Category: Family

Appetite control technology

Appetite control technology

Iwasaki Y, Sendo M, Dezaki K, Appteite T, Sato Contrl, Nakata M, et al. Correspondence to Jie YinLiang Chen or Hongfu Zhang. Deep Dive. Article Talk. STAT reporter Meghana Keshavan speaks with Prof.

Appetite control technology -

Gastric bypass surgery recruits a gut PPAR-alpha-striatal D1R pathway to reduce fat appetite in obese rats. Breton J, Tennoune N, Lucas N, Francois M, Legrand R, Jacquemot J, et al. Gut commensal E. coli proteins activate host satiety pathways following nutrient-induced bacterial growth.

Breton J, Tirelle P, Hasanat S, Pernot A, L'Huillier C, do Rego JC, et al. Gut microbiota alteration in a mouse model of anorexia nervosa. Greenhill C. Gut microbiota: proteins released by E. coli in the gut influence host appetite control.

PubMed Google Scholar. Sharon G, Cruz NJ, Kang DW, Gandal MJ, Wang B, Kim YM, et al. Human gut microbiota from autism spectrum disorder promote behavioral symptoms in mice. Grasset E, Puel A, Charpentier J, Collet X, Christensen JE, Terce F, et al. A specific gut microbiota dysbiosis of type 2 diabetic mice induces GLP-1 resistance through an enteric NO-dependent and gut-brain axis mechanism.

Morita C, Tsuji H, Hata T, Gondo M, Takakura S, Kawai K, et al. Gut dysbiosis in patients with anorexia nervosa. Plos One. Article PubMed PubMed Central CAS Google Scholar.

Kleiman SC, Watson HJ, Bulik-Sullivan EC, Huh EY, Tarantino LM, Bulik CM, et al. The intestinal microbiota in acute anorexia nervosa and during renourishment: relationship to depression, anxiety, and eating disorder psychopathology. Psychosom Med. Mack I, Cuntz U, Gramer C, Niedermaier S, Pohl C, Schwiertz A, et al.

Weight gain in anorexia nervosa does not ameliorate the faecal microbiota, branched chain fatty acid profiles, and gastrointestinal complaints. Sci Rep. Breton J, Legrand R, Akkermann K, Järv A, Harro J, Déchelotte P, et al. Elevated plasma concentrations of bacterial ClpB protein in patients with eating disorders.

Int J Eat Disord. Yin J, Han H, Li Y, Liu Z, Zhao Y, Fang R, et al. Lysine restriction affects feed intake and amino acid metabolism via gut microbiome in piglets. Cell Physiol Biochem. Khera AV, Emdin CA, Drake I, Natarajan P, Bick AG, Cook NR, et al. Genetic risk, adherence to a healthy lifestyle, and coronary disease.

N Engl J Med. Barabási A-L, Menichetti G, Loscalzo J. The unmapped chemical complexity of our diet. Nature Food. Article Google Scholar. van de Wouw M, Schellekens H, Dinan TG and Cryan JF. Microbiota-Gut-Brain Axis: Modulator of Host Metabolism and Appetite.

J Nutr. TO K, Carli JF, Skowronski AA, Sun Q, Kriebel J, Feitosa MF, et al. Genome-wide meta-analysis uncovers novel loci influencing circulating leptin levels. Nat Commun. Article CAS Google Scholar. Park S, Aintablian A, Coupe B, Bouret SG. The endoplasmic reticulum stress-autophagy pathway controls hypothalamic development and energy balance regulation in leptin-deficient neonates.

Rasmussen BA, Breen DM, Duca FA, Côté CD, Zadeh-Tahmasebi M, Filippi BM, et al. Jejunal leptin-PI3K signaling lowers glucose production. Sobhani I, Bado A, Vissuzaine C, Buyse M, Kermorgant S, Laigneau JP, et al. Leptin secretion and leptin receptor in the human stomach.

Banks WA. The blood-brain barrier as an endocrine tissue. Elefteriou F, Ahn JD, Takeda S, Starbuck M, Yang X, Liu X, et al. Leptin regulation of bone resorption by the sympathetic nervous system and CART. Yang Y, van der Klaauw AA, Zhu L, Cacciottolo TM, He Y, Stadler LKJ, et al.

Steroid receptor coactivator-1 modulates the function of Pomc neurons and energy homeostasis. Xu J, Bartolome CL, Low CS, Yi X, Chien CH, Wang P, et al.

Genetic identification of leptin neural circuits in energy and glucose homeostases. Cedernaes J, Huang W, Ramsey KM, Waldeck N, Cheng L, Marcheva B, et al. Transcriptional basis for rhythmic control of hunger and metabolism within the AgRP neuron.

Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, et al. Richness of human gut microbiome correlates with metabolic markers.

Article PubMed CAS Google Scholar. Amar J, Chabo C, Waget A, Klopp P, Vachoux C, Bermúdez-Humarán LG, et al. Intestinal mucosal adherence and translocation of commensal bacteria at the early onset of type 2 diabetes: molecular mechanisms and probiotic treatment.

EMBO Mol Med. Massier L, Chakaroun R, Tabei S, Crane A, Didt KD, Fallmann J, et al. Adipose tissue derived bacteria are associated with inflammation in obesity and type 2 diabetes.

Schéle E, Grahnemo L, Anesten F, Hallén A, Bäckhed F, Jansson J-O. The gut microbiota reduces leptin sensitivity and the expression of the obesity-suppressing neuropeptides proglucagon Gcg and brain-derived neurotrophic factor Bdnf in the central nervous system.

Yao H, Fan C, Fan X, Lu Y, Wang Y, Wang R, et al. Effects of gut microbiota on leptin expression and body weight are lessened by high-fat diet in mice. Br J Nutr. Backhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, et al.

The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A. Lee J, Jang JY, Kwon MS, Lim SK, Kim N, Lee J, et al.

Mixture of two Lactobacillus plantarum strains modulates the gut microbiota structure and regulatory T cell response in diet-induced obese mice.

Mol Nutr Food Res. Song X, Zhong L, Lyu N, Liu F, Li B, Hao Y, et al. Genomics Proteomics Bioinformatics. Al-Muzafar HM, Amin KA.

Probiotic mixture improves fatty liver disease by virtue of its action on lipid profiles, leptin, and inflammatory biomarkers. BMC Complement Altern Med. Bagarolli RA, Tobar N, Oliveira AG, Araujo TG, Carvalho BM, Rocha GZ, et al. Probiotics modulate gut microbiota and improve insulin sensitivity in DIO mice.

J Nutr Biochem. Swisa A, Avrahami D, Eden N, Zhang J, Feleke E, Dahan T, et al. PAX6 maintains β cell identity by repressing genes of alternative islet cell types.

Cowley MA, Smith RG, Diano S, Tschöp M, Pronchuk N, Grove KL, et al. The distribution and mechanism of action of ghrelin in the CNS demonstrates a novel hypothalamic circuit regulating energy homeostasis. Date Y, Murakami N, Toshinai K, Matsukura S, Niijima A, Matsuo H, et al.

The role of the gastric afferent vagal nerve in ghrelin-induced feeding and growth hormone secretion in rats.

Panaro BL, Tough IR, Engelstoft MS, Matthews RT, Digby GJ, Moller CL, 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.

Kuhnen P, Krude H, Biebermann H. Melanocortin-4 receptor signalling: importance for weight regulation and obesity treatment. Trends Mol Med. Suarez AN, Liu CM, Cortella AM, Noble EE, Kanoski SE. Ghrelin and orexin interact to increase meal size through a descending hippocampus to hindbrain signaling pathway.

Biol Psychiatry. Theander-Carrillo C, Wiedmer P, Cettour-Rose P, Nogueiras R, Perez-Tilve D, Pfluger P, et al. Ghrelin action in the brain controls adipocyte metabolism. Perry RJ, Peng L, Barry NA, Cline GW, Zhang D, Cardone RL, et al.

Acetate mediates a microbiome-brain-β-cell axis to promote metabolic syndrome. Torres-Fuentes C, Golubeva AV, Zhdanov AV, Wallace S, Arboleya S, Papkovsky DB, et al. Short-chain fatty acids and microbiota metabolites attenuate ghrelin receptor signaling. FASEB J. Parnell JA, Reimer RA. Weight loss during oligofructose supplementation is associated with decreased ghrelin and increased peptide YY in overweight and obese adults.

Am J Clin Nutr. Cani PD, Lecourt E, Dewulf EM, Sohet FM, Pachikian BD, Naslain D, et al. Gut microbiota fermentation of prebiotics increases satietogenic and incretin gut peptide production with consequences for appetite sensation and glucose response after a meal.

Hume MP, Nicolucci AC, Reimer RA. Prebiotic supplementation improves appetite control in children with overweight and obesity: a randomized controlled trial. Berthoud HR. Metabolic and hedonic drives in the neural control of appetite: who is the boss? Curr Opin Neurobiol. Leidmaa E, Gazea M, Patchev AV, Pissioti A, Christian Gassen N, Kimura M, et al.

Blunted leptin sensitivity during hedonic overeating can be reinstated by activating galanin 2 receptors Gal2R in the lateral hypothalamus.

Acta Physiol Oxf. Zhang X, van den Pol AN. Rapid binge-like eating and body weight gain driven by zona incerta GABA neuron activation. Murphy KG, Bloom SR. Gut hormones and the regulation of energy homeostasis. van Golen LW, Ijzerman RG, Huisman MC, Hensbergen JF, Hoogma RP, Drent ML, et al.

Cerebral blood flow and glucose metabolism in appetite-related brain regions in type 1 diabetic patients after treatment with insulin detemir and NPH insulin: a randomized controlled crossover trial.

Diabetes Care. Loh K, Zhang L, Brandon A, Wang Q, Begg D, Qi Y, et al. Insulin controls food intake and energy balance via NPY neurons.

Mol Metab. Hallschmid M, Higgs S, Thienel M, Ott V, Lehnert H. Postprandial administration of intranasal insulin intensifies satiety and reduces intake of palatable snacks in women. Kim HK, Youn BS, Shin MS, Namkoong C, Park KH, Baik JH, et al. Suarez-Zamorano N, Fabbiano S, Chevalier C, Stojanovic O, Colin DJ, Stevanovic A, et al.

Microbiota depletion promotes browning of white adipose tissue and reduces obesity. Nat Med. Zarrinpar A, Chaix A, Xu ZZ, Chang MW, Marotz CA, Saghatelian A, et al. Antibiotic-induced microbiome depletion alters metabolic homeostasis by affecting gut signaling and colonic metabolism.

Li X, Wang E, Yin B, Fang D, Chen P, Wang G, et al. Effects of Lactobacillus casei CCFM on insulin resistance and gut microbiota in type 2 diabetic mice. Benef Microbes. Mao JH, Kim YM, Zhou YX, Hu D, Zhong C, Chang H, et al. Genetic and metabolic links between the murine microbiome and memory.

de Clercq NC, Frissen MN, Davids M, Groen AK and Nieuwdorp M. Weight Gain after Fecal Microbiota Transplantation in a Patient with Recurrent Underweight following Clinical Recovery from Anorexia Nervosa. Psychother Psychosom.

Yang H, Yang M, Fang S, Huang X, He M, Ke S, et al. Evaluating the profound effect of gut microbiome on host appetite in pigs. BMC Microbiol. Liu JL, Segovia I, Yuan XL and 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. Acetate mediates a microbiome-brain-beta-cell axis to promote metabolic syndrome. Priyadarshini M, Navarro G and Layden BT. Gut Microbiota: FFAR Reaching Effects on Islets.

Chambers ES, Viardot A, Psichas A, Morrison DJ, Murphy KG, Zac-Varghese SE, et al. Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults. Bolognini D, Barki N, Butcher AJ, Hudson BD, Sergeev E, Molloy C, et al.

Chemogenetics defines receptor-mediated functions of short chain free fatty acids. Nat Chem Biol. Li Z, Yi C-X, Katiraei S, Kooijman S, Zhou E, Chung CK, et al.

Butyrate reduces appetite and activates brown adipose tissue via the gut-brain neural circuit. Kimura I, Ozawa K, Inoue D, Imamura T, Kimura K, Maeda T, et al.

The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR Iwasaki Y, Sendo M, Dezaki K, Hira T, Sato T, Nakata M, et al. GLP-1 release and vagal afferent activation mediate the beneficial metabolic and chronotherapeutic effects of D-allulose.

Song Y, Koehler JA, Baggio LL, Powers AC, Sandoval DA, Drucker DJ. Gut-proglucagon-derived peptides are essential for regulating glucose homeostasis in mice. Gero D, Steinert RE, Hosa H, Cummings DE, Bueter M.

Appetite, glycemia, and entero-insular hormone responses differ between oral, gastric-remnant, and duodenal administration of a mixed-meal test after Roux-en-Y gastric bypass. Lu VB, Rievaj J, O'Flaherty EA, Smith CA, Pais R, Pattison LA, et al.

Adenosine triphosphate is co-secreted with glucagon-like peptide-1 to modulate intestinal enterocytes and afferent neurons. A long-acting PYY analog mediates robust anorectic efficacy with minimal emesis in nonhuman primates. Lerche S, Brock B, Rungby J, Bøtker HE, Møller N, Rodell A, et al.

Glucagon-like peptide-1 inhibits blood-brain glucose transfer in humans. Sam AH, Gunner DJ, King A, Persaud SJ, Brooks L, Hostomska K, et al.

Selective ablation of peptide YY cells in adult mice reveals their role in beta cell survival. Schirra J, Nicolaus M, Roggel R, Katschinski M, Storr M, Woerle HJ, et al. Endogenous glucagon-like peptide 1 controls endocrine pancreatic secretion and antro-pyloro-duodenal motility in humans.

Hagve M, Gjessing PF, Hole MJ, Jansen KM, Fuskevag OM, Mollnes TE, et al. Perioperative infusion of glucagon like peptide-1 prevents insulin resistance after surgical trauma in female pigs. Cox HM, Tough IR, Woolston AM, Zhang L, Nguyen AD, Sainsbury A, et al.

Peptide YY is critical for acylethanolamine receptor Gprinduced activation of gastrointestinal mucosal responses. Byrne CS, Chambers ES, Alhabeeb H, Chhina N, Morrison DJ, Preston T, et al. Increased colonic propionate reduces anticipatory reward responses in the human striatum to high-energy foods.

Frost G, Sleeth ML, Sahuri-Arisoylu M, Lizarbe B, Cerdan S, Brody L, et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Jesus P, Ouelaa W, Francois M, Riachy L, Guerin C, Aziz M, et al.

Alteration of intestinal barrier function during activity-based anorexia in mice. Mörkl S, Lackner S, Meinitzer A, Mangge H, Lehofer M, Halwachs B, et al. Gut microbiota, dietary intakes and intestinal permeability reflected by serum zonulin in women. Eur J Nutr. Schroeder M, Jakovcevski M, Polacheck T, Drori Y, Luoni A, Roh S, et al.

Placental miR mediates vulnerability to activity based anorexia in mice. Cani PD, Possemiers S, Van de Wiele T, Guiot Y, Everard A, Rottier O, et al.

Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLPdriven improvement of gut permeability. Cani PD, Osto M, Geurts L and Everard A. Involvement of gut microbiota in the development of low-grade inflammation and type 2 diabetes associated with obesity.

Gut Microbes. Everard A, Lazarevic V, Derrien M, Girard M, Muccioli GG, Muccioli GM, et al. Responses of gut microbiota and glucose and lipid metabolism to prebiotics in genetic obese and diet-induced leptin-resistant mice.

Dominique M, Legrand R, Galmiche M, Azhar S, Deroissart C, Guérin C, et al. Changes in Microbiota and Bacterial Protein Caseinolytic Peptidase B During Food Restriction in Mice: Relevance for the Onset and Perpetuation of Anorexia Nervosa.

Cryan JF, O'Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, et al. The Microbiota-Gut-Brain Axis. Physiol Rev. Cao J, Peng J, An H, He Q, Boronina T, Guo S, et al.

Endotoxemia-mediated activation of acetyltransferase P impairs insulin signaling in obesity. Anitha M, Vijay-Kumar M, Sitaraman SV, Gewirtz AT and Srinivasan S. Gut microbial products regulate murine gastrointestinal motility via Toll-like receptor 4 signaling.

Jing Y, Yu Y, Bai F, Wang L, Yang D, Zhang C, et al. Effect of fecal microbiota transplantation on neurological restoration in a spinal cord injury mouse model: involvement of brain-gut axis.

Wanders AJ, van den Borne JJ, de Graaf C, Hulshof T, Jonathan MC, Kristensen M, et al. Effects of dietary fibre on subjective appetite, energy intake and body weight: a systematic review of randomized controlled trials.

Obes Rev. CAS PubMed Google Scholar. Serena C, Ceperuelo-Mallafré V, Keiran N, Queipo-Ortuño MI, Bernal R, Gomez-Huelgas R, et al. Elevated circulating levels of succinate in human obesity are linked to specific gut microbiota. Isme j. De Vadder F, Kovatcheva-Datchary P, Zitoun C, Duchampt A, Bäckhed F, Mithieux G.

Microbiota-produced succinate improves glucose homeostasis via intestinal gluconeogenesis. Mills EL, Pierce KA, Jedrychowski MP, Garrity R, Winther S, Vidoni S, et al.

Accumulation of succinate controls activation of adipose tissue thermogenesis. Wang K, Liao M, Zhou N, Bao L, Ma K, Zheng Z, et al. Parabacteroides distasonis alleviates obesity and metabolic dysfunctions via production of succinate and secondary bile acids. Cell Rep. Mithieux G.

A novel function of intestinal gluconeogenesis: central signaling in glucose and energy homeostasis. Soto M, Herzog C, Pacheco JA, Fujisaka S, Bullock K, Clish CB, et al. Gut microbiota modulate neurobehavior through changes in brain insulin sensitivity and metabolism.

Israelyan N, Del Colle A, Li Z, Park Y, Xing A, Jacobsen JPR, et al. Effects of serotonin and slow-release 5-hydroxytryptophan on gastrointestinal motility in a mouse model of depression.

Clarke G, Grenham S, Scully P, Fitzgerald P, Moloney RD, Shanahan F, et al. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner.

Bhattarai Y, Williams BB, Battaglioli EJ, Whitaker WR, Till L, Grover M, et al. Gut microbiota-produced tryptamine activates an epithelial G-protein-coupled receptor to increase colonic secretion. Cell Host Microbe. Gartner SN, Aidney F, Klockars A, Prosser C, Carpenter EA, Isgrove K, et al.

Intragastric preloads of l-tryptophan reduce ingestive behavior via oxytocinergic neural mechanisms in male mice. Miao J, Adewole D, Liu S, Xi P, Yang C, Yin Y.

Tryptophan supplementation increases reproduction performance, milk yield, and milk composition in lactating sows and growth performance of their piglets. J Agric Food Chem. Zhao Y, Wu XY, Xu SX, Xie JY, Xiang KW, Feng L, et al. Dietary tryptophan affects growth performance, digestive and absorptive enzyme activities, intestinal antioxidant capacity, and appetite and GH-IGF axis-related gene expression of hybrid catfish Pelteobagrus vachellifemale symbol x Leiocassis longirostrismale symbol.

Fish Physiol Biochem. Ullrich SS, Fitzgerald PCE, Giesbertz P, Steinert RE, Horowitz M, Feinle-Bisset C. Effects of intragastric administration of tryptophan on the blood glucose response to a nutrient drink and energy intake, in Lean and Obese Men. Steinert RE, Luscombe-Marsh ND, Little TJ, Standfield S, Otto B, Horowitz M, et al.

Effects of intraduodenal infusion of L-tryptophan on ad libitum eating, antropyloroduodenal motility, glycemia, insulinemia, and gut peptide secretion in healthy men. J Clin Endocrinol Metab. McVeay C, Fitzgerald PCE, Ullrich SS, Steinert RE, Horowitz M, Feinle-Bisset C.

Effects of intraduodenal administration of lauric acid and L-tryptophan, alone and combined, on gut hormones, pyloric pressures, and energy intake in healthy men. Ayaso R, Ghattas H, Abiad M, Obeid O. Meal pattern of male rats maintained on amino acid supplemented diets: the effect of tryptophan, lysine, arginine, proline and threonine.

Zapata RC, Singh A, Ajdari NM, Chelikani PK. Dietary tryptophan restriction dose-dependently modulates energy balance, gut hormones, and microbiota in obesity-prone rats. Obesity Silver Spring. Dodd D, Spitzer MH, Van Treuren W, Merrill BD, Hryckowian AJ, Higginbottom SK, et al.

A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Devlin AS, Marcobal A, Dodd D, Nayfach S, Plummer N, Meyer T, et al. Modulation of a circulating uremic solute via rational genetic manipulation of the gut microbiota.

Chimerel C, Emery E, Summers DK, Keyser U, Gribble FM, Reimann F. Bacterial metabolite indole modulates incretin secretion from intestinal enteroendocrine L cells. Scott SA, Fu J, Chang PV. Microbial tryptophan metabolites regulate gut barrier function via the aryl hydrocarbon receptor.

Wu Q, Clark MS, Palmiter RD. Deciphering a neuronal circuit that mediates appetite. Capello AE, Markus CR. Effect of sub chronic tryptophan supplementation on stress-induced cortisol and appetite in subjects differing in 5-HTTLPR genotype and trait neuroticism.

De Vadder F, Grasset E, Mannerås Holm L, Karsenty G, Macpherson AJ, Olofsson LE, et al. Gut microbiota regulates maturation of the adult enteric nervous system via enteric serotonin networks.

Yano JM, Yu K, Donaldson GP, Shastri GG, Ann P, Ma L, et al. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Reigstad CS, Salmonson CE, Rainey JF 3rd, Szurszewski JH, Linden DR, Sonnenburg JL, et al.

Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells.

Martin AM, Lumsden AL, Young RL, Jessup CF, Spencer NJ, Keating DJ. Regional differences in nutrient-induced secretion of gut serotonin. Physiol Rep. Ye L, Bae M, Cassilly CD, Jabba SV, Thorpe DW, Martin AM, et al. Enteroendocrine cells sense bacterial tryptophan catabolites to activate enteric and vagal neuronal pathways.

Williams BB, Van Benschoten AH, Cimermancic P, Donia MS, Zimmermann M, Taketani M, et al. Discovery and characterization of gut microbiota decarboxylases that can produce the neurotransmitter tryptamine.

Kopeikina E, Dukhinova M, Yung AWY, Veremeyko T, Kuznetsova IS, Lau TYB, et al. Platelets promote epileptic seizures by modulating brain serotonin level, enhancing neuronal electric activity, and contributing to neuroinflammation and oxidative stress.

Prog Neurobiol. Nectow AR, Schneeberger M, Zhang H, Field BC, Renier N, Azevedo E, et al. Identification of a brainstem circuit controlling feeding.

van Lingen M, Sidorova M, Alenina N, Klempin F. Lack of brain serotonin affects feeding and differentiation of newborn cells in the adult hypothalamus. Front Cell Dev Biol. D'Agostino G, Lyons D, Cristiano C, Lettieri M, Olarte-Sanchez C, Burke LK, et al.

Nucleus of the solitary tract serotonin 5-HT2C receptors modulate food intake. Crane JD, Palanivel R, Mottillo EP, Bujak AL, Wang H, Ford RJ, et al. Inhibiting peripheral serotonin synthesis reduces obesity and metabolic dysfunction by promoting brown adipose tissue thermogenesis.

Savastano DM, Covasa M. Intestinal nutrients elicit satiation through concomitant activation of CCK 1 and 5-HT 3 receptors.

Physiol Behav. 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. Rhoades JL, Nelson JC, Nwabudike I, Yu SK, McLachlan IG, Madan GK, et al. ASICs mediate food responses in an enteric serotonergic neuron that controls foraging behaviors.

Heisler LK, Jobst EE, Sutton GM, Zhou L, Borok E, Thornton-Jones Z, et al. Serotonin reciprocally regulates melanocortin neurons to modulate food intake. Valles-Colomer M, Falony G, Darzi Y, Tigchelaar EF, Wang J, Tito RY, et al.

The neuroactive potential of the human gut microbiota in quality of life and depression. Nat Microbiol. Yunes RA, Poluektova EU, Dyachkova MS, Klimina KM, Kovtun AS, Averina OV, et al.

Barrett E, Ross RP, O'Toole PW, Fitzgerald GF, Stanton C. gamma-Aminobutyric acid production by culturable bacteria from the human intestine. J Appl Microbiol. Liu R, Hong J, Xu X, Feng Q, Zhang D, Gu Y, et al.

Gut microbiome and serum metabolome alterations in obesity and after weight-loss intervention. Matsumoto M, Ooga T, Kibe R, Aiba Y, Koga Y and Benno Y. Colonic Absorption of Low-Molecular-Weight Metabolites Influenced by the Intestinal Microbiome: A Pilot Study.

PLoS One. Fujisaka S, Avila-Pacheco J, Soto M, Kostic A, Dreyfuss JM, Pan H, et al. Diet, Genetics, and the Gut Microbiome Drive Dynamic Changes in Plasma Metabolites.

Kootte RS, Levin E, Salojarvi J, Smits LP, Hartstra AV, Udayappan SD, et al. Improvement of Insulin Sensitivity after Lean Donor Feces in Metabolic Syndrome Is Driven by Baseline Intestinal Microbiota Composition.

Xu J, Bartolome CL, Low CS, Yi X, Chien C-H, Wang P, et al. For subjective assessment of appetite, participants reported their sensations of hunger, fullness, and desire to eat using visual analog scales VASs every 2 hours during the day and at 0, 30, 60, and minutes postprandial on intervention days during EB, CR, and OF.

A subsample of eight subjects additionally completed VASs on desire for something sweet, salty, and fatty at the same time points.

Higher ratings indicate greater experienced sensations. Total area under the curve was calculated for appetite hormones for 14 hours am to pm and for VASs for 15 hours am to pm using the trapezoidal rule Results on appetite hormones and VASs were therefore adjusted for differences in absolute intake of calories.

Blood samples were taken every 2 hours during the day from am to pm at EB, CR, and OF and 0, 30, 60, and minutes postprandially.

Samples were stored on ice after collection and then centrifuged immediately. Total ghrelin, total GLP-1, and insulin were measured using a Bio-Plex Pro TM human diabetes 3-plex kit Bio-Rad Laboratories, Hercules, CA containing antibody-conjugated microspheres.

Twenty-four hour urea excretion was determined using photometry Beckman Coulter, Brea, CA; AU Body weight was also measured at the beginning and end of each EB condition using a calibrated electronic scale Seca mBCA ; Seca.

All measurements were conducted in the fasting state and in underwear. Prior to the study start, which walking time and speed were needed to reach the predetermined PALs was tested. Medium and high ET were achieved through different treadmill protocols. On medium ET days, participants walked three times for 55 minutes 11 km on a treadmill Kettler Track 9 with software World Tours 2.

On high ET days, time on the treadmill was doubled to three times for minutes 22 km at the same speed. Besides the prescribed time on the treadmill participants remained sedentary sitting and lying but awake until bedtime. On the low ET days participants remained sedentary all of the time. SEE was measured as reported by Schrauwen et al.

Owing to technical failure, one subject had no valid data of physical activity for the whole intervention and two participants had missing data for one EB condition each.

Primary outcome variables for the present analysis are ghrelin, GLP-1, and insulin concentrations. Secondary outcomes are subjective appetite sensations VAS and EB during ad libitum EI. Faul, University of Kiel, Kiel, Germany for ghrelin concentration as the primary outcome variable.

King et al. The statistical software R was used to analyze the data. Data evaluation started with the definition of an appropriate statistical mixed model 24 , The data were assumed to be normally distributed and to be heteroscedastic with respect to the different levels of EB and ET.

These assumptions were based on a graphical residual analysis. The statistical model included EB ad libitum EI, EB, CR, OF and ET low, medium, high , as well as their interaction term as fixed factors. The subject identity was regarded as a random factor. Additionally, the correlations of the measurement values due to the several intervention days were taken into account autocorrelation.

Based on this model, a pseudo R 2 was calculated 26 and an ANOVA was conducted, followed by multiple contrast tests [ e. Taking this model into account, Pearson correlations were calculated for all levels of ET combined comparing daylong insulin and ghrelin.

Sixteen adults 3 women, 13 men aged 20 to 32 years According to World Health Organization criteria, one women and four men were overweight and one women was obese. Taking body composition into account, two women and two men had an FMI above the age- and sex-adjusted 95th percentile Components of EB are shown in Table 1.

Parameters differed between interventions according to study protocol. Results for daylong concentrations of appetite hormones are shown in Fig.

Comparison of daylong appetite hormone responses for ghrelin a , GLP-1 b , and insulin c as tAUC for 14 h between interventions with differing ET and differing EB. tAUC, total area under the curve.

Results for subjective appetite ratings are shown in Table 2. Comparison of Daylong Subjective Appetite Feelings From Visual Analog Scales All tAUCs Between Interventions With Differing ET and Differing EB. The current study confirms the hypothesis that appetite control is improved at higher levels of ET achieved by low-intensity physical activity.

The results are independent of EB and are verified by changes in endocrine signals, subjective appetite ratings, and ad libitum EI: ghrelin concentrations, hunger, and desire to eat were higher, GLP-1 concentrations were lower, and EB during ad libitum EI was positive with low ET compared with high ET.

In a prospective study by Hume et al. However, the study was criticized for not measuring real ET but simply examining baseline energy expenditure of weight-stable persons against long-term changes in body fat Nevertheless, the results by Hume et al.

are in line with the present randomized intervention study that carefully measured ET at different levels of EB in a whole-room calorimeter and that has demonstrated improved appetite control with increased ET. The present findings indicate a relationship between decreasing ghrelin levels in the observed concentration range and a reduced food consumption.

These findings are confirmed by a meal test trial where physiological ghrelin levels were closely correlated with hunger during the postprandial period and predicted the amount of subsequent food consumption Ghrelin concentrations are known to rise in the fasted state until meal initiation and to fall afterward proportional to the ingested energy content Reduced ghrelin concentrations with increasing ET could therefore be due to higher energy content of meals with increasing ET.

It was shown that stomach distention does not directly affect declining ghrelin concentrations 34 , indicating that the size or volume per se has no effect on postprandial ghrelin suppression. Because insulin is essential for postprandial ghrelin suppression 35 , the inverse correlation between insulin and ghrelin concentrations in the present trial supports an indirect anorexigenic effect of insulin via ghrelin suppression.

Because daylong insulin secretion decreased with higher ET [ Fig. The anorexigenic effect implied by high GLP-1 [ Fig. These effects were also found with CR and OF [ Fig. Beyond ET, the type of energy deficit might also impact appetite control. A similar effect in women was already reported earlier The current study confirms these findings despite a lower intensity of physical activity because a high ET achieved by brisk walking led to lower appetite and ad libitum EI compared with low ET.

Similarly, no compensatory increases in acylated ghrelin, appetite, or EI were observed after an acute brisk walking session The results of the present trial confirm these findings, as ghrelin was reduced and GLP-1 increased with high ET.

At a low ET, the macronutrient intake may have been too small and therefore not sufficient to stimulate GLP-1 secretion and ghrelin suppression.

A threshold effect could therefore explain the lack in satiety Table 2 and energy overconsumption during ad libitum EI Fig. This assumption is supported by the finding of higher prospective weight gain with snacking behavior frequent consumption of small amounts of food under free-living conditions In line with our findings, Stubbs et al.

In the current study, during ad libitum EI, EB was Assuming Therefore, an inactive lifestyle facilitates caloric overconsumption and thus weight gain. Low-intensity physical activity was chosen to increase the PAL, simulating daily activities without the effects of vigorous exercise on metabolism and avoiding fatigue or pain in persons with very low fitness.

Furthermore, low-intensity physical activity is considered to play an important role in promoting health and may be implemented more easily in public health strategies than high-intensity exercise In the current study, a PAL of 1.

The setting of a 9-m 2 metabolic chamber only allows physical activity on a treadmill and not manifold physical activities like housekeeping or taking the stairs as compared with free-living conditions. Under free-living conditions, the appropriate amount of steps per day would therefore be much lower.

Because the difference between a PAL of 1. As an outcome of the International Association for the Study of Obesity First Stock Conference, a consensus statement was published that confirmed the necessary PAL of 1.

Future studies need to investigate whether a higher intensity of physical activity at a lower duration is equally sufficient for optimal control of EB. Importantly, note that our study was conducted in mostly lean young subjects and a typical Western diet was used energy density 5.

Therefore, the PAL necessary to obtain EB may be different in subjects with obesity and a less energy-dense diet. The role that exercise can play in energy expenditure is commonly exaggerated Our findings show that the positive impact of physical activity on weight control is partially independent of burning up more calories and is additionally explained by an improvement in appetite sensations.

Furthermore, increasing physical activity may be easier for most people than eating less [for review, see 50 ]. Therefore, maintaining an equal EB due to achieving a high ET seems to be easier compared with obtain a low ET.

However, a high level of energy expenditure and thus a high ET may also be a risk factor for future weight gain when ET cannot be maintained in the long term. Thus, a gain in FM is observed with detraining in athletes 51 or postpregnancies Additionally, increasing ET by exercise in people with obesity led to a compensatory increase in EI that alleviated the beneficial effect of exercise on body weight The extensive study protocol in randomized crossover design and the highly standardized metabolic ward conditions are a strength of our study.

Nevertheless, there are also some limitations that should be addressed. We measured total ghrelin concentrations, which may mask relevant changes in acylated ghrelin that seems to be essential for appetite control However, it was found that the ratio of acylated and total ghrelin is relatively robust, rendering total ghrelin an appropriate surrogate Because only acute effects of changes in ET were investigated in the present trial, the results cannot be transferred to long-term habitual high or low ET.

In daily life, ET is highly variable from day to day. For example, brief periods where EI far exceeds energy expenditure last from one meal to several days and regularly occur over the weekend In line with our results, others have found that individuals with higher habitual PALs better adjust EI in response to EB perturbations compared with habitually inactive individuals 48 , However, long-term interventions are needed to confirm the present findings.

Because only three women were investigated in the current study, sex differences in appetite control could not be analyzed. The impact of sex on appetite control remains unclear, with some studies showing sex differences [for review, see 39 ] whereas others did not In conclusion, a high ET improves appetite control, reflected by changes in hormonal biomarkers, including increased GLP-1 and decreased ghrelin and insulin concentrations independent of EB.

These results indicate an asymmetric control of appetite where reduced energy expenditure was not compensated by an appropriate adaptation in EI.

In contrast to the prevailing concept of body weight control, the positive impact of physical activity is not completely explained by burning up more calories but, at certain volumes, also involves improving appetite control.

Importantly, for prevention of weight gain a PAL of 1. gov no. NCT registered 1 December designed research; F. conducted research; F. analyzed data; A. analyzed metabolic chamber data; F. and A. wrote the paper and had primary responsibility for the final content; M.

gave statistical advice and wrote R code; A. discussed the data; and all authors read and approved the final manuscript. Disclosure Summary: The authors have nothing to disclose. Edholm OG , Fletcher JG , Widdowson EM , McCance RA. The energy expenditure and food intake of individual men.

Br J Nutr. Google Scholar. Mayer J , Roy P , Mitra KP. Relation between caloric intake, body weight, and physical work: studies in an industrial male population in West Bengal. Am J Clin Nutr. Blundell JE , Gibbons C , Caudwell P , Finlayson G , Hopkins M.

Appetite control and energy balance: impact of exercise. Obes Rev. Stubbs RJ , Sepp A , Hughes DA , Johnstone AM , Horgan GW , King N , Blundell J.

The effect of graded levels of exercise on energy intake and balance in free-living men, consuming their normal diet. Eur J Clin Nutr. Westerterp KR. Physical activity, food intake, and body weight regulation: insights from doubly labeled water studies.

Nutr Rev. Myers A , Gibbons C , Finlayson G , Blundell J. Associations among sedentary and active behaviours, body fat and appetite dysregulation: investigating the myth of physical inactivity and obesity. Br J Sports Med. Unick JL , Lang W , Williams SE , Bond DS , Egan CM , Espeland MA , Wing RR , Tate DF ; SNAP Research Group.

Objectively-assessed physical activity and weight change in young adults: a randomized controlled trial. Search MIT. Search websites, locations, and people. Enter keywords to search for news articles: Submit.

Browse By. Breadcrumb MIT News A new player in appetite control. A new player in appetite control. Brain cells that provide structural support also influence feeding behavior, study shows. Anne Trafton MIT News Office. Publication Date :. Press Inquiries. Press Contact : Abby Abazorius.

Email: abbya mit. Phone: Caption : Scientists have identified two key groups of neurons within the hypothalamus that help regulate appetite. Caption :. Credits :. Turning on appetite It has long been known that the hypothalamus, an almond-sized structure located deep within the brain, controls appetite as well as energy expenditure, body temperature, and circadian rhythms including sleep cycles.

Share this news article on: X Facebook LinkedIn Reddit Print. STAT STAT reporter Meghana Keshavan speaks with Prof. Related Links Paper: "Direct modulation of GFAP-expressing glia in the arcuate nucleus bi-directionally regulates feeding.

Related Topics Research Brain and cognitive sciences McGovern Institute School of Science Biology Neuroscience Health Obesity.

By Brigham tfchnology Women's Hospital April 26, Blood glucose control have developed an ingestible Blood glucose control technollogy FLASH Fluid-wicking Active Stimulation Appettie Immune system boosters Improving immune system function that can electronically stimulate the tefhnology hormone ghrelin in pigs. Inspired by the fluid-wicking skin of the Australian thorny devil lizard, the capsule emits electronic signals and moves through the body, eventually being excreted. This proof-of-concept study demonstrates the potential of ingestible electroceuticals for treating gastrointestinal, neuropsychiatric, and metabolic disorders. The team plans to continue researching for human application and explore treatment options for eating disorders and metabolic diseases. Credit: Giancarlo Traverso GT Reel Productions. Suggestions Technoolgy feedback? Technokogy for download on the MIT News office website are made cojtrol to non-commercial controol, Immune system boosters and the general ccontrol under a Creative Commons Cholesterol level and overall well-being Non-Commercial No Derivatives license. You may not alter the technooogy provided, other than to crop them to size. A credit line must be used when reproducing images; if one is not provided below, credit the images to "MIT. Previous image Next image. MIT neuroscientists have discovered that brain cells called glial cells play a critical role in controlling appetite and feeding behavior. In a study of mice, the researchers found that activating these cells stimulates overeating, and that when the cells are suppressed, appetite is also suppressed.

Author: Tygomuro

5 thoughts on “Appetite control technology

  1. Ich meine, dass Sie sich irren. Es ich kann beweisen. Schreiben Sie mir in PM, wir werden besprechen.

  2. Absolut ist mit Ihnen einverstanden. Mir scheint es die ausgezeichnete Idee. Ich bin mit Ihnen einverstanden.

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com