Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Available online at www.sciencedirect.

com

ScienceDirect

Targeting gastrointestinal nutrient sensing mechanisms


to treat obesity
Mariana Norton and Kevin G Murphy

Gut hormones have important roles in the regulation of appetite Carbohydrate


and glucose homeostasis. Understanding how macronutrient Glucose, a common carbohydrate and break-down prod-
sensing in the gastrointestinal tract modulates gut hormone uct of larger carbohydrates, potently stimulates the
release may reveal novel pharmacological or dietary release of the incretin and appetite-regulating hormones
approaches to metabolic disease. In this short review we gastric-inhibitory peptide (GIP) and glucagon-like pep-
discuss the mechanisms by which the gut senses tide 1 (GLP-1) when administered orally but not intrave-
macronutrients and the products of macronutrient digestion, nously [4!,5,6]. In the small intestine, this effect is pri-
and their putative utility in treating obesity and related marily mediated by the sodium-dependent glucose co-
conditions. transporter SGLT-1, which is expressed in both K-cells
and L-cells; GIP and GLP-1 expressing EECs, respec-
Address
tively [5]. The entry of sodium, coupled to glucose,
Section of Endocrinology and Investigative Medicine, Department of depolarizes the cell membrane, opening voltage gated
Medicine, Imperial College London, W12 0NN, UK calcium channels and stimulating hormone exocytosis
[4!,7!]. Knockout of SGLT-1 in mice abolishes peak
Corresponding author: Murphy, Kevin G (k.g.murphy@imperial.ac.uk) glucose-induced GIP and GLP-1 secretion [8,9], but at
later time points, circulating levels of GLP-1 and the
Current Opinion in Pharmacology 2017, 37:16–23 anorectic gut hormone peptide YY (PYY) are increased in
This review comes from a themed issue on Endocrine and metabolic
both SGLT-1"/" and antagonist treated mice [9], sug-
diseases gesting alternative glucose sensing mechanisms in the
Edited by Gavin Bewick
distal gut. Specifically inhibiting intestinal SGLT-1 in
mice also resulted in a trend for increased GLP-1 follow-
ing a glucose challenge, but importantly highlighted
potential gastrointestinal side effects consistent with
http://dx.doi.org/10.1016/j.coph.2017.07.005
the malabsorption observed in knockout mice and
humans with SGLT-1 inactivating mutations [8,10].
1471-4892/ã 2017 Elsevier Ltd. All rights reserved.

In humans, SGLT-1 inhibition resulted in decreased


postprandial GIP and increased postprandial GLP-1
levels [11]. Longer term human studies have used
antagonists such as LX4211 that also target SGLT-2.
Introduction Inhibiting SGLT-1/2 in diabetic patients improved gly-
Obesity is associated to comorbidities including cancer cemic control and resulted in a trend for decreased
and diabetes [1]. The most effective therapy is gastric weight and increased GLP-1 and PYY levels [12].
bypass but it is expensive and invasive. Some of the Decreased weight gain, regardless of increased food
benefits of gastric bypass are attributed to changes in intake was observed in animal models [13]. As an adjunct
patients’ gut hormone profiles [2]. Gut hormones, therapy to insulin in diabetic patients, LX4211 improved
secreted by enteroendocine cells (EECs), are crucial in glycemic control, increased post-prandial PYY levels and
appetite regulation. They respond to a variety of stimuli, promoted weight loss [14]. Some FDA approved SGLT-
such as nutrients in the lumen, which are detected by a 2 inhibitors also lead to weight loss [15]. Whether the gut
wide range of sensors [3!]. These sensors may be useful hormone effects resulting from SGLT-1 antagonism
anti-obesity targets capable of modulating endogenous contribute to this needs further investigation. Overall,
gut hormone secretion to promote weight loss. This SGLT-1 partial antagonism, to minimise malabsorption
review will focus on extracellular nutrient sensors side effects, may be a better anti-obesity strategy than
expressed in the gut, their role in gut hormone secretion SGLT-1 agonism.
and their potential use as anti-obesity targets (Figure 1).
Glucose transporter 2 (GLUT-2) may also mediate glu-
Macronutrients are classified as carbohydrates, fats or cose-induced gut hormone secretion. GLUT-2 inhibition
proteins. Upon ingestion, these macronutrients are con- and knockout in mice reduced glucose-stimulated GLP-1
verted by endogenous and bacterial enzymes to metab- secretion by #11% and 55% respectively [4!,16]. Intesti-
olites that are detected by sensors in the gut. nal specific GLUT-2 knockout did not alter basal GLP-1

Current Opinion in Pharmacology 2017, 37:16–23 www.sciencedirect.com


Targeting nutrient sensing mechanisms to treat obesity Norton and Murphy 17

Figure 1

GPRC6A
Orexigenic
Amino Acids T1R1/T1R3

B0AT1 Ghrelin
Protein
CaSR
Peptides and P/D1-cell
Oligopeptides GPR93

PEPT-1 Anorectic

FFAR1 CCK
LCFA
FFAR4
I-cell
Fat (TG)
MAG GPR119

GIP
SGLT-1
K-cell
Glucose GLUT-2

TIR2/TIR3
PYY
GLUT5 GLP-1
Carbohydrate Fructose

FFAR2 L-cell
SCFA
FFAR3
Current Opinion in Pharmacology

Simplified overview of gastrointestinal nutrient sensors of the gut. Macronutrients are broken-down by endogenous and bacterial enzymes in
the gut, producing metabolites which can be detected by G-protein coupled receptors and transporters. This leads to the modulation of the
secretion of appetite-regulating hormones by enteroendocrine cells. This schematic does not include intracellular nutrient sensors. Abbreviations:
long chain fatty acids (LCFA), 2-monoacylglycerols (MAG), short chain fatty acids (SCFA), G protein-coupled receptor family C group 6 member A
(GPRC6A), amino acid sensing heterodimer taste receptor type 1 member 1/taste receptor type 1 member 3 (T1R1/T1R3), neutral amino acid
transporter (B0AT1), calcium sensing receptor (CaSR), G protein-coupled receptor 93 (GPR93), peptide transporter 1 (PEPT-1), free fatty acid
receptor 1 (FFAR1), free fatty acid receptor (FFAR 4), G protein-coupled receptor 119 (GPR119), sodium-dependent glucose co-transporter 1
(SGLT-1), glucose transporter 2 (GLUT-2), sweet taste receptor taste receptor type 1 member 3/taste receptor type 1 member 3 (T1R2/T1R3),
glucose transporter 5 (GLUT-5), free fatty acid receptor 2 (FFAR 2), free fatty acid receptor 3 (FFAR 3), cholecystokinin (CCK), gastric inhibitory
polypeptide (GIP), glucagon-like peptide 1 (GLP-1), peptide YY (PYY).

levels but resulted in lower L-cell density, compensated The clinical utility of targeting the heterodimer sweet
for with an 80% increase in GLP-1 content per cell [17!], taste receptor T1R2/T1R3 may also be limited. Expres-
suggesting the long-term effects of targeting GLUT-2 sion of its subunits is low in K-cells and L-cells, and
may differ from the acute effects. Additionally, human studies in perfused rat intestine required toxic levels of
mutations in GLUT-2 result in Fanconi-Bickel syn- sweeteners to stimulate GLP-1 release [20]. In humans,
drome, suggesting it may not be a useful target in the sweeteners which activate the receptor failed to increase
treatment of obesity [18]. gut hormone release [21]. Additionally, lactisole, a T1R2/
T1R3 antagonist, failed to suppress the secretion of
The essential role of SGLT-1 and the contributory role of incretins in response to glucose in one study [22] but
GLUT-2 in GLP-1 secretion is in agreement with math- not in another [23].
ematical spatial models and transporter knockout models,
but the effect of GLUT-2 knockout on glucose-induced ATP-sensitive potassium channels (KATP) on EECs have
GIP secretion is less clear [8,16,19]. Overall, SGLT-1 also been suggested as potential sensors due to their role
appears more important than GLUT-2 in peak glucose- in b-cell glucose sensing. In rat perfused intestine, KATP
induced gut hormone secretion. inhibition increases GLP-1 release [4!]. However,

www.sciencedirect.com Current Opinion in Pharmacology 2017, 37:16–23


18 Endocrine and metabolic diseases

sulfonylurea treatment, which leads to KATP channel high fat diet (HFD) in DIO but not DIO-resistant rats
closure, failed to affect gut hormone secretion in humans [39], and in humans, expression is positively correlated
during an oral glucose tolerance test [24]. In addition, with BMI [34].
individuals with KATP channel mutations have neonatal
diabetes, but normal baseline and glucose-stimulated FFAR4 agonists inhibit ghrelin secretion in vitro; an
GLP-1 levels, questioning its importance in mediating effect lost following FFAR4 knock-down [40]. In vivo,
gut hormone release [25]. FFAR4"/" mice have raised fasting plasma ghrelin levels
but are still responsive to triglyceride’s inhibitory effect
There is also evidence of fructose sensing in the gut [26] on ghrelin secretion. FFAR4 may therefore provide a
and short chain fatty acids, products of carbohydrate baseline inhibitory tone on ghrelin secretion, but other
fermentation in the colon, [27] influencing gut hormone systems may mediate triglyceride-induced ghrelin sup-
release and appetite. pression [31]. FFAR4"/" mice also have blunted CCK
and GIP responses, but a normal GLP-1 response to orally
Fat gavaged oil [29,41]. In humans, an FFAR4 loss-of-func-
Fatty acid products of fat digestion can act intracellularly tion mutation is associated with increased obesity risk
to alter metabolic processes [28]. However, there is cur- [42].
rently much interest in the gut-based cell surface receptor
systems which detect lipids. Triglycerides from dietary Overall, FFAR4 may be a possible target to modulate gut
fat are metabolised by pancreatic lipase into long chain hormones. FFAR4 internalisation is observed in response
fatty acids, which are detected by fatty acid receptors 1 to agonist administration in vitro, but the system appears
(FFAR1) and 4 (FFAR4), and 2-monoacylglycerol, to re-sensitise quickly [43]. Additionally, though mice
detected by G-protein coupled receptor 119 (GPR119). with FFAR4 agonist supplemented food did not show
These receptors are expressed in EECs and are thought changes in basal GLP-1 levels or body weight
to modulate the release of GLP-1, PYY, GIP and chole- [29,41,44,45], colonic administration of an alternative
cystokinin (CCK), which is anorectic, and perhaps agonist in mice did increase plasma GLP-1 levels [46],
ghrelin, which is orexigenic, in response to dietary fat suggesting the region of the gastrointestinal tract targeted
[29–31]. They thus may also have potential as anti-obesity is important.
targets.
GPR119, a constitutively active receptor, is expressed in
FFAR1"/" mice have blunted CCK, GIP and perhaps K-cells and L-cells [47,48]. In humans, GPR119 duodenal
GLP-1 responses to fat ingestion, FFAR1 is therefore expression is inversely correlated to BMI [34], though
important in mediating the gut hormone response to intraduodenal administration of lipids increases duodenal
dietary fat but is not essential, and likely functions expression [49].
alongside other sensors [29,32,33]. In humans, FFAR1
is expressed more than FFAR4 and GPR119 in the small GPR119 agonists stimulate GLP-1 and GIP release. The
intestine [32,34]. receptor is also thought to act in synergy with FFARs to
potentiate GLP-1 secretion [47,48]. The proportion of L-
Known endogenous ligands of FFAR1 signal via Gaq. In cells responsive to GPR119 agonists varies along the
vivo FFAR1 is thought to synergistically interact with Gas length of the gut, suggesting GPR119 may be expressed
coupled receptors such as GPR119, the bile acid receptor in a specific L-cell subpopulation [50]. In accord with this,
TGR5 and the GIP receptor to potentiate GLP-1 (but not GPR119 agonists-stimulated GLP-1 release is blunted in
GIP) release. This can be recapitulated in vitro using ileal and colonic cultures, but not in proximal gut cultures,
second generation FFAR1 agonists that signal via both from mice lacking GPR119 specifically in L-cells. In vivo,
Gaq and Gas [30,35]. Second generation agonists may L-cell specific knockout of GPR119 reduced the GLP-1
therefore be more effective at promoting weight loss than response to lipid gastric gavage [50] and global knockout
Gaq coupled first generation agonists such as TAK-875, blunted GLP-1 and GIP responses to oral triglycerides
which failed to show any clinically significant effects on [48].
body weight in clinical trials stopped due to liver toxicity
concerns [36]. Gut hormone release is likely driven by The incretin effects of GPR119 agonism have made
basolaterally expressed FFAR1 in EECs, thus fatty acid GPR119 a popular therapeutic target [51]. However, thus
absorption is an important step in lipid sensing; an impor- far, these agonists have failed to translate to the clinic.
tant consideration for the design of drugs aimed at The agonist JNJ-38431055 increased GLP-1 and GIP
exploiting FFAR1 [37!]. secretion after a meal challenge, but the resulting glucose
lowering effect was insufficient to be clinically useful, and
FFAR4 is one of the most highly expressed nutrient there was no effect on appetite [52]. Another agonist,
receptors in the large intestine [38!]. Like FFAR1, its GDK263, increased PYY levels by fivefold but did not
expression in the small intestine increases in response to a affect food intake or hunger [53]. Several other clinical

Current Opinion in Pharmacology 2017, 37:16–23 www.sciencedirect.com


Targeting nutrient sensing mechanisms to treat obesity Norton and Murphy 19

candidates have also been discontinued [54], questioning The umami taste receptor T1R1/T1R3 in humans is
the potential of GPR119 as an anti-obesity drug target. particularly sensitivity to L-glutamate, whereas the rodent
receptor responds to a wider range of amino acids [78,79].
Amino acids can stimulate CCK release via T1R1/T1R3
Protein in rodent in vitro models [79]. In pigs, signalling compo-
Protein, the most satiating macronutrient, stimulates the nents of T1Rs are up-regulated in response to HPDs [80].
release of various anorectic hormones including: PYY Whether the human receptor is also able to mediate gut
[55,56], CCK [57,58], GLP-1 [56,59–61] and GIP [57], hormone secretion is unknown. Additionally, there is
which are thought to be important in protein’s satiating redundancy in the system; taste cells from T1R1"/" or
effect. Therefore protein metabolite nutrient sensors are T1R3"/" mice partially responded to L-amino acids,
potential anti-obesity targets. which may reflect the detection of L-amino acids by
metabotropic glutamate receptors [81–83], and similar
Three well characterised promiscuous L-amino acid sens- redundancy is possible in the gut.
ing receptors: G protein-coupled receptor family C group
6 member A (GPRC6A), umami taste receptor T1R1/ GPR142 is particularly activated by the essential aro-
T1R3 and the calcium sensing receptor (CaSR); a more matic amino acids L-phenylalanine and L-tryptophan.
recently identified amino acid receptor: GPR142; and the The concentrations of luminal amino acids following
peptide receptor GPR93 are all present in the gut and protein digestion are within the range of receptor activa-
appear to sense the products of protein digestion. tion [84!!]. GPR142"/" mice have a similar food intake
and body weight to control mice but the effect of agonists
GPRC6A is activated by basic amino acids which trigger on food intake and body weight remain to be explored
GLP-1 secretion in vitro in the GLUTag L-cell line, but [85!!]. There is evidence that GPR142 activation can
not in primary cultures of the mouse small intestine modulate gut hormone release. In mice it is expressed in
[62,63]. Data from GPRC6A"/" mice suggest it is not gastrin-releasing G-cells and in K-cells [31,85!!,86]. L-
necessary for the appetite-reducing effects of high protein tryptophan simulates GIP and insulin secretion, and
diets or L-arginine, or for L-arginine-induced or L-Orni- improves glucose tolerance in rodents via GPR142
thine-induced GLP-1 secretion [64,65!,66]. Additionally, [85!!,87]. Additionally, oral administration of a
in the majority of humans, particularly Europeans and GPR142 agonist increased plasma GLP-1 in control mice
Asians, the receptor tends to be retained intracellularly, but not GPR142"/" mice [85!!]. The importance of
putting into question its physiological role [67]. GPR142 in energy and glucose homeostasis thus requires
further investigation.
GPRC6A is closely related to the CaSR, making it possi-
ble to target both receptors with one agent. The CaSR is GPR93 is expressed more than other amino acid or fatty
allosterically modulated by aromatic amino acids and acid nutrient sensor in the small intestine [38!]. It is found
oligopeptides. CaSR activation has been associated with on EECs including L-cells and G-cells [38!,88,89]. Over-
the secretion of various anorectic hormones in vitro, expression in STC-1 cells increases the transcription and
including CCK and GLP-1 [68,69,70,71!!]. CaSR antag- release of CCK in response to protein hydrolysate [90]
onism decreased GLP-1, GIP and PYY secretion from and in vivo its expression may be responsive to changes in
small intestinal loops [72]. However, variable effects of diet [89]. However, in primary cell culture, receptor
CaSR activation on ghrelin secretion have been reported knockout does not impair peptone-induced GLP-1 secre-
[73]. In vivo oral administration of a CaSR agonist tion [70].
decreased food intake in mice [74], but at high concen-
trations caused emesis in mink, highlighting a potential Some amino acid and peptide transporters can act as
side effect of targeting this receptor [75]. nutrient sensors that stimulate gut hormone release,
including peptide transporter 1 (PEPT-1) and the
The CaSR undergoes agonist-driven insertional signal- sodium-dependent transporter of neutral amino acids,
ling, making it highly resistant to functional desensitisa- B0AT1, which is enriched in L-cells [91,92].
tion [76]. This, alongside its ability to stimulate the
secretion of a variety of anorectic hormones, make it PEPT-1 is a proton coupled di-peptide and tri-peptide
an interesting anti-obesity pharmacological target. How- transporter expressed on the intestinal brush border.
ever, it is widely expressed, crucial in calcium homeosta- Peptide transport through PEPT1 can lead to membrane
sis and has multiple roles in the gastrointestinal tract depolarisation, opening of calcium channels and subse-
which could lead to several side effects if targeted quent secretion of GLP-1 in vitro [70,93]. It may also
pharmacologically. In addition, mice with a gain-of-func- indirectly mediate the secretion of gut hormones such as
tion mutation showed no difference in food intake or CCK [94]. Expression and activity of the transporter is
body weight, but did have impaired glucose tolerance influenced by dietary protein content [95] and may influ-
and hypocalcaemia [77]. ence food intake in response to a HPD [96].

www.sciencedirect.com Current Opinion in Pharmacology 2017, 37:16–23


20 Endocrine and metabolic diseases

B0AT1"/" animals fed a HFD have lower abdominal 2. Meek CL, Lewis HB, Reimann F, Gribble FM, Park AJ: The effect
of bariatric surgery on gastrointestinal and pancreatic peptide
white adipose tissue and weight gain, increased basal hormones. Peptides 2016, 77:28-37.
levels of GIP and GLP-1, and improved glycaemic con- 3. Gribble FM, Reimann F: Signalling in the gut endocrine axis.
trol. This phenotype may reflect the reduced absorption ! Physiol Behav 2017, 176:183-188.
Recent update on EECs and L-cell nutrient sensing.
of protein digest products. These animals are also slightly
hyperphagic, perhaps in an attempt to meet their amino 4. Kuhre RE, Frost CR, Svendsen B, Holst JJ: Molecular
! mechanisms of glucose-stimulated GLP-1 secretion from
acid requirements [97]. B0AT1 human mutations result in perfused rat small intestine. Diabetes 2015, 64:370-382.
Hartnup disorder, suggesting it is perhaps a less than Experiments deciphering the mechanism by which glucose stimulates
GLP-1 secretion.
optimal anti-obesity therapy target [98].
5. Saltiel MY, Kuhre RE, Christiansen CB, Eliasen R, Conde-
Frieboes KW, Rosenkilde MM, Holst JJ: Sweet taste receptor
Conclusion activation in the gut is of limited importance for glucose-
stimulated GLP-1 and GIP secretion. Nutrients 2017, 9.
The weight loss observed in gastric bypass patients and
patients treated with incretin mimetics suggest gut hor- 6. Herrmann C, Goke R, Richter G, Fehmann HC, Arnold R, Goke B:
Glucagon-like peptide-1 and glucose-dependent insulin-
mone signalling is a viable anti-obesity target. Evidence releasing polypeptide plasma levels in response to nutrients.
suggests that exploiting signals from multiple gut hor- Digestion 1995, 56:117-126.
mones concurrently would cause greater appetite sup- 7. Kuhre RE, Bechmann LE, Wewer Albrechtsen NJ, Hartmann B,
pression, and perhaps fewer side effects [99,100]. Target- ! Holst JJ: Glucose stimulates neurotensin secretion from the
rat small intestine by mechanisms involving SGLT1 and
ing nutrient sensors to stimulate the endogenous GLUT2, leading to cell depolarization and calcium influx. Am J
production of multiple gut hormones thus seems a prom- Physiol Endocrinol Metab 2015, 308:E1123-E1130.
Elucidating the mechanisms underlying glucose induced neurotensin
ising avenue. However, there are a number of challenges secretion.
to overcome, including understanding where in the gut 8. Gorboulev V, Schurmann A, Vallon V, Kipp H, Jaschke A,
such nutrients are acting, how they or mimetics can be Klessen D, Friedrich A, Scherneck S, Rieg T, Cunard R, Veyhl-
delivered to these regions, administration regimens that Wichmann M et al.: Na(+)-D-glucose cotransporter SGLT1 is
pivotal for intestinal glucose absorption and glucose-
avoid desensitisation of the system, and the potential for dependent incretin secretion. Diabetes 2012, 61:187-196.
side effects. Further work is required to better understand 9. Powell DR, Smith M, Greer J, Harris A, Zhao S, DaCosta C,
these nutrient sensing systems to enable their effective Mseeh F, Shadoan MK, Sands A, Zambrowicz B, Ding Z-M:
exploitation for the prevention or treatment of obesity. LX4211 increases serum glucagon-like peptide 1 and peptide
YY levels by reducing sodium/glucose cotransporter 1
(SGLT1)-mediated absorption of intestinal glucose. J
Pharmacol Exp Ther 2013, 345:250-259.
Funding sources
M.N. is funded by the Imperial College Ghatei Student- 10. Powell DR, Smith MG, Doree DD, Harris AL, Greer J, DaCosta CM,
Thompson A, Jeter-Jones S, Xiong W, Carson KG, Goodwin NC
ship. The Section of Endocrinology and Investigative et al.: LX2761, a sodium/glucose cotransporter 1 inhibitor
Medicine is funded by grants from the Medical Research restricted to the intestine, improves glycemic control in mice.
J Pharmacol Exp Ther 2017, 362:85-97.
Council, Biotechnology and Biological Sciences Research
Council, National Institute for Health Research, an Inte- 11. Dobbins RL, Greenway FL, Chen L, Liu Y, Breed SL, Andrews SM,
Wald JA, Walker A, Smith CD: Selective sodium-dependent
grative Mammalian Biology (IMB) Capacity Building glucose transporter 1 inhibitors block glucose absorption and
Award, an FP7-HEALTH-2009-241592 EuroCHIP grant impair glucose-dependent insulinotropic peptide release. Am
J Physiol — Gastrointest Liver Physiol 2015, 308:G946-G954.
and is supported by the NIHR Biomedical Research
12. Zambrowicz B, Freiman J, Brown PM, Frazier KS, Turnage A,
Centre Funding Scheme. The views expressed are those Bronner J, Ruff D, Shadoan M, Banks P, Mseeh F, Rawlins DB
of the authors and not necessarily those of the NHS, the et al.: LX4211, a dual SGLT1/SGLT2 inhibitor, improved
NIHR or the Department of Health. glycemic control in patients with type 2 diabetes in a
randomized, placebo-controlled trial. Clin Pharmacol Ther
2012, 92:158-169.
Conflict of interest statement 13. Powell DR, DaCosta CM, Smith M, Doree D, Harris A, Buhring L,
Nothing declared. Heydorn W, Nouraldeen A, Xiong W, Yalamanchili P, Mseeh F
et al.: Effect of LX4211 on glucose homeostasis and body
composition in preclinical models. J Pharmacol Exp Ther 2014,
350:232-242.
Acknowledgements
14. Sands AT, Zambrowicz BP, Rosenstock J, Lapuerta P, Bode BW,
A special thank you to David Leonard for his drawing of the gut. Garg SK, Buse JB, Banks P, Heptulla R, Rendell M, Cefalu WT
et al.: Sotagliflozin, a dual SGLT1 and SGLT2 inhibitor, as
adjunct therapy to insulin in type 1 diabetes. Diabetes Care
References and recommended reading 2015, 38:1181-1188.
Papers of particular interest, published within the period of review,
have been highlighted as: 15. Ridderstråle M, Andersen KR, Zeller C, Kim G, Woerle HJ,
Broedl UC: Comparison of empagliflozin and glimepiride as
! of special interest add-on to metformin in patients with type 2 diabetes: a 104-
!! of outstanding interest week randomised, active-controlled, double-blind, phase
3 trial. Lancet Diabetes Endocrinol 2014, 2:691-700.
1. Trends in adult body-mass index in 200 countries from 1975 to 16. Cani PD, Holst JJ, Drucker DJ, Delzenne NM, Thorens B,
2014: a pooled analysis of 1698 population-based Burcelin R, Knauf C: GLUT2 and the incretin receptors are
measurement studies with 19 2 million participants. Lancet involved in glucose-induced incretin secretion. Mol Cell
2016, 387:1377-1396. Endocrinol 2007, 276:18-23.

Current Opinion in Pharmacology 2017, 37:16–23 www.sciencedirect.com


Targeting nutrient sensing mechanisms to treat obesity Norton and Murphy 21

17. Schmitt CC, Aranias T, Viel T, Chateau D, Le Gall M, Waligora- directly mediates long-chain fatty acid-induced secretion of
! Dupriet A-J, Melchior C, Rouxel O, Kapel N, Gourcerol G, cholecystokinin. Gastroenterology 2011, 140:903-912.
Tavitian B et al.: Intestinal invalidation of the glucose
transporter GLUT2 delays tissue distribution of glucose and 34. Little TJ, Isaacs NJ, Young RL, Ott R, Nguyen NQ, Rayner CK,
reveals an unexpected role in gut homeostasis. Mol Metab Horowitz M, Feinle-Bisset C: Characterization of duodenal
2017, 6:61-72. expression and localization of fatty acid-sensing receptors in
First intestinal specific GLUT-2 knockout model demonstrating impor- humans: relationships with body mass index. Am J Physiol
tance of GLUT-2 in L-cell function. Gastrointest Liver Physiol 2014, 307:G958-G967.

18. Santer R, Steinmann B, Schaub J: Fanconi-bickel syndrome — a 35. Hauge M, Vestmar MA, Husted AS, Ekberg JP, Wright MJ, Di
congenital defect of facilitative glucose transport. Curr Mol Salvo J, Weinglass AB, Engelstoft MS, Madsen AN, Luckmann M,
Med 2002, 2:213-227. Miller MW et al.: Gpr40 (FFAR1) — combined Gs and Gq
signaling in vitro is associated with robust incretin
19. Roder PV, Geillinger KE, Zietek TS, Thorens B, Koepsell H, secretagogue action ex vivo and in vivo. Mol Metab 2015, 4:3-
Daniel H: The role of SGLT1 and GLUT2 in intestinal glucose 14.
transport and sensing. PLOS ONE 2014, 9:e89977.
36. Kaku K, Enya K, Nakaya R, Ohira T, Matsuno R: Efficacy and
20. Saltiel MY, Kuhre RE, Christiansen CB, Eliasen R, Conde- safety of fasiglifam (tak-875), a g protein-coupled receptor
Frieboes KW, Rosenkilde MM, Holst JJ: Sweet taste receptor 40 agonist, in Japanese patients with type 2 diabetes
activation in the gut is of limited importance for glucose- inadequately controlled by diet and exercise: a randomized,
stimulated GLP-1 and GIP secretion. Nutrients 2017, 9:418. double-blind, placebo-controlled, phase III trial. Diabetes Obes
Metab 2015, 17:675-681.
21. Ford HE, Peters V, Martin NM, Sleeth ML, Ghatei MA, Frost GS,
Bloom SR: Effects of oral ingestion of sucralose on gut 37. Christensen LW, Kuhre RE, Janus C, Svendsen B, Holst JJ:
hormone response and appetite in healthy normal-weight ! Vascular, but not luminal, activation of ffar1 (gpr40) stimulates
subjects. Eur J Clin Nutr 2011, 65:508-513. GLP-1 secretion from isolated perfused rat small intestine.
Physiol Rep 2015, 3.
22. Karimian Azari E, Smith KR, Yi F, Osborne TF, Bizzotto R, Mari A, Findings suggest FFAR1 acts on the basolateral side of EECs and
Pratley RE, Kyriazis GA: Inhibition of sweet chemosensory therefore that absorption is required for detection of long chain fatty
receptors alters insulin responses during glucose ingestion in acids.
healthy adults: a randomized crossover interventional study.
Am J Clin Nutr 2017, 105:1001-1009. 38. Symonds EL, Peiris M, Page AJ, Chia B, Dogra H, Masding A,
! Galanakis V, Atiba M, Bulmer D, Young RL, Blackshaw LA:
23. Gerspach AC, Steinert RE, Schönenberger L, Graber-Maier A, Mechanisms of activation of mouse and human
Beglinger C: The role of the gut sweet taste receptor in enteroendocrine cells by nutrients. Gut 2015, 64:618-626.
regulating GLP-1, PYY, and CCK release in humans. Am J Examines the distribution of nutrient sensors in the gut of both humans
Physiol — Endocrinol Metab 2011, 301:E317-E325. and mice and their colocalisation with gut hormones. Demonstrates that
essential amino acids recruit specific GPR signalling pathways in human
24. Stephens JW, Bodvarsdottir TB, Wareham K, Prior SL, EECs.
Bracken RM, Lowe GD, Rumley A, Dunseath G, Luzio S,
Deacon CF, Holst JJ et al.: Effects of short-term therapy with 39. Duca FA, Swartz TD, Sakar Y, Covasa M: Decreased intestinal
glibenclamide and repaglinide on incretin hormones and nutrient response in diet-induced obese rats: role of gut
oxidative damage associated with postprandial peptides and nutrient receptors. Int J Obes (Lond) 2013, 37:375-
hyperglycaemia in people with type 2 diabetes mellitus. 381.
Diabetes Res Clin Pract 2011, 94:199-206.
40. Gong Z, Yoshimura M, Aizawa S, Kurotani R, Zigman JM, Sakai T,
25. Pearson ER, Flechtner I, Njolstad PR, Malecki MT, Flanagan SE, Sakata I: G protein-coupled receptor 120 signaling regulates
Larkin B, Ashcroft FM, Klimes I, Codner E, Iotova V, Slingerland AS ghrelin secretion in vivo and in vitro. Am J Physiol — Endocrinol
et al.: Switching from insulin to oral sulfonylureas in patients Metab 2014, 306:E28-E35.
with diabetes due to kir6.2 mutations. N Engl J Med 2006,
355:467-477. 41. Iwasaki K, Harada N, Sasaki K, Yamane S, Iida K, Suzuki K,
Hamasaki A, Nasteska D, Shibue K, Joo E, Harada T et al.: Free
26. Ochoa M, Lallès J-P, Malbert C-H, Val-Laillet D: Dietary sugars: fatty acid receptor gpr120 is highly expressed in
their detection by the gut-brain axis and their peripheral and enteroendocrine k cells of the upper small intestine and has a
central effects in health and diseases. Eur J Nutr 2015, 54:1-24. critical role in GIP secretion after fat ingestion. Endocrinology
2015, 156:837-846.
27. Byrne CS, Chambers ES, Morrison DJ, Frost G: The role of short
chain fatty acids in appetite regulation and energy 42. Ichimura A, Hirasawa A, Poulain-Godefroy O, Bonnefond A,
homeostasis. Int J Obes (Lond) 2015, 39:1331-1338. Hara T, Yengo L, Kimura I, Leloire A, Liu N, Iida K, Choquet H et al.:
28. Georgiadi A, Kersten S: Mechanisms of gene regulation by fatty Dysfunction of lipid sensor gpr120 leads to obesity in both
acids. Adv Nutr: Int Rev J 2012, 3:127-134. mouse and human. Nature 2012, 483:350-354.

29. Sankoda A, Harada N, Iwasaki K, Yamane S, Murata Y, Shibue K, 43. Hudson BD, Shimpukade B, Mackenzie AE, Butcher AJ,
Thewjitcharoen Y, Suzuki K, Harada T, Kanemaru Y, Shimazu- Pediani JD, Christiansen E, Heathcote H, Tobin AB, Ulven T,
Kuwahara S et al.: Long chain free fatty acid receptor gpr120 Milligan G: The pharmacology of tug-891, a potent and
mediates oil-induced GIP secretion through CCK in male mice. selective agonist of the free fatty acid receptor 4 (ffa4/gpr120),
Endocrinology 2017. demonstrates both potential opportunity and possible
challenges to therapeutic agonism. Mol Pharmacol 2013,
30. Hauge M, Ekberg JP, Engelstoft MS, Timshel P, Madsen AN, 84:710-725.
Schwartz TW: Gq and Gs signaling acting in synergy to control
GLP-1 secretion. Mol Cell Endocrinol 2017, 449:64-73. 44. Oh DY, Walenta E, Akiyama TE, Lagakos WS, Lackey D,
Pessentheiner AR, Sasik R, Hah N, Chi TJ, Cox JM, Powels MA
31. Engelstoft MS, Park W-M, Sakata I, Kristensen LV, Husted AS, et al.: A gpr120-selective agonist improves insulin resistance
Osborne-Lawrence S, Piper PK, Walker AK, Pedersen MH, and chronic inflammation in obese mice. Nat Med 2014, 20:942-
Nøhr MK, Pan J et al.: Seven transmembrane g protein-coupled 947.
receptor repertoire of gastric ghrelin cells. Mol Metab 2013,
2:376-392. 45. Quesada-Lopez T, Cereijo R, Turatsinze JV, Planavila A, Cairo M,
Gavalda-Navarro A, Peyrou M, Moure R, Iglesias R, Giralt M,
32. Edfalk S, Steneberg P, Edlund H: Gpr40 is expressed in Eizirik DL et al.: The lipid sensor gpr120 promotes brown fat
enteroendocrine cells and mediates free fatty acid stimulation activation and fgf21 release from adipocytes. Nat Commun
of incretin secretion. Diabetes 2008, 57:2280-2287. 2016, 7:13479.

33. Liou AP, Lu X, Sei Y, Zhao X, Pechhold S, Carrero RJ, 46. Sun Q, Hirasawa A, Hara T, Kimura I, Adachi T, Awaji T, Ishiguro M,
Raybould HE, Wank S: The G-protein-coupled receptor gpr40 Suzuki T, Miyata N, Tsujimoto G: Structure-activity relationships

www.sciencedirect.com Current Opinion in Pharmacology 2017, 37:16–23


22 Endocrine and metabolic diseases

of gpr120 agonists based on a docking simulation. Mol 62. Wellendorph P, Hansen KB, Balsgaard A, Greenwood JR,
Pharmacol 2010, 78:804-810. Egebjerg J, Bräuner-Osborne H: Deorphanization of GPRC6a: a
promiscuous L-a-amino acid receptor with preference for
47. Engelstoft MS, Norn C, Hauge M, Holliday ND, Elster L, basic amino acids. Mol Pharmacol 2005, 67:589-597.
Lehmann J, Jones RM, Frimurer TM, Schwartz TW: Structural
basis for constitutive activity and agonist-induced activation 63. Oya M, Kitaguchi T, Pais R, Reimann F, Gribble F, Tsuboi T: The g
of the enteroendocrine fat sensor gpr119. Br J Pharmacol 2014, protein-coupled receptor family c group 6 subtype a (GPRC6a)
171:5774-5789. receptor is involved in amino acid-induced glucagon-like
peptide-1 secretion from glutag cells. J Biol Chem 2013,
48. Ekberg JH, Hauge M, Kristensen LV, Madsen AN, Engelstoft MS, 288:4513-4521.
Husted AS, Sichlau R, Egerod KL, Timshel P, Kowalski TJ,
Gribble FM et al.: Gpr119, a major enteroendocrine sensor of 64. Clemmensen C, Jorgensen CV, Smajilovic S, Brauner-Osborne H:
dietary triglyceride metabolites coacting in synergy with ffa1 Robust GLP-1 secretion by basic l-amino acids does not
(gpr40). Endocrinology 2016, 157:4561-4569. require the GPRC6a receptor. Diabetes Obes Metab 2017,
19:599-603.
49. Cvijanovic N, Isaacs NJ, Rayner CK, Feinle-Bisset C, Young RL,
Little TJ: Duodenal fatty acid sensor and transporter 65. Kinsey-Jones JS, Alamshah A, McGavigan AK, Spreckley E,
expression following acute fat exposure in healthy lean ! Banks K, Cereceda Monteoliva N, Norton M, Bewick GA,
humans. Clin Nutr 2017, 36:564-569. Murphy KG: Gprc6a is not required for the effects of a high-
protein diet on body weight in mice. Obesity (Silver Spring) 2015,
50. Moss CE, Glass LL, Diakogiannaki E, Pais R, Lenaghan C, 23:1194-1200.
Smith DM, Wedin M, Bohlooly YM, Gribble FM, Reimann F: Lipid First study to look at the effect of HPD on GPRC6a knock out mice,
derivatives activate gpr119 and trigger GLP-1 secretion in demonstrates that the GPRC6a is not necessary for the effects of high
primary murine l-cells. Peptides 2016, 77:16-20. protein diets on energy homeostasis.
51. Hassing HA, Engelstoft MS, Sichlau RM, Madsen AN, Rehfeld JF, 66. Alamshah A, McGavigan AK, Spreckley E, Kinsey-Jones JS,
Pedersen J, Jones RM, Holst JJ, Schwartz TW, Rosenkilde MM, Amin A, Tough IR, O’Hara HC, Moolla A, Banks K, France R,
Hansen HS: Oral 2-oleyl glyceryl ether improves glucose Hyberg G et al.: L-Arginine promotes gut hormone release and
tolerance in mice through the gpr119 receptor. Biofactors 2016, reduces food intake in rodents. Diabetes Obes Metab 2016,
42:665-673. 18:508-518.
52. Katz LB, Gambale JJ, Rothenberg PL, Vanapalli SR, Vaccaro N, 67. Jorgensen S, Have CT, Underwood CR, Johansen LD,
Xi L, Sarich TC, Stein PP: Effects of jnj-38431055, a novel gpr119 Wellendorph P, Gjesing AP, Jorgensen CV, Quan S, Rui G, Inoue A,
receptor agonist, in randomized, double-blind, placebo- Linneberg A et al.: Genetic variations in the human g protein-
controlled studies in subjects with type 2 diabetes. Diabetes coupled receptor class c, group 6, member a (GPRC6a) control
Obes Metab 2012, 14:709-716. cell surface expression and function. J Biol Chem 2017,
292:1524-1534.
53. Nunez DJ, Bush MA, Collins DA, McMullen SL, Gillmor D,
Apseloff G, Atiee G, Corsino L, Morrow L, Feldman PL: Gut 68. Wang Y, Chandra R, Samsa LA, Gooch B, Fee BE, Cook JM,
hormone pharmacology of a novel gpr119 agonist Vigna SR, Grant AO, Liddle RA: Amino acids stimulate
(gsk1292263), metformin, and sitagliptin in type 2 diabetes cholecystokinin release through the Ca2+-sensing receptor.
mellitus: results from two randomized studies. PLOS ONE Am J Physiol Gastrointest Liver Physiol 2011, 300:G528-G537.
2014, 9:e92494.
69. Liou AP, Sei Y, Zhao X, Feng J, Lu X, Thomas C, Pechhold S,
54. Ritter K, Buning C, Halland N, Poverlein C, Schwink L: G protein- Raybould HE, Wank SA: The extracellular calcium-sensing
coupled receptor 119 (gpr119) agonists for the treatment of receptor is required for cholecystokinin secretion in response
diabetes: recent progress and prevailing challenges. J Med to L-phenylalanine in acutely isolated intestinal I cells. Am J
Chem 2016, 59:3579-3592. Physiol Gastrointest Liver Physiol 2011, 300:G538-G546.
55. Batterham RL, Heffron H, Kapoor S, Chivers JE, Chandarana K, 70. Diakogiannaki E, Pais R, Tolhurst G, Parker HE, Horscroft J,
Herzog H, Le Roux CW, Thomas EL, Bell JD, Withers DJ: Critical Rauscher B, Zietek T, Daniel H, Gribble FM, Reimann F:
role for peptide YY in protein-mediated satiation and body- Oligopeptides stimulate glucagon-like peptide-1 secretion in
weight regulation. Cell Metab 2006, 4:223-233. mice through proton-coupled uptake and the calcium-sensing
receptor. Diabetologia 2013, 56:2688-2696.
56. van der Klaauw AA, Keogh JM, Henning E, Trowse VM, Dhillo WS,
Ghatei MA, Farooqi IS: High protein intake stimulates 71. Pais R, Gribble FM, Reimann F: Signalling pathways involved in
postprandial GLP1 and PYY release. Obesity (Silver Spring) !! the detection of peptones by murine small intestinal
2013, 21:1602-1607. enteroendocrine l-cells. Peptides 2015.
Examines the mechanisms underlying peptone induced gut hormone
57. Blom WA, Lluch A, Stafleu A, Vinoy S, Holst JJ, Schaafsma G, secretion, suggesting that the CaSR is important in peptone stimulated
Hendriks HF: Effect of a high-protein breakfast on the release and that the CaSR may recruit TRP channels.
postprandial ghrelin response. Am J Clin Nutr 2006, 83.
72. Mace OJ, Schindler M, Patel S: The regulation of k- and l-cell
58. Brennan IM, Luscombe-Marsh ND, Seimon RV, Otto B, activity by GLUT2 and the calcium-sensing receptor CaSR in
Horowitz M, Wishart JM, Feinle-Bisset C: Effects of fat, protein, rat small intestine. J Physiol 2012, 590:2917-2936.
and carbohydrate and protein load on appetite, plasma
cholecystokinin, peptide YY, and ghrelin, and energy intake in 73. Vancleef L, Van Den Broeck T, Thijs T, Steensels S, Briand L,
lean and obese men. Am J Physiol Gastrointest Liver Physiol Tack J, Depoortere I: Chemosensory signalling pathways
2012, 303:G129-G140. involved in sensing of amino acids by the ghrelin cell. Sci Rep
2015, 5:15725.
59. Lejeune MP, Westerterp KR, Adam TC, Luscombe-Marsh ND,
Westerterp-Plantenga MS: Ghrelin and glucagon-like peptide 74. Wu W, Zhou HR, Pestka JJ: Potential roles for calcium-sensing
1 concentrations, 24-h satiety, and energy and substrate receptor (CaSR) and transient receptor potential ankyrin-1
metabolism during a high-protein diet and measured in a (trpa1) in murine anorectic response to deoxynivalenol
respiration chamber. Am J Clin Nutr 2006, 83:89-94. (vomitoxin). Arch Toxicol 2017, 91:495-507.
60. Belza A, Ritz C, Sorensen MQ, Holst JJ, Rehfeld JF, Astrup A: 75. Wu W, Zhou H-R, Bursian SJ, Link JE, Pestka JJ: Calcium-
Contribution of gastroenteropancreatic appetite hormones to sensing receptor and transient receptor ankyrin-1 mediate
protein-induced satiety. Am J Clin Nutr 2013, 97:980-989. emesis induction by deoxynivalenol (vomitoxin). Toxicol Sci
2017, 155:32-42.
61. Veldhorst MA, Nieuwenhuizen AG, Hochstenbach-Waelen A,
Westerterp KR, Engelen MP, Brummer RJ, Deutz NE, Westerterp- 76. Grant MP, Stepanchick A, Cavanaugh A, Breitwieser GE: Agonist-
Plantenga MS: Effects of complete whey-protein breakfasts driven maturation and plasma membrane insertion of calcium-
versus whey without GMP-breakfasts on energy intake and sensing receptors dynamically control signal amplitude. Sci
satiety. Appetite 2009, 52:388-395. Signal 2011, 4:ra78.

Current Opinion in Pharmacology 2017, 37:16–23 www.sciencedirect.com


Targeting nutrient sensing mechanisms to treat obesity Norton and Murphy 23

77. Babinsky VN, Hannan FM, Ramracheya RD, Zhang Q, Nesbit MA, 88. Choi S, Lee M, Shiu AL, Yo SJ, Aponte GW: Identification of a
Hugill A, Bentley L, Hough TA, Joynson E, Stewart M, Aggarwal A protein hydrolysate responsive g protein-coupled receptor in
et al.: Mutant mice with calcium-sensing receptor (CaSR) enterocytes. Am J Physiol — Gastrointest Liver Physiol 2007, 292:
activation have hyperglycemia, that is rectified by calcilytic G98-G112.
therapy. Endocrinology 2017.
89. Rettenberger AT, Schulze W, Breer H, Haid D: Analysis of the
78. Nelson G, Chandrashekar J, Hoon MA, Feng L, Zhao G, Ryba NJP, protein related receptor GPR92 in g-cells. Front Physiol 2015,
Zuker CS: An amino-acid taste receptor. Nature 2002, 416:199- 6:261.
202.
90. Choi S, Lee M, Shiu AL, Yo SJ, Hallden G, Aponte GW: GPR93
79. Daly K, Al-Rammahi M, Moran A, Marcello M, Ninomiya Y, Shirazi- activation by protein hydrolysate induces CCK transcription
Beechey SP: Sensing of amino acids by the gut-expressed and secretion in STC-1 cells. Am J Physiol Gastrointest Liver
taste receptor t1r1-t1r3 stimulates CCK secretion. Am J Physiol 2007, 292:G1366-G1375.
Physiol — Gastrointest Liver Physiol 2013, 304:G271-G282.
91. Bröer S, Bröer A: Amino acid homeostasis and signalling in
80. De Giorgio R, Mazzoni M, Vallorani C, Latorre R, Bombardi C, mammalian cells and organisms. Biochem J 2017, 474:1935-
Bacci ML, Forni M, Falconi M, Sternini C, Clavenzani P: 1963.
Regulation of alpha-transducin and alpha-gustducin 92. Joshi S, Tough IR, Cox HM: Endogenous PYY and GLP-1
expression by a high protein diet in the pig gastrointestinal mediate L-glutamine responses in intestinal mucosa. Br J
tract. PLOS ONE 2016, 11:e0148954. Pharmacol 2013, 170:1092-1101.
81. Pal Choudhuri S, Delay RJ, Delay ER: L-amino acids elicit diverse 93. Zietek T, Rath E, Haller D, Daniel H: Intestinal organoids for
response patterns in taste sensory cells: a role for multiple assessing nutrient transport, sensing and incretin secretion.
receptors. PLOS ONE 2015, 10:e0130088. Sci Rep 2015, 5:16831.
82. Pal Choudhuri S, Delay RJ, Delay ER: Metabotropic glutamate 94. Liou AP, Chavez DI, Espero E, Hao S, Wank SA, Raybould HE:
receptors are involved in the detection of imp and L-amino Protein hydrolysate-induced cholecystokinin secretion from
acids by mouse taste sensory cells. Neuroscience 2016, 316:94- enteroendocrine cells is indirectly mediated by the intestinal
108. oligopeptide transporter PEPT1. Am J Physiol — Gastrointest
Liver Physiol 2011, 300:G895-G902.
83. Yasumatsu K, Manabe T, Yoshida R, Iwatsuki K, Uneyama H,
Takahashi I, Ninomiya Y: Involvement of multiple taste 95. Shiraga T, Miyamoto K, Tanaka H, Yamamoto H, Taketani Y,
receptors in umami taste: analysis of gustatory nerve Morita K, Tamai I, Tsuji A, Takeda E: Cellular and molecular
responses in metabotropic glutamate receptor 4 knockout mechanisms of dietary regulation on rat intestinal H+/peptide
mice. J Physiol 2015, 593:1021-1034. transporter PEPT1. Gastroenterology 1999, 116:354-362.

84. Husted AS, Trauelsen M, Rudenko O, Hjorth SA, Schwartz TW: 96. Nässl A-M, Rubio-Aliaga I, Sailer M, Daniel H: The intestinal
!! GPCR-mediated signaling of metabolites. Cell Metab 2017, peptide transporter PEPT1 is involved in food intake
25:777-796. regulation in mice fed a high-protein diet. PLoS ONE 2011, 6:
Extensive and thorough review of metabolite sensing. e26407.

85. Lin HV, Efanov AM, Fang X, Beavers LS, Wang X, Wang J, 97. Jiang Y, Rose AJ, Sijmonsma TP, Bröer A, Pfenninger A, Herzig S,
!! Gonzalez Valcarcel IC, Ma T: GPR142 controls tryptophan- Schmoll D, Bröer S: Mice lacking neutral amino acid transporter
induced insulin and incretin hormone secretion to improve B0AT1 (Slc6a19) have elevated levels of FGF21 and GLP-1 and
glucose metabolism. PLOS ONE 2016, 11:e0157298. improved glycaemic control. Mol Metab 2015, 4:406-417.
The first publication to examine GPR142 activity in the gut in vivo found
98. Cheng Q, Shah N, Broer A, Fairweather S, Jiang Y, Schmoll D,
that this receptor can drive the secretion of incretins.
Corry B, Broer S: Identification of novel inhibitors of the amino
86. Sommer CA, Mostoslavsky G: RNA-seq analysis of acid transporter B0 AT1 (Slc6a19), a potential target to induce
enteroendocrine cells reveals a role for FABP5 in the control of protein restriction and to treat type 2 diabetes. Br J Pharmacol
GIP secretion. Mol Endocrinol 2014, 28:1855-1865. 2017, 174:468-482.
99. Choudhury SM, Tan TM, Bloom SR: Gastrointestinal hormones
87. Toda N, Hao X, Ogawa Y, Oda K, Yu M, Fu Z, Chen Y, Kim Y,
and their role in obesity. Curr Opin Endocrinol Diabetes Obes
Lizarzaburu M, Lively S, Lawlis S et al.: Potent and orally
2016, 23:18-22.
bioavailable GPR142 agonists as novel insulin secretagogues
for the treatment of type 2 diabetes. ACS Med Chem Lett 2013, 100. McGavigan AK, Murphy KG: Gut hormones: the future of obesity
4:790-794. treatment? Br J Clin Pharmacol 2012, 74:911-919.

www.sciencedirect.com Current Opinion in Pharmacology 2017, 37:16–23

You might also like