1,044
Views
0
CrossRef citations to date
0
Altmetric
Review

From gut microbiota to brain: implications on binge eating disorders

&
Article: 2357177 | Received 30 Dec 2023, Accepted 15 May 2024, Published online: 23 May 2024

References

  • Silén Y, Keski-Rahkonen A. Worldwide prevalence of DSM-5 eating disorders among young people. Curr Opin Psychiatry. 2022;35(6):362–18. doi:10.1097/YCO.0000000000000818.
  • van Hoeken D, Hoek HW. Review of the burden of eating disorders: mortality, disability, costs, quality of life, and family burden. Curr Opin Psychiatry. 2020;33(6):521–527. doi:10.1097/YCO.0000000000000641.
  • American Psychiatric Association. Diagnostic and statistical manual of mental disorders. 5th ed. Washington, DC: American Psychiatric Publishing; 2013.
  • World Health Organization. ICD-11: International classification of diseases 11th revision. ICD; 2019. https://icd.who.int/.
  • Udo T, Grilo CM. Psychiatric and medical correlates of DSM-5 eating disorders in a nationally representative sample of adults in the United States. Int J Eat Disord. 2019;52(1):42–50. doi:10.1002/eat.23004.
  • Kessler RC, Berglund PA, Chiu WT, Deitz AC, Hudson JI, Shahly V, Aguilar-Gaxiola S, Alonso J, Angermeyer MC, Benjet C. et al. The prevalence and correlates of binge eating disorder in the World Health Organization world mental health surveys. Biol Psychiatry. 2013;73(9):904–914. doi:10.1016/j.biopsych.2012.11.020.
  • Hay P, Girosi F, Mond J. Prevalence and sociodemographic correlates of DSM-5 eating disorders in the Australian population. J Eat Disord. 2015;3(1):19. doi:10.1186/s40337-015-0056-0.
  • Giel KE, Bulik CM, Fernandez-Aranda F, Hay P, Keski-Rahkonen A, Schag K, Schmidt U, Zipfel S. Binge eating disorder. Nat Rev Dis Primers. 2022;8(1):16. doi:10.1038/s41572-022-00344-y.
  • Treasure J, Duarte TA, Schmidt U. Eating disorders. Lancet. 2020;395(10227):899–911. doi:10.1016/S0140-6736(20)30059-3.
  • Sommer F, Bäckhed F. The gut microbiota–masters of host development and physiology. Nat Rev Microbiol. 2013;11(4):227–238. doi:10.1038/nrmicro2974.
  • Morais LH, Schreiber HL 4th, Mazmanian SK. The gut microbiota-brain axis in behaviour and brain disorders. Nat Rev Microbiol. 2021;19(4):241–255. doi:10.1038/s41579-020-00460-0.
  • Fan S, Guo W, Xiao D, Guan M, Liao T, Peng S, Feng A, Wang Z, Yin H, Li M. et al. Microbiota-gut-brain axis drives overeating disorders. Cell Metab. 2023;35(11):2011–2027.e7. doi:10.1016/j.cmet.2023.09.005.
  • Ousey J, Boktor JC, Mazmanian SK. Gut microbiota suppress feeding induced by palatable foods. Curr Biol. 2023;33(1):147–157.e7. doi:10.1016/j.cub.2022.10.066.
  • David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA. et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505(7484):559–563. doi:10.1038/nature12820.
  • Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, Bewtra M, Knights D, Walters WA, Knight R. et al. Linking long-term dietary patterns with gut microbial enterotypes. Science. 2011;334(6052):105–108. doi:10.1126/science.1208344.
  • Gubert C, Kong G, Renoir T, Hannan AJ. Exercise, diet and stress as modulators of gut microbiota: Implications for neurodegenerative diseases. Neurobiol Dis. 2020;134:104621. doi:10.1016/j.nbd.2019.104621.
  • Mathes WF, Brownley KA, Mo X, Bulik CM. The biology of binge eating. Appetite. 2009;52(3):545–553. doi:10.1016/j.appet.2009.03.005.
  • Raevuori A, Lukkariniemi L, Suokas JT, Gissler M, Suvisaari JM, Haukka J. Increased use of antimicrobial medication in bulimia nervosa and binge-eating disorder prior to the eating disorder treatment. Int J Eat Disord. 2016;49(6):542–552. doi:10.1002/eat.22497.
  • Carbone EA, D’Amato P, Vicchio G, De Fazio P, Segura-Garcia C. A systematic review on the role of microbiota in the pathogenesis and treatment of eating disorders. Eur Psychiatry. 2020;64(1):e2. doi:10.1192/j.eurpsy.2020.109.
  • Leyrolle Q, Cserjesi R, Mulders MDGH, Zamariola G, Hiel S, Gianfrancesco MA, Rodriguez J, Portheault D, Amadieu C, Leclercq S. et al. Specific gut microbial, biological, and psychiatric profiling related to binge eating disorders: A cross-sectional study in obese patients. Clinical Nutrition. 2021;40(4):2035–2044. doi:10.1016/j.clnu.2020.09.025.
  • Monteleone AM, Troisi J, Serena G, Fasano A, Dalle Grave R, Cascino G, Marciello F, Calugi S, Scala G, Corrivetti G. et al. The gut microbiome and metabolomics profiles of restricting and binge-purging type anorexia nervosa. Nutrients. 2021;13(2):507. doi:10.3390/nu13020507.
  • Castellini G, Cassioli E, Vitali F, Rossi E, Dani C, Melani G, Flaccomio D, D’Andria M, Mejia Monroy M, Galli A. et al. Gut microbiota metabolites mediate the interplay between childhood maltreatment and psychopathology in patients with eating disorders. Sci Rep. 2023;13(1):11753. doi:10.1038/s41598-023-38665-x.
  • Bulik CM, Butner JE, Tregarthen J, Thornton LM, Flatt RE, Smith T, Carroll IM, Baucom BRW, Deboeck PR. The binge eating genetics initiative (BEGIN): study protocol. BMC Psychiatry. 2020;20(1):307. doi:10.1186/s12888-020-02698-7.
  • Herman A, Bajaka A. The role of the intestinal microbiota in eating disorders - bulimia nervosa and binge eating disorder. Psychiatry Res. 2021;300:113923. doi:10.1016/j.psychres.2021.113923.
  • Koh A, De Vadder F, Kovatcheva-Datchary P, Bäckhed F. From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell. 2016;165(6):1332–1345. doi:10.1016/j.cell.2016.05.041.
  • Fernandes J, Su W, Rahat-Rozenbloom S, Wolever TM, Comelli EM. Adiposity, gut microbiota and faecal short chain fatty acids are linked in adult humans. Nutr Diabetes. 2014;4(6):e121. doi:10.1038/nutd.2014.23.
  • Han H, Yi B, Zhong R, Wang M, Zhang S, Ma J, Yin Y, Yin J, Chen L, Zhang H. From gut microbiota to host appetite: gut microbiota-derived metabolites as key regulators. Microbiome. 2021;9(1):162. doi:10.1186/s40168-021-01093-y.
  • Liu JL, Segovia I, Yuan XL, Gao ZH. Controversial roles of gut microbiota-derived short-chain fatty Acids (SCFAs) on pancreatic β-cell growth and insulin secretion. Int J Mol Sci. 2020;21(3):910. doi:10.3390/ijms21030910.
  • Perry RJ, Peng L, Barry NA, Cline GW, Zhang D, Cardone RL, Petersen KF, Kibbey RG, Goodman AL, Shulman GI. Acetate mediates a microbiome-brain-β-cell axis to promote metabolic syndrome. Nature. 2016;534(7606):213–217. doi:10.1038/nature18309.
  • Bolognini D, Barki N, Butcher AJ, Hudson BD, Sergeev E, Molloy C, Moss CE, Bradley SJ, Le Gouill C, Bouvier M. et al. Chemogenetics defines receptor-mediated functions of short chain free fatty acids. Nat Chem Biol. 2019;15(5):489–498. doi:10.1038/s41589-019-0270-1.
  • Li Z, Yi CX, Katiraei S, Kooijman S, Zhou E, Chung CK, Gao Y, van den Heuvel JK, Meijer OC, Berbée JFP. et al. Butyrate reduces appetite and activates brown adipose tissue via the gut-brain neural circuit. Gut. 2018;67(7):1269–1279. doi:10.1136/gutjnl-2017-314050.
  • Chambers ES, Viardot A, Psichas A, Morrison DJ, Murphy KG, Zac-Varghese SE, MacDougall K, Preston T, Tedford C, Finlayson GS. et al. Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults. Gut. 2015;64(11):1744–1754. doi:10.1136/gutjnl-2014-307913.
  • Frost G, Sleeth ML, Sahuri-Arisoylu M, Lizarbe B, Cerdan S, Brody L, Anastasovska J, Ghourab S, Hankir M, Zhang S. et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat Commun. 2014;5(1):3611. doi:10.1038/ncomms4611.
  • Torres-Fuentes C, Golubeva AV, Zhdanov AV, Wallace S, Arboleya S, Papkovsky DB, Aidy SE, Ross P, Roy BL, Stanton C. et al. Short-chain fatty acids and microbiota metabolites attenuate ghrelin receptor signaling. Faseb J. 2019;33(12):13546–13559. doi:10.1096/fj.201901433R.
  • Otto B, Cuntz U, Fruehauf E, Wawarta R, Folwaczny C, Riepl RL, Heiman ML, Lehnert P, Fichter M, Tschöp M. Weight gain decreases elevated plasma ghrelin concentrations of patients with anorexia nervosa. Eur J Endocrinol. 2001;145(5):669–673. doi:10.1530/EJE-1450669.
  • Tong J, D’Alessio D. Eating disorders and gastrointestinal peptides. Curr Opin Endocrinol Diabetes Obes. 2011;18(1):42–49. doi:10.1097/MED.0b013e328341e12b.
  • Kojima S, Nakahara T, Nagai N, Muranaga T, Tanaka M, Yasuhara D, Masuda A, Date Y, Ueno H, Nakazato M. et al. Altered ghrelin and peptide YY responses to meals in bulimia nervosa. Clin Endocrinol (Oxf). 2005;62(1):74–78. doi:10.1111/j.1365-2265.2004.02176.x.
  • Monteleone P, Martiadis V, Rigamonti AE, Fabrazzo M, Giordani C, Muller EE, Maj M. Investigation of peptide YY and ghrelin responses to a test meal in bulimia nervosa. Biol Psychiatry. 2005;57(8):926–931. doi:10.1016/j.biopsych.2005.01.004.
  • Matafome P, Eickhoff H, Letra L, Seiça R. Neuroendocrinology of adipose tissue and gut-brain axis. Adv Neurobiol. 2017;19:49–70. doi:10.1007/978-3-319-63260-5_3.
  • Fetissov SO, Hamze Sinno M, Coëffier M, Bole-Feysot C, Ducrotté P, Hökfelt T, Déchelotte P. Autoantibodies against appetite-regulating peptide hormones and neuropeptides: putative modulation by gut microflora. Nutrition. 2008;24(4):348–359. doi:10.1016/j.nut.2007.12.006.
  • Panaro BL, Tough IR, Engelstoft MS, Matthews RT, Digby GJ, Møller CL, Svendsen B, Gribble F, Reimann F, Holst JJ. et al. The melanocortin-4 receptor is expressed in enteroendocrine L cells and regulates the release of peptide YY and glucagon-like peptide 1 in vivo. Cell Metab. 2014;20(6):1018–1029. doi:10.1016/j.cmet.2014.10.004.
  • Breton J, Tennoune N, Lucas N, Francois M, Legrand R, Jacquemot J, Goichon A, Guérin C, Peltier J, Pestel-Caron M. et al. Gut commensal E. coli proteins activate host satiety pathways following nutrient-induced bacterial growth. Cell Metab. 2016;23(2):324–334. doi:10.1016/j.cmet.2015.10.017.
  • Breton J, Legrand R, Akkermann K, Järv A, Harro J, Déchelotte P, Fetissov SO. Elevated plasma concentrations of bacterial ClpB protein in patients with eating disorders. Int J Eat Disord. 2016;49(8):805–808. doi:10.1002/eat.22531.
  • Tennoune N, Chan P, Breton J, Legrand R, Chabane YN, Akkermann K, Järv A, Ouelaa W, Takagi K, Ghouzali I. et al. Bacterial ClpB heat-shock protein, an antigen-mimetic of the anorexigenic peptide α-MSH, at the origin of eating disorders. Transl Psychiatry. 2014;4(10):e458. doi:10.1038/tp.2014.98.
  • Lucas N, Legrand R, Bôle-Feysot C, Breton J, Coëffier M, Akkermann K, Järv A, Harro J, Déchelotte P, Fetissov SO. Immunoglobulin G modulation of the melanocortin 4 receptor signaling in obesity and eating disorders. Transl Psychiatry. 2019;9(1):87. doi:10.1038/s41398-019-0422-9.
  • Fetissov SO, Harro J, Jaanisk M, Järv A, Podar I, Allik J, Nilsson I, Sakthivel P, Lefvert AK, Hökfelt T. Autoantibodies against neuropeptides are associated with psychological traits in eating disorders. Proc Natl Acad Sci USA. 2005;102(41):14865–14870. doi:10.1073/pnas.0507204102.
  • Auteri M, Zizzo MG, Serio R. GABA and GABA receptors in the gastrointestinal tract: from motility to inflammation. Pharmacol Res. 2015;93:11–21. doi:10.1016/j.phrs.2014.12.001.
  • Xu J, Bartolome CL, Low CS, Yi X, Chien CH, Wang P, Kong D. Genetic identification of leptin neural circuits in energy and glucose homeostases. Nature. 2018;556(7702):505–509. doi:10.1038/s41586-018-0049-7.
  • Prochazkova P, Roubalova R, Dvorak J, Kreisinger J, Hill M, Tlaskalova-Hogenova H, Tomasova P, Pelantova H, Cermakova M, Kuzma M. et al. The intestinal microbiota and metabolites in patients with anorexia nervosa. Gut Microbes. 2021;13(1):1–25. doi:10.1080/19490976.2021.1902771.
  • Zhang X, van den Pol AN. Rapid binge-like eating and body weight gain driven by zona incerta GABA neuron activation. Science. 2017;356(6340):853–859. doi:10.1126/science.aam7100.
  • Yano JM, Yu K, Donaldson GP, Shastri GG, Ann P, Ma L, Nagler CR, Ismagilov RF, Mazmanian SK, Hsiao EY. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell. 2015;161(2):264–276. doi:10.1016/j.cell.2015.02.047.
  • Kim HJ, Kim JH, Noh S, Hur HJ, Sung MJ, Hwang JT, Park JH, Yang HJ, Kim MS, Kwon DY. et al. Metabolomic analysis of livers and serum from high-fat diet induced obese mice. J Proteome Res. 2011;10(2):722–731. doi:10.1021/pr100892r.
  • Janssen P, Vos R, Van Oudenhove L, Tack J. Influence of the 5-HT3 receptor antagonist ondansetron on gastric sensorimotor function and nutrient tolerance in healthy volunteers. Neurogastroenterol Motil. 2011;23(5):444–9, e175. doi:10.1111/j.1365-2982.2010.01655.x.
  • Savastano DM, Covasa M. Intestinal nutrients elicit satiation through concomitant activation of CCK(1) and 5-HT(3) receptors. Physiol Behav. 2007;92(3):434–442. doi:10.1016/j.physbeh.2007.04.017.
  • Rhoades JL, Nelson JC, Nwabudike I, Yu SK, McLachlan IG, Madan GK, Abebe E, Powers JR, Colón-Ramos DA, Flavell SW. ASICs mediate food responses in an enteric serotonergic neuron that controls foraging behaviors. Cell. 2019;176(1–2):85–97.e14. doi:10.1016/j.cell.2018.11.023.
  • Kopeikina E, Dukhinova M, Yung AWY, Veremeyko T, Kuznetsova IS, Lau TYB, Levchuk K, Ponomarev ED. Platelets promote epileptic seizures by modulating brain serotonin level, enhancing neuronal electric activity, and contributing to neuroinflammation and oxidative stress. Prog Neurobiol. 2020;188:101783. doi:10.1016/j.pneurobio.2020.101783.
  • D’Agostino G, Lyons D, Cristiano C, Lettieri M, Olarte-Sanchez C, Burke LK, Greenwald-Yarnell M, Cansell C, Doslikova B, Georgescu T. et al. Nucleus of the solitary tract serotonin 5-HT2C receptors modulate food intake. Cell Metab. 2018;28(4):619–630.e5. doi:10.1016/j.cmet.2018.07.017.
  • He Y, Cai X, Liu H, Conde KM, Xu P, Li Y, Wang C, Yu M, He Y, Liu H. et al. 5-HT recruits distinct neurocircuits to inhibit hunger-driven and non-hunger-driven feeding. Mol Psychiatry. 2021;26(12):7211–7224. doi:10.1038/s41380-021-01220-z.
  • Price AE, Anastasio NC, Stutz SJ, Hommel JD, Cunningham KA. Serotonin 5-HT2C receptor activation suppresses binge intake and the reinforcing and motivational properties of high-fat food. Front Pharmacol. 2018;9:821. doi:10.3389/fphar.2018.00821.
  • Xu P, He Y, Cao X, Valencia-Torres L, Yan X, Saito K, Wang C, Yang Y, Hinton A Jr, Zhu L. et al. Activation of serotonin 2C receptors in dopamine neurons inhibits binge-like eating in mice. Biol Psychiatry. 2017;81(9):737–747. doi:10.1016/j.biopsych.2016.06.005.
  • Jimerson DC, Lesem MD, Kaye WH, Hegg AP, Brewerton TD. Eating disorders and depression: is there a serotonin connection? Biol Psychiatry. 1990;28(5):443–454. doi:10.1016/0006-3223(90)90412-u.
  • Steiger H, Koerner N, Engelberg MJ, Israël M, Ng Ying Kin NM, Young SN. Self-destructiveness and serotonin function in bulimia nervosa. Psychiatry Res. 2001;103(1):15–26. doi:10.1016/s0165-1781(01)00264-5.
  • Kaye W. Neurobiology of anorexia and bulimia nervosa. Physiol Behav. 2008;94(1):121–135. doi:10.1016/j.physbeh.2007.11.037.
  • Dingemans A, Danner U, Parks M. Emotion regulation in binge eating disorder: a review. Nutrients. 2017;9(11):1274. doi:10.3390/nu9111274.
  • Kaye WH, Gendall KA, Fernstrom MH, Fernstrom JD, McConaha CW, Weltzin TE. Effects of acute tryptophan depletion on mood in bulimia nervosa. Biol Psychiatry. 2000;47(2):151–157. doi:10.1016/s0006-3223(99)00108-0.
  • Hilbert A, Petroff D, Herpertz S, Pietrowsky R, Tuschen-Caffier B, Vocks S, Schmidt R. Meta-analysis on the long-term effectiveness of psychological and medical treatments for binge-eating disorder. Int J Eat Disord. 2020;53(9):1353–1376. doi:10.1002/eat.23297.
  • Raybould HE. Gut chemosensing: interactions between gut endocrine cells and visceral afferents. Auton Neurosci. 2010;153(1–2):41–46. doi:10.1016/j.autneu.2009.07.007.
  • Lo Sauro C, Ravaldi C, Cabras PL, Faravelli C, Ricca V. Stress, hypothalamic-pituitary-adrenal axis and eating disorders. Neuropsychobiology. 2008;57(3):95–115. doi:10.1159/000138912.
  • Wisse BE, Ogimoto K, Tang J, Harris MK Jr, Raines EW, Schwartz MW. Evidence that lipopolysaccharide-induced anorexia depends upon central, rather than peripheral, inflammatory signals. Endocrinology. 2007;148(11):5230–5237. doi:10.1210/en.2007-0394.
  • von Meyenburg C, Hrupka BH, Arsenijevic D, Schwartz GJ, Landmann R, Langhans W. Role for CD14, TLR2, and TLR4 in bacterial product-induced anorexia. Am J Physiol Regul Integr Comp Physiol. 2004;287(2):R298–305. doi:10.1152/ajpregu.00659.2003.
  • Banks WA, Erickson MA. The blood-brain barrier and immune function and dysfunction. Neurobiol Dis. 2010;37(1):26–32. doi:10.1016/j.nbd.2009.07.031.
  • Kaelberer MM, Buchanan KL, Klein ME, Barth BB, Montoya MM, Shen X, Bohórquez DV. A gut-brain neural circuit for nutrient sensory transduction. Science. 2018;361(6408):eaat5236. doi:10.1126/science.aat5236.
  • Bellono NW, Bayrer JR, Leitch DB, Castro J, Zhang C, O’Donnell TA, Brierley SM, Ingraham HA, Julius D. Enterochromaffin cells are gut chemosensors that couple to sensory neural pathways. Cell. 2017;170(1):185–198.e16. doi:10.1016/j.cell.2017.05.034.
  • Williams EK, Chang RB, Strochlic DE, Umans BD, Lowell BB, Liberles SD. Sensory neurons that detect stretch and nutrients in the digestive system. Cell. 2016;166(1):209–221. doi:10.1016/j.cell.2016.05.011.
  • Bai L, Mesgarzadeh S, Ramesh KS, Huey EL, Liu Y, Gray LA, Aitken TJ, Chen Y, Beutler LR, Ahn JS. et al. Genetic identification of vagal sensory neurons that control feeding. Cell. 2019;179(5):1129–1143.e23. doi:10.1016/j.cell.2019.10.031.
  • Kupari J, Häring M, Agirre E, Castelo-Branco G, Ernfors P. An atlas of vagal sensory neurons and their molecular specialization. Cell Rep. 2019;27(8):2508–2523.e4. doi:10.1016/j.celrep.2019.04.096.
  • Chen H, Nwe PK, Yang Y, Rosen CE, Bielecka AA, Kuchroo M, Cline GW, Kruse AC, Ring AM, Crawford JM. et al. A forward chemical genetic screen reveals gut microbiota metabolites that modulate host physiology. Cell. 2019;177(5):1217–1231.e18. doi:10.1016/j.cell.2019.03.036.
  • Colosimo DA, Kohn JA, Luo PM, Piscotta FJ, Han SM, Pickard AJ, Rao A, Cross JR, Cohen LJ, Brady SF. Mapping interactions of microbial metabolites with human G-Protein-coupled receptors. Cell Host Microbe. 2019;26(2):273–282.e7. doi:10.1016/j.chom.2019.07.002.
  • Johnson EL, Heaver SL, Waters JL, Kim BI, Bretin A, Goodman AL, Gewirtz AT, Worgall TS, Ley RE. Sphingolipids produced by gut bacteria enter host metabolic pathways impacting ceramide levels. Nat Commun. 2020;11(1):2471. doi:10.1038/s41467-020-16274-w.
  • Wu X, Li JY, Lee A, Lu YX, Zhou SY, Owyang C. Satiety induced by bile acids is mediated via vagal afferent pathways. JCI Insight. 2020;5(14):e132400. doi:10.1172/jci.insight.132400.
  • Wahlström A, Sayin SI, Marschall HU, Bäckhed F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab. 2016;24(1):41–50. doi:10.1016/j.cmet.2016.05.005.
  • Buckley MM, O’Brien R, Brosnan E, Ross RP, Stanton C, Buckley JM, O’Malley D. Glucagon-like peptide-1 secreting L-Cells coupled to sensory nerves translate microbial signals to the host rat nervous system. Front Cell Neurosci. 2020;14:95. doi:10.3389/fncel.2020.00095.
  • Psichas A, Sleeth ML, Murphy KG, Brooks L, Bewick GA, Hanyaloglu AC, Ghatei MA, Bloom SR, Frost G. The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. Int J Obes (Lond). 2015;39(3):424–429. doi:10.1038/ijo.2014.153.
  • Caengprasath N, Gonzalez-Abuin N, Shchepinova M, Ma Y, Inoue A, Tate EW, Frost G, Hanyaloglu AC. Internalization-dependent free fatty acid receptor 2 signaling is essential for propionate-induced anorectic gut hormone release. iScience. 2020;23(9):101449. doi:10.1016/j.isci.2020.101449.
  • Shackley M, Ma Y, Tate EW, Brown AJH, Frost G, Hanyaloglu AC. Short chain fatty acids enhance expression and activity of the umami taste receptor in enteroendocrine cells via a Gαi/o pathway. Front Nutr. 2020;7:568991. doi:10.3389/fnut.2020.568991.
  • Goswami C, Iwasaki Y, Yada T. Short-chain fatty acids suppress food intake by activating vagal afferent neurons. J Nutr Biochem. 2018;57:130–135. doi:10.1016/j.jnutbio.2018.03.009.
  • Waise TMZ, Dranse HJ, Lam TKT. The metabolic role of vagal afferent innervation. Nat Rev Gastroenterol Hepatol. 2018;15(10):625–636. doi:10.1038/s41575-018-0062-1.
  • Aklan I, Sayar Atasoy N, Yavuz Y, Ates T, Coban I, Koksalar F, Filiz G, Topcu IC, Oncul M, Dilsiz P. et al. NTS catecholamine neurons mediate hypoglycemic hunger via medial hypothalamic feeding pathways. Cell Metab. 2020;31(2):313–326.e5. doi:10.1016/j.cmet.2019.11.016.
  • Han W, Tellez LA, Perkins MH, Perez IO, Qu T, Ferreira J, Ferreira TL, Quinn D, Liu ZW, Gao XB. et al. A neural circuit for gut-induced reward. Cell. 2018;175(3):887–888. doi:10.1016/j.cell.2018.10.018.
  • Kessler RM, Hutson PH, Herman BK, Potenza MN. The neurobiological basis of binge-eating disorder. Neurosci Biobehav Rev. 2016;63:223–238. doi:10.1016/j.neubiorev.2016.01.013.
  • Donnelly B, Touyz S, Hay P, Burton A, Russell J, Caterson I. Neuroimaging in bulimia nervosa and binge eating disorder: a systematic review. J Eat Disord. 2018;6(1):3. doi:10.1186/s40337-018-0187-1.
  • Goldschmidt AB, Smith KE, Crosby RD, Boyd HK, Dougherty E, Engel SG, Haedt-Matt A. Ecological momentary assessment of maladaptive eating in children and adolescents with overweight or obesity. Int J Eat Disord. 2018;51(6):549–557. doi:10.1002/eat.22864.
  • Witt AA, Lowe MR. Hedonic hunger and binge eating among women with eating disorders. Int J Eat Disord. 2014;47(3):273–280. doi:10.1002/eat.22171.
  • Dawe S, Loxton NJ. The role of impulsivity in the development of substance use and eating disorders. Neurosci Biobehav Rev. 2004;28(3):343–351. doi:10.1016/j.neubiorev.2004.03.007.
  • Galanti K, Gluck ME, Geliebter A. Test meal intake in obese binge eaters in relation to impulsivity and compulsivity. Int J Eat Disord. 2007;40(8):727–732. doi:10.1002/eat.20441.
  • Hege MA, Stingl KT, Kullmann S, Schag K, Giel KE, Zipfel S, Preissl H. Attentional impulsivity in binge eating disorder modulates response inhibition performance and frontal brain networks. Int J Obes (Lond). 2015;39(2):353–360. doi:10.1038/ijo.2014.99.
  • Wu M, Brockmeyer T, Hartmann M, Skunde M, Herzog W, Friederich HC. Set-shifting ability across the spectrum of eating disorders and in overweight and obesity: a systematic review and meta-analysis. Psychol Med. 2014;44(16):3365–3385. doi:10.1017/S0033291714000294.
  • Cox J, Witten IB. Striatal circuits for reward learning and decision-making. Nat Rev Neurosci. 2019;20(8):482–494. doi:10.1038/s41583-019-0189-2.
  • Voon V, Morris LS, Irvine MA, Ruck C, Worbe Y, Derbyshire K, Rankov V, Schreiber LR, Odlaug BL, Harrison NA. et al. Risk-taking in disorders of natural and drug rewards: neural correlates and effects of probability, valence, and magnitude. Neuropsychopharmacology. 2015;40(4):804–812. doi:10.1038/npp.2014.242.
  • Mestre-Bach G, Fernández-Aranda F, Jiménez-Murcia S, Potenza MN. Decision-making in gambling disorder, problematic pornography use, and binge-eating disorder: similarities and differences. Curr Behav Neurosci Rep. 2020;7(3):97–108. doi:10.1007/s40473-020-00212-7.
  • Manwaring JL, Green L, Myerson J, Strube MJ, Wilfley DE. Discounting of various types of rewards by women with and without binge eating disorder: evidence for general rather than specific differences. Psychol Rec. 2011;61(4):561–582. doi:10.1007/BF03395777.
  • Appelhans BM, Woolf K, Pagoto SL, Schneider KL, Whited MC, Liebman R. Inhibiting food reward: delay discounting, food reward sensitivity, and palatable food intake in overweight and obese women. Obesity (Silver Spring). 2011;19(11):2175–2182. doi:10.1038/oby.2011.57.
  • Steward T, Mestre-Bach G, Vintró-Alcaraz C, Agüera Z, Jiménez-Murcia S, Granero R, Fernández-Aranda F. Delay discounting of reward and impulsivity in eating disorders: from anorexia nervosa to binge eating disorder. Eur Eat Disord Rev. 2017;25(6):601–606. doi:10.1002/erv.2543.
  • Steward T, Mestre-Bach G, Fernández-Aranda F, Granero R, Perales JC, Navas JF, Soriano-Mas C, Baño M, Fernández-Formoso JA, Martín-Romera V. et al. Delay discounting and impulsivity traits in young and older gambling disorder patients. Addict Behav. 2017;71:96–103. doi:10.1016/j.addbeh.2017.03.001.
  • Amlung M, Marsden E, Holshausen K, Morris V, Patel H, Vedelago L, Naish KR, Reed DD, McCabe RE. Delay discounting as a transdiagnostic process in psychiatric disorders: a meta-analysis. JAMA Psychiarty. 2019;76(11):1176–1186. doi:10.1001/jamapsychiatry.2019.2102.
  • Friedman NP, Robbins TW. The role of prefrontal cortex in cognitive control and executive function. Neuropsychopharmacology. 2022;47(1):72–89. doi:10.1038/s41386-021-01132-0.
  • Cury MEG, Berberian A, Scarpato BS, Kerr-Gaffney J, Santos FH, Claudino AM. Scrutinizing domains of executive function in binge eating disorder: a systematic review and meta-analysis. Front Psychiatry. 2020;11:288. doi:10.3389/fpsyt.2020.00288.
  • Iceta S, Rodrigue C, Legendre M, Daoust J, Flaudias V, Michaud A, Bégin C. Cognitive function in binge eating disorder and food addiction: A systematic review and three-level meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry. 2021;111:110400. doi:10.1016/j.pnpbp.2021.110400.
  • Balodis IM, Molina ND, Kober H, Worhunsky PD, White MA, Sinha R, Grilo CM, Potenza MN. Divergent neural substrates of inhibitory control in binge eating disorder relative to other manifestations of obesity. Obesity (Silver Spring). 2013;21(2):367–377. doi:10.1002/oby.20068.
  • Pearson CM, Wonderlich SA, Smith GT. A risk and maintenance model for bulimia nervosa: From impulsive action to compulsive behavior. Psychol Rev. 2015;122(3):516–535. doi:10.1037/a0039268.
  • Leenaerts N, Jongen D, Ceccarini J, Van Oudenhove L, Vrieze E. The neurobiological reward system and binge eating: A critical systematic review of neuroimaging studies. Int J Eat Disord. 2022;55(11):1421–1458. doi:10.1002/eat.23776.
  • Mond JM, Arrighi A. Gender differences in perceptions of the severity and prevalence of eating disorders. Early Interv Psychiatry. 2011;5(1):41–49. doi:10.1111/j.1751-7893.2010.00257.x.
  • Culbert KM, Sisk CL, Klump KL. A narrative review of sex differences in eating disorders: is there a biological basis? Clin Ther. 2021;43(1):95–111. doi:10.1016/j.clinthera.2020.12.003.
  • Culbert KM, Sinclair EB, Hildebrandt BA, Klump KL, Sisk CL. Perinatal testosterone contributes to mid-to-post pubertal sex differences in risk for binge eating in male and female rats. J Abnorm Psychol. 2018;127(2):239–250. doi:10.1037/abn0000334.
  • Bergman L, Eriksson E. Marked symptom reduction in two women with bulimia nervosa treated with the testosterone receptor antagonist flutamide. Acta Psychiatr Scand. 1996;94(2):137–139. doi:10.1111/j.1600-0447.1996.tb09838.x.
  • Sundblad C, Landén M, Eriksson T, Bergman L, Eriksson E. Effects of the androgen antagonist flutamide and the serotonin reuptake inhibitor citalopram in bulimia nervosa: a placebo-controlled pilot study. J Clin Psychopharmacol. 2005;25(1):85–88. doi:10.1097/01.jcp.0000150222.31007.a9.
  • Asarian L, Geary N. Modulation of appetite by gonadal steroid hormones. Phil Trans R Soc B. 2006;361(1471):1251–1263. doi:10.1098/rstb.2006.1860.
  • Micioni Di Bonaventura MV, Lutz TA, Romano A, Pucci M, Geary N, Asarian L, Cifani C. Estrogenic suppression of binge-like eating elicited by cyclic food restriction and frustrative-nonreward stress in female rats. Int J Eat Disord. 2017;50(6):624–635. doi:10.1002/eat.22687.
  • Sinha T, Vich Vila A, Garmaeva S, Jankipersadsing SA, Imhann F, Collij V, Bonder MJ, Jiang X, Gurry T, Alm EJ. et al. Analysis of 1135 gut metagenomes identifies sex-specific resistome profiles. Gut Microbes. 2019;10(3):358–366. doi:10.1080/19490976.2018.1528822.
  • Kwa M, Plottel CS, Blaser MJ, Adams S. The intestinal microbiome and estrogen receptor-positive female breast cancer. J Natl Cancer Inst. 2016;108(8):djw029. doi:10.1093/jnci/djw029.
  • Gomez A, Luckey D, Taneja V. The gut microbiome in autoimmunity: Sex matters. Clin Immunol. 2015;159(2):154–162. doi:10.1016/j.clim.2015.04.016.
  • Russell H, Aouad P, Le A, Marks P, Maloney D, Aouad P, Barakat S, Boakes R, Brennan L, Bryant E. et al. Psychotherapies for eating disorders: findings from a rapid review. J Eat Disord. 2023;11(1):175. doi:10.1186/s40337-023-00886-w.
  • National Institute for Health and Care Excellence. Eating disorders: recognition and treatment. NICE; 2017. https://www.nice.org.uk/guidance/ng69/evidence/full-guideline-pdf-161214767896.
  • Hilbert A, Hoek HW, Schmidt R. Evidence-based clinical guidelines for eating disorders: international comparison. Curr Opin Psychiatry. 2017;30(6):423–437. doi:10.1097/YCO.0000000000000360.
  • Wilson GT, Zandberg LJ. Cognitive-behavioral guided self-help for eating disorders: effectiveness and scalability. Clin Psychol Rev. 2012;32(4):343–357. doi:10.1016/j.cpr.2012.03.001.
  • Hilbert A, Petroff D, Herpertz S, Pietrowsky R, Tuschen-Caffier B, Vocks S, Schmidt R. Meta-analysis of the efficacy of psychological and medical treatments for binge-eating disorder. J Consult Clin Psychol. 2019;87(1):91–105. doi:10.1037/ccp0000358.
  • Lammers MW, Vroling MS, Crosby RD, van Strien T. Dialectical behavior therapy adapted for binge eating compared to cognitive behavior therapy in obese adults with binge eating disorder: a controlled study. J Eat Disord. 2020;8(1):27. doi:10.1186/s40337-020-00299-z.
  • Reas DL, Grilo CM. Review and meta-analysis of pharmacotherapy for binge-eating disorder. Obesity (Silver Spring). 2008;16(9):2024–2038. doi:10.1038/oby.2008.333.
  • Heo YA, Duggan ST. Lisdexamfetamine: A Review in Binge Eating Disorder. CNS Drugs. 2017;31(11):1015–1022. doi:10.1007/s40263-017-0477-1.
  • Ghaderi A, Odeberg J, Gustafsson S, Råstam M, Brolund A, Pettersson A, Parling T. Psychological, pharmacological, and combined treatments for binge eating disorder: a systematic review and meta-analysis. PeerJ. 2018;6:e5113. doi:10.7717/peerj.5113.
  • Monteleone AM, Pellegrino F, Croatto G, Carfagno M, Hilbert A, Treasure J, Wade T, Bulik CM, Zipfel S, Hay P. et al. Treatment of eating disorders: A systematic meta-review of meta-analyses and network meta-analyses. Neurosci Biobehav Rev. 2022;142:104857. doi:10.1016/j.neubiorev.2022.104857.
  • Reas DL, Grilo CM. Psychotherapy and medications for eating disorders: better together? Clin Ther. 2021;43(1):17–39. doi:10.1016/j.clinthera.2020.10.006.
  • Terry SM, Barnett JA, Gibson DL. A critical analysis of eating disorders and the gut microbiome. J Eat Disord. 2022;10(1):154. doi:10.1186/s40337-022-00681-z.
  • de Clercq NC, Frissen MN, Davids M, Groen AK, Nieuwdorp M. Weight gain after fecal microbiota transplantation in a patient with recurrent underweight following clinical recovery from anorexia nervosa. Psychother Psychosom. 2019;88(1):58–60. doi:10.1159/000495044.
  • Currò D, Ianiro G, Pecere S, Bibbò S, Cammarota G. Probiotics, fibre and herbal medicinal products for functional and inflammatory bowel disorders. Br J Pharmacol. 2017;174(11):1426–1449. doi:10.1111/bph.13632.
  • Ford AC, Quigley EM, Lacy BE, Lembo AJ, Saito YA, Schiller LR, Soffer EE, Spiegel BM, Moayyedi P. Efficacy of prebiotics, probiotics, and synbiotics in irritable bowel syndrome and chronic idiopathic constipation: systematic review and meta-analysis. Am J Gastroenterol. 2014;109(10):1547–1561. doi:10.1038/ajg.2014.202.
  • Kho ZY, Lal SK. The human gut microbiome - a potential controller of wellness and disease. Front Microbiol. 2018;9:1835. doi:10.3389/fmicb.2018.01835.
  • Möhle L, Mattei D, Heimesaat MM, Bereswill S, Fischer A, Alutis M, French T, Hambardzumyan D, Matzinger P, Dunay IR. et al. Ly6C(hi) monocytes provide a link between antibiotic-induced changes in gut microbiota and adult hippocampal neurogenesis. Cell Rep. 2016;15(9):1945–1956. doi:10.1016/j.celrep.2016.04.074.
  • McKean J, Naug H, Nikbakht E, Amiet B, Colson N. Probiotics and subclinical psychological symptoms in healthy participants: a systematic review and meta-analysis. J Altern Complement Med. 2017;23(4):249–258. doi:10.1089/acm.2016.0023.
  • Carlos LO, Ramos MRZ, Wagner NRF, Freitas LAC, Felicidade I, Campos ACL. Probiotic supplementation attenuates binge eating and food addiction 1 year after ROUX-EN-Y gastric bypass: a randomized, double-blind, placebo-controlled trial. Arq Bras Cir Dig. 2022;35:e1659. doi:10.1590/0102-672020210002e1659.
  • Pirbaglou M, Katz J, de Souza RJ, Stearns JC, Motamed M, Ritvo P. Probiotic supplementation can positively affect anxiety and depressive symptoms: a systematic review of randomized controlled trials. Nutr Res. 2016;36(9):889–898. doi:10.1016/j.nutres.2016.06.009.
  • Huang R, Wang K, Hu J. Effect of probiotics on depression: a systematic review and meta-analysis of randomized controlled trials. Nutrients. 2016;8(8):483. doi:10.3390/nu8080483.