Register      Login
Animal Production Science Animal Production Science Society
Food, fibre and pharmaceuticals from animals
REVIEW

Role of the gut, melanocortin system and malonyl-CoA in control of feed intake in non-ruminant animals

Frank R. Dunshea A D , Evan P. Bittner A , John R. Pluske B and John L. Black C
+ Author Affiliations
- Author Affiliations

A Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, Vic. 3010, Australia.

B School of Veterinary and Life Sciences, Murdoch University, Murdoch, WA 6150, Australia.

C John L. Black Consulting, Warrimoo, NSW 2774, Australia.

D Corresponding author. Email: fdunshea@unimelb.edu.au

Animal Production Science 58(4) 627-639 https://doi.org/10.1071/AN17273
Submitted: 1 May 2017  Accepted: 9 October 2017   Published: 7 February 2018

Abstract

Regulation of feed intake is under complex control, involving physical, chemical, hormonal and neuronal responses. Understanding the regulation of feed intake in farm animals is key to optimisation of intake to meet production and profitability goals. Fundamental mechanisms regulating feed intake include constraints imposed by the gut, systems monitoring current and long-term energy status to increase or decrease intake, and hedonic, reward-related drives. Feed intake is closely related to the rate of passage of digesta and the capacity of the gastrointestinal tract. Indigestible fibre increases the rate of digesta passage and feed intake until excess distension sends signals of satiety to the brain. The presence of partially digested nutrients and products of microbial fermentation in the distal intestines releases peptides (PYY, OXM, GPL-1, Apo A-IV, amylin) from gut and pancreas to activate the intestinal brake, which slows the rate of passage and reduces feed intake. These peptides also act on orexigenic (NPY, AgRP) and anorexigenic (POMC, CART) peptides of the melanocortin system of the hypothalamus to reduce intake over the long term. Immediate energy status of the animal is monitored through the ratio of AMP : ATP via adenosine monophosphate-activated kinase and mammalian target of rapamycin, whereas the overall animal energy status is monitored by insulin, leptin and ghrelin. These energy-monitoring systems control short- and long-term intakes through the melanocortin system of the hypothalamus, primarily via malonyl-CoA, to alter the relative expression of orexigenic and anorexigenic peptides. Gut and hypothalamic control of feed intake can be over-ridden by hedonic, reward-related centres of the brain, predominantly through the release of dopamine. These hedonic responses can lead to over-consumption and obesity under some circumstances or reduced feed intake under stressful or other negative environmental situations. Knowledge of these mechanisms can be used to identify practical strategies for either increasing or decreasing voluntary intake in pigs.

Additional keywords: ileal brake, lauric acid, particle size, resistant starch, soluble fibre.


References

Al-Rabadi GJ, Gilbert RG, Gidley MJ (2009) Effect of particle size on kinetics of starch digestion in milled barley and sorghum grains by porcine alpha-amylase. Journal of Cereal Science 50, 198–204.
Effect of particle size on kinetics of starch digestion in milled barley and sorghum grains by porcine alpha-amylase.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXhtVKlsr3O&md5=f3034196c5f8f91ea205da8ccfe508c8CAS |

Alonso-Alonso M, Woods SC, Pelchat M, Grigson PS, Stice E, Farooqi S, Khoo CS, Mattes RD, Beauchamp GK (2015) Food reward system: current perspectives and future research needs. Nutrition Reviews 73, 296–307.
Food reward system: current perspectives and future research needs.Crossref | GoogleScholarGoogle Scholar |

Anderson GH, Moore SE (2004) Dietary proteins in the regulation of food intake and body weight in humans. The American Society for Nutritional Sciences 134, 974S–979S.

Barb CR, Yan Z, Azain MJ, Kraeling RR, Rampacek GB, Ramsay TG (1998) Recombinant porcine leptin reduces feed intake and stimulates growth hormone secretion in swine. Domestic Animal Endocrinology 15, 77–86.
Recombinant porcine leptin reduces feed intake and stimulates growth hormone secretion in swine.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1cXktlaquw%3D%3D&md5=c24afa3b4866a50f26e29d3fa52cf12cCAS |

Barb CR, Robertson AS, Barnett JB, Kraeling RR, Houseknecht KL (2004) The role of elanocortin-3 and -4 receptor in regulating appetite, energy homeostasis and neuroendocrine function in the pig. The Journal of Endocrinology 181, 39–52.
The role of elanocortin-3 and -4 receptor in regulating appetite, energy homeostasis and neuroendocrine function in the pig.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXjvVaqsr0%3D&md5=91d807752966bbfe188d80dde5b997b4CAS |

Berthoud H-R (2011) Metabolic and hedonic drives in the neural control of appetite: Who’s the boss? Current Opinion in Neurobiology 21, 888–896.
Metabolic and hedonic drives in the neural control of appetite: Who’s the boss?Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XlsFSltA%3D%3D&md5=3ae8d1aa3f22447c4004ebc8681b3608CAS |

Black JL (1983) Growth and development of lambs. In ‘Sheep production’. (Ed. W Haresign) pp. 21–58. (Butterworths: London)

Black JL (2009) Models to predict feed intake. In ‘Voluntary feed intake in pigs’. (Eds D Torrallardona, E Roura) pp. 323–351. (Wageningen Academic Publishers: Wageningen, The Netherlands)

Black JL, Campbell RG, Williams IH, James KJ, Davies GT (1986) Simulation of energy and amino acid utilization in the pig. Research and Development in Agriculture 3, 121–145.

Black JL, Giles LR, Wynn PC, Knowles AG, Kerr CA, Jones MR, Strom AD, Gallagher NL, Eamens GJ (2001). Factors limiting the performance of growing pigs in commercial environments. In ‘Manipulating pig production VIII’. (Ed. PD Cranwell) pp. 9–36. (Australasian Pig Science Association: Melbourne)

Black JL, Williams BA, Gidley MJ (2009) Metabolic regulation of feed intake in monogastric animals. In ‘Voluntary feed intake in pigs’. (Eds D Torrallardona, E Roura) pp. 189–213. (Wageningen Academic Press: Wageningen, The Netherlands)

Black JL, Williams BA, Roura E, Gidley MJ (2011) Physiological and metabolic regulation of feed intake. In ‘Manipulating pig production XIII’. (Ed. RJ van Barneveld) pp. 118–127. (Australasian Pig Science Association: Melbourne)

Boutry C, El-Kadi SW, Suryawan A, Steinhoff-Wagner J, Stoll B, Orellana RA, Nguyen HV, Kimball SR, Fiorotto ML, Davis TA (2016) Pulsatile delivery of a leucine supplement during long-term continuous enteral feeding enhances lean growth in term neonatal pigs. American Journal of Physiology. Endocrinology and Metabolism 310, E699–E713.

Burton-Freeman B, Schneeman BO (1996) Lipid infused into the duodenum at varied rates influences food intake and body weight gain. The Journal of Nutrition 126, 2934–2939.

Campbell R, Taverner M, Mullaney P (1975) The effect of dietary concentrations of digestible energy on the performance and carcass characteristics of early-weaned pigs. Animal Science 21, 285–294.

Cave NAG (1982) Effect of dietary short- and medium-chain fatty acids on feed intake in chicks. Poultry Science 61, 1147–1153.
Effect of dietary short- and medium-chain fatty acids on feed intake in chicks.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaL38Xkt1als7c%3D&md5=5ae3963833be651789123e7721820151CAS |

Celi P, Cowieson AJ, Fru-Nji F, Steinert RE, Kluenter A-M, Verlhac V (2017) Gastrointestinal functionality in animal nutrition and health: new opportunities for sustainable animal production. Animal Feed Science and Technology 234, 88–100.
Gastrointestinal functionality in animal nutrition and health: new opportunities for sustainable animal production.Crossref | GoogleScholarGoogle Scholar |

Cha SH, Hu Z, Lane MD (2004) Long-term effects of a fatty acid synthase inhibitor on obese mice: food intake, hypothalamic neuropeptides, and UCP3. Biochemical and Biophysical Research Communications 317, 301–308.
Long-term effects of a fatty acid synthase inhibitor on obese mice: food intake, hypothalamic neuropeptides, and UCP3.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXivVCrtro%3D&md5=ed2cb4bc32efe3fea714addf8eecf3f3CAS |

Chaudhri O, Small C, Bloom S (2006) Gastrointestinal hormones regulating appetite. Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences 361, 1187–1209.
Gastrointestinal hormones regulating appetite.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XhtVygtL7M&md5=f484a2b0ac6aafc38991e74e0532ef77CAS |

Chaudhri OB, Salem V, Murphy KG, Bloom SR (2008) Gastrointestinal satiety signals. Annual Review of Physiology 70, 239–255.
Gastrointestinal satiety signals.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXkt1eqtrs%3D&md5=b98d0fea93669eb2130db417e62c4359CAS |

Chelikani PK, Keisler DH, Kennelly JJ (2003) Response of plasma leptin concentration to jugular infusion of glucose or lipid is dependent on the stage of lactation of Holstein cows. The Journal of Nutrition 133, 4163–4171.

Clement K, Vaisse C, Lahlou N, Cabrol S, Pelloux V, Cassuto D, Gourmelen M, Dina C, Chambaz J, Lacorte JM, Basdevant A, Bougneres P, Lebouc Y, Froguel P, Guy-Crand B (1998) A mutation in the human leptin receptor gene causes obesity and pituitary dysfunction. Nature 392, 398–401.
A mutation in the human leptin receptor gene causes obesity and pituitary dysfunction.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1cXitlShtrs%3D&md5=11c96c0d78ec1152096f8b8c60c2d1d9CAS |

Clouard C Jouhanneau M Meunier-Salaün M-C Malbert C-H Val-Lailet D 2012 Exposure to conditioned flavours with different hedonic values induce contrasted behavioural and brain responses in pigs. Plos One 7 e37968 https://doi.org/10.1371/journal.pone.0037968

Cota D, Proulx K, Blake Smith KA, Kozma SC, Thomas G, Woods SC, Seeley RJ (2006) Hypothalamic mTOR signaling regulates food intake. Science 312, 927–930.
Hypothalamic mTOR signaling regulates food intake.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28Xktlyhs7Y%3D&md5=c20742e2a2ba4eaa85c99dc1f49bc897CAS |

Cox JE, Tyler WJ, Randich A, Kelm GR, Bharaj SS, Jandacek RJ, Meller ST (2000) Suppression of food intake, body weight, and body fat by jejunal fatty acid infusions. The American Journal of Physiology 278, R604–R610.

Crowe TC, Seligman SA, Copeland L (2000) Inhibition of enzymic digestion of amylose by free fatty acids in vitro contributes to resistant starch formation. The Journal of Nutrition 130, 2006–2008.

Cummings DE (2006) Ghrelin and the short- and long-term regulation of appetite and body weight. Physiology & Behavior 89, 71–84.
Ghrelin and the short- and long-term regulation of appetite and body weight.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XotFyktL4%3D&md5=60c59d1fd22a0efacca2757bb910b3b1CAS |

De Silva A, Bloom SR (2012) Gut hormones and appetite control: a focus on PYY and GLP-1 as therapeutic targets in obesity. Gut and Liver 6, 10–20.
Gut hormones and appetite control: a focus on PYY and GLP-1 as therapeutic targets in obesity.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38Xmt1ylsLo%3D&md5=d824abba4f9e0a6bcfa1e51772844b81CAS |

Delporte C (2013) Structure and physiological actions of ghrelin. Scientifica
Structure and physiological actions of ghrelin.Crossref | GoogleScholarGoogle Scholar |

Druce MR, Wren AM, Park AJ, Milton JE, Patterson M, Frost G, Ghatei MA, Small C, Bloom SR (2005) Ghrelin increases food intake in obese as well as lean subjects. International Journal of Obesity 29, 1130–1136.
Ghrelin increases food intake in obese as well as lean subjects.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXnt1Sht7s%3D&md5=aa5b86c99ade50733810f82e658701d3CAS |

Dunshea FR, Gannon NJ, van Barneveld RJ, Mullan BP, Campbell RG, King RH (2001) Dietary lupins (Lupinus angustifolius and Lupinus albus) can increase digesta retention in the gastrointestinal tract of pigs. Australian Journal of Agricultural Research 52, 593–602.
Dietary lupins (Lupinus angustifolius and Lupinus albus) can increase digesta retention in the gastrointestinal tract of pigs.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXktlCktLw%3D&md5=0de8e2420cd318d582db45e4c07548b8CAS |

Dunshea FR, Ostrowska E, Ferrari JM, Gill HS (2007) Dairy proteins and the control of satiety and obesity. Australian Journal of Experimental Agriculture 47, 1051–1058.
Dairy proteins and the control of satiety and obesity.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXos1Kgur8%3D&md5=988fb9a584b3388503ca3c117c92470cCAS |

Edmonds MS, Baker DH (1987) Amino acid excesses for young pigs: effects of excess methionine, tryptophan, threonine or leucine. Journal of Animal Science 64, 1664–1671.
Amino acid excesses for young pigs: effects of excess methionine, tryptophan, threonine or leucine.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaL2sXkslKlt74%3D&md5=267fa30887fdba137ee01322ee4f5e86CAS |

Emmans GC (1989) The growth of turkeys. In ‘Recent advances in turkey science’. (Eds C Nixey, TC Grey) pp. 135–166. (Butterworths: London)

Epstein LH, Temple JL, Roemmich JN, Bouton ME (2009) Habituation as a determinant of food intake. Physiological Reviews 116, 384–407.

Farooqi IS, Jebb SA, Langmack G, Lawrence E, Cheetham CH, Prentice AM, Hughes IA, McCamish MA, O’Rahilly S (1999) Effects of recombinant leptin therapy in a child with congenital leptin deficiency. The New England Journal of Medicine 341, 879–884.
Effects of recombinant leptin therapy in a child with congenital leptin deficiency.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK1MvgsVyrtA%3D%3D&md5=42d220e4a31650811c4870a303a8fae2CAS |

Faulconbridge LF, Cummings DE, Kap-lan JM, Grill HJ (2003) Hyperphagic effects of brainstem ghrelin administration. Diabetes 52, 2260–2265.
Hyperphagic effects of brainstem ghrelin administration.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXntFeqtbw%3D&md5=94cc1c5b8fc3374f03826e73aead387fCAS |

Feltrin KL, Little TJ, Meyer JH, Horowitz M, Smout AJPM, Wishart J, Pilichiewicz AN, Rades T, Chapman IM, Feinle-Bisset C (2004) Effects of intraduodenal fatty acids on appetite, antropyloroduodenal motility, and plasma CCK and GLP-1 in humans vary with their chain length. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology 287, R524–R533.
Effects of intraduodenal fatty acids on appetite, antropyloroduodenal motility, and plasma CCK and GLP-1 in humans vary with their chain length.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXnvVaqtL4%3D&md5=0ecd2d17c65db44252747675b3f0e0cdCAS |

Feltrin KL, Little TJ, Meyer JH, Horowitz M, Rades T, Wishart J, Feinle-Bisset C (2007) Effects of lauric acid n upper gut motility, plasma cholecystokinin and peptide YY, and energy intake are load, but not concentration dependent in humans. The Journal of Physiology 581, 767–777.
Effects of lauric acid n upper gut motility, plasma cholecystokinin and peptide YY, and energy intake are load, but not concentration dependent in humans.Crossref | GoogleScholarGoogle Scholar |

Figlewicz DP (2003) Adiposity signals and food reward: expanding the CNS roles of insulin and leptin. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology 284, R882–R892.
Adiposity signals and food reward: expanding the CNS roles of insulin and leptin.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXjtlyks7o%3D&md5=73d819caffa6d884185526fccaa709ffCAS |

Figueroa J, Solâ-Oriol D, Manteca X, Pérez JF, Dwyer DM (2015) Anhedonia in pigs? Effects of social and restraint stress on sucrose preference. Physiology & Behavior 151, 509–515.
Anhedonia in pigs? Effects of social and restraint stress on sucrose preference.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2MXhsVens7vF&md5=737196f33cf81980101e546d0dd0c6b7CAS |

Flannery BM, Clark ES, Pestka JJ (2012) Anorexia induced by the trichothecene deoxynivalenol (vomitoxin) is mediated by the release of the gut satiety hormone peptide YY. Toxicological Sciences 130, 289–297.
Anorexia induced by the trichothecene deoxynivalenol (vomitoxin) is mediated by the release of the gut satiety hormone peptide YY.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38Xhs12ltb3K&md5=2790966cbaaa4a2ac6edc8e7567c0811CAS |

Fleming SE, Lee B (1983) Growth performance and intestinal transit time of rats fed purified and natural fibers. The Journal of Nutrition 113, 592–601.

Furness JB Cottrell JJ 2017 Signalling from the gut lumen. Animal Production Science 57 2175 2187 10.1071/AN17276

Gao S, Howard S, LoGrasso PV (2017) Pharmacological inhibition of c-Jun N-terminal kinase reduces food intake and sensitizes leptin’s anorectic signaling actions. Scientific Reports 7,
Pharmacological inhibition of c-Jun N-terminal kinase reduces food intake and sensitizes leptin’s anorectic signaling actions.Crossref | GoogleScholarGoogle Scholar |

Geliebter A (1988) Gastric distension and gastric capacity in relation to food intake in humans. Physiology & Behavior 44, 665–668.
Gastric distension and gastric capacity in relation to food intake in humans.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaL1M7osFSitw%3D%3D&md5=c22634b4e67da53263b8a1f1253e29b7CAS |

Giles LR, Lorschy ML, Bray HJ, Black JL (1998) Predicting feed intake in growing pigs. In ‘Progress in pig science’. (Eds J Wiseman, MA Varley, JP Chadwick) pp. 209–228. (Nottingham University Press: Nottingham, UK)

Gloaguen M, Le Floc’h N, Corrent E, Primot Y, van Milgen J (2012) Providing a diet deficient in valine but with excess leucine results in a rapid decrease in feed intake and modifies the postprandial plasma amino acid and α-keto acid concentrations in pigs. Journal of Animal Science 90, 3135–3142.
Providing a diet deficient in valine but with excess leucine results in a rapid decrease in feed intake and modifies the postprandial plasma amino acid and α-keto acid concentrations in pigs.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XhsVKku7nO&md5=1e8fbdd95bc46bbc7076c855cb4fb736CAS |

Goldstone AP, Prechtl de Hernandez CG, Beaver JD, Muhammed K, Croese C, Bell G, Durighel G, Hughes E, Waldman AD, Frost G, Bell JD (2009) Fasting biases brain reward systems towards high-calorie foods. The European Journal of Neuroscience 30, 1625–1635.
Fasting biases brain reward systems towards high-calorie foods.Crossref | GoogleScholarGoogle Scholar |

Gous RM (2007) Methodologies for modelling energy and amino acid responses in poultry. Revista Brasileira de Zootecnia 36, 263–275.
Methodologies for modelling energy and amino acid responses in poultry.Crossref | GoogleScholarGoogle Scholar |

Gregory PC, Rayner DV (1987) The influence of gastrointestinal infusion of fats on regulation of food intake in pigs. The Journal of Physiology 385, 471–481.
The influence of gastrointestinal infusion of fats on regulation of food intake in pigs.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaL2sXhvFGhsr8%3D&md5=8ed061c8255c5f5bd43926502f9963bbCAS |

Haissaguerre M, Saucisse N, Cota D (2014) Influence of mTOR in energy and metabolic homeostasis. Molecular and Cellular Endocrinology 397, 67–77.
Influence of mTOR in energy and metabolic homeostasis.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXhtlGnsLfK&md5=60a7329d9c1289e4bfee4b98f0fba2ecCAS |

Hardie DG (2016) Regulation of AMP-activated kinase by natural and synthetic activators. Acta Pharmaceutica Sinica. B
Regulation of AMP-activated kinase by natural and synthetic activators.Crossref | GoogleScholarGoogle Scholar |

Hooda S, Metzler-Zebeli BU, Vasabthan T, Zijlstra RT (2011) Effects of viscosity and fermentability of dietary fibre on nutrient digestibility and digesta characteristics in ileal-cannulated grower pigs. British Journal of Nutrition 106, 664–674.
Effects of viscosity and fermentability of dietary fibre on nutrient digestibility and digesta characteristics in ileal-cannulated grower pigs.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXht1SrsL7I&md5=c8179dcfc03a9a765edc79cd77e6061aCAS |

Horvath TL, Sarman B, Garcia-Cáceres C, Enriori PJ, Sotonyi P, Shanabrough M, Borok E, Argente J, Chowen JA, Perez-Tilve D, Pfluger PT, Brönneke HS, Levin BE, Diano S, Cowley MA, Tschöp MH (2010) Synaptic input organization of the melanocortin system predicts diet-induced hypothalamic reactive gliosis and obesity. Proceedings of the National Academy of Sciences, USA 107, 14875–14880.
Synaptic input organization of the melanocortin system predicts diet-induced hypothalamic reactive gliosis and obesity.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXhtVKqu7zI&md5=1ceb54f061e0648bfce57f6f5d5d31b3CAS |

Howick K, Griffin BT, Cryan JF, Schellekens H (2017) From belly to brain: targeting the ghrelin receptor in appetite and food intake regulation. International Journal of Molecular Sciences 18, 273
From belly to brain: targeting the ghrelin receptor in appetite and food intake regulation.Crossref | GoogleScholarGoogle Scholar |

Hu Z, Cha SH, Chohnan S, Lane MD (2003) Hypothalamic malonyl–CoA as a mediator of feeding behaviour. Proceedings of the National Academy of Sciences, USA 100, 12624–12629.
Hypothalamic malonyl–CoA as a mediator of feeding behaviour.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXoslKntrs%3D&md5=51b310f8133f46926430ddf10467e8bcCAS |

Huge A, Weber E, Ehrlein H-J (1995) Effects of central feedback inhibition on motility, luminal flow, and absorption of nutrients in proximal gut of minipigs. Digestive Diseases and Sciences 40, 1024–1034.
Effects of central feedback inhibition on motility, luminal flow, and absorption of nutrients in proximal gut of minipigs.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK2M3ks1GmsA%3D%3D&md5=8746ab99395223e3a65ea2f3b4fcec9bCAS |

Huynh MKQ, Kinyua AW, Yang DJ, Kim KW (2016) Hypothalmic AMPK as a regulator of energy homeostasis. Neural Plasticity
Hypothalmic AMPK as a regulator of energy homeostasis.Crossref | GoogleScholarGoogle Scholar |

Ito T, Thidarmyint H, Murata T, Inoue H, Neyra RM, Kuwayama H (2006) Effects of peripheral administration of PYY3–36 on feed intake and plasma acyl-ghrelin levels in pigs. The Journal of Endocrinology 191, 113–119.
Effects of peripheral administration of PYY3–36 on feed intake and plasma acyl-ghrelin levels in pigs.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28Xht1eju7nJ&md5=dcaa6651bafd3e39011104a9a73bf064CAS |

Kask A, Rago L, Mutulis F, Pahkla R, Wikberg JES, Schioth HB (1998) Selective antagonist for melanocortin receptor 4 receptor (HSO14) increases food intake in free-feeding rats. Biochemical and Biophysical Research Communications 245, 90–93.
Selective antagonist for melanocortin receptor 4 receptor (HSO14) increases food intake in free-feeding rats.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1cXisVWls78%3D&md5=9e73bde2ce7c12f65be45de7bca8e2a2CAS |

Kendall GPN, Thompson DG, Day SJ (1987) Motor responses of the small intestines to intraluminal distension in normal volunteers and a patient with visceral neuropathy. Gut 28, 714–720.
Motor responses of the small intestines to intraluminal distension in normal volunteers and a patient with visceral neuropathy.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaL2szgsFOmtA%3D%3D&md5=47c4163626ab186dd310fc8995a08effCAS |

Kim E-K, Miller I, Aja S, Landree LE, Pinn M, McFadden J, Kuhajda FP, Moran TH, Ronnett GV (2004) C75, a fatty acid synthase inhibitor, reduces food intake via hypothalamic AMP-activated protein kinase. The Journal of Biological Chemistry 279, 19970–19976.
C75, a fatty acid synthase inhibitor, reduces food intake via hypothalamic AMP-activated protein kinase.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXjs1Whsrg%3D&md5=9807d2f00a6e1c018d982adc2f1ee7ceCAS |

Koch M, Horvath TL (2014) Molecular and cellular regulation of hypothalamic melanocortin neurons controlling food intake and energy metabolism. Molecular Psychiatry 19, 752–761.
Molecular and cellular regulation of hypothalamic melanocortin neurons controlling food intake and energy metabolism.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXmsVKktL4%3D&md5=16b59140bdd55dead000633fdbd60f2dCAS |

Kola B, Hubina E, Tucci SA, Kirkham TC, Garcia EA, Mitchell SE, Williams LM, Hawley SA, Hardie DG, Grossman AB, Korbonits M (2005) Cannabinoids and ghrelin have central and peripheral metabolic and cardiac effects via AMP-activated protein kinase. The Journal of Biological Chemistry 280, 25196–25201.
Cannabinoids and ghrelin have central and peripheral metabolic and cardiac effects via AMP-activated protein kinase.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXlsFyqtL8%3D&md5=0de2fe79f3d5f1460abd101370f6d573CAS |

Korek E, Krauss H, Gibas-Dorna M, Kupsz J, Piatek M, Piatek J (2013) Fasting and postprandial levels of ghrelin, leptin and insulin in lean, obese and anorexic subjects. Przegląd Gastroenterologiczny 8, 383–389.
Fasting and postprandial levels of ghrelin, leptin and insulin in lean, obese and anorexic subjects.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXpt12msbw%3D&md5=5bc906d9e478fb27a7fb1fef4c7b7e6bCAS |

Krügel U, Schraft T, Kittner H, Kiess W, Illes P (2003) Basal and feeding-evoked dopamine release in the rat nucleus accumbens is depressed by leptin. European Journal of Pharmacology 482, 185–187.
Basal and feeding-evoked dopamine release in the rat nucleus accumbens is depressed by leptin.Crossref | GoogleScholarGoogle Scholar |

Kyriazakis I, Emmans GC (1998) Voluntary feed intake and diet selection. In ‘A quantitative biology of the pig’. (Ed. I Kyriazakis) pp. 229–248. (CAB International: Wallingford, UK)

Kyriazakis I, Emmans GC (1995) The voluntary feed intake of pigs given feeds based on wheat bran, dried citrus pulp and grass meal in relation to measurements of feed bulk. British Journal of Nutrition 73, 191–207.
The voluntary feed intake of pigs given feeds based on wheat bran, dried citrus pulp and grass meal in relation to measurements of feed bulk.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2MXktFGltL4%3D&md5=2d05b4f082990c7e94646a84472f325fCAS |

Layman DK, Baum JI (2004) Dietary protein impact on glycemic control during weight loss. The Journal of Nutrition 134, 968S–973S.

Le Goff G, van Milgen J, Noblet J (2002) Influence of dietary fibre on digestive utilization and rate of passage in growing pigs, finisher pigs and adult sows. Animal Science 74, 303–315.

Lee B-H, Bello-Pérez LA, Lin AH-M, Kim CY, Hamaker BR (2013) Importance of location of digestion and colonic fermentation of starch related to its quality. Cereal Chemistry 90, 335–343.
Importance of location of digestion and colonic fermentation of starch related to its quality.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXhtlClsrzF&md5=14acf9601ae851a24f9b913bdd54c5f7CAS |

Leinninger GM, Jo YH, Leshan RL, Louis GW, Yang H, Barrera JG, Wilson H, Opland DM, Faouzi MA, Gong Y, Jones JC, Rhodes CJ, Chua S, Diano S, Horvath TL, Seeley RJ, Becker JB, Münzberg H, Myers MG (2009) Leptin acts via leptin receptor-expressing lateral hypothalamic neurons to modulate the mesolimbic dopamine system and suppress feeding. Cell Metabolism 10, 89–98.
Leptin acts via leptin receptor-expressing lateral hypothalamic neurons to modulate the mesolimbic dopamine system and suppress feeding.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXhsVChsLrP&md5=296abd5996f21986b69f0e15629bd83fCAS |

Lin HC, Perdomo OL, Zhao XT (2001) Intestinal transit in dogs is accelerated by volume distension during fat-induced jejuna brake. Digestive Diseases and Sciences 46, 19–23.
Intestinal transit in dogs is accelerated by volume distension during fat-induced jejuna brake.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD3M7nsFOgtw%3D%3D&md5=0b77d01b0a0c2c4911961d3543bf75eeCAS |

Luick BR, Penner MH (1991) Nominal response of passage rates of fiber to particle size in rats. The Journal of Nutrition 121, 1940–1947.

Maljaars J, Peters HPF, Masclee AM (2007) Review article: the gastrointestinal tract: neuroendocrine regulation of satiety and food intake. Alimentary Pharmacology & Therapeutics 26, 241–250.
Review article: the gastrointestinal tract: neuroendocrine regulation of satiety and food intake.Crossref | GoogleScholarGoogle Scholar |

Maljaars PWJ, Peters HPF, Mela DJ, Masclee AAM (2008) Ileal brake: a sensible food target for appetite control. A review. Physiology & Behavior 95, 271–281.
Ileal brake: a sensible food target for appetite control. A review.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXhtFOlsL%2FE&md5=ef658665b48ef2fcbdc44839b7e7ff6aCAS |

Maljaars PWJ, Romeyn EA, Haddeman E, Peters HPF, Masclee AAM (2009) Effect of fat saturation on satiety, hormone release, and food intake. The American Journal of Clinical Nutrition 89, 1019–1024.
Effect of fat saturation on satiety, hormone release, and food intake.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXjslSksrk%3D&md5=117bea94588f5a2f530d719c66815dafCAS |

Marić G, Gazibara T, Zaletel I, Labudovic Borovic M, Tomanovic N, Ciric M, Puskas N (2014) The role of gut hormones in appetite regulation. Acta Physiologica Hungarica 101, 395–407.
The role of gut hormones in appetite regulation.Crossref | GoogleScholarGoogle Scholar |

Martinez-Miró S, Tecles F, Ramón M, Escribano D, Hernández F, Madrid J, Orengo J, Martinez-Subiela S, Manteca X, Cerón JJ (2016) Causes, consequences and biomarkers of stress in swine: an update. BioMedical Central Veterinary Research 12, 171
Causes, consequences and biomarkers of stress in swine: an update.Crossref | GoogleScholarGoogle Scholar |

Mateos GG, Martin F, Latorre MA, Vicente B, Lazaro R (2007) Inclusion of oat hulls in diets of young pigs based on cooked maize or cooked rice. Animal Science 82, 57–63.

Meyer JH, Hlinka M, Khatibi A, Raybould HE, Tso P (1998a) Role of small intestine caloric compensations to oil premeals in rats. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology 275, R1320–R1333.

Meyer JH, Hlinka M, Tabrizi Y, DiMaso N, Raybould HE (1998b) Chemical specificities and intestinal distributions of nutrient-driven satiety. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology 275, R1293–R1307.

Moore K, Mullan B, Kim JC, Dunshea F (2016) The effect of Lupinus albus and calcium chloride on growth performance, body composition, plasma biochemistry and meat quality of male pigs immunized against gonadotrophin releasing factor. Animals (Basel) 6, 78
The effect of Lupinus albus and calcium chloride on growth performance, body composition, plasma biochemistry and meat quality of male pigs immunized against gonadotrophin releasing factor.Crossref | GoogleScholarGoogle Scholar |

Moore K, Mullan B, Kim JC, Dunshea F (2017) The effect of Lupinus albus on growth performance, body composition and satiety hormones of male pigs immunized against gonadotrophin releasing hormone. Animals (Basel) 7, 15
The effect of Lupinus albus on growth performance, body composition and satiety hormones of male pigs immunized against gonadotrophin releasing hormone.Crossref | GoogleScholarGoogle Scholar |

Morgan K, Obici S, Rossetti L (2004) Hypothalmic responses to long-chain fatty acids are nutritionally regulated. The Journal of Biological Chemistry 279, 31139–31148.
Hypothalmic responses to long-chain fatty acids are nutritionally regulated.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXlslOkurY%3D&md5=29e0c3a0107813cf7b098326afb18868CAS |

Nuzzaci D, Laderriére A, Lwmoine A, Nédélec E, Pénicaud L, Rigault C, Benani A (2015) Plasticity of the melanocortin system: determinants and possible consequences on food intake. Frontiers in Endocrinology 6, 143–156.
Plasticity of the melanocortin system: determinants and possible consequences on food intake.Crossref | GoogleScholarGoogle Scholar |

Ohno T, Mochiki E, Kuwano H (2010) The roles of motilin and ghrelin in gastrointestinal motility. International Journal of Peptides
The roles of motilin and ghrelin in gastrointestinal motility.Crossref | GoogleScholarGoogle Scholar |

Owusu-Asiedu A, Patience JF, Laarveld B, Van Kessel AG, Simmins PH, Zijlstra RT (2006) Effects of guar gum and cellulose on digesta passage rate, ileal microbial populations, energy and protein digestibility, and performance of grower pigs. Journal of Animal Science 84, 843–852.
Effects of guar gum and cellulose on digesta passage rate, ileal microbial populations, energy and protein digestibility, and performance of grower pigs.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XjtlKktr0%3D&md5=462dc0084c60474082c036612553319fCAS |

Paz-Filho G, Mastronard C, Delibasi T, Wong M-L, Licinio J (2010) Congenital leptin deficiency: diagnosis and effects of leptin replacement therapy. Arquivos Brasileiros de Endocrinologia & Metabologia 54, 690–697.
Congenital leptin deficiency: diagnosis and effects of leptin replacement therapy.Crossref | GoogleScholarGoogle Scholar |

Perry B Wang Y 2012 Appetite regulation and weight control: the role of gut hormones. Nutrition and Diabetics 2 e26 10.1038/nutd.2011.21

Pluske JR, Black JL, Kim JC, Dunshea FR (2015) Suppressing feed intake of finisher pigs: a preliminary study. Animal Production Science 55, 1546

Potkins ZV, Lawrence TLJ, Thomlinson JR (1991) Effects of structural and non-structural polysaccharides in the diet of the growing pig on gastric emptying rate and rate of passage of digesta to the terminal ileum and through the total gastrointestinal tract. British Journal of Nutrition 65, 391–413.
Effects of structural and non-structural polysaccharides in the diet of the growing pig on gastric emptying rate and rate of passage of digesta to the terminal ileum and through the total gastrointestinal tract.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK3MXltFentr0%3D&md5=3cb430cb564a1eb8cb7d94c3f8f9ad4dCAS |

Qin X, Tso P (2005) The role of apolipoprotein AIV on the control of food intake. Current Drug Targets 6, 145–151.
The role of apolipoprotein AIV on the control of food intake.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXisVSqurs%3D&md5=33fc156c720c11cf56fa73ebcac4bbacCAS |

Ratanpaul V, Zhang D, Diffey S, Black JL, Gidley MJ, Williams BA (2017a) Effects of different amounts of wheat bran and oat hulls on production of short chain fatty acids in the hindgut of pigs. Animal Production Science 57, 2413
Effects of different amounts of wheat bran and oat hulls on production of short chain fatty acids in the hindgut of pigs.Crossref | GoogleScholarGoogle Scholar |

Ratanpaul V, Zhang D, Williams BA, Diffey S, Black JL, Gidley MJ (2017b) Effects of different amounts of wheat bran and oat hulls in a starch-based diet on voluntary feed intake in grower pigs. Animal Production Science 57, 2436
Effects of different amounts of wheat bran and oat hulls in a starch-based diet on voluntary feed intake in grower pigs.Crossref | GoogleScholarGoogle Scholar |

Rhoads JM, Niu X, Odle J, Graves LM (2006) Role of mTOR signaling in intestinal cell migration. American Journal of Physiology. Gastrointestinal and Liver Physiology 291, G510–G517.
Role of mTOR signaling in intestinal cell migration.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XhtVSksr7L&md5=d72817a4ff0ed253a4c13d7c5d9e83ecCAS |

Ropert A, Cherbut C, Roze C, Le Quellec A, Holst JJ, Fu-Cheng X, des Varannes BS, Galmiche JP (1996) Colonic fermentation and proximal gastric tone in humans. Gastroenterology 111, 289–296.
Colonic fermentation and proximal gastric tone in humans.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK28zgsFSqtA%3D%3D&md5=2c637efc497f12b4121840d9d8e29510CAS |

Roth JD, Roland BL, Cole RL, Trevaskis JL, Weyer C, Koda JE, Anderson CM, Parkes DG, Baron AD (2008) Leptin responsiveness restored by amylin agonism in diet-induced obesity: evidence from nonclinical and clinical studies. Proceedings of the National Academy of Sciences, USA 105, 7257–7262.
Leptin responsiveness restored by amylin agonism in diet-induced obesity: evidence from nonclinical and clinical studies.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXmsFCqsLw%3D&md5=46ec13836b7619435e7533bf094c9185CAS |

Roura E, Navarro M (2017) Physiological and metabolic control of diet selection. Animal Production Science

Sakata I, Sakai T (2010) Ghrelin cells in the gastrointestinal tract. International Journal of Peptides
Ghrelin cells in the gastrointestinal tract.Crossref | GoogleScholarGoogle Scholar |

Salfen BE, Carroll JA, Keisler DH, Strauch TA (2004) Effects of exogenous ghrelin on feed intake, weight gain, behaviour and endocrine responses in weanling pigs. Journal of Animal Science 82, 1957–1966.
Effects of exogenous ghrelin on feed intake, weight gain, behaviour and endocrine responses in weanling pigs.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXlsFWksrk%3D&md5=fc093b45d3ed32d42d5ce5d9cbc73345CAS |

Sandoval D, Cota D, Seeley RJ (2008) The integrative role of CNS fuel-sensing mechanisms in energy balance and glucose regulation. Annual Review of Physiology 70, 513–535.
The integrative role of CNS fuel-sensing mechanisms in energy balance and glucose regulation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXkt1eqt7g%3D&md5=0a6426eae2f8408d321938f5f5d31ca1CAS |

Sartin JL, Whitlock BK, Daniel JA (2011) Triennial growth symposium: neural regulation of feed intake: modification by hormones, fasting, and disease. Journal of Animal Science 89, 1991–2003.
Triennial growth symposium: neural regulation of feed intake: modification by hormones, fasting, and disease.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXoslCisrg%3D&md5=3d188a63afd9816ac332dd3f56b87c52CAS |

Seeley RJ, Yogaloff KA, Fisher SL, Burn P, Thiele TE, van Dijk G, Baskin DG, Schwartz MW (1997) Melanocortin receptors in leptin effects. Nature 390, 349
Melanocortin receptors in leptin effects.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2sXnslSitrw%3D&md5=60b6b71d947fececead8ddf975527e2cCAS |

Shin HS, Ingram JR, McGill A-T, Poppitt SD (2013) Lipids, CHOs, proteins: can all macronutrients put a ‘brake’ on eating? Physiology & Behavior 120, 114–123.
Lipids, CHOs, proteins: can all macronutrients put a ‘brake’ on eating?Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXhsF2gtb7P&md5=4ed318af17d303ead1a7b16c038a09dbCAS |

Small DM, Jones-Gotman M, Dagher A (2003) Feeding-induced dopamine release in dorsal striatum correlates with meal pleasantness ratings in healthy human volunteers. NeuroImage 19, 1709–1715.
Feeding-induced dopamine release in dorsal striatum correlates with meal pleasantness ratings in healthy human volunteers.Crossref | GoogleScholarGoogle Scholar |

Sopade PA, Nguyen GT, Gidley MJ (2016) Optimising particle size distribution for grains and protein sources. Final report to the Co-operative Research Centre for High Integrity Australian Pork, University of Adelaide, Roseworthy, SA.

Spiller RC, Trotman IF, Higgins BE, Ghatei MA, Lee YC, Bloom SR, Misiewicz JJ, Silk DB (1984) The ileal brake-inhibition of jejunal motility after ileal fat perfusion in man. Gut 25, 365–374.
The ileal brake-inhibition of jejunal motility after ileal fat perfusion in man.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaL2cXitVOgu78%3D&md5=7c387c3740c0e42f5713be491d218a5dCAS |

Stanogias G, Pearce GR (1985a) The digestion of fibre by pigs. 1. The effects of amount and type of fibre on apparent digestibility, nitrogen balance and rate of passage. British Journal of Nutrition 53, 513–530.
The digestion of fibre by pigs. 1. The effects of amount and type of fibre on apparent digestibility, nitrogen balance and rate of passage.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaL2MXksFeisrY%3D&md5=5d4694886a677c1992e331d949bfb925CAS |

Stanogias G, Pearce GR (1985b) The digestion of fibre by pigs. 3. Effects of the amount and type of fibre on physical characteristics of segments of the gastrointestinal tract. British Journal of Nutrition 53, 537–548.
The digestion of fibre by pigs. 3. Effects of the amount and type of fibre on physical characteristics of segments of the gastrointestinal tract.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaL28%2FltFCjtw%3D%3D&md5=1ee6c8472ce35d39f7b7463b8c44bc75CAS |

Steinberg GR, Kemp BE (2009) AMPK in health and disease. Physiological Reviews 89, 1025–1078.
AMPK in health and disease.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXpslyjsb4%3D&md5=312e3e949d35370215b25e9deed8a857CAS |

Steinert RE, Feinle-Bisset C, Geary N, Beglinger C (2013) Digestive physiology of the pig symposium: Secretion of gastrointestinal hormones and eating control. Journal of Animal Science 91, 1963–1973.
Digestive physiology of the pig symposium: Secretion of gastrointestinal hormones and eating control.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXotl2gsro%3D&md5=8d22a7d367c4eac6470c683ecad30cd8CAS |

Talsania T, Anini Y, Siu S, Drucker DJ, Brubaker PL (2005) Peripheral extendin-4 and peptide YY (3–36) synergistically reduce food intake through different mechanisms in mice. Endocrinology 146, 3748–3756.
Peripheral extendin-4 and peptide YY (3–36) synergistically reduce food intake through different mechanisms in mice.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXpsVehu7c%3D&md5=20dce44a1c4814a3963f47f2feb142f2CAS |

Torrazza RM, Suryawan A, Gazzaneo MC, Orellana RA, Frank JW, Nguyen HV, Fiorott ML, ElKadi S, Davis TA (2010) Leucine supplementation of a low-protein meal increases skeletal muscle and visceral tissue protein synthesis in neonatal pigs by stimulating mTOR-dependent translation initiation. Journal of Nutrition 140, 2145–2152.
Leucine supplementation of a low-protein meal increases skeletal muscle and visceral tissue protein synthesis in neonatal pigs by stimulating mTOR-dependent translation initiation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXhsFGgtLjO&md5=b4432907a9767bdf817e43e0c669ff4eCAS |

Tsaousidou E, Paeger L, Belgardt BF, Pal M, Wundelich CM, Brönneke H, Collienne U, Hampel B, Wunderlich FT, Schmidt-Supprian M, Kloppenburg P, Brüning JC (2014) Distinct role for JNK and IKK activation in agouti-related peptide neurons in the development of obesity and insulin resistance. Cell Reports 9, 1495–1506.
Distinct role for JNK and IKK activation in agouti-related peptide neurons in the development of obesity and insulin resistance.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXhvFGitr%2FJ&md5=6b8fc0d7e5e56bc35b0c0050b2f7d410CAS |

Unger EK, Olofsson LE, Xu AW (2010) Functional role of c-Jun-N-terminal kinase in feeding regulation. Endocrinology 151, 671–682.
Functional role of c-Jun-N-terminal kinase in feeding regulation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXhsVOqurg%3D&md5=4c49cdb412bbe7b29126c7ac0df8f45cCAS |

van Avesaat M, Troost FJ, Ripken D, Hendriks HF, Masclee AA (2015) Ileal brake activation: macronutrient-specific effects on eating behavior? International Journal of Obesity 39, 235–243.
Ileal brake activation: macronutrient-specific effects on eating behavior?Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXht1Ghsr3M&md5=04e3593cead201b43919a612dff1766fCAS |

van Nevel C, Seynaeve M, Van De Voorde G, De Smet S, Van Driessche E, de Wilde R (2000) Effects of increasing amounts of Lupinus albus seeds without or with whole egg powder in the diet of growing pigs on performance. Animal Feed Science and Technology 83, 89–101.
Effects of increasing amounts of Lupinus albus seeds without or with whole egg powder in the diet of growing pigs on performance.Crossref | GoogleScholarGoogle Scholar |

van Thuijl H, Kola B, Korbonits M (2008) Appetite and metabolic effects of ghrelin and cannabinoids: involvement of AMP-activated protein kinase. In ‘Ghrelin, vitamins and hormones, vol. 77’. (Ed. G Litwack) pp. 121–148. (Elsevier Inc.: Amsterdam, Netherlands)

Vicente B, Valencia DG, Perez-Serrano M, Lazaro R, Mateos GG (2008) The effects of feeding rice in substitution of corn and the degree of starch gelatinization of rice on the digestibility of dietary components and productive performance of young pigs. Journal of Animal Science 86, 119–126.
The effects of feeding rice in substitution of corn and the degree of starch gelatinization of rice on the digestibility of dietary components and productive performance of young pigs.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXhsVOru7bE&md5=7659f87c0ba4cc074822e1a5378e9991CAS |

Viollet B, Andreelli F, Jorgensen SB, Perrin C, Geloen A, Flamez D, Mu J, Lenzner C, Baud O, Bennoun M, Gomas E, Nicolas G, Wojtaszewski JF, Kahn A, Carling D, Schuit FC, Birnbaum MJ, Richter EA, Burcelin R, Vaulont S (2003) The AMP-activated protein kinase alpha2 catalytic subunit controls whole-body insulin sensitivity. The Journal of Clinical Investigation 111, 91–98.
The AMP-activated protein kinase alpha2 catalytic subunit controls whole-body insulin sensitivity.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXitVKntg%3D%3D&md5=4edfcd65fab79082f547f4c4ddcd1866CAS |

Volkow ND, Wang G-J, Baler RD (2011) Reward, dopamine and the control of food intake: implications for obesity. Trends in Cognitive Sciences 15, 37–46.
Reward, dopamine and the control of food intake: implications for obesity.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXms1Cr&md5=0fb8d7f73b1e86009cb05006977a775aCAS |

Welch IM, Sepple CP, Read NW (1988) Comparisons of the effects on satiety and eating behaviour of infusion on lipid into different regions of the small intestine. Gut 29, 306–311.
Comparisons of the effects on satiety and eating behaviour of infusion on lipid into different regions of the small intestine.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaL1cXksFekuro%3D&md5=2f3de397d67d1487ff16e5df44dad589CAS |

Whited KL, Lu D, Tso P, Kent Lloyd KC, Raybould HE (2005) Apolipoprotein A-IV is involved in detection of lipid in the rat intestine. The Journal of Physiology 569, 949–958.
Apolipoprotein A-IV is involved in detection of lipid in the rat intestine.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XivVOjsQ%3D%3D&md5=27003f6730350a8a59a45e6863ae120bCAS |

Whited KL, Tso P, Raybould HE (2007) Involvement of apolipoprotein A-IV and cholecystokinin1 receptors in exogenous peptide YY3–36-induced stimulation of intestinal feedback. Endocrinology 148, 4695–4703.
Involvement of apolipoprotein A-IV and cholecystokinin1 receptors in exogenous peptide YY3–36-induced stimulation of intestinal feedback.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXhtFWls7vI&md5=ace5cbfc9d8146123c7ea8bd2abd7dc4CAS |

Wilfart A, Montague L, Simmins H, Noblet J, van Milgen J (2007) Digesta transit in different segments of the gastrointestinal tract of pigs as affected by insoluble fibre supplied by wheat bran. British Journal of Nutrition 98, 54–62.
Digesta transit in different segments of the gastrointestinal tract of pigs as affected by insoluble fibre supplied by wheat bran.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXosVOjs7Y%3D&md5=e34cf3ccfe7d872651885c7b4a2dc73cCAS |

Woods SC, D’Alessio DA, Tso P, Rushing PA, Clegg DJ, Benoit SC, Gotoh K, Liu M, Seeley RJ (2004) Consumption of a high-fat diet alters the homeostatic regulation of energy balance. Physiology & Behavior 83, 573–578.
Consumption of a high-fat diet alters the homeostatic regulation of energy balance.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXhtFCqtLjM&md5=780edb412e7738b68ae19acc7dcfbbfbCAS |

Woods SC, Benoit SC, Clegg DJ (2006) The brain–gut–islet connection. Diabetes 55, S114–S121.
The brain–gut–islet connection.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XhtlSisr%2FF&md5=f7de86a544f889dcdfb5f8f4de939244CAS |

Woods SC, Seeley RJ, Cota D (2008) Regulation of food intake through hypothalamic signaling networks involving mTOR. Annual Review of Nutrition 28, 295–311.
Regulation of food intake through hypothalamic signaling networks involving mTOR.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXhtV2isL3M&md5=9219124dc58a2c162930b49f05fb3a0aCAS |

Wullschleger S, Loewith R, Hall MN (2006) TOR signaling in growth and metabolism. Cell 124, 471–484.
TOR signaling in growth and metabolism.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28Xhslaqs74%3D&md5=6656399e3662aa5013778c7ed2635f9eCAS |

Wynne K, Bloom SR (2006) Review: the role of oxyntomodulin and peptide tyrosine-tyrosine (PYY) in appetite control. Nature Clinical Practice. Endocrinology & Metabolism 2, 612–620.
Review: the role of oxyntomodulin and peptide tyrosine-tyrosine (PYY) in appetite control.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28Xht1aktbnF&md5=78730381749155a9ed6ed0b70e6f33f8CAS |

Xu J, Ji J, Yan XH (2012) Cross-talk between AMPK and mTOR in regulating energy balance. Critical Reviews in Food Science and Nutrition 52, 373–381.
Cross-talk between AMPK and mTOR in regulating energy balance.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38Xms1WgtLw%3D&md5=63dd7d067bd84bcea401a3eef71b53bdCAS |

Zhang Y, Guo K, LeBlanc RE, Loh D, Schwartz GJ, Yu Y-H (2007) Increasing dietary leucine intake reduces diet-induced obesity and improves glucose and cholesterol metabolism in mice via multimechanisms. Diabetes 56, 1647–1654.
Increasing dietary leucine intake reduces diet-induced obesity and improves glucose and cholesterol metabolism in mice via multimechanisms.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXmtlKhs70%3D&md5=ea12882aee6d04292ad3c9af8da7b294CAS |