ebook img

Review Article Obesity and Appetite Control - Hindawi Publishing Corporation PDF

20 Pages·2014·1.38 MB·English
by  
Save to my drive
Quick download
Download
Most books are stored in the elastic cloud where traffic is expensive. For this reason, we have a limit on daily download.

Preview Review Article Obesity and Appetite Control - Hindawi Publishing Corporation

HindawiPublishingCorporation ExperimentalDiabetesResearch Volume2012,ArticleID824305,19pages doi:10.1155/2012/824305 Review Article Obesity and Appetite Control KeisukeSuzuki,ChannaN.Jayasena,andStephenR.Bloom SectionofInvestigativeMedicine,ImperialCollegeLondon,CommonwealthBuilding,DuCaneRoad,LondonW120NN,UK CorrespondenceshouldbeaddressedtoStephenR.Bloom,[email protected] Received15March2012;Accepted20June2012 AcademicEditor:BernardThorens Copyright©2012KeisukeSuzukietal.ThisisanopenaccessarticledistributedundertheCreativeCommonsAttributionLicense, whichpermitsunrestricteduse,distribution,andreproductioninanymedium,providedtheoriginalworkisproperlycited. Obesity is one of the major challenges to human health worldwide; however, there are currently no effective pharmacological interventions for obesity. Recent studies have improved our understanding of energy homeostasis by identifying sophisticated neurohumoralnetworkswhichconveysignalsbetweenthebrainandgutinordertocontrolfoodintake.Thehypothalamusisa keyregionwhichpossessesreciprocalconnectionsbetweenthehighercorticalcentressuchasreward-relatedlimbicpathways,and thebrainstem.Furthermore,thehypothalamusintegratesanumberofperipheralsignalswhichmodulatefoodintakeandenergy expenditure.Guthormones,suchaspeptideYY,pancreaticpolypeptide,glucagon-likepeptide-1,oxyntomodulin,andghrelin,are modulatedbyacutefoodingestion.Incontrast,adipositysignalssuchasleptinandinsulinareimplicatedinbothshort-andlong- termenergyhomeostasis.Inthispaper,wefocusontheroleofguthormonesandtheirrelatedneuronalnetworks(thegut-brain axis)inappetitecontrol,andtheirpotentialsasnoveltherapiesforobesity. 1.Introduction In addition to local paracrine actions and peripheral endocrine effects mediated through the bloodstream, gut Despite recent progress in our understanding of the phys- hormonesplayapivotalrolerelayinginformationonnutri- iological mechanisms regulating body weight and energy tionalstatustoimportantappetitecontrollingcentreswithin expenditure, obesity remains a major worldwide health thecentralnervoussystem(CNS),suchasthehypothalamus crisiswithanarrayofvascular,metabolic,andpsychosocial andthebrainstem. consequences[1,2].Overweightorobeseindividuals(body In this article, we will summarise our current under- mass index 25–30) have an increased risk of developing standing of the physiological interactions between the gut diabetes, coronary heart disease, and hypertension [2, 3]. andbrain,termedthe“gut-brainaxis,”focussingparticularly Adults with a body mass index of 40 or higher have been ontheinteractionsofguthormoneswiththeCNSandvagus associatedwithahighriskofdevelopingdiabetes,hyperten- nerve[6].Wewillnotdiscusssignaltransductionpathways, sion,dyslipidaemia,asthma,arthritis,andpoorhealthstatus, enteric nervous systems related to controlling food intake, whencomparedwithnormalweightindividuals[4]. or neural signalling pathways in organs associated with the Bodyweightistightlyregulatedbycomplexhomeostatic gastrointestinaltractsuchasliverorpancreas. mechanisms. Obesity is a state in which energy intake chronically exceeds energy expenditure. Even a subtle mis- match(lessthan0.5%)incaloricintakeoverexpenditureis 2.GutHormones sufficient to cause weight gain [5]. The rising prevalence of obesityislikelytoresultfromcontemporaryenvironmental 2.1.PancreaticPolypeptide-FoldPeptides. ThePP-foldfamily and lifestyle factors such as increased access to palatable comprises neuropeptide Y (NPY), peptide YY (PYY), and foodsandreducedrequirementsforphysicalexercise,when pancreaticpolypeptide(PP).Theyarecomposedofachain compared with ancient hunter-gatherer lifestyles charac- of 36 amino acids residue and share amino acid homology, terisedbyunpredictableperiodsoffeastandfamine. amidatedC-terminalends.ThetertiarystructurePP-foldis 2 ExperimentalDiabetesResearch Ushapedwithanextendedpolyprolinehelixandanαhelix nervosa when compared withcontrol subjects[24]. Studies connectedbyaβturn[7].Inaddition,ahairpin-likePP-fold of circulating levels of PYY in obese and lean people have motifisvitalforreceptorbinding.PYYandPParesecreted yielded inconsistent results [25, 26]; however, a blunted from gastrointestinal tract, whereas NPY is predominantly, postprandialriseinPYYinobesesubjectssuggestsapossible widelydistributedinCNS[8].ThisfamilyactsviaGprotein- associationwithimpairedpostprandialsatietyduringobesity coupledreceptors;Y1,Y2,Y4,Y5,andY6[9].TheY3receptor [21]. hasnotyetbeencloned,andtheY5receptorhasbeenfound PYY exerts anorectic effects via a direct action in 3–36 asanonfunctionaltruncatedform. thehypothalamicarcuatenucleus(ARC).Peripheraladmin- istration of PYY increases c-fos expression (a marker 3–36 2.2.PeptideTyrosineTyrosine(PYY). PYYisanappetitesup- of neuronal activation) in the ARC and direct injection of pressing hormone, which was isolated originally from PYY intotheARCinhibitsfoodintake.Thiseffectislikely 3–36 porcine upper small intestine [8]. Its name is derived from to be mediated through the Y2 receptor since the anorectic its characteristic tyrosine (Y) residues at both the C and N effect of peripheral PYY administration is blocked in 3–36 terminals.PYYisreleasedfromtheLcellsofthedistalgutin Y2 receptor-null mice, and intraarcuate injection of a Y2 responsetoingestednutrientswithtwootherguthormones, receptor selective agonist also supresses food intake [16]. GLP-1 and OXM. PYY immunoreactivity is highest in the Although conflicting results have been reported [27], the rectum, and decreases proximally to low levels in the duo- vagal-brainstem may also signal the actions of PYY on denumandjejunum.PYYimmunoreactivityisalsofoundin food intake. Two independent laboratories have observed the CNS regions such as the hypothalamus, medulla, pons, that vagotomy abolishes anorexia c-fos activation following and spinal cord [10]. Two endogenous circulating forms, peripheralPYY administration[28,29]. 3–36 PYY andPYY ,aresynthesizedwithinthegut.PYY Incontrasttotheanorecticeffectsobservedbyperipheral 1–36 3–36 1–36 is the biologically active major circulating form, which is andintraarcuatePYY administration,directadministra- 3–36 produced by cleavage of the N-terminal tyrosine-proline tion of PYY into the third ventricle of the brain [30] or 3–36 residues from PYY by the enzyme dipeptidyl-peptidase paraventricularnucleus(PVN)[31]increasesinfoodintake. 1–36 IV (DPP-IV) [11]. PYY has affinity to all Y receptors, This paradoxical action may be explained by considering 1–36 whilePYY actsmainlyviathehigh-affinityhypothalamic that such effects might be endogenously mediated by the 3–36 Y2receptor. orexigenicCNS-distributedpeptide,NPY,throughanaction The PYY secretion pattern suggests a role in satiety. on Y1 receptor and Y5 receptors [32]. PYY may also act in Circulating PYY concentrations are low in fasted state and thebrainareasotherthanthehypothalamusandbrainstem. increase rapidly following a meal with a peak at 1-2 hours In a clinical study using functional MRI by Batterham et and remain elevated for several hours [12]. PYY release is al. [33], PYY infusion modulated neural activity within 3–36 increased in proportion to calorie intake [12]. PYY may corticolimbicandhighercorticalbrainregions. have a role in the pathogenesis of a number of anorectic ExogenousPYY andexendin-4,aGLP-1receptor conditionssuchasinflammatoryboweldisease,steatorrhea, agonist, have syner3g–i3s6tiNcHe2ffects to suppress food intake in tropical sprue, and cardiac cachexia, since plasma PYY mice [34]. Furthermore a recent study utilizing functional levels are elevated in patients with these conditions [13– MRIbyDeSilvaetal.[35]showedthatcoadministrationof 15]. Peripheral PYY administration shows a reduction PYY andGLP-1 tofastedhumansubjectsresults 3–36 3–36 7–36amide in food intake and body weight gain in rats [16]. In both insimilarreductionsinsubsequentenergyintakeandbrain leanandobesehumansubjects,intravenousadministration activity,asobservedphysiologicallyfollowingfeeding. of PYY reduces appetite and food intake [16, 17] with NeuropeptideY2receptorshavecardiovasculareffectsin 3–36 observed plasma PYY levels similar to the physiological additiontotheirmetaboliceffects.Y2agonismisimplicated 3–36 levels after a meal; this data suggests that the physiological inthepathogenesisofhypertensioninhypertensiverats[36]. effectofPYYistosuppressfoodintake.Ofnote,nonausea Nordheim and Hofbauer [37] reported that Y2 receptor was reported in subjects following PYY administration. stimulation by PYY demonstrated cardiovascular effects 3–36 3–36 Thissuggeststhat,unlikeleptin,thesensitivityofsubjectsto of endogenous NPY in rats on different dietary regimens. PYYispreservedinobesesubjects.Someinvestigatorsfailed In food-restricted rats, PYY increased mean arterial 3–36 toshowananorecticeffectofPYY,possiblyduetoinadequate pressure and heart rate, whereas PYY did not influence 3–36 acclimatizationofcontrolandtreatedanimals[18]. mean arterial pressure and heart rate in high-fat diet rats. The “ileal brake” is the negative feedback mechanism However,humanstudiesthusfarhavenotdemonstratedany in which the presence of nutrients into the colon inhibits hypertensivechangesasaresultofPYYadministration. motility and transit of further nutrients within the upper gastrointestinaltract[19].Fatisknowntobethemostpotent 2.3.PancreaticPolypeptide(PP). PPissecretedfromPPcells triggeroftheilealbrake.GLP-1andPYYmaycontributeto inthepancreaticisletsofLangerhansinresponsetoameal. thisphenomenon[20]. AnorecticeffectsofPParethoughttobemediatedbydirectly PYY has been reported to regulate energy expenditure, throughtheY4receptorinthebrainstemandhypothalamus. delay gastric emptying, reduce acid secretion, and inhibit In addition, it may act also via the vagus nerve, as the gallbladder contraction and pancreatic exocrine secretions anorecticeffectsofPPareabolishedbyvagotomyinrodents [21, 22]. Circulating PYY levels are low in obese subjects [38]. PP has a high affinity for the Y4 receptor, of which [17, 23], and they are higher in patients with anorexia expression is found in the area postrema (AP), nucleus ExperimentalDiabetesResearch 3 of the tractus solitarius (NTS), dorsal motor nucleus of In addition, GLP-1 is distributed within the CNS. 7–36amide vagus (DVN), ARC, and PVN [39]. An autoradiography Immunoreactive neurons for GLP-1 are located 7–36amide study also identified saturable PP binding sites at the in the PVN, DMN, NTS, dorsal vagal complex (DVC), interpeduncularnucleus,AP,NTS,andDVN[40].LikePYY, pituitary, and thalamus [57]. GLP-1 receptor mRNA is paradoxical effects on food intake are observed following distributedthroughouttherostrocaudalhypothalamus,with PP injection, depending on its route of administration. In denseaccumulationintheARC,PVN,andsupraopticnuclei contrast to the anorectic effects observed with peripheral [58].WhileperipheraladministrationofGLP-1inratsleads PP administration, central PP administration stimulates to increased c-fos expression in the ARC [28], intracere- food intake [41]. Although the exact mechanism of this broventricular (ICV) administration results in increased c- phenomenon is unclear, these differential effects may be fos expression in the PVN, NTS, and AP [59]. Ascending mediated by activation of distinct populations of receptors. NTS-PVNprojectionscontainGLP-1[60]areimplicatedin PP also has other physiological effects, such as delaying controllingfoodintake.IntheCNS,leptinreceptor(Ob-Rb) gastricemptying,attenuatingpancreaticexocrinesecretion, was expressed in GLP-1-containing neurons in the NTS in andinhibitinggallbladdercontraction[42]. animals and leptin activated GLP-1 containing neurons in PlasmaPPlevelsshowdiurnalvariations:lowestlevelsare the NTS [61]. Signals arising from the hepatoportal GLP- observed in the early morning and highest in the evening. 1R promote glucose clearance, which are independent of The release of postprandial PP is biphasic. Circulating PP changesininsulinsecretion[62,63]. concentrations increase after a meal in proportion to the GLP-1 exerts its effect by activation of the GLP-1R caloric intake, and increased levels remain for up to 6 to stimulate adenylyl cyclase activity and thereby cAMP hours postprandially [43]. Circulating PP levels seem to be production [64]. GLP-1R is widely distributed particularly inverselyproportionaltoadiposity;higherlevelsarereported in the brain, gastrointestinal tract, and pancreas [64, 65]. in subjects with anorexia nervosa [44]. Some, but not all In the brain, binding sites for GLP-1Rs have been found in [45, 46], studies have demonstrated significant reductions thehypothalamus,striatum,brainstem,substantianigra,and in circulating levels of PP in obese subjects [47, 48]. subventricular zone among other structures [64, 66]. GLP- Furthermore, obese patients with Prader-Willi syndrome 1Rs are present on both glia and neuronal cell types [66]. (PWS) have been reported to have reduced PP release both In addition, GLP-1Rs are expressed in the nodose ganglion basallyandpostprandially[49]. [67]. Furthermore bilateral subdiaphragmatic total truncal Inmice,acuteandchronicperipheralPPadministration vagotomyorbrainstem-hypothalamicpathwaytransetioning resultsinreducedfoodintake.Inleptin-deficientob/obmice, abolishes the suppressing actions of GLP-1 on food intake repeatedintraperitonealPPinjectiondecreasesbodyweight [28];thissuggeststhatthevaguscontributestotheactionsof gain and improves insulin resistance and hyperlipidaemia GLP-1onfoodintake. [38]. Furthermore, transgenic mice overexpressing PP have Circulating GLP-1 levels rise postprandially and fall in reduced food intake when compared with wild-type con- the fasted state. Recent evidence also suggests that GLP- trols [50]. In normal-weight human subjects, intravenous 1 levels rise in anticipation of a meal [68]. GLP-1 not infusion of PP achieved three times higher circulating PP only reduces food intake, but also suppresses glucagon concentrations when compared with postprandial levels in secretion and delays gastric emptying [69]. Intravenous the same subjects after a buffet lunch (which reduced food administrationofGLP-1isassociatedwithadose-dependent intakeby25%over24hours)[51].Furthermore,twice-daily reduction of food intake in both normal weight and obese infusion of PP in volunteers with PWS resulted in a 12% subjects[70],althoughobesesubjectsmaybelessresponsive reduction in food intake [52]. Agonists to the Y4 receptor [64]. designedtomimictheactionsofPPhavebeendevelopedand GLP-1possessesapotentincretineffectinadditiontoits are under further investigation as potential novel therapies anorecticaction;itstimulatesinsulinsecretioninaglucose- forobesity. dependent manner following ingestion of carbohydrate. However, its use as obesity treatment was limited for many yearsbyitsshortplasmahalf-lifeof1-2minutes[71],which 2.4. Proglucagon-Derived Peptides. The proglucagon gene is ispartlyattributedtoenzymaticdegradationbyDPP-IVand expressedinthepancreas,intheL-cellsofthesmallintestine renal clearance that rapidly inactivate and remove GLP-1 and in the NTS of the brainstem [53, 54]. GLP-1, GLP- fromplasmacirculation[72,73].Continuoussubcutaneous 2, OXM, and glucagon are proglucagon-derived peptides. infusionofGLP-1topatientswithtype2diabetesfor6weeks Glucagonisthemainproductinthepancreas,whereasOXM, reducesappetite,andbodyweight,andimprovesglycaemic GLP-1, and GLP-2 are the major products in the brain and control [74]. However, DPP-IV-resistant analogues of GLP- intestine[55]. 1 have been developed. Exendin-4 (exenatide), a naturally occurring peptide originally isolated from the saliva of the 2.4.1.Glucagon-LikePeptide-1(GLP-1). GLP-1iscosecreted Gila monster lizard, is a DPP-IV-resistant GLP-1R agonist with PYY from the L cells in the intestine in response to [75]. Exenatide improves glycaemic control and decreases nutrientingestion.GLP-1hastwobiologicallyactiveforms, body weight in patients with type 2 diabetes. [76]. GLP-1 GLP-1 and GLP-1 . The latter truncated form possesses trophic effects on pancreatic beta cells in animal 7–37 7–36amide is the major circulating form in humans, although both models[77].GLP-1andexendin-4havebeenrecentlyshown active isoforms of GLP-1 have equivalent potency [56]. topromotecellulargrowthandreduceapoptosisinnervous 4 ExperimentalDiabetesResearch tissues[78],buttrophiceffectsonpancreaticbetacellshave in response to hypoglycaemia. Glucagon enhances the not been demonstrated clinically in human subjects. GLP- body’sphysiologicalresponsetostress,byincreasingenergy 1 agonists are, therefore, a good example of how research expenditure[95,96].However,glucagonadministrationalso in this area has been translated into clinical practice. A decreases food intake, possibly by modulating vagal tone three-year duration of treatment with exenatide has been andgastricemptying[97,98].Schulmanetal.[99]reported reported to improve beta cell function; however, when that glucagon reduces food intake and body weight but adjusting for weight loss associated with exenatide therapy, caused hyperglycemia. However, the administration of the thiseffectremainsspeculative[79].DPP-IVinhibitors,such dualagonistsstimulatingbothglucagonandGLP-1receptors as sitagliptin and vildagliptin, which are licensed for the achieved improvement of diet-induced obesity and glucose treatmentoftype2diabetes,donotresultindecreaseinbody intolerance [100, 101]. It is, therefore, plausible that dual weight. This may be explained by considering that DPPIV agonism of glucagon and GLP-1 receptors may offer novel is also involved in the modification of other gut hormones targetsforantiobesitytreatment. suchasPYY,andcytokineswhichmayhaveoppositeeffects toGLP-1[80]. GLP-1-based therapies are promising novel treatments 2.5.Ghrelin. Ghrelinwasidentifiedoriginallyasanendoge- for type 2 diabetes, however, long-term outcome data nous ligand for the growth hormone secretagogue receptor are not yet available. The reported side effects of GLP-1 (GHS-R) in rat stomach [102]. Ghrelin comprises a chain agonistsarenauseaandvomiting.Animalsafetystudieswith of 28 amino acids with esterification of the hydroxyl group liraglutide have identified C-cell carcinoma of the thyroid. of the third serine residue by octanoic acid, and it is the Acutepancreatitishasbeenreportedinhumanstreatedwith only known orexigenic gut hormone. Ghrelin is principally liraglutide or exenatide [81]. Further outcome data will, secreted from X/A-like cells within gastric oxyntic glands therefore, be important in confirming the long-term safety [103]. In keeping with this, gastrectomy results in an 80% ofGLP-1-basedtherapies. reductionofplasmaghrelinlevels;theremainderissecreted from the intestine, pancreas, pituitary, and colon [104]. Ghrelin also acts as a neurotransmitter, being expressed 2.4.2. Oxyntomodulin (OXM). OXM is a 37-amino acid withintheARCandperiventricularareaofthehypothalamus peptideoriginallyisolatedfromporcinejejunoilealcellsand [102,105]. is found to show glucagon-like activity in the liver [82]. Serum ghrelin levels are increased by fasting and OXM is another product of the proglucagon gene and is decreased by refeeding or oral glucose administration, but cosecreted with GLP-1 and PYY by the L-cells of the distal they are not decreased by water ingestion [106]. In rats, gastrointestinal tract, in response to ingested food and in ghrelinlevelsshowadiurnalpattern,withthebimodalpeaks proportiontocaloricintake[83].OXMhasanorecticeffects occurring before dark and light periods [107]. In humans, andshowsincretinactivitywithamuchlowerpotencywhen ghrelin levels have a diurnal rhythm which is identical to compared with GLP-1 [84]. OXM also inhibits gastric acid the diurnal rhythm of leptin, with both hormones rising secretionanddelaysingastricemptying[85]. throughout the day to a zenith at 0100h, then falling Administration of OXM is associated with decreased overnighttoanadirat0900h[108]. foodintakeandincreasesenergyexpenditureinbothrodents Levels of circulating ghrelin rise preprandially and fall andhumans[86–88].TheanorecticeffectofOXMisblocked rapidly in the postprandial period [108]. Both central and by the GLP-1R antagonist, exendin [89], and is not 9–39 peripheral administration of ghrelin increase food intake observed in GLP-1R null mice [90]; this suggests that the andbodyweightalongwithareductioninfatutilisationin anorectic effects of OXM may be mediated by the GLP-1R. rodents[106,109].Negativecorrelationsbetweencirculating However, OXM has relatively low in vitro affinity for the ghrelin levels and body mass index are found in human. GLP-1R which is 50 folds lower than the affinity of GLP- Fasting plasma levels of ghrelin are reported to be high in 1 for GLP1R, despite the anorectic potency of OXM being patientswithanorexianervosa[110]andsubjectswithdiet- comparable to the potency of GLP-1 [91]. Several actions induced weight loss [111]. In contrast, obese subjects show of OXM seem independent of the GLP-1R [87, 92, 93]; a less marked drop in plasma ghrelin after meal ingestion the cardiovascular effects of OXM are preserved in GLP- [112]. In patients with heart failure, increased levels of 1R knockout mice [92]. These data suggest that a further plasma ghrelin are reported in cachectic patients when receptor through which OXM mediates its anorectic effect compared with noncachectic patients [113]. Furthermore, hasyettobeidentified. Furthermore,directadministration in patients with PWS, elevated circulating ghrelin levels are of the GLP-1R antagonist, exendin , to the ARC fails to 9–39 found,whencomparedwithindividualswithnonsyndromic inhibit the anorectic effects of OXM but inhibits that of formsofobesity[114]. GLP-1 [87]. Like GLP-1, OXM is inactivated by DPP-IV. Ghrelin mediates its orexigenic action via stimulation OXM analogues resistant to DPP-IV degradation are being ofNPY/agouti-relatedpeptide(AgRP)coexpressingneurons developedaspotentialobesitytreatments[94]. withintheARCofhypothalamus.Peripheraladministration ofghrelinincreasesc-fosexpressionintheARCNPY/AgRP 2.4.3.Glucagon. Theroleofglucagoninglucosehomeostasis neurons[115]andablationofbothAgRPandNPYneurons is well established; glucagon is produced by alpha cells of completely abolishes the orexigenic effect of ghrelin [116]. the pancreatic islets and increases glucose concentration The brainstem and vagus nerve may also contribute to the ExperimentalDiabetesResearch 5 effects of ghrelin on food intake. ICV injection of ghrelin 2.8.Amylin. Amyliniscoreleasedwithinsulininresponseto induces c-fos expression in the NTS and AP [117]. GHS-R mealingestion,anditmayfunctionasananorectichormone. is found to be expressed in the vagus nerve. Furthermore, Circulatinglevelsofamylinarefoundtobehigherinobese blockade of gastric vagal afferents in rats abolishes ghrelin- than lean subjects [133, 134]. Administration of amylin inducedfeedingandpreventstheghrelin-inducedriseinc- is associated with reduced food intake and body weight fosexpressionwithintheARC[118].Inadditiontoitspotent [135]. The anorectic effects of amylin may be mediated orexigenic property, ghrelin also increases gastric motility, by modulating activity of the serotonin, histamine, and upstimulates the hypothalamo-pituitary-adrenal axis, and dopaminergic system in the brain as well as inhibition possesses cardiovascular effects such as vasodilatation and of NPY release [133]. Administration of pramlintide, a enhancedcardiaccontractility[104]. synthetic analogue of human amylin, improves glycaemic Ghrelin may promote food intake in part by enhancing control and causes weight loss in type 2 diabetes patients the hedonic responses to food cues, which is demonstrated using insulin [136]. Therefore, amylin replacement with by the recent study by Malik et al. [119]. In their study, pramlintide as an adjunct to insulin has been reported as functional MRI was performed during exposure to food a novel physiological approach toward improved long-term pictures, and the study results demonstrated increased glycaemicandbodyweightcontrolinpatientswithdiabetes activation in the amygdala, orbitofrontal cortex, anterior [137]. insula,andstriatum,duringintravenousinfusionofghrelin. 3.PeripheralAdipositySignals 2.6. Obestatin. Obestatin is a 23-amino acid peptide hor- mone which is derived from posttranslational cleavage of 3.1.Insulin. Circulatinglevelsofinsulinandleptinpositively preproghrelin, and released from the stomach [120]. In correlate with adipose tissue mass within the body. Both contrasttoghrelinwhichhasorexigenicproperties,obestatin insulinandleptinareimplicatedinthelong-termregulation may have anorectic effects by decreasing food intake, of energy balance. Insulin is synthesized in the ß cells of delaying gastric emptying, and reducing body weight in thepancreasandissecretedrapidlyafterameal,withwell- rodents[121].However,thepotentialanorecticofobestatin characterisedhypoglycaemiceffects[138].However,insulin remainscontroversial,sinceotherinvestigatorshavefailedto alsoactsasananorecticsignalwithintheCNS.ICVadmin- demonstrateeffectsonfoodintakeinleanorobeserodents istration of insulin results in a dose-dependent suppression [122]. offoodintakeandbodyweightgaininbaboonsandrodents [139, 140]. Intrahypothalamic insulin injection to the PVN alsoresultsindecreasedfoodintake[141].Insulinentersthe 2.7.Cholecystokinin(CCK). CCKwasthefirstguthormone CNS through a saturable and receptor-mediated transport found to be implicated in appetite control [123]. CCK is process [142]. Insulin receptors are widely expressed in the secreted postprandially by the I cell of the small intestine brain,particularlyinhypothalamicnuclei,suchastheARC, intocirculation[124],withashortplasmahalf-lifeofafew DMN, and PVN, which are involved in control of food minutes. Plasma CCK levels rise within 15 minutes after intake [143]. Although the mechanism of insulin-mediated mealingestion[124].InfusionofC-terminaloctapeptideof anorexia has not been fully elucidated, hypothalamic NPY CCKdecreasedfoodintakein12leanmen[125].However, seemstobeinvolved.ICVadministrationofinsulininhibits intermittent prandial CCK infusion reduces meal size in thefasting-inducedincreaseinNPYmRNAexpressioninthe rats but causes a compensatory increase in meal frequency PVN and ARC in rats. This suggests that fasting increases [126].A2-weekcontinuousintraperitonealinfusionofCCK NPY biosynthesis through an ARC-PVN pathway in the failed to suppress food intake at any time point [127]. hypothalamusviaamechanism whichisdependentonlow Other physiological functions of CCK include stimulating insulinlevels[144]. the release of enzymes from the pancreas and gall bladder, promotingintestinalmotility,anddelayinggastricemptying. There are two CCK receptor subtypes known; CCK1 and 3.2. Leptin. Leptin is the product of the ob gene, and it is CCK2receptors,previouslyclassifiedasCCKAandCCKB. predominantlysecretedbyadipocyteswithcirculatinglevels TheanorecticactionofCCKappearstobemostlymediated proportional to fat mass [145]. Levels of circulating leptin via CCK1 receptors on the vagal nerve [128, 129]. CCK 1 haveadiurnalandpulsatilepattern,withpeaklevelsatnight and2receptorsarewidelydistributedinbrainincludingthe [146]. Leptin is transported across the BBB by a saturable brainstemandhypothalamus[130]. transportersystem[147],anditexertsitsanorecticeffectvia Some studies suggest that leptin and CCK may interact theARC,wherebothNPY/AgRPandpro-opiomelanocortin synergisticallytoinduceshort-terminhibitionoffoodintake (POMC)/cocaine- and amphetamine-regulated transcript and long-term reduction of body weight [131]. Leptin- (CART) neurons express leptin receptors [148]. Leptin deficientmiceareinsensitive tothemeal-terminatingeffect inhibits NPY/AgRP neurons and activates POMC/CART ofCCKadministration.Furthermore,leptinsignallingpath- neurons [149, 150], resulting in reduced food intake [149] ways to brain are dampened in the absence of interaction and increased energy expenditure [151]. The effects of gut with CCK release after a meal or in the setting of CCK-A satiation signals such as CCK can be amplified by leptin receptorblockade[132]. whichactsintheCNS,includingtheARCinparticular[152]. 6 ExperimentalDiabetesResearch Therearethreetypesofleptinreceptorsidentified:long, Cleavageofaprecursorprotein,calledPOMC,produces short,andsecretedform[153].Amongthose,Ob-Rbrecep- α-melanocyte-stimulating hormone (α-MSH), which binds tor, which is highly expressed in the hypothalamus [154], to melanocortin-3 receptor (MC3R) and melanocortin-4 is thought to act as the main receptor involved in appetite receptor(MC4R)tosuppressfoodintake[167].TheMC4R control.Thedb/dbmouse,withaninactivatingmutationin is highly expressed in the hypothalamus and is thought to theOb-Rbreceptor,hasanobesephenotype[155,156],and have a major role in suppressing food intake compared to leptin-deficientob/obmiceexhibithyperphagiaandobesity, the MC3R. MC4R knock-out mice have hyperphagia and whichcanbereversedbyleptinadministration[157]. obesity [167]. MC3R-deficient mice also have increased fat Subcutaneous administration of recombinant leptin mass and reduced lean body mass [168]; however, selective reduces fat mass, hyperinsulinaemia, and hyperlipidaemia MC3Ragonistsfailtosuppressfeeding[169]. in obese children with congenital leptin deficiency [158]. CART is the third most abundant transcript identified However, obese individuals often have high leptin levels, within the hypothalamus and is mostly colocalized with which result in a failure to respond to exogenous leptin. POMCintheARC.ICVadministrationofCARTsuppresses Thisleptinresistanceseverelylimitsthetherapeuticutilityof feeding,whereasICVinjectionofCARTantiserumincreases leptin,anditislikelytoresultfromreducedleptinreceptor foodintake[170].However,CARTinjecteddirectlyintothe signaltransduction[159]oranimpairedabilityoftheBBB PVNorARCoffastedratscausesanincreaseinfoodintake totransportleptin[160]. at 1-2 hours postinjection [171], which suggests opposing effects of CART on food intake can be observed depending on the site of administration. Hence, the physiological role 4.NeuralPathwaysRelatedto ofCARTinenergyhomeostasisisunclear. theAppetiteControl NPY/AgRP neurons extensively project to the adjacent hypothalamic nuclei, such as the PVN, DMN, and LHA. Feeding and energy expenditure are controlled by com- AgRPandNPYareexclusivelycolocalizedinARCneurons, plex neural networks distributed throughout the forebrain bothofwhichexertorexigeniceffects[172].NPYisthemost and brainstem. Reward-related neural brain regions have abundant neuropeptide in the CNS [173]. ICV injection been implicated in the nonhomeostatic control of feeding of NPY stimulates food intake in rats [41] and repeated behaviour[161].Bycontrast,homeostaticfeedingbehaviour daily bilateral PVN injection of NPY for 10 days causes an isintegratedwithinthehypothalamus.Keyperipheralsignals approximately two-fold increase in daily food intake and of energy status such as gut hormones and adipokines a six-fold increase in the rate of body weight gain [174]. either signal to the hypothalamus directly or signal to The orexigenic effect of NPY appears to be mediated by the hypothalamus indirectly via the brainstem and vagal afferent fibres. Adiposity signals such as insulin and leptin stimulationofhypothalamicY1andY5receptors.AGRPwas isolatedbyitshigh-sequencehomologywiththeAgouticoat are involved in the long-term energy homeostasis, and gut colourgenewhichisassociatedwithyellowcoat,obesity,and hormones such as PYY, GLP-1, PP, OXM, and ghrelin are increased body length in mice. AgRP is a potent-selective implicated in the short-term regulation of meal ingestion [162–164]. antagonistattheMC3RandMC4R[175]. The PVN receives projections of NPY/AgRP and POMC/CART from the ARC and contains neurons which 5.TheHypothalamus express the anorectic factors, thyrotropin-releasing hor- mone,andcorticotropin-releasinghormone.Microinjection Thehypothalamuscontrolsfeedingbyintegratingperipheral oforexigenicoranorexigenicsignals,suchasghrelin,orexin- humoral signals that influence food intake and energy A, CCK, leptin, and GLP-1 into the PVN alter food intake expenditure, with neural signals from the brainstem and and body weight [163]. While ICV injection of NPY into highercorticalcentres.Theimportanceofthehypothalamus thePVNcauseshyperphagiaandobesity[174],destruction inenergyhomeostasiswasfirstsuggestedbyclassiclesioning of the PVN causes hyperphagia and obesity [176]. This experiments in rodents [165]; subsequent studies have finding implies that the PVN may have an inhibitory role suggested a role of hypothalamic nuclei, such as arcuate in food intake and body weight. The LHA also receives nucleus (ARC), paraventricular nucleus (PVN), ventrome- projectionsfromtheARCandcontainstwoorexigenicneu- dial nucleus (VMN), dorsomedial nucleus (DMN), and ropeptides, melanin-concentrating hormone (MCH), and lateralhypothalamicarea(LHA)inenergyhomeostasis. orexin (hypocretin). Transgenic mice overexpressing MCH In the ARC, there are two important discrete neu- develop obesity and insulin resistance [177]. Furthermore, ronal populations: neurons which coexpress neuropeptide transgenic mice which are deficient in the prohormone Y (NPY) and agouti-related peptide (AgRP) stimulate food intake,whereasneuronscoexpressingpro-opiomelanocortin precursor of MCH or the MCH 1 receptor are lean [178]. (POMC) and cocaine- and amphetamine-regulated tran- Hypocretins1and2areproducedbythegroupsofneurons script (CART) suppress food intake (Figure1). The ARC in the LHA [179]. These neurons project extensively to is located at the base of median eminence which may be theolfactorybulb,cerebralcortex,thalamus,hypothalamus, accessed by circulating hormones likely due to its deficient brainstem, locus coeruleus, tuberomamillary nucleus, and blood-brain barrier (BBB) [166] or by carrier-mediated raphenucleus.Inadditiontotheorexigeniceffectsonfood transport. intake,orexinseemstoalsohavearoleinotherphysiological ExperimentalDiabetesResearch 7 Higher brain centres Hunger ARC Brainstem PVN ↑ONPreYx/iAgegnRi Pc ↓APOnoMreC x/iCgeAnRi cT Brain BBB Periphery V a g u s PP from PYY, GLP-1 Ghrelin ↓ pancreas ↓OXM from ↑ from intestine stomach Fasting/preprandial state (a) Higher brain centres Satiety ARC PVN ↓Orexigenic ↑Anorexigenic Brainstem NPY/AgRP POMC/CART Brain BBB Periphery V a g us PP from PYY, GLP-1 Ghrelin ↑pancreas ↑OXM from ↓ from intestine stomach Postprandial state (b) Figure1:Theguthormonesignallingtothebrainunderfasted(a)andfedstates(b).(a)Duringthefasting/preprandialstate,ghrelinrelease fromthestomachactsupontheARCandvagustostimulatehunger.(b)Inthepostprandialstate,releaseofanorectichormones,PYY,GLP-1, OXM,andPPfromintestineactupontheARC,brainstem,andvagustocausesatiety.ARC,arcuatenucleus;NPY/AgRP,neuropeptideYand agoutirelatedpeptide;POMC/CART,pro-opiomelanocortin,andcocaine-andamphetamine-regulatedtranscript;PVN,paraventricular nucleus;GLP-1,glucagon-likepeptide-1;PP,pancreaticpolypeptide;PYY,peptideYY;OXM,oxyntomodulin. functions such as regulation of blood pressure, the neu- DMNlesionscausehyperphagiaandobesity,whichsuggests roendocrinesystem,bodytemperature,andthesleep-waking a suppressive role in appetite [185]. In diet-induced mice, cycle[180].Animpairmentofhypocretinneurotransmission an approximately 40-fold increase in NPY expression is hasbeenassociatedwiththepathologyofhumannarcolepsy, observed in the DMN and VMN when compared with which is a chronic sleep disorder characterized by excessive controls [186]. In the VMN, brain-derived neurotrophic daytime sleepiness, cataplexy, hypnagogic hallucinations, factor (BDNF) is highly expressed, and VMN BDNF neu- and sleep paralysis [181]. MCH-R1 antagonists may have rons suppress food intake through MC4R signalling [187]. therapeuticpotentialforthetreatmentofobesity[182],but Increased signalling in the VMN following an oral glucose further work is required to determine if their use would load has been observed [162]. Selective deletion of BDNF be associated with adverse effects attributable to the other neurons in the VMN and DMN of adult mice results in biologicalactionsoforexin. hyperphagiaandobesity[188]. TheDMNreceivesNPY/AgRPprojectionsfromtheARC Glucose sensing plays an important role of the brain. [183]andprojectstheα-MSHfibretothePVN[162,184]. Conventionally, glucose sensing is thought to involve 8 ExperimentalDiabetesResearch glucokinase-dependentmetabolismofglucosetoATP,which to that seen in the hypothalamus [193]. POMC neurons then alters membrane excitability by modulating ATP- exist within the NTS, which show STAT-3 activation in dependent channels or transporters, such as ATP-inhibited response to leptin administration to suppress food intake K+ channels (KATP). Recent studies, however, suggest that [196].AdministrationofleptinintotheDVCsuppressesfood glucose-excited and glucose-inhibited neurones are able intake[193]. to sense glucose irrespective of such metabolic pathways. Inadditiontothehypothalamus,thevagusnerveplaysa Brain glucose sensors, specialized neurones which respond centralroleinregulatingthefeeding.Vagalafferentneurons to fluctuations in local extracellular glucose concentration, have been shown to express a variety of receptors within have been found only in a few brain regions, in particular, thebrainstem,whichincludecholecystokinin(CCK)1Rand the hypothalamus and brainstem. Hypothalamic glucose- CCK2R (at which both CCK and gastrin act [197]), Ob- sensing neurones are found in the LHA, ARC, and VMN, R [198], Y2R [29], GLP-1 [67], and GLP-2R [199], growth and responsive neurons have been identified which either hormone secretagogue receptor (GHS)-R1 where ghrelin increase firing rate (glucose-excited neurones) or decrease acts[118],andtheorexinreceptor,OX-R1[200]. firing rate (glucose-inhibited neurones) in response to ex- The vagal stretch and tension sensors detect nutrients tracellularglucose[189]. stored in the stomach. The vagus nerve also helps to transmit gut hormones signals such as CCK, ghrelin, PYY, 6.Brainstem PP, and GLP-1, which are released by anticipation of meals and the presence of food in the upper gastrointestinal Withinthebrainstem,thedorsalvagalcomplex(DVC)plays tract. Cell bodies of afferent fibres of the abdominal vagus an important role in relaying peripheral signals via vagal nerve are located in the nodose ganglia, which project afferent fibres from the gut to hypothalamus. The DVC to the DVC of brainstem. In rats, infusion of saline into has projections to the hypothalamus and higher cortical the stomach has been observed to reduce food intake to centres [190] and comprises the dorsal motor nucleus of similar extents to infusion of nutrients into the stomach vagus (DVN), area postrema (AP), and the nucleus of the [201]; this phenomenon is likely to be attributable to vagal tractussolitarius(NTS).NTSisanidealpositiontointegrate nervefunction.Thevagusnerveparticipatesintransmitting peripheralsignalsduetoitscloseproximitytotheAP,which the food-induced negative-feedback signals important for hasanincompleteBBB[163]. determining meal size. Transection of all gut sensory vagal Other than ascending brainstem-hypothalamus path- fibres results in increased meal size and meal duration, but ways,descendinghypothalamicprojectionstothebrainstem doesnotblockgastricpreload-inducedfeedingsuppression, are also important in control of food intake. α-MSH implyingthatvagalafferentsignalshaveasignificantrolein projections from POMC neurons in the ARC terminate satietyduringspontaneousmeals[202,203].Randichetal. in close anatomical proximity to neurons in the NTS, [204]utilizedextracellularrecordingsfromthevagusnerve, which respond to gastric distension [191]. Furthermore, andfoundthatittransmitsasatietysignalfromthejejunum, descending projections from the LHA to the NTS contain followingactivationbyinfusionoffattyacids. orexinandMCH,anddescendingARC-parabrachialnucleus The importance of the role of the vagus nerve in pathways have been identified [152]. The PVN projects transmitting peripheral signals has been demonstrated by regions of the midbrain such as the ventral tegmental area, vagotomyorcapsaicintreatmenttoabolishitseffect,andby Edinger-Westphalnucleus,ventrolateralperiaqueductalgray vagusnervestimulation(VNS)toenhanceitsactivity[205]. matter, reticular formation, pedunculopontine tegmental Low-frequency VNS in rats fed with a standard diet results nucleus, and dorsal raphe nucleus. The PVN also projects indecreasedfoodintakeandbodyweight[206].Compared to the prelocus coeruleus in the dorsal pons as well as the withtheshamgroup, obeseminipigs receivedVNS did not nucleus ambiguous and NTS in the ventral medulla. The gain body weight and showed decreased food intake by medial NTS receives the most extensive projections of the 18%—the effects lasted for 14 weeks [207]. Gil et al. [208] PVN,substantiallymorethantheDVNorAP[192]. reported that chronic VNS with 10Hz electrical impulses Theimportanceofthehindbraininenergyhomeostasis in rats fed with a high-fat diet significantly decreased food is highlighted by considering that chronically maintained intake and body weight gain. In their study, significant decerebrate rats, with complete high mesencephalic tran- neuronalresponsesintheNTSanddecreasedserumleptin, section, remain responsive to taste stimuli and respond to butincreasedghrelinlevels,wereobservedandalsonesfatin- intake-inhibitory feedback from the gut; however, hyper- 1levelstendedtoincreasefollowingVNS.Thissuggeststhat phagicresponsetofooddeprivationisnotobservedinthese VNSresultsinreductionsinfoodintakeandbodyweightby animals [193]. Direct delivery of leptin to the lateral or increasingbrainsatietysignalsthroughthevagalafferents. third ventricle as well as the fourth ventricle significantly The close association of the vagal efferent, sympa- suppresses food intake up to 24h after treatment [193]. The effects of various gut hormones on food intake are thetic, and enteric systems makes it difficult to selec- attenuatedbylesionsoftheareapostrema[194]orvagotomy tively manipulate the vagus nerve. Genetic approaches to [28, 29, 118, 195]. Taken together, these findings suggest modulate signalling of neurotrophin factors (e.g., BDNF brainstem-mediatedmechanismsoncontrollingfoodintake. and neurotrophin-3), which are essential for vagal afferent The expression of leptin and insulin receptors, and of development, may help to further elucidate the regulatory glucose sensing mechanisms in the brainstem, is similar roleofthevagusnerveingutphysiology[209]. ExperimentalDiabetesResearch 9 7.RewardSystems to odours and fat rich food when compared with lean subjects[220]. Inhumans,environmentalcues,cognitive,reward,andemo- tional factors play an important role in food intake which mayoverridehomeostaticrequirements[210].Thecorticol- 8.NutrientsanditsRelatedSignals imbicpathwaysareresponsibleforreward-associatedfeeding ModulatingAppetite behaviour, which include the striatum, ventral tegmental area, nucleus accumbens, insular cortex, anterior cingulate Althoughlow-energydensitydietsinduceshort-termweight cortex, and orbitofrontal cortex. The orbitofrontal cortex loss [221], they are usually associated with rebound weight is associated with regulating gustatory, olfactory, visual, gain.SomedietarypatternssuchastheMediterranean-type and somatosensory function, and sensory factors, such as dietareassociatedwithadecreasedrateofcardiacdeathand taste and smell, and has an important role in reward- nonfatal myocardial infarction compared with a Northern relatedfeeding[211].Inpatientswithfrontotemporallobar European or North American dietary pattern [222]. In the degeneration,hyperphagiaisreportedtobeassociatedwith Mediterranean diet, monounsaturated fat is substituted for atrophyintheanterolateralorbitofrontalcortex[212]. saturated and trans-fats, and intake of fruits, vegetables, The endocannabinoid and opioid systems have wide fibre,andwholegrainsarehigh,isrecommended[223].Itis, receptor distributions within the CNS and play important therefore, interesting to consider whether specific nutrients rolesinreward-relatedfeeding[213].Administrationofaμ- withindietsareabletomodulatefoodintake,inadditionto opioid receptor agonist into the nucleus accumbens prefer- effectsassociatedwiththeirdirectnutritionalvalue. entially stimulates intake of high-fat diet regardless of diet The amino acid L-Glutamate is involved in multiple preferenceatbaseline,whenbothfatandcarbohydratediets physiologic functions, which include taste perception, car- are displayed simultaneously [214]. Increased expression of bohydratemetabolism,andexcitatoryneurotransmissionin orexin in the hypothalamus has been observed following the brain [224]. L-Glutamate stimulates its receptors in gut administrationofopioidμ-receptoragonistsintothenucleus epithelialcells,whichactivatecerebralregionssuchasbasal accumbens[215].Preadministrationofacannabinoidrecep- ganglia, limbic systems, and hypothalamus through vagal tor (CB1) antagonist prevents the orexigenic effect of the afferent nerve [225]. Kondoh et al. [226] reported that rats endocannabinoidagonist,anandamideonfoodintake[213]. with chronic ad libitum administration of monosodium Leptin has been shown to reduce endocannabinoid levels L-glutamate had reduced weight gain, fat deposition, and in the hypothalamus [216]. This suggests that hypothala- plasmaleptinconcentrationswhencomparedwithcontrols. mic endocannabinoids may act via CB1 to increase food In rodents, long-chain omega-3 polyunsaturated fatty intake through a leptin-regulated mechanism. The nucleus acids supplementation has been shown to improve obesity accumbens (NAs) is a key region of limbic pathway and [227]. In a recent study using a mouse model of diet- may be implicated in regulation of hedonistic feeding and induced obesity, ICV administration of unsaturated fatty homeostaticfeeding[210]. acids reduced hypothalamic inflammation, hypothalamic The ventral striatum and population of dopamine neu- andwholebodyinsulinresistance,andbodyadiposity[228]. ronsofthesubstantianigraareinvolvedintherewardsystem Free fatty acids exert insulin-like effects in key brain areas in human and nonhuman primates. The ventral striatum for energy homeostasis, including the ARC, possibly by receivesinputfromtheorbitofrontalcortexandanteriorcin- favouringintracellularaccumulationofthelong-chainfatty gulatecortex,whichincludetheNAandthebroadcontinuity acyl-CoA(LCFA-CoA)[229]. Unsaturated free fatty acid may, therefore, be beneficial betweenthecaudatenucleusandtheputamenandtheolfac- totreatobesity,althoughevidenceinhumanisstilllimited. torytubercle[217].Dopamineappearstobeassociatedwith Fructose is being increasingly used in processed foods reward-related food intake and with behaviours required withintheWesterndiet.However,itseffectsmaybedistinct to maintain feeding essential for survival. Mice lacking to those of the related sugar, glucose. As glucose levels dopamine, caused by the selective inactivation of tyrosine enteringtothebrainincrease,foodintakeissuppressed.In hydroxylase, develop fatal hypophagia; and replacement of contrast, fructose increases food intake when metabolized dopamine in these animals to the caudate putamen or NA in the brain. Fructose has the opposite effect of glucose on restores preference for sucrose or palatable chow [218]. theAMPactivatedkinase/malonyl-CoAsignalingsystemand However, dopamine may have more complex effects on therebyenhancesfeedingbehaviour[230]. feeding,sincedopaminesignallingintheDMNandARCof Serotoninhasaroleinappetitecontrol.5HT-containing hypothalamusmayinhibitfoodintake[149]. neurons are organized into nine nuclei (B1–B9) which In a recent positron emission tomography study, meal are located in the midbrain and hindbrain. In particular, ingestion was associated with greater activation of the the midbrain dorsal raphe (B7) contains a substantial midbrain and middle-dorsal insula, and lesser activation portion of the total brain 5HT neurons, with distinct in the posterior cingulate cortex, temporal cortex, and or- projectionstohypothalamicnucleiandotherfeeding-related bitofrontal cortex following a meal in obese individuals forebrain areas [231]. 5-HT-stimulating drugs reduce food when compared with lean individuals [219]. In addition, intake partly mediated through the 5-HT receptor [232]. 2C a study utilizing functional magnetic resonance imaging Although its effects on eating behaviour remain to be (MRI)suggestedthatobeseindividualshadgreaterresponses characterised,lorcaserin,aselective5-HT receptoragonist 2C 10 ExperimentalDiabetesResearch Table1:Thesummaryoftheroleofguthormonesonappetiteregulationandotheractions. Guthormones Feeding Receptor Majorsecretionsite Otheractions Delaysgastricemptying,inhibitsgallbladdercontraction,pancreatic PYY ↓ Y2 Lcellsingut 3–36 exocrinesecretions,andgastricacidsecretion Delaysgastricemptying,attenuatespancreaticexocrinesecretion, PP ↓ Y4,Y5 PPcellsinpancreas andinhibitsgallbladdercontraction Incretin,decreasesbloodglucose,delaysgastricemptying,and GLP-1 ↓ GLP-1 Lcellsingut neurotrophiceffect OXM ↓ GLP-1 Lcellsingut Inhibitsgastricacidsecretionandgastricemptying Glucagon ↓ GCGR Pancreaticαcells Enhancingphysiologicalresponsetostress Gallbladdercontraction,relaxationofsphincterofOddi,and CCK ↓ CCK1,2 Icellofsmallintestine pancreaticenzymesecretion Growthhormonesecretion,promotesgastricmotility,vasodilatation, Ghrelin ↑ GHS stomach andincreasescardiaccontractility Amylin ↓ AMY1-3 pancreaticβcells Adipositysignals PYY:peptideYY,PP:pancreaticpolypeptide,GLP-1:glucagon-likepeptide-1,OXM:oxyntomodulin,CCK:cholecystokinin,GCGR:glucagonreceptor. isreportedtobeanovelantiobesitydrugreducingbothfood of weight loss with an acceptable level of risk. RYGB intakeandbodyweight. is thought to achieve its beneficial effects through the Tasteaffectsfoodpreferenceandintake.Lingualproteins BRAVE effects: Bile flow alteration, Reduction of gastric CD36 and GPR120 are reported to be responsible for size, Anatomical gut rearrangement and altered flow of the spontaneous preference for lipid-rich foods [233] and nutrients, Vagal manipulation, and subsequent Enteric gut have been identified in human taste buds [234]. The gut hormone modulation [241]. A decrease in levels of the hormones such as GLP-1 and CCK and neurotransmitters orexigenichormoneghrelin,andanincreaseinlevelsofthe are also produced locally in taste buds [235]. Sweet and anorectic hormones PYY and GLP-1 have been observed umami taste are mediated by T1R family (T1R1, T1R2, following bypass surgery [111, 242, 243]. An increase in T1R3) which belongs to family C of GPCRs including energy expenditure may play a role in part in weight loss metabotropic glutamate receptors, calcium sensing recep- aftergastricbypasssurgery[244].Moreover,microbialshifts tors, and V2r pheromone receptors. In the intestine, there towards substantially higher concentrations of Proteobace- are different sweet taste cells (enteroendocrine, brush cells) ria, specifically Enterobacter hormaechei, are demonstrated within the epithelial layer. These sweet taste receptors may following RYGB surgery [241]. Gastric bypass surgery is signal through vagal afferent fibres to alter food intake and associated with greater improvements in glycaemic control delay gastric emptying [236]. It has been shown that leptin inpatientswithtype2diabetes,whencomparedwithgastric selectively suppresses sweet taste sensitivity or taste cells bandingprocedures[245].Furthermoretheseimprovements through Ob-RB, whereas endocannabinoids enhance sweet inglycaemicstatusoftenprecedeweightloss,whichimplies tastesensitivityoftastecellsviaCB1receptor[237]. that bypass surgery may have effects in ameliorating type 2 diabeteswhichareadditionaltotheireffectsonbodyweight. 9.BariatricSurgery Whereas pharmacological and behavioural treatments are 10.GutMicrobiota usuallyassociatedwithweightlossfollowedbyweightregain, bariatricsurgeryprovidesweightlossforatleast15years,in A potential association between gut microbiota and the patientswithobesity[238,239].Bariatricsurgeryisclassified pathogenesis of obesity has been recently recognised [246]. into 3 types of surgical procedures; malabsorptive surgery, The human gut harbours a large number of 1000 to 1150 restrictive surgery, and mixed procedures. Malabsorption- bacterial species collectively termed gut microbiota [247]. based procedures include the jejuno-ileal bypass, which Adult germ-free mice have 40% less total body fat than results in decreased nutrients absorption by shortening the micewithnormalmicrobiota;andreplacingthemicrobiota functional small bowel length, and by allowing nutrients in adult germ-free mice is associated with a 60% increase to pass directly from the proximal jejunum to the terminal in body fat content and insulin resistance within 14 days ileum. Restrictive bariatric surgery includes the laparo- of replacement [248]. In contrast to mice with normal scopic application of an adjustable gastric band, which is gut microbiota, germ-free mice may be protected against associated with lower comorbidity when compared with high fat diet-induced metabolic changes; increased fatty malabsorption-based procedures [240]. Roux-en-Y gastric acid metabolism, elevated levels of fasting-induced adipose bypass (RYGB) is a combined restrictive and malabsorp- factor,Fiaf,knownasangiopoietin-likeprotein-4,asecreted tive procedure, which yields long-term, sustained period lipoprotein lipase inhibitor, and increased AMP-activated

Description:
Y2 receptor. The PYY secretion pattern suggests a role in satiety. Circulating PYY . renal clearance that rapidly inactivate and remove GLP-1 from plasma . rats but causes a compensatory increase in meal frequency. [126]. A 2-week . blood-brain barrier (BBB) [166] or by carrier-mediated transport.
See more

The list of books you might like

Most books are stored in the elastic cloud where traffic is expensive. For this reason, we have a limit on daily download.