Skip to main content

Advertisement

Log in

Dissecting ultra-processed foods and drinks: Do they have a potential to impact the brain?

  • Published:
Reviews in Endocrine and Metabolic Disorders Aims and scope Submit manuscript

Abstract

Ultra-processed foods and drinks (UPF) are formulation of ingredients, mostly of exclusive industrial use, that result from a series of industrial processes. They usually have a low nutrient but high energy density, with a high content of saturated and trans fats, and added sugars. In addition, they have characteristic organoleptic properties, and usually contain sophisticated additives, including artificial sweeteners, to intensify their sensory qualities and imitate the appearance of minimally processed foods. In addition, recent research has warned about the presence of chemicals (e.g., bisphenol) and neo-formed contaminants in these products. UPF production and consumption growth have been spectacular in the last decades, being specially consumed in children and adolescents. UPF features have been associated with a range of adverse health effects such as overeating, the promotion of inflammatory and oxidative stress processes, gut dysbiosis, and metabolic dysfunction including problems in glucose regulation. The evidence that these UPF-related adverse health effects may have on the neural network implicated in eating behavior are discussed, including the potential impact on serotonergic and dopaminergic neurotransmission, brain integrity and function. We end this review by placing UPF in the context of current food environments, by suggesting that an increased exposure to these products through different channels, such as marketing, may contribute to the automatic recruitment of the brain regions associated with food consumption and choice, with a detrimental effect on inhibitory-related prefrontal cortices. While further research is essential, preliminary evidence point to UPF consumption as a potential detrimental factor for brain health and eating behavior.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

Availability of data and material

Not applicable.

Code availability

Not applicable.

Abbreviations

ADI:

Acceptable daily intake

BBB:

Blood Brain Barrier

BMI:

Body Mass Index

BPA:

Bisphenol-A

FDA:

US Food and Drug Administration

GLP-1:

Glucagon like peptide-1

KYN:

Kynurenine

LNCSs:

Low-/-non calorie sweeteners

PHO:

Partially hydrogenated vegetable oils

PYY:

Peptide tyrosine-tyrosine

SCFA:

Short-chain fatty acids

TiO2:

Titanium dioxide

TCS:

Triclosan

Trp:

Tryptophan

UPF:

Ultra-processed foods and drinks

5-HT:

Serotonin (5-hydroxytryptamine)

5-HIAA:

5-Hydroxyindoleacetic acid

References

  1. Monteiro CA, Cannon G, Levy RB, Moubarac JC, Louzada MLC, Rauber F, et al. Ultra-processed foods: What they are and how to identify them. Public Health Nutr. 2019;22:936–41.

    Article  PubMed  Google Scholar 

  2. Monteiro CA, Cannon G, Levy R, Moubarac J-C. The Food System.NOVA. The star shines bright. Public Health. 2016;7:28–38.

  3. Gupta S, Hawk T, Aggarwal A, Drewnowski A. Characterizing ultra-processed foods by energy density, nutrient density, and cost. Front Nutr. 2019;6:1–9.

    Article  Google Scholar 

  4. Steele EM, Baraldi LG, Da Costa Louzada ML, Moubarac JC, Mozaffarian D, Monteiro CA. Ultra-processed foods and added sugars in the US diet: Evidence from a nationally representative cross-sectional study. BMJ Open. 2016;6:e009892.

  5. Martínez Steele E, Khandpur N, da Costa Louzada ML, Monteiro CA. Association between dietary contribution of ultra-processed foods and urinary concentrations of phthalates and bisphenol in a nationally representative sample of the US population aged 6 years and older. PLoS ONE. 2020;15:1–21.

    Article  CAS  Google Scholar 

  6. Baraldi LG, Martinez Steele E, Canella DS, Monteiro CA. Consumption of ultra-processed foods and associated sociodemographic factors in the USA between 2007 and 2012: Evidence from a nationally representative cross-sectional study. BMJ Open. 2018;8:e020574.

  7. Moubarac JC, Batal M, Louzada ML, Martinez Steele E, Monteiro CA. Consumption of ultra-processed foods predicts diet quality in Canada. Appetite. 2017;108:512–20.

    Article  PubMed  Google Scholar 

  8. Rauber F, Louzada ML da C, Steele EM, Millett C, Monteiro CA, Levy RB. Ultra-processed food consumption and chronic non-communicable diseases-related dietary nutrient profile in the UK (2008–2014). Nutrients. 2018;10:587.

  9. Da Costa Louzada ML, Ricardo CZ, Steele EM, Levy RB, Cannon G, Monteiro CA. The share of ultra-processed foods determines the overall nutritional quality of diets in Brazil. Public Health Nutr. 2018;21:94–102.

    Article  Google Scholar 

  10. Marrón-Ponce JA, Sánchez-Pimienta TG, Da Costa Louzada ML, Batis C. Energy contribution of NOVA food groups and sociodemographic determinants of ultra-processed food consumption in the Mexican population. Public Health Nutr. 2018;21:87–93.

    Article  PubMed  Google Scholar 

  11. Cediel G, Reyes M, Da Costa Louzada ML, Martinez Steele E, Monteiro CA, Corvalán C, et al. Ultra-processed foods and added sugars in the Chilean diet (2010). Public Health Nutr. 2018;21:125–33.

    Article  PubMed  Google Scholar 

  12. Baker P, Machado P, Santos T, Sievert K, Backholer K, Hadjikakou M, et al. Ultra-processed foods and the nutrition transition: Global, regional and national trends, food systems transformations and political economy drivers. Obes Rev. 2020;21:e13126.

  13. Zhong G-C, Gu H-T, Peng Y, Wang K, Wu Y-Q-L, Hu T-Y, et al. Association of Ultra-processed Food Consumption With Cardiovascular Mortality in the US Population: Long-term Results From a Large Prospective Multicenter Study. Int J Behav Nutr Phys Act. 2021;18:21.

  14. Bawaked RA, Fernández-Barrés S, Navarrete-Muñoz EM, González-Palacios S, Guxens M, Irizar A, et al. Impact of lifestyle behaviors in early childhood on obesity and cardiometabolic risk in children: Results from the Spanish INMA birth cohort study. Pediatr Obes. 2020;15:1–15.

    Article  Google Scholar 

  15. Fiolet T, Srour B, Sellem L, Kesse-Guyot E, Allès B, Méjean C, et al. Consumption of ultra-processed foods and cancer risk: Results from NutriNet-Santé prospective cohort. BMJ. 2018;360:k322.

  16. Romaguera D, Fernández-Barrés S, Gracia-Lavedán E, Vendrell E, Azpiri M, Ruiz-Moreno E, et al. Consumption of ultra-processed foods and drinks and colorectal, breast, and prostate cancer. Clin Nutr. 2021;40:1537–45.

    Article  CAS  PubMed  Google Scholar 

  17. Narula N, Wong ECL, Dehghan M, Mente A, Rangarajan S, Lanas F, et al. Association of ultra-processed food intake with risk of inflammatory bowel disease: Prospective cohort study. BMJ. 2021;374:n1554.

  18. Schnabel L, Kesse-Guyot E, Allès B, Touvier M, Srour B, Hercberg S, et al. Association between Ultraprocessed Food Consumption and Risk of Mortality among Middle-aged Adults in France. JAMA Intern Med. 2019;179:490–8.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Sandoval-Insausti, H Blanco-Rojo R, Graciani A, López-García E, Moreno-Franco B, Laclaustra M, Donat-Vargas C, et al. Ultra-processed Food Consumption and Incident Frailty: A Prospective Cohort Study of Older Adults. J Gerontol A Biol Sci Med Sci. 2020;75:1126–1133.

  20. Elizabeth L, Machado P, Zinöcker M, Baker P, Lawrence M. Ultra-processed foods and health outcomes: A narrative review. Nutrients. 2020;12:1–36.

    Article  CAS  Google Scholar 

  21. Sadler CR, Grassby T, Hart K, Raats M, Sokolović M, Timotijevic L. Processed food classification: Conceptualisation and challenges. Trends Food Sci Technol. 2021;112:149–62.

    Article  CAS  Google Scholar 

  22. Cordain L, Eaton S, Sebastian A, Mann N, Lindeberg S, Watkins B, et al. Origins and evolution of the Western diet: health implications for the 21st century. Am J Clin Nutr. 2005;81:341–54.

    Article  CAS  PubMed  Google Scholar 

  23. Poti JM, Braga B, Qin B. Ultra-processed Food Intake and Obesity: What Really Matters for Health-Processing or Nutrient Content? Curr Obes Rep. 2017;6:420–31.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Pagliai G, Dinu M, Madarena MP, Bonaccio M, Iacoviello L, Sofi F. Consumption of ultra-processed foods and health status: A systematic review and meta-Analysis. Br J Nutr. 2021;125:308–18.

    Article  CAS  PubMed  Google Scholar 

  25. Hall KD, Ayuketah A, Brychta R, Cai H, Cassimatis T, Chen KY, et al. Ultra-Processed Diets Cause Excess Calorie Intake and Weight Gain: An Inpatient Randomized Controlled Trial of Ad Libitum Food Intake. Cell Metab. 2020;32:690.

    Article  CAS  PubMed  Google Scholar 

  26. Wang L, Martínez Steele E, Du M, Pomeranz JL, O’Connor LE, Herrick KA, et al. Trends in Consumption of Ultraprocessed Foods among US Youths Aged 2–19 Years, 1999–2018. JAMA - J Am Med Assoc. 2021;326:519–30.

    Article  Google Scholar 

  27. Fox EL, Timmer A. Children’s and adolescents’ characteristics and interactions with the food system. Glob Food Sec. 2020;27:0–5.

  28. Bhutta ZA, Guerrant RL, Nelson CA. Neurodevelopment, nutrition, and inflammation: The evolving global child health landscape. Pediatrics. 2017;139:S12–22.

    Article  PubMed  Google Scholar 

  29. Gogtay N, Giedd JN, Lusk L, Hayashi KM, Greenstein D, Vaituzis AC, et al. Dynamic mapping of human cortical development during childhood through early adulthood. Proc Natl Acad Sci U S A. 2004;101:8174–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Sydnor VJ, Larsen B, Bassett DS, Alexander-Bloch A, Fair DA, Liston C, et al. Neurodevelopment of the association cortices: Patterns, mechanisms, and implications for psychopathology. Neuron. 2021;109:1–27.

    Article  CAS  Google Scholar 

  31. Adjibade M, Julia C, Allès B, Touvier M, Lemogne C, Srour B, et al. Prospective association between ultra-processed food consumption and incident depressive symptoms in the French NutriNet-Santé cohort. BMC Med. 2019;17:78.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Gómez-Donoso C, Sánchez-Villegas A, Martínez-González MA, Gea A, Mendonça R de D, Lahortiga-Ramos F, et al. Ultra-processed food consumption and the incidence of depression in a Mediterranean cohort: the SUN Project. Eur J Nutr. 2020;59:1093–1103.

  33. Lane MM, Davis JA, Beattie S, Gómez-Donoso C, Loughman A, O’Neil A, et al. Ultraprocessed food and chronic noncommunicable diseases: A systematic review and meta-analysis of 43 observational studies. Obes Rev. 2021;22:1–19.

    Article  Google Scholar 

  34. Schulte EM, Avena NM, Gearhardt AN. Which foods may be addictive? The roles of processing, fat content, and glycemic load. PLoS ONE. 2015;10:1–18.

    Article  CAS  Google Scholar 

  35. Filgueiras AR, Pires de Almeida VB, Koch Nogueira PC, Alvares Domene SM, Eduardo da Silva C, Sesso R, et al. Exploring the consumption of ultra-processed foods and its association with food addiction in overweight children. Appetite. 2019;135:137–145.

  36. Pursey KM, Davis C, Burrows TL. Nutritional Aspects of Food Addiction. Curr Addict Reports. 2017;4:142–50.

    Article  Google Scholar 

  37. Ayton A, Ibrahim A, Dugan J, Galvin E, Wright OW. Ultra-processed foods and binge eating: A retrospective observational study. Nutrition. 2021;84:111023.

  38. Anjos LA Dos, Vieira DiADS, Siqueira BNF, Voci SM, Botelho AJ, Silva DG Da. Low adherence to traditional dietary pattern and food preferences of low-income preschool children with food neophobia. Public Health Nutr. 2021;24:2859–2866.

  39. Vedovato GM, Vilela S, Severo M, Rodrigues S, Lopes C, Oliveira A. Ultra-processed food consumption, appetitive traits and BMI in children: A prospective study. Br J Nutr. 2021;125:1427–36.

    Article  CAS  PubMed  Google Scholar 

  40. Oliveira N, Coelho GM de O, Cabral MC, Bezerra FF, Faerstein E, Canella DS. Association of body image (dis)satisfaction and perception with food consumption according to the NOVA classification: Pró-Saúde Study. Appetite. 2020;144:104464.

  41. De Deus MR, Souza Lopes AC, Pimenta AM, Gea A, Martinez-Gonzalez MA, Bes-Rastrollo M. Ultra-processed food consumption and the incidence of hypertension in a mediterranean cohort: The seguimiento universidad de navarra project. Am J Hypertens. 2017;30:358–66.

    Google Scholar 

  42. Martines RM, Machado PP, Neri DA, Levy RB, Rauber F. Association between watching TV whilst eating and children’s consumption of ultraprocessed foods in United Kingdom. Matern Child Nutr. 2019;15:1–10.

    Article  Google Scholar 

  43. Costa C dos S, Flores TR, Wendt A, Neves RG, Assunção MCF, Santos IS. Sedentary behavior and consumption of ultra-processed foods by Brazilian adolescents: Brazilian National School Health Survey (PeNSE), 2015. Cad Saude Publica. 2018;34:e00021017.

  44. Werneck AO, Hoare E, Silva DR. Do TV-viewing and frequency of ultra-processed food consumption share mediators in relation to adolescent anxiety-induced sleep disturbance? Public Health Nutr. 2021;27:1–7.

    Google Scholar 

  45. Calder PC, Ahluwalia N, Brouns F, Buetler T, Clement K, Cunningham K, et al. Dietary factors and low-grade inflammation in relation to overweight and obesity. Br J Nutr. 2011;106:S5-78.

    Article  CAS  PubMed  Google Scholar 

  46. Guillemot-Legris O, Muccioli GG. Obesity-Induced Neuroinflammation: Beyond the Hypothalamus. Trends Neurosci. 2017;40:237–53.

    Article  CAS  PubMed  Google Scholar 

  47. Dagbasi A, Lett AM, Murphy K, Frost G. Understanding the interplay between food structure, intestinal bacterial fermentation and appetite control. Proc Nutr Soc. 2020;79:514–30.

    Article  CAS  Google Scholar 

  48. De Graaf C, Kok FJ. Slow food, fast food and the control of food intake. Nat Rev Endocrinol. 2010;6:290–3.

    Article  PubMed  Google Scholar 

  49. Lasschuijt MP, de Graaf K, Mars M. Effects of oro-sensory exposure on satiation and underlying neurophysiological mechanisms—what do we know so far? Nutrients. 2021;13:1391.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Kokkinos A, Le Roux CW, Alexiadou K, Tentolouris N, Vincent RP, Kyriaki D, et al. Eating slowly increases the postprandial response of the anorexigenic gut hormones, peptide YY and glucagon-like peptide-1. J Clin Endocrinol Metab. 2010;95:333–7.

    Article  CAS  PubMed  Google Scholar 

  51. Krop EM, Hetherington MM, Nekitsing C, Miquel S, Postelnicu L, Sarkar A. Influence of oral processing on appetite and food intake – A systematic review and meta-analysis. Appetite. 2018;125:253–69.

    Article  PubMed  Google Scholar 

  52. Viskaal-van Dongen M, Kok FJ, de Graaf C. Eating rate of commonly consumed foods promotes food and energy intake. Appetite. 2011;56:25–31.

    Article  PubMed  Google Scholar 

  53. Bolhuis DP, Forde CG, Cheng Y, Xu H, Martin N, De Graaf C. Slow food: Sustained impact of harder foods on the reduction in energy intake over the course of the day. PLoS ONE. 2014;9:1–7.

    Article  CAS  Google Scholar 

  54. Zhu Y, Hsu WH, Hollis JH. The effect of food form on satiety. Int J Food Sci Nutr. 2013;64:385–91.

    Article  CAS  PubMed  Google Scholar 

  55. Chambers L, McCrickerd K, Yeomans MR. Optimising foods for satiety. Trends Food Sci Technol. 2015;41:149–60.

    Article  CAS  Google Scholar 

  56. May CE, Dus M. Confection Confusion: Interplay Between Diet, Taste, and Nutrition. Trends Endocrinol Metab. 2021;32:95–105.

    Article  CAS  PubMed  Google Scholar 

  57. Liu D, Archer N, Duesing K, Hannan G, Keast R. Mechanism of fat taste perception: Association with diet and obesity. Prog Lipid Res. 2016;63:41–9.

    Article  CAS  PubMed  Google Scholar 

  58. Archer N, Shaw J, Cochet-Broch M, Bunch R, Poelman A, Barendse W, et al. Obesity is associated with altered gene expression in human tastebuds. Int J Obes. 2019;43:1475–84.

    Article  CAS  Google Scholar 

  59. Ramos-Lopez O, Arpón A, Riezu-Boj JI, Milagro FI, Mansego ML, Martinez JA, et al. DNA methylation patterns at sweet taste transducing genes are associated with BMI and carbohydrate intake in an adult population. Appetite. 2018;120:230–9.

    Article  CAS  PubMed  Google Scholar 

  60. FDA. Food and drug administration, department of health and human services. Animal drugs, feeds, and related products. Part 570 food additives. https://www.AccessdataFda.Gov/Scripts/Cdrh/Cfdocs/CfCFR/CFRSearchCfm?CFRPart=570&ShowFR=1 Accessed 20 Sept 2021.

  61. Medina-Reyes EI, Rodríguez-Ibarra C, Déciga-Alcaraz A, Díaz-Urbina D, Chirino YI, Pedraza-Chaverri J. Food additives containing nanoparticles induce gastrotoxicity, hepatotoxicity and alterations in animal behavior: The unknown role of oxidative stress. Food Chem Toxicol. 2020;146:111814.

  62. Laster J, Frame LA. Beyond the Calories—Is the Problem in the Processing? Curr Treat Options Gastroenterol. 2019;17:577–86.

    Article  PubMed  Google Scholar 

  63. Edalati S, Bagherzadeh F, Asghari Jafarabadi M, Ebrahimi-Mamaghani M. Higher ultra-processed food intake is associated with higher DNA damage in healthy adolescents. Br J Nutr. 2021;125:568–76.

    Article  CAS  PubMed  Google Scholar 

  64. Alonso-Pedrero L, Ojeda-Rodríguez A, Martínez-González M, Zalba G, Bes-Rastrollo M, Marti A. Association between diet quality indexes and the risk of short telomeres in an elderly population of the SUN project. Am J Clin Nutr. 2020;111:1259–66.

    Article  PubMed  Google Scholar 

  65. Freitas-Simoes TM, Ros E, Sala-Vila A. Telomere length as a biomarker of accelerated aging: Is it influenced by dietary intake? Curr Opin Clin Nutr Metab Care. 2018;21:430–6.

    Article  CAS  PubMed  Google Scholar 

  66. García-Sánchez A, Gámez-Nava JI, Díaz-De La Cruz EN, Cardona-Muñoz EG, Becerra-Alvarado IN, Aceves-Aceves JA, et al. The effect of visceral abdominal fat volume on oxidative stress and proinflammatory cytokines in subjects with normal weight, overweight and obesity. Diabetes, Metab Syndr Obes. 2020;13:1077–1087.

  67. Cinkajzlová A, Mráz M, Haluzík M. Adipose tissue immune cells in obesity, type 2 diabetes mellitus and cardiovascular diseases. J Endocrinol. 2021;252:R1–22.

    Article  PubMed  Google Scholar 

  68. Bian X, Chi L, Gao B, Tu P, Ru H, Lu K. Gut microbiome response to sucralose and its potential role in inducing liver inflammation in mice. Front Physiol. 2017;8:1–13.

    Article  Google Scholar 

  69. Vos MB, Lavine JE. Dietary fructose in nonalcoholic fatty liver disease. Hepatology. 2013;57:2525–31.

    Article  CAS  PubMed  Google Scholar 

  70. Shil A, Chichger H. Artificial sweeteners negatively regulate pathogenic characteristics of two model gut bacteria, E. Coli and E. faecalis. Int J Mol Sci. 2021;22:5228.

  71. Lee JA, Kim MK, Song JH, Jo MR, Yu J, Kim KM, et al. Biokinetics of food additive silica nanoparticles and their interactions with food components. Colloids Surfaces B Biointerfaces. 2017;150:384–92.

    Article  CAS  PubMed  Google Scholar 

  72. Bancil AS, Sandall AM, Rossi M, Chassaing B, Lindsay JO, Whelan K. Food Additive Emulsifiers and Their Impact on Gut Microbiome, Permeability, and Inflammation: Mechanistic Insights in Inflammatory Bowel Disease. J Crohns Colitis. 2021 Jun 22;15(6):1068–1079.

  73. Cao Y, Liu H, Qin N, Ren X, Zhu B, Xia X. Impact of food additives on the composition and function of gut microbiota: A review. Trends Food Sci Technol. 2020;99(March):295-310.

  74. Laudisi F, Stolfi C, Monteleone G. Impact of Food Additives on Gut Homeostasis. Nutrients. 2019 Oct1;11(10):2334.

  75. Nettleton JE, Reimer RA, Shearer J. Reshaping the gut microbiota: Impact of low calorie sweeteners and the link to insulin resistance? Physiol Behav. 2016;164:488–93.

    Article  CAS  PubMed  Google Scholar 

  76. Plaza-Diaz J, Pastor-Villaescusa B, Rueda-Robles A, Abadia-Molina F, Ruiz-Ojeda FJ. Plausible biological interactions of low- and non-calorie sweeteners with the intestinal microbiota: An update of recent studies. Nutrients. 2020;12:1–15.

    Article  Google Scholar 

  77. FDA. Additional Information about High-Intensity Sweeteners Permitted for Use in Food in the United States. https://www.Fda.Gov/Food/Food-Additives-Petitions/Additional-Information-about-High-Intensity-Sweeteners-Permitted-Use-Food-United-States.

  78. Lohner S, Toews I, Meerpohl JJ. Health outcomes of non-nutritive sweeteners: Analysis of the research landscape. Nutr J. 2017;16:1–21.

    Article  Google Scholar 

  79. Pepino MY. Physiology & behavior metabolic effects of non-nutritive sweeteners. Physiol Behav. 2015;152:450–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Magnuson BA, Carakostas MC, Moore NH, Poulos SP, Renwick AG. Biological fate of low-calorie sweeteners. Nutr Rev. 2016;74:670–89.

    Article  PubMed  Google Scholar 

  81. Smeets PAM, Erkner A, De Graaf C. Cephalic phase responses and appetite. Nutr Rev. 2010;68:643–55.

    Article  PubMed  Google Scholar 

  82. Silva YP, Bernardi A, Frozza RL. The Role of Short-Chain Fatty Acids From Gut Microbiota in Gut-Brain Communication. Front Endocrinol (Lausanne). 2020;11:1–14.

    Article  Google Scholar 

  83. Dalile B, Van Oudenhove L, Vervliet B, Verbeke K. The role of short-chain fatty acids in microbiota–gut–brain communication. Nat Rev Gastroenterol Hepatol. 2019;16:461–78.

    Article  PubMed  Google Scholar 

  84. Suez J, Korem T, Zeevi D, Zilberman-Schapira G, Thaiss CA, Maza O, et al. Artificial sweeteners induce glucose intolerance by altering the gut microbiota. Nature. 2014;514:181–6.

    Article  CAS  PubMed  Google Scholar 

  85. Alfhili MA, Lee MH. Triclosan: An update on biochemical and molecular mechanisms. Oxid Med Cell Longev. 2019;2019:1607304.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  86. Yang H, Wang W, Romano KA, Gu M, Katherine Z, Kim D, et al. A common antimicrobial additive increases colonic inflammation and colitis-associated colon tumorigenesis in mice. Sci Transl Med. 2018;10:eaan4116.

  87. Calafat AM, Ye X, Wong LY, Reidy JA, Needham LL. Urinary concentrations of triclosan in the U.S. population: 2003–2004. Environ Health Perspect. 2008;116:303–307.

  88. Wang DD, Hu FB. Dietary Fat and Risk of Cardiovascular Disease: Recent Controversies and Advances. Annu Rev Nutr. 2017;37:423–46.

    Article  PubMed  CAS  Google Scholar 

  89. Okamura T, Hashimoto Y, Majima S, Senmaru T, Ushigome E, Nakanishi N, et al. Trans Fatty Acid Intake Induces Intestinal Inflammation and Impaired Glucose Tolerance. Front Immunol. 2021;12:1–14.

    Article  Google Scholar 

  90. Shoshtari-Yeganeh B, Zarean M, Mansourian M, Riahi R, Poursafa P, Teiri H, et al. Systematic review and meta-analysis on the association between phthalates exposure and insulin resistance. Environ Sci Pollut Res. 2019;26:9435–42.

    Article  CAS  Google Scholar 

  91. Pacyga DC, Sathyanarayana S, Strakovsky RS. Dietary Predictors of Phthalate and Bisphenol Exposures in Pregnant Women. Adv Nutr. 2019;10:803–15.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Cao XL. Phthalate Esters in Foods: Sources, Occurrence, and Analytical Methods. Compr Rev Food Sci Food Saf. 2010;9:21–43.

    Article  CAS  PubMed  Google Scholar 

  93. Calafat AM, Longnecker MP, Koch HM, Swan SH, Hauser R, Goldman LR, et al. Optimal Exposure Biomarkers for Nonpersistent Chemicals in Environmental Epidemiology. Environ Health Perspect. 2015;123:166–8.

    Article  CAS  Google Scholar 

  94. Gore AC, Chappell VA, Fenton SE, Flaws JA, Nadal A, Prins GS, et al. EDC-2: The Endocrine Society’s Second Scientific Statement on Endocrine-Disrupting Chemicals. Endocr Rev. 2015;36:1–150.

    Article  Google Scholar 

  95. Charisiadis P, Andrianou XD, Van Der Meer TP, Den Dunnen WFA, Swaab DF, Wolffenbuttel BHR, et al. Possible obesogenic effects of bisphenols accumulation in the human brain. Sci Rep. 2018;8:1–10.

    Article  CAS  Google Scholar 

  96. Radke EG, Galizia A, Thayer KA, Cooper GS. Phthalate exposure and metabolic effects: a systematic review of the human epidemiological evidence. Environ Int. 2019;132:1–17.

    Article  CAS  Google Scholar 

  97. Hetherington AW, Ranson SW. Hypothalamic lesions and adiposity in the rat. Anat Rec. 1940;78:149–72.

    Article  Google Scholar 

  98. Berthoud H-R. Homeostatic and Non-homeostatic Pathways Involved in the Control of Food Intake and Energy Balance. Obesity. 2006;14(197S):200S.

    Google Scholar 

  99. Kullmann S, Heni M, Linder K, Zipfel S, Häring HU, Veit R, et al. Resting-state functional connectivity of the human hypothalamus. Hum Brain Mapp. 2014;35:6088–96.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Dagher A, Neseliler S, Han JE. Appetite as motivated choice: Hormonal and environmental influences. Decis Neurosci An Integr Perspect. 2017:397–409.

  101. Avery JA, Liu AG, Ingeholm JE, Riddell CD, Gotts SJ, Martin A. Taste quality representation in the human brain. BioRxiv. 2019;40:1042–52.

    Google Scholar 

  102. Rolls ET. Taste, olfactory and food texture reward processing in the brain and the control of appetite. Proc Nutr Soc. 2012;71:488–501.

    Article  PubMed  Google Scholar 

  103. Li J, An R, Zhang Y, Li X, Wang S. Correlations of macronutrient-induced functional magnetic resonance imaging signal changes in human brain and gut hormone responses. Am J Clin Nutr. 2012;96:275–82.

    Article  CAS  PubMed  Google Scholar 

  104. Kyle Simmons W, Rapuano KM, Kallman SJ, Ingeholm JE, Miller B, Gotts SJ, et al. Category-specific integration of homeostatic signals in caudal but not rostral human insula. Nat Publ Gr. 2013;16:1551–2.

    Google Scholar 

  105. Nitschke JB, Dixon GE, Sarinopoulos I, Short SJ, Cohen JD, Smith EE, et al. Altering expectancy dampens neural response to aversive taste in primary taste cortex. Nat Neurosci. 2006;9:435–42.

    Article  CAS  PubMed  Google Scholar 

  106. Wurtman J, Wurtman R. The Trajectory from Mood to Obesity. Curr Obes Rep. 2018;7:1–5.

    Article  PubMed  Google Scholar 

  107. Berthoud H, Münzberg H, Morrison C. Blaming the brain for obesity: Integration of hedonic and homeostatic mechanisms. Gastroenterology. 2017;152:1728–38.

    Article  PubMed  Google Scholar 

  108. Dallman MF. Stress-induced obesity and the emotional nervous system. Trends Endocrinol Metab. 2010;21:159–65.

    Article  CAS  PubMed  Google Scholar 

  109. Contreras-Rodríguez O, Martín-Pérez C, Vilar-López R, Verdejo-Garcia A. Ventral and Dorsal Striatum Networks in Obesity: Link to Food Craving and Weight Gain. Biol Psychiatry. 2017;81:789–96.

    Article  PubMed  Google Scholar 

  110. Lowe C, Reichelt A, HAll P. The Prefrontal Cortex and Obesity: A Health Neuroscience Perspective. Trends Cogn Sci. 2019;23:349–361.

  111. Jansen JM, Daams JG, Koeter MWJ, Veltman DJ, Van Den Brink W, Goudriaan AE. Effects of non-invasive neurostimulation on craving: A meta-analysis. Neurosci Biobehav Rev. 2013;37:2472–80.

    Article  PubMed  Google Scholar 

  112. Jensen CD, Kirwan CB. Functional brain response to food images in successful adolescent weight losers compared with normal-weight and overweight controls. Obesity. 2015;23:630–6.

    Article  PubMed  Google Scholar 

  113. Volkow ND, Wise RA, Baler R. The dopamine motive system: implications for drug and food addiction. Nat Rev Neurosci. 2017;18:741–52.

    Article  CAS  PubMed  Google Scholar 

  114. Strasser B, Gostner JM, Fuchs D. Mood, food, and cognition: Role of tryptophan and serotonin. Curr Opin Clin Nutr Metab Care. 2016;19:55–61.

    Article  CAS  PubMed  Google Scholar 

  115. Parsons LH, Hurd YL. Endocannabinoid signalling in reward and addiction. Nat Rev Neurosci. 2015;16:579–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Lovelace MD, Varney B, Sundaram G, Lennon MJ, Lim CK, Jacobs K, et al. Recent evidence for an expanded role of the kynurenine pathway of tryptophan metabolism in neurological diseases. Neuropharmacology. 2017;112:373–88.

    Article  CAS  PubMed  Google Scholar 

  117. O’Farrell K, Harkin A. Stress-related regulation of the kynurenine pathway: Relevance to neuropsychiatric and degenerative disorders. Neuropharmacology. 2017;112:307–23.

    Article  PubMed  CAS  Google Scholar 

  118. Maes M, Yirmyia R, Noraberg J, Brene S, Hibbeln J, Perini G, et al. The inflammatory & neurodegenerative (I&ND) hypothesis of depression: Leads for future research and new drug developments in depression. Metab Brain Dis. 2009;24:27–53.

    Article  CAS  PubMed  Google Scholar 

  119. Luppino F, de Wit L, Bouvy P, Stijnen T, Cuijpers P, Penninx BW-J-H, et al. Overweight, Obesity, and Depression. A Systematic Review and Meta-analysis of Longitudinal Studies. Arch Gen Psychiatry. 2010;67:220–229.

  120. Castro B, Sánchez P, Torres JM, Ortega E. Bisphenol A, bisphenol F and bisphenol S affect differently 5α-reductase expression and dopamine-serotonin systems in the prefrontal cortex of juvenile female rats. Environ Res. 2015;142:281–7.

    Article  CAS  PubMed  Google Scholar 

  121. Matsuda S, Matsuzawa D, Ishii D, Tomizawa H, Sajiki J, Shimizu E. Perinatal exposure to bisphenol A enhances contextual fear memory and affects the serotoninergic system in juvenile female mice. Horm Behav. 2013;63:709–16.

    Article  CAS  PubMed  Google Scholar 

  122. Matsuda S, Matsuzawa D, Ishii D, Tomizawa H, Sutoh C, Nakazawa K, et al. Effects of perinatal exposure to low dose of bisphenol A on anxiety like behavior and dopamine metabolites in brain. Prog Neuro-Psychopharmacology Biol Psychiatry. 2012;39:273–9.

    Article  CAS  Google Scholar 

  123. Naderi M, Kwong RWM. A comprehensive review of the neurobehavioral effects of bisphenol S and the mechanisms of action: New insights from in vitro and in vivo models. Environ Int. 2020;145:106078.

  124. Matsumoto M, Kibe R, Ooga T, Aiba Y, Sawaki E, Koga Y, et al. Cerebral low-molecular metabolites influenced by intestinal microbiota: A pilot study. Front Syst Neurosci. 2013;7:1–19.

    Article  Google Scholar 

  125. Desbonnet L, Garrett L, Clarke G, Bienenstock J, Dinan TG. The probiotic Bifidobacteria infantis: An assessment of potential antidepressant properties in the rat. J Psychiatr Res. 2008;43:164–74.

    Article  PubMed  Google Scholar 

  126. Heidari Z, Mohammadipour A, Haeri P, Ebrahimzadeh-Bideskan A. The effect of titanium dioxide nanoparticles on mice midbrain substantia nigra. Iran J Basic Med Sci. 2019;22:745–51.

    PubMed  PubMed Central  Google Scholar 

  127. Hu R, Gong X, Duan Y, Li N, Che Y, Cui Y, et al. Neurotoxicological effects and the impairment of spatial recognition memory in mice caused by exposure to TiO2 nanoparticles. Biomaterials. 2010;31:8043–50.

    Article  CAS  PubMed  Google Scholar 

  128. Button EB, Mitchell AS, Domingos MM, Chung JHJ, Bradley RM, Hashemi A, et al. Microglial cell activation increases saturated and decreases monounsaturated fatty acid content, but both lipid species are proinflammatory. Lipids. 2014;49:305–16.

    Article  CAS  PubMed  Google Scholar 

  129. Gupta S, Knight AG, Gupta S, Keller JN, Bruce-Keller AJ. Saturated long-chain fatty acids activate inflammatory signaling in astrocytes. J Neurochem. 2012;120:1060–71.

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Ze Y, Sheng L, Zhao X, Hong J, Ze X, Yu X, et al. TiO2 nanoparticles induced hippocampal neuroinflammation in mice. PLoS ONE. 2014;9:1–8.

    Article  CAS  Google Scholar 

  131. Hu R, Zheng L, Zhang T, Gao G, Cui Y, Cheng Z, et al. Molecular mechanism of hippocampal apoptosis of mice following exposure to titanium dioxide nanoparticles. J Hazard Mater. 2011;191:32–40.

    Article  CAS  PubMed  Google Scholar 

  132. Van Der Zande M, Vandebriel RJ, Van Doren E, Kramer E, Herrera Rivera Z, Serrano-Rojero CS, et al. Distribution, elimination, and toxicity of silver nanoparticles and silver ions in rats after 28-day oral exposure. ACS Nano. 2012;6:7427–42.

    Article  PubMed  CAS  Google Scholar 

  133. Węsierska M, Dziendzikowska K, Gromadzka-Ostrowska J, Dudek J, Polkowska-Motrenko H, Audinot JN, et al. Silver ions are responsible for memory impairment induced by oral administration of silver nanoparticles. Toxicol Lett. 2018;290:133–44.

    Article  PubMed  CAS  Google Scholar 

  134. Waegeneers N, De Vos S, Verleysen E, Ruttens A, Mast J. Estimation of the uncertainties related to the measurement of the size and quantities of individual silver nanoparticles in confectionery. Materials (Basel). 2019;12:2677.

    Article  CAS  Google Scholar 

  135. Shelby M. NTP-CERHR monograph on the potential human reproductive and developmental effects of bisphenol A. NTP CERHR MON. 2008;v:1‐64 passim.

  136. Patisaul HB. Achieving CLARITY on bisphenol A, brain and behaviour. J Neuroendocrinol. 2020;32:1–12.

    Article  CAS  Google Scholar 

  137. Völkel W, Colnot T, Csanády GA, Filser JG, Dekant W. Metabolism and kinetics of bisphenol a in humans at low doses following oral administration. Chem Res Toxicol. 2002;15:1281–7.

    Article  PubMed  CAS  Google Scholar 

  138. Stahlhut RW, Welshons WV, Swan SH. Bisphenol A data in NHANES suggest longer than expected half-life, substantial nonfood exposure, or both. Environ Health Perspect. 2009;117:784–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Pase CS, Metz VG, Roversi K, Roversi K, Vey LT, Dias VT, et al. Trans fat intake during pregnancy or lactation increases anxiety-like behavior and alters proinflammatory cytokines and glucocorticoid receptor levels in the hippocampus of adult offspring. Brain Res Bull. 2021;166:110–7.

    Article  CAS  PubMed  Google Scholar 

  140. Trevizol F, Roversi K, Dias VT, Roversi K, Pase CS, Barcelos RCS, et al. Influence of lifelong dietary fats on the brain fatty acids and amphetamine-induced behavioral responses in adult rat. Prog Neuro-Psychopharmacology Biol Psychiatry. 2013;45:215–22.

    Article  CAS  Google Scholar 

  141. Trevizol F, Roversi KR, Dias VT, Roversi K, Barcelos RCS, Kuhn FT, et al. Cross-generational trans fat intake facilitates mania-like behavior: Oxidative and molecular markers in brain cortex. Neuroscience. 2015;286:353–63.

    Article  CAS  PubMed  Google Scholar 

  142. Morris M, Tangney C. Dietary fat composition and dementia risk. Neurobiol Aging. 2014;35:S59–64.

    Article  CAS  PubMed  Google Scholar 

  143. Ginter E, Simko V. New data on harmful effects of trans-fatty acids. Bratisl Med J. 2016;117:251–3.

    Article  CAS  Google Scholar 

  144. Tang W, Zhu H, Feng Y, Guo R, Wan D. The impact of gut microbiota disorders on the blood–brain barrier. Infect Drug Resist. 2020;13:3351–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Burger KS. Frontostriatal and behavioral adaptations to daily sugar-sweetened beverage intake: A randomized controlled trial. Am J Clin Nutr. 2017;105:555–63.

    Article  PubMed  PubMed Central  Google Scholar 

  146. Burger KS, Stice E. Frequent ice cream consumption is associated with reduced striatal response to receipt of an ice cream-based milkshake. Am J Clin Nutr. 2012;95:810–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Johnson PM, Kenny PJ. Addiction-like reward dysfunction and compulsive eating in obese rats: Role for dopamine D2 receptors. Nat Neurosci. 2010;13:635–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Yunker AG, Patel R, Page KA. Effects of Non-nutritive Sweeteners on Sweet Taste Processing and Neuroendocrine Regulation of Eating Behavior. Curr Nutr Rep. 2020;9:278–89.

    Article  CAS  PubMed  Google Scholar 

  149. Yeung AWK, Wong NSM. How does our brain process sugars and non-nutritive sweeteners differently: A systematic review on functional magnetic resonance imaging studies. Nutrients. 2020;12:1–14.

    Article  CAS  Google Scholar 

  150. Smeets PAM, De Graaf C, Stafleu A, Van Osch MJP, Van Der Grond J. Functional magnetic resonance imaging of human hypothalamic responses to sweet taste and calories. Am J Clin Nutr. 2005;82:1011–6.

    Article  CAS  PubMed  Google Scholar 

  151. van Opstal AM, Kaal I, van den Berg-Huysmans AA, Hoeksma M, Blonk C, Pijl H, et al. Dietary sugars and non-caloric sweeteners elicit different homeostatic and hedonic responses in the brain. Nutrition. 2019;60:80–6.

    Article  PubMed  CAS  Google Scholar 

  152. Van Opstal AM, Hafkemeijer A, van den Berg-Huysmans AA, Hoeksma M, Mulder TPJ, Pijl H, et al. Brain activity and connectivity changes in response to nutritive natural sugars, non-nutritive natural sugar replacements and artificial sweeteners. Nutr Neurosci. 2021;24:395–405.

    Article  PubMed  Google Scholar 

  153. Rudenga K, Small D. Amygdala response to sucrose consumption is inversely related to artificial sweetener use. Appetite. 2012;58:504–7.

    Article  CAS  PubMed  Google Scholar 

  154. Crézé C, Candal L, Cros J, Knebel JF, Seyssel K, Stefanoni N, et al. The impact of caloric and non-caloric sweeteners on food intake and brain responses to food: A randomized crossover controlled trial in healthy humans. Nutrients. 2018;10:615.

    Article  PubMed Central  CAS  Google Scholar 

  155. Crézé C, Notter-Bielser ML, Knebel JF, Campos V, Tappy L, Murray M, et al. The impact of replacing sugar- by artificially-sweetened beverages on brain and behavioral responses to food viewing – An exploratory study. Appetite. 2018;123:160–8.

    Article  PubMed  Google Scholar 

  156. Folkvord F, Hermans RCJ. Food Marketing in an Obesogenic Environment: a Narrative Overview of the Potential of Healthy Food Promotion to Children and Adults. Curr Addict Reports. 2020;7:431–6.

    Article  Google Scholar 

  157. Motoki K, Suzuki S. Extrinsic Factors Underlying Food Valuation in the Human Brain. Front Behav Neurosci. 2020;14:1–7.

    Article  Google Scholar 

  158. Cairns G, Angus K, Hastings G, Caraher M. Systematic reviews of the evidence on the nature, extent and effects of food marketing to children. A retrospective summary Appetite. 2013;62:209–15.

    PubMed  Google Scholar 

  159. Story M, French S. International Journal of Behavioral Food Advertising and Marketing Directed at Children and Adolescents in the US. Int J Behav Nutr Phys Act. 2004;17:1–17.

    Google Scholar 

  160. Khonje MG, Ecker O, Qaim M. Effects of modern food retailers on adult and child diets and nutrition. Nutrients. 2020;12:1–17.

    Article  Google Scholar 

  161. Kelly B, Vandevijvere S, Ng SH, Adams J, Allemandi L, Bahena-Espina L, et al. Global benchmarking of children’s exposure to television advertising of unhealthy foods and beverages across 22 countries. Obes Rev. 2019;20:116–28.

    Article  PubMed  PubMed Central  Google Scholar 

  162. Spence C, Okajima K, Cheok AD, Petit O, Michel C. Eating with our eyes: From visual hunger to digital satiation. Brain Cogn. 2016;110:53–63.

    Article  PubMed  Google Scholar 

  163. Pelchat ML, Johnson A, Chan R, Valdez J, Ragland JD. Images of desire: Food-craving activation during fMRI. Neuroimage. 2004;23:1486–93.

    Article  PubMed  Google Scholar 

  164. Simmons WK, Martin A, Barsalou LW. Pictures of appetizing foods activate gustatory cortices for taste and reward. Cereb Cortex. 2005;15:1602–8.

    Article  PubMed  Google Scholar 

  165. Burger KS, Stice E. Neural responsivity during soft drink intake, anticipation, and advertisement exposure in habitually consuming youth. Obes (Silver Spring). 2014;22:441–50.

    Article  CAS  Google Scholar 

  166. Bruce AS, Lepping RJ, Bruce JM, Cherry JBC, Martin LE, Davis AM, et al. Brain responses to food logos in obese and healthy weight children. J Pediatr. 2013;162:759-764.e2.

    Article  PubMed  Google Scholar 

  167. Bruce AS, Bruce JM, Black WR, Lepping RJ, Henry JM, Bradley J, et al. Branding and a child’s brain: an fMRI study of neural responses to logos. Soc Cogn Affect Neurosci. 2014;9:118–22.

    Article  PubMed  Google Scholar 

  168. Masterson TD, Stein WM, Beidler E, Bermudez M, English LK, Keller KL. Brain response to food brands correlates with increased intake from branded meals in children: an fMRI study. Brain Imaging Behav. 2019;13:1035–48.

    Article  PubMed  PubMed Central  Google Scholar 

  169. Gearhardt AN, Yokum S, Harris JL, Epstein LH, Lumeng JC. Neural response to fast food commercials in adolescents predicts intake. Am J Clin Nutr. 2020;111:493–502.

    Article  PubMed  PubMed Central  Google Scholar 

  170. Gearhardt AN, Yokum S, Stice E, Harris JL, Brownell KD. Relation of obesity to neural activation in response to food commercials. Soc Cogn Affect Neurosci. 2014;9:932–8.

    Article  PubMed  Google Scholar 

  171. McClure SM, Li J, Tomlin D, Cypert KS, Montague LM, Montague PR. Neural correlates of behavioral preference for culturally familiar drinks. Neuron. 2004;44:379–87.

    Article  CAS  PubMed  Google Scholar 

  172. Stice E, Burger KS, Yokum S. Reward Region Responsivity Predicts Future Weight Gain and Moderating Effects of the TaqIA Allele. J Neurosci. 2015;35:10316–3024.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Hall PA, Lowe C, Vincent C. Executive control resources and snack food consumption in the presence of restraining versus facilitating cues. J Behav Med. 2014;37:587–94.

    Article  PubMed  Google Scholar 

  174. Jensen CD, Duraccio KM, Carbine KA, Barnett KA, Kirwan CB. Motivational impact of palatable food correlates with functional brain responses to food images in adolescents. J Pediatr Psychol. 2017;42:578–87.

    PubMed  Google Scholar 

  175. Lawrence NS, Hinton EC, Parkinson JA, Lawrence AD. Nucleus accumbens response to food cues predicts subsequent snack consumption in women and increased body mass index in those with reduced self-control. Neuroimage. 2012;63:415–22.

    Article  PubMed  Google Scholar 

  176. Liang P, Jiang J, Ding Q, Tang X, Roy S. Memory load influences taste sensitivities. Front Psychol. 2018;9:1–7.

    Article  Google Scholar 

  177. Hansen H, Melbye EL. Negative Information, Cognitive Load, and Taste Perceptions. J Food Prod Mark. 2020;26:185–96.

    Article  Google Scholar 

  178. Machín L, Antúnez L, Curutchet MR, Ares G. The heuristics that guide healthiness perception of ultra-processed foods: A qualitative exploration. Public Health Nutr. 2020;23:2932–40.

    Article  PubMed  Google Scholar 

  179. Grabenhorst F, Rolls ET, Bilderbeck A. How cognition modulates affective responses to taste and flavor: Top-down influences on the orbitofrontal and pregenual cingulate cortices. Cereb Cortex. 2008;18:1549–59.

    Article  PubMed  Google Scholar 

  180. Patton GC, Sawyer SM, Santelli JS, Ross DA, Afifi R, Allen NB, et al. Our future: a Lancet commission on adolescent health and wellbeing. Lancet. 2016;387:2423–78.

    Article  PubMed  PubMed Central  Google Scholar 

  181. Tymula A, Rosenberg Belmaker LA, Roy AK, Ruderman L, Manson K, Glimcher PW, et al. Adolescents’ risk-taking behavior is driven by tolerance to ambiguity. Proc Natl Acad Sci U S A. 2012;109:17135–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This study has received support from the Intramural Translational Projects (2021) from the CIBERSAM granted to O Contreras-Rodriguez, and the project PID2020-119391GB-I00 granted M Solanas and RM Escorihuela.O Contreras-Rodriguez is funded by a “Miguel Servet” contract (CP20/00165) from the Health Institute Carlos III (ISCIII), Spain.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Oren Contreras-Rodriguez.

Ethics declarations

Conflicts of interest/Competing interests

The authors declare no conflict of interest.

Research involving human participants and/or animals

Informed consent no applicable. This work focused on previously published studies and no own new studies were undertook.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Contreras-Rodriguez, O., Solanas, M. & Escorihuela, R.M. Dissecting ultra-processed foods and drinks: Do they have a potential to impact the brain?. Rev Endocr Metab Disord 23, 697–717 (2022). https://doi.org/10.1007/s11154-022-09711-2

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11154-022-09711-2

Keywords

Navigation