanghai Men’s Health Study and a meta-analysis. Int. J. Cancer 2021, 148, 77–89. [Google Scholar] [CrossRef] [PubMed]
- Michels, N.; Specht, I.O.; Heitmann, B.L.; Chajès, V.; Huybrechts, I. Dietary trans-fatty acid intake in relation to cancer risk: A systematic review and meta-analysis. Nutr. Rev. 2021, 79, 758–776. [Google Scholar] [CrossRef] [PubMed]
- Downs, S.M.; Loehr, S.; Wu, J.H.Y. Trans Fatty Acids: A Summary of the Evidence Relating Consumption to Cardiovascular Outcomes and the Efficacy of Prevention Policy to Reduce Levels in the Food Supply. In Preventive Nutrition: The Comprehensive Guide for Health Professionals; Springer: Cham, Switzerland, 2015; pp. 273–296. [Google Scholar]
- Han, S.N.; Leka, L.S.; Lichtenstein, A.H.; Ausman, L.M.; Schaefer, E.J.; Meydani, S.N. Effect of hydrogenated and saturated, relative to polyunsaturated, fat on immune and inflammatory responses of adults with moderate hypercholesterolemia. J. Lipid Res. 2002, 43, 445–452. [Google Scholar] [CrossRef]
- Mozaffarian, D.; Pischon, T.; E Hankinson, S.; Rifai, N.; Joshipura, K.; Willett, W.C.; Rimm, E.B. Dietary intake of trans fatty acids and systemic inflammation in women. Am. J. Clin. Nutr. 2004, 79, 606–612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seyyedsalehi, M.S.; Collatuzzo, G.; Rashidian, H.; Hadji, M.; Gholipour, M.; Mohebbi, E.; Kamangar, F.; Pukkala, E.; Huybrechts, I.; Gunter, M.J.; et al. Dietary Ruminant and Industrial Trans-Fatty Acids Intake and Colorectal Cancer Risk. Nutrients 2022, 14, 4912. [Google Scholar] [CrossRef] [PubMed]
- Supabphol, S.; Seubwai, W.; Wongkham, S.; Saengboonmee, C. High glucose: An emerging association between diabetes mellitus and cancer progression. J. Mol. Med. 2021, 99, 1175–1193. [Google Scholar] [CrossRef]
- Vaupel, P.; Schmidberger, H.; Mayer, A. The Warburg effect: Essential part of metabolic reprogramming and central contributor to cancer progression. Int. J. Radiat. Biol. 2019, 95, 912–919. [Google Scholar] [CrossRef]
- Czekajło, A.; Rozanska, D.; Mandecka, A.; Konikowska, K.; Madalińska, M.; Szuba, A.; Regulska-Ilow, B. Glycemic load and carbohydrates content in the diets of cancer patients. Rocz. Panstw. Zakl. Hig. 2017, 68, 261–268. [Google Scholar]
- Dang, C.V. Rethinking the Warburg effect with Myc micromanaging glutamine metabolism. Cancer Res. 2010, 70, 859–862. [Google Scholar] [CrossRef] [Green Version]
- DeBerardinis, R.J.; Cheng, T. Q’s next: The diverse functions of glutamine in metabolism, cell biology and cancer. Oncogene 2010, 29, 313–324. [Google Scholar] [CrossRef] [Green Version]
- de Groot, S.; Vreeswijk, M.P.; Welters, M.J.; Gravesteijn, G.; Boei, J.J.; Jochems, A.; Houtsma, D.; Putter, H.; van der Hoeven, J.J.; Nortier, J.W.; et al. The effects of short-term fasting on tolerance to (neo) adjuvant chemotherapy in HER2-negative breast cancer patients: A randomized pilot study. BMC Cancer 2015, 15, 652. [Google Scholar] [CrossRef] [Green Version]
- Derr, R.L.; Ye, X.; Islas, M.U.; Desideri, S.; Saudek, C.D.; Grossman, S.A. Association between hyperglycemia and survival in patients with newly diagnosed glioblastoma. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2009, 27, 1082–1086. [Google Scholar] [CrossRef] [PubMed]
- Fiolet, T.; Srour, B.; Sellem, L.; Kesse-Guyot, E.; Allès, B.; Méjean, C.; Deschasaux, M.; Fassier, P.; Latino-Martel, P.; Beslay, M.; et al. Consumption of ultra-processed foods and cancer risk: Results from NutriNet-Santé prospective cohort. BMJ 2018, 360, k322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tian, Y.; Su, L.; Wang, J.; Duan, X.; Jiang, X. Fruit and vegetable consumption and risk of the metabolic syndrome: A meta-analysis. Public Health Nutr. 2018, 21, 756–765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maino Vieytes, C.A.; Taha, H.M.; Burton-Obanla, A.A.; Douglas, K.G.; Arthur, A.E. Carbohydrate Nutrition and the Risk of Cancer. Curr. Nutr. Rep. 2019, 8, 230–239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Böhm, V.; Lietz, G.; Olmedilla-Alonso, B.; Phelan, D.; Reboul, E.; Bánati, D.; Borel, P.; Corte-Real, J.; de Lera, A.R.; Desmarchelier, C.; et al. From carotenoid intake to carotenoid blood and tissue concentrations—Implications for dietary intake recommendations. Nutr. Rev. 2021, 79, 544–573. [Google Scholar] [CrossRef]
- Ornish, D.; Magbanua, M.J.M.; Weidner, G.; Weinberg, V.; Kemp, C.; Green, C.; Mattie, M.D.; Marlin, R.; Simko, J.; Shinohara, K.; et al. Changes in prostate gene expression in men undergoing an intensive nutrition and lifestyle intervention. Proc. Natl. Acad. Sci. USA 2008, 105, 8369–8374. [Google Scholar] [CrossRef] [Green Version]
- Slavin, J.L. Mechanisms for the impact of whole grain foods on cancer risk. J. Am. Coll. Nutr. 2000, 19, 300S–307S. [Google Scholar] [CrossRef]
- Carlsen, M.H.; Halvorsen, B.L.; Holte, K.; Bøhn, S.K.; Dragland, S.; Sampson, L.; Willey, C.; Senoo, H.; Umezono, Y.; Sanada, C.; et al. The total antioxidant content of more than 3100 foods, beverages, spices, herbs and supplements used worldwide. Nutr. J. 2010, 9, 3. [Google Scholar] [CrossRef]
- Halvorsen, B.L.; Holte, K.; Myhrstad, M.C.W.; Barikmo, I.; Hvattum, E.; Remberg, S.F.; Wold, A.-B.; Haffner, K.; Baugerød, H.; Andersen, L.F.; et al. A systematic screening of total antioxidants in dietary plants. J. Nutr. 2002, 132, 461–471. [Google Scholar] [CrossRef] [Green Version]
- Comba, A.; Maestri, D.M.; A Berra, M.; Garcia, C.P.; Das, U.N.; Eynard, A.R.; E Pasqualini, M. Effect of ω-3 and ω-9 fatty acid rich oils on lipoxygenases and cyclooxygenases enzymes and on the growth of a mammary adenocarcinoma model. Lipids Health Dis. 2010, 9, 112. [Google Scholar] [CrossRef] [Green Version]
- Labrecque, L.; Lamy, S.; Chapus, A.; Mihoubi, S.; Durocher, Y.; Cass, B.; Bojanowski, M.W.; Gingras, D.; Béliveau, R. Combined inhibition of PDGF and VEGF receptors by ellagic acid, a dietary-derived phenolic compound. Carcinogenesis 2005, 26, 821–826. [Google Scholar] [CrossRef] [Green Version]
- Sung, B.; Pandey, M.K.; Ahn, K.S.; Yi, T.; Chaturvedi, M.M.; Liu, M.; Aggarwal, B.B. Anacardic acid (6-nonadecyl salicylic acid), an inhibitor of histone acetyltransferase, suppresses expression of nuclear factor-kappaB-regulated gene products involved in cell survival, proliferation, invasion, and inflammation through inhibition of the inhibitory subunit of nuclear factor-kappaBalpha kinase, leading to potentiation of apoptosis. Blood 2008, 111, 4880–4891. [Google Scholar]
- Carvalho, A.L.; Annoni, R.; Torres, L.H.; Durão, A.C.; Shimada, A.L.; Almeida, F.M.; Hebeda, C.B.; Lopes, F.D.; Dolhnikoff, M.; Martins, M.A.; et al. Anacardic acids from cashew nuts ameliorate lung damage induced by exposure to diesel exhaust particles in mice. Evid. Based Complement. Altern. Med. eCAM 2013, 2013, 549879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tsoukas, M.A.; Ko, B.-J.; Witte, T.R.; Dincer, F.; Hardman, W.E.; Mantzoros, C.S. Dietary walnut suppression of colorectal cancer in mice: Mediation by miRNA patterns and fatty acid incorporation. J. Nutr. Biochem. 2015, 26, 776–783. [Google Scholar] [CrossRef]
- Kanoski, S.E.; Davidson, T.L. Western diet consumption and cognitive impairment: Links to hippocampal dysfunction and obesity. Physiol. Behav. 2011, 103, 59–68. [Google Scholar] [CrossRef] [Green Version]
- Poutahidis, T.; Kleinewietfeld, M.; Smillie, C.; Levkovich, T.; Perrotta, A.; Bhela, S.; Varian, B.; Ibrahim, Y.; Lakritz, J.; Kearney, S.; et al. Microbial reprogramming inhibits Western diet-associated obesity. PLoS ONE 2013, 8, e68596. [Google Scholar] [CrossRef] [Green Version]
- Christ, A.; Latz, E. The Western lifestyle has lasting effects on metaflammation. Nat. Rev. Immunol. 2019, 19, 267–268. [Google Scholar] [CrossRef] [PubMed]
- Vuik, S.; Lerouge, A.; Guillemette, Y.; Feigl, A.; Aldea, A. The Economic Burden of Obesity; OECD iLibrary: Paris, France, 2019. [Google Scholar]
- Cecchini, M.; Vuik, S. The Heavy Burden of Obesity; OECD iLibrary: Paris, France, 2019. [Google Scholar]
- Peng, W.; Zhang, J.; Zhou, H.; Zhang, A.; Wang, Y.; Tian, X.; Wen, D.; Wang, Y. Obesity intervention efforts in China and the 2022 World Obesity Day. Glob. Health J. 2022, 6, 118–121. [Google Scholar] [CrossRef]
- Gooey, M.; Bacus, C.A.; Ramachandran, D.; Piya, M.K.; Baur, L.A. Health service approaches to providing care for people who seek treatment for obesity: Identifying challenges and ways forward. Public Health Res. Pract. 2022, 32, e3232228. [Google Scholar] [CrossRef]
- Shaw, J.E.; Zimmet, P.Z.; McCarty, D.; de Courten, M. Type 2 diabetes worldwide according to the new classification and criteria. Diabetes Care 2000, 23 (Suppl. 2), B5–B10. [Google Scholar]
- Fazeli Farsani, S.; van der Aa, M.P.; van der Vorst MM, J.; Knibbe CA, J.; de Boer, A. Global trends in the incidence and prevalence of type 2 diabetes in children and adolescents: A systematic review and evaluation of methodological approaches. Diabetologia 2013, 56, 1471–1488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zheng, Y.; Ley, S.H.; Hu, F.B. Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nat. Rev. Endocrinol. 2018, 14, 88–98. [Google Scholar] [CrossRef] [PubMed]
- Hussain, S.; Chowdhury, T.A. The Impact of Comorbidities on the Pharmacological Management of Type 2 Diabetes Mellitus. Drugs 2019, 79, 231–242. [Google Scholar] [CrossRef]
- Kim, H.S.; Shin, A.M.; Kim, M.K.; Kim, Y.N. Comorbidity study on type 2 diabetes mellitus using data mining. Korean J. Intern. Med. 2012, 27, 197–202. [Google Scholar] [CrossRef] [PubMed]
- Pantalone, K.M.; Hobbs, T.M.; Wells, B.J.; Kong, S.X.; Kattan, M.W.; Bouchard, J.; Yu, C.; Sakurada, B.; Milinovich, A.; Weng, W.; et al. Clinical characteristics, complications, comorbidities and treatment patterns among patients with type 2 diabetes mellitus in a large integrated health system. BMJ Open Diabetes Res. Care 2015, 3, e000093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nittari, G.; Scuri, S.; Petrelli, F.; Pirillo, I.; Di Luca, N.M.; Grappasonni, I. Fighting obesity in children from European World Health Organization member states. Epidemiological data, medical-social aspects, and prevention programs. La Clin. Ter. 2019, 170, e223–e230. [Google Scholar]
- Afolabi, H.A.; bin Zakariya, Z.; Shokri, A.B.A.; Hasim, M.N.B.M.; Vinayak, R.; Afolabi-Owolabi, O.T.; Elesho, R.F. The relationship between obesity and other medical comorbidities. Obes. Med. 2020, 17, 100164. [Google Scholar] [CrossRef]
- Seuring, T.; Archangelidi, O.; Suhrcke, M. The Economic Costs of Type 2 Diabetes: A Global Systematic Review. PharmacoEconomics 2015, 33, 811–831. [Google Scholar] [CrossRef] [Green Version]
- Hernan, W.H.; Brandle, M.; Zhang, P.; Williamson, D.F.; Matulik, M.J.; Ratner, R.E.; Lachin, J.M.; Engelgau, M.M.; Diabetes Prevention Program Research Group. Costs associated with the primary prevention of type 2 diabetes mellitus in the diabetes prevention program. Diabetes Care 2003, 26, 36–47. [Google Scholar]
- Dall, T.M.; Mann, S.E.; Zhang, Y.; Quick, W.W.; Seifert, R.F.; Martin, J.; Huang, E.A.; Zhang, S. Distinguishing the economic costs associated with type 1 and type 2 diabetes. Popul. Health Manag. 2009, 12, 103–110. [Google Scholar] [CrossRef]
- Kennedy-Martin, T.; Boye, K.S.; Peng, X. Cost of medication adherence and persistence in type 2 diabetes mellitus: A literature review. Patient Prefer. Adherence 2017, 11, 1103–1117. [Google Scholar] [CrossRef] [Green Version]
- Sikter, A. Psychosomatic Molecular Mechanisms of Metabolic Syndrome and Type 2 Diabetes. Part 2. Psychosomatic Mechanism of Metabolic Syndrome (a Theory). Acta Sci. Med. Sci. 2020, 4, 98–107. [Google Scholar]
- Harada, M.D. The Role of Health and Diet in the Development of Metabolic Syndrome Stratified by Race, Sex, and Age. Ph.D. Thesis, Walden University, Minneapolis, MN, USA, 2022. [Google Scholar]
- Boudreau, D.; Malone, D.; Raebel, M.; Fishman, P.; Nichols, G.; Feldstein, A.; Boscoe, A.; Ben-Joseph, R.; Magid, D.; Okamoto, L. Health care utilization and costs by metabolic syndrome risk factors. Metab. Syndr. Relat. Disord. 2009, 7, 305–314. [Google Scholar] [CrossRef]
- Belete, R.; Ataro, Z.; Abdu, A.; Sheleme, M. Global prevalence of metabolic syndrome among patients with type I diabetes mellitus: A systematic review and meta-analysis. Diabetol. Metab. Syndr. 2021, 13, 25. [Google Scholar] [CrossRef]
- Al-Rubeaan, K.; Bawazeer, N.; Al Farsi, Y.; Youssef, A.M.; Al-Yahya, A.A.; AlQumaidi, H.; Al-Malki, B.M.; Naji, K.A.; Al-Shehri, K.; Al Rumaih, F.I. Prevalence of metabolic syndrome in Saudi Arabia—A cross sectional study. BMC Endocr. Disord. 2018, 18, 16. [Google Scholar] [CrossRef]
- Amini, M.; Zayeri, F.; Salehi, M. Trend analysis of cardiovascular disease mortality, incidence, and mortality-to-incidence ratio: Results from global burden of disease study 2017. BMC Public Health 2021, 21, 401. [Google Scholar] [CrossRef]
- Lee, H.; Lee, Y.-H.; Kim, S.U.; Kim, H.C. Metabolic Dysfunction-Associated Fatty Liver Disease and Incident Cardiovascular Disease Risk: A Nationwide Cohort Study. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2021, 19, 2138–2147.e10. [Google Scholar] [CrossRef]
- Cainzos-Achirica, M.; Fedeli, U.; Sattar, N.; Agyemang, C.; Jenum, A.K.; McEvoy, J.W.; Murphy, J.D.; Brotons, C.; Elosua, R.; Bilal, U.; et al. Epidemiology, risk factors, and opportunities for prevention of cardiovascular disease in individuals of South Asian ethnicity living in Europe. Atherosclerosis 2019, 286, 105–113. [Google Scholar] [CrossRef] [Green Version]
- Vikulova, D.; Grubisic, M.; Zhao, Y.; Lynch, K.; Humphries, K.H.; Pimstone, S.N.; Brunham, L.R. Premature Atherosclerotic Cardiovascular Disease: Trends in Incidence, Risk Factors, and Sex-Related Differences, 2000 to 2016. J. Am. Heart Assoc. 2019, 8, e012178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Townsend, N.; Kazakiewicz, D.; Wright, F.L.; Timmis, A.; Huculeci, R.; Torbica, A.; Gale, C.P.; Achenbach, S.; Weidinger, F.; Vardas, P. Epidemiology of cardiovascular disease in Europe. Nat. Rev. Cardiol. 2022, 19, 133–143. [Google Scholar] [CrossRef] [PubMed]
- Dermadi, D.; Valo, S.; Ollila, S.; Soliymani, R.; Sipari, N.; Pussila, M.; Sarantaus, L.; Linden, J.; Baumann, M.; Nyström, M. Western Diet Deregulates Bile Acid Homeostasis, Cell Proliferation, and Tumorigenesis in Colon. Cancer Res. 2017, 77, 3352–3363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hintze, K.J.; Benninghoff, A.D.; Cho, C.E.; Ward, R.E. Modeling the Western Diet for Preclinical Investigations. Adv. Nutr. 2018, 9, 263–271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Steele, C.B.; Thomas, C.C.; Henley, S.J.; Massetti, G.M.; Galuska, D.A.; Agurs-Collins, T.; Puckett, M.; Richardson, L.C. Vital Signs: Trends in Incidence of Cancers Associated with Overweight and Obesity—United States, 2005–2014. MMWR. Morb. Mortal. Wkly. Rep. 2017, 66, 1052–1058. [Google Scholar] [CrossRef] [Green Version]
- Sung, H.; Siegel, R.L.; Torre, L.A.; Pearson-Stuttard, J.; Islami, F.; Fedewa, S.A.; Sauer, A.G.; Shuval, K.; Gapstur, S.M.; Jacobs, E.J.; et al. Global patterns in excess body weight and the associated cancer burden. CA Cancer J. Clin. 2019, 69, 88–112. [Google Scholar] [CrossRef] [PubMed]
- López-Suárez, A. Burden of cancer attributable to obesity, type 2 diabetes and associated risk factors. Metab. Clin. Exp. 2019, 92, 136–146. [Google Scholar] [CrossRef] [PubMed]
- Mirsepasi-Lauridsen, H.C.; Vallance, B.A.; Krogfelt, K.A.; Petersen, A.M. Escherichia coli Pathobionts Associated with Inflammatory Bowel Disease. Clin. Microbiol. Rev. 2019, 32, e00060-18. [Google Scholar] [CrossRef] [Green Version]
- Seyedian, S.S.; Nokhostin, F.; Malamir, M.D. A review of the diagnosis, prevention, and treatment methods of inflammatory bowel disease. J. Med. Life 2019, 12, 113–122. [Google Scholar] [CrossRef]
- Chan, S.S.M.; Chen, Y.; Casey, K.; Olen, O.; Ludvigsson, J.F.; Carbonnel, F.; Oldenburg, B.; Gunter, M.J.; Tjønneland, A.; Grip, O.; et al. Obesity is Associated with Increased Risk of Crohn’s disease, but not Ulcerative Colitis: A Pooled Analysis of Five Prospective Cohort Studies. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2022, 20, 1048–1058. [Google Scholar] [CrossRef]
- Szilagyi, A.; Smith, B.E.; Sebbag, N.; Leighton, H.; Xue, X. Changing Patterns of Relationships Between Geographic Markers and IBD: Possible Intrusion of Obesity. Crohn’s Colitis 360 2020, 2, otaa044. [Google Scholar] [CrossRef]
- Park, S.; Kang, B.; Kim, S.; Choi, S.; Suh, H.R.; Kim, E.S.; Park, J.H.; Kim, M.J.; Choe, Y.H.; Lee, Y.J.; et al. Comparison between Pediatric Crohn’s Disease and Ulcerative Colitis at Diagnosis in Korea: Results from a Multicenter, Registry-Based, Inception Cohort Study. Gut Liver 2022, 16, 921–929. [Google Scholar] [CrossRef]
- Szilagyi, A. Relationship(s) between obesity and inflammatory bowel diseases: Possible intertwined pathogenic mechanisms. Clin. J. Gastroenterol. 2020, 13, 139–152. [Google Scholar] [CrossRef] [Green Version]
- El-Dallal, M.; Stein, D.J.; Raita, Y.; Feuerstein, J.D. The impact of obesity on hospitalized patients with ulcerative colitis. Ann. Gastroenterol. 2021, 34, 196–201. [Google Scholar] [CrossRef] [PubMed]
- Milajerdi, A.; Abbasi, F.; Esmaillzadeh, A. A systematic review and meta-analysis of prospective studies on obesity and risk of inflammatory bowel disease. Nutr. Rev. 2022, 80, 479–487. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Zhang, L.; Gao, X.; Dai, C.; Huang, Y.; Wu, Y.; Zhou, W.; Cao, Q.; Jing, X.; Jiang, H.; et al. Validation of the GLIM criteria for diagnosis of malnutrition and quality of life in patients with inflammatory bowel disease: A multicenter, prospective, observational study. Clin. Nutr. 2022, 41, 1297–1306. [Google Scholar] [CrossRef]
- Carreira-Míguez, M.; Ramos-Campo, D.J.; Clemente-Suárez, V.J. Differences in Nutritional and Psychological Habits in Hypertension Patients. BioMed Res. Int. 2022, 2022, 1920996. [Google Scholar] [CrossRef] [PubMed]
- Carreira-Míguez, M.; Belinchón-deMiguel, P.P.; Clemente-Suárez, V.J. Behavioural, odontological and physical activity patterns of hypertense and control population. Physiol. Behav. 2022, 252, 113841. [Google Scholar] [CrossRef]
- Martín-Rodríguez, A.; Tornero-Aguilera, J.F.; López-Pérez, P.J.; Clemente-Suárez, V.J. Overweight and executive functions, psychological and behavioral profile of Spanish adolescents. Physiol. Behav. 2022, 254, 113901. [Google Scholar] [CrossRef]
- Clemente-Suárez, V.J.; Martín-Rodríguez, A.; Redondo-Flórez, L.; Ruisoto, P.; Navarro-Jiménez, E.; Ramos-Campo, D.J.; Tornero-Aguilera, J.F. Metabolic Health, Mitochondrial Fitness, Physical Activity, and Cancer. Cancers 2023, 15, 814. [Google Scholar] [CrossRef]
- Clemente-Suárez, V.J.; Martín-Rodríguez, A.; Yáñez-Sepúlveda, R.; Tornero-Aguilera, J.F. Mitochondrial Transfer as a Novel Therapeutic Approach in Disease Diagnosis and Treatment. Int. J. Mol. Sci. 2023, 24, 8848. [Google Scholar] [CrossRef]
- Gu, P.; Luo, J.; Kim, J.; Paul, P.; Limketkai, B.; Sauk, J.S.; Park, S.; Parekh, N.; Zheng, K.; Rudrapatna, V.; et al. Effect of Obesity on Risk of Hospitalization, Surgery, and Serious Infection in Biologic-Treated Patients With Inflammatory Bowel Diseases: A CA-IBD Cohort Study. Am. J. Gastroenterol. 2022, 117, 1639–1647. [Google Scholar] [CrossRef]
- Clemente-Suárez, V.J.; Navarro-Jiménez, E.; Jimenez, M.; Hormeño-Holgado, A.; Martinez-Gonzalez, M.B.; Benitez-Agudelo, J.C.; Perez-Palencia, N.; Laborde-Cárdenas, C.C.; Tornero-Aguilera, J.F. Impact of COVID-19 Pandemic in Public Mental Health: An Extensive Narrative Review. Sustainability 2021, 13, 3221. [Google Scholar] [CrossRef]
- Tornero-Aguilera, J.F.; Sánchez-Molina, J.; Parraca, J.A.; Morais, A.; Clemente-Suárez, V.J. Are Crohn’s Disease Patients Limited in Sport Practise? An UltraEndurance Case–Control Study Response. Int. J. Environ. Res. Public Health 2022, 19, 10007. [Google Scholar] [CrossRef] [PubMed]
- Clemente-Suárez, V.J.; Bustamante-Sanchez, Á.; Tornero-Aguilera, J.F.; Ruisoto, P.; Mielgo-Ayuso, J. Inflammation in COVID-19 and the Effects of Non-Pharmacological Interventions during the Pandemic: A Review. Int. J. Mol. Sci. 2022, 23, 15584. [Google Scholar] [CrossRef] [PubMed]
- Clemente-Suárez, V.J.; Redondo-Flórez, L.; Beltrán-Velasco, A.I.; Martín-Rodríguez, A.; Martínez-Guardado, I.; Navarro-Jiménez, E.; Laborde-Cárdenas, C.C.; Tornero-Aguilera, J.F. The Role of Adipokines in Health and Disease. Biomedicines 2023, 11, 1290. [Google Scholar] [CrossRef]
- Clemente-Suárez, V.J.; Ramírez-Goerke, M.I.; Redondo-Flórez, L.; Beltrán-Velasco, A.I.; Martín-Rodríguez, A.; Ramos-Campo, D.J.; Navarro-Jiménez, E.; Yáñez-Sepúlveda, R.; Tornero-Aguilera, J.F. The Impact of Anorexia Nervosa and the Basis for Non-Pharmacological Interventions. Nutrients 2023, 15, 2594. [Google Scholar] [CrossRef]
Study Design | Ethnic Majority Population, n | Study Participants | Type of Diet | Results | |
---|---|---|---|---|---|
Shah et al. [202] | Randomized Controlled Trial | White, n = 100 | Male and female, 63 years, 30.5 kg/m2 | Vegan | Lower C-reactive protein |
Djekic et al. [203] | Randomized Controlled Trial | White, n = 31 | Male, 67 years, 28 kg/m2 | Vegetarian | Decreased total cholesterol, LDL-C, and body weight |
Djekic et al. [204] | Randomized Controlled Trial | White, n = 31 | Male, 67 years, 28 kg/m2 | Lacto- ovo vegetarian | Increased plasma lipid profiles (TGs, PCs, O-PCs, and SMs) |
Mishra et al. [205] | Randomized Controlled Trial | White, n = 291 | Male and female, 44 years, 34.7 kg/m2 | Plant-based diet | Decreased total cholesterol and LDL-C |
Turner-McGrievy et al. [206] | Randomized Controlled Trial | Black, n = 159 | Male and female, 48 years, 25 kg/m2 | Plant-based diet | Decreased total cholesterol, LDL-C, and body weight |
Wright et al. [207] | Randomized Controlled Trial | White, n = 65 | Male and female, 46 years, 34.5 kg/m2 | Plant-based diet | Decreased cholesterol and BMI |
Jenkins et al. [208] | Randomized Controlled Trial | White, n = 39 | Male and female, 55 years, 30.5 kg/m2 | Low-carbohydratesdiet | Total cholesterol and LDL-C decreased |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Clemente-Suárez, V.J.; Beltrán-Velasco, A.I.; Redondo-Flórez, L.; Martín-Rodríguez, A.; Tornero-Aguilera, J.F. Global Impacts of Western Diet and Its Effects on Metabolism and Health: A Narrative Review. Nutrients 2023, 15, 2749. https://doi.org/10.3390/nu15122749
Clemente-Suárez VJ, Beltrán-Velasco AI, Redondo-Flórez L, Martín-Rodríguez A, Tornero-Aguilera JF. Global Impacts of Western Diet and Its Effects on Metabolism and Health: A Narrative Review. Nutrients. 2023; 15(12):2749. https://doi.org/10.3390/nu15122749
Chicago/Turabian StyleClemente-Suárez, Vicente Javier, Ana Isabel Beltrán-Velasco, Laura Redondo-Flórez, Alexandra Martín-Rodríguez, and José Francisco Tornero-Aguilera. 2023. "Global Impacts of Western Diet and Its Effects on Metabolism and Health: A Narrative Review" Nutrients 15, no. 12: 2749. https://doi.org/10.3390/nu15122749
APA StyleClemente-Suárez, V. J., Beltrán-Velasco, A. I., Redondo-Flórez, L., Martín-Rodríguez, A., & Tornero-Aguilera, J. F. (2023). Global Impacts of Western Diet and Its Effects on Metabolism and Health: A Narrative Review. Nutrients, 15(12), 2749. https://doi.org/10.3390/nu15122749