Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Med Chem ; 66(13): 8745-8766, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37339079

ABSTRACT

Positive allosteric modulators targeting the Y4 receptor (Y4R), a G protein-coupled receptor (GPCR) involved in the regulation of satiety, offer great potential in anti-obesity research. In this study, we selected 603 compounds by using quantitative structure-activity relationship (QSAR) models and tested them in high-throughput screening (HTS). Here, the novel positive allosteric modulator (PAM) VU0506013 was identified, which exhibits nanomolar affinity and pronounced selectivity toward the Y4R in engineered cell lines and mouse descending colon mucosa natively expressing the Y4R. Based on this lead structure, we conducted a systematic SAR study in two regions of the scaffold and presented a series of 27 analogues with modifications in the N- and C-terminal heterocycles of the molecule to obtain insight into functionally relevant positions. By mutagenesis and computational docking, we present a potential binding mode of VU0506013 in the transmembrane core of the Y4R. VU0506013 presents a promising scaffold for developing in vivo tools to move toward anti-obesity drug research focused on the Y4R.


Subject(s)
Neuropeptide Y , Receptors, Neuropeptide Y , Animals , Mice , Receptors, Neuropeptide Y/metabolism , Structure-Activity Relationship , Quantitative Structure-Activity Relationship , High-Throughput Screening Assays , Obesity , Allosteric Regulation
2.
Neurogastroenterol Motil ; 35(8): e14589, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37010838

ABSTRACT

BACKGROUND: Enterochromaffin (EC) cell-derived 5-hydroxytryptamine (5-HT) is a mediator of toxin-induced reflexes, initiating emesis via vagal and central 5-HT3 receptors. The amine is also involved in gastrointestinal (GI) reflexes that are prosecretory and promotile, and recently 5-HT's roles in chemosensation in the distal bowel have been described. We set out to establish the efficacy of 5-HT signaling, local 5-HT levels and pharmacology in discrete regions of the mouse small and large intestine. We also investigated the inter-relationships between incretin hormones, glucagon-like peptide-1 (GLP-1) and gastric inhibitory polypeptide (GIP) and endogenous 5-HT in mucosal and motility assays. METHODS: Adult mouse GI mucosae were mounted in Ussing chambers and area-specific studies were performed to establish the 5-HT3 and 5-HT4 pharmacology, the sidedness of responses, and the inter-relationships between incretins and endogenous 5-HT. Natural fecal pellet transit in vitro and full-length GI transit in vivo were also measured. KEY RESULTS: We observed the greatest level of tonic and exogenous 5-HT-induced ion transport and highest levels of 5-HT in ascending colon mucosa. Here both 5-HT3 and 5-HT4 receptors were involved but elsewhere in the GI tract epithelial basolateral 5-HT4 receptors mediate 5-HT's prosecretory effect. Exendin-4 and GIP induced 5-HT release in the ascending colon, while L cell-derived PYY also contributed to GIP mucosal effects in the descending colon. Both peptides slowed colonic transit. CONCLUSIONS & INFERENCES: We provide functional evidence for paracrine interplay between 5-HT, GLP-1 and GIP, particularly in the colonic mucosal region. Basolateral epithelial 5-HT4 receptors mediated both 5-HT and incretin mucosal responses in healthy colon.


Subject(s)
Incretins , Serotonin , Mice , Animals , Serotonin/pharmacology , Incretins/pharmacology , Gastric Inhibitory Polypeptide , Colon , Intestinal Mucosa , Glucagon-Like Peptide 1
3.
Diabetes ; 71(8): 1623-1635, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35594379

ABSTRACT

Bariatric surgery improves glucose homeostasis, but the underlying mechanisms are not fully elucidated. Here, we show that the expression of sodium-glucose cotransporter 2 (SGLT2/Slc5a2) is reduced in the kidney of lean and obese mice following vertical sleeve gastrectomy (VSG). Indicating an important contribution of altered cotransporter expression to the impact of surgery, inactivation of the SGLT2/Slc5a2 gene by clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 attenuated the effects of VSG, with glucose excursions following intraperitoneal injection lowered by ∼30% in wild-type mice but by ∼20% in SGLT2-null animals. The effects of the SGLT2 inhibitor dapaglifozin were similarly blunted by surgery. Unexpectedly, effects of dapaglifozin were still observed in SGLT2-null mice, consistent with the existence of metabolically beneficial off-target effects of SGLT2 inhibitors. Thus, we describe a new mechanism involved in mediating the glucose-lowering effects of bariatric surgery.


Subject(s)
Blood Glucose , Insulin-Secreting Cells , Sodium-Glucose Transporter 2 Inhibitors , Sodium-Glucose Transporter 2/metabolism , Animals , Blood Glucose/metabolism , Gastrectomy , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Kidney/metabolism , Mice , Mice, Knockout , Sodium-Glucose Transporter 2/genetics , Sodium-Glucose Transporter 2 Inhibitors/pharmacology
4.
Mol Metab ; 49: 101207, 2021 07.
Article in English | MEDLINE | ID: mdl-33711555

ABSTRACT

OBJECTIVES: Obesity is a complex disease associated with a high risk of comorbidities. Gastric bypass surgery, an invasive procedure with low patient eligibility, is currently the most effective intervention that achieves sustained weight loss. This beneficial effect is attributed to alterations in gut hormone signaling. An attractive alternative is to pharmacologically mimic the effects of bariatric surgery by targeting several gut hormonal axes. The G protein-coupled receptor 39 (GPR39) expressed in the gastrointestinal tract has been shown to mediate ghrelin signaling and control appetite, food intake, and energy homeostasis, but the broader effect on gut hormones is largely unknown. A potent and efficacious GPR39 agonist (Cpd1324) was recently discovered, but the in vivo function was not addressed. Herein we studied the efficacy of the GPR39 agonist, Cpd1324, on metabolism and gut hormone secretion. METHODS: Body weight, food intake, and energy expenditure in GPR39 agonist-treated mice and GPR39 KO mice were studied in calorimetric cages. Plasma ghrelin, glucose-dependent insulinotropic polypeptide (GIP), glucagon-like peptide-1 (GLP-1), and peptide YY (PYY) levels were measured. Organoids generated from murine and human small intestine and mouse colon were used to study GLP-1 and PYY release. Upon GPR39 agonist administration, dynamic changes in intracellular GLP-1 content were studied via immunostaining and changes in ion transport across colonic mucosa were monitored in Ussing chambers. The G protein activation underlying GPR39-mediated selective release of gut hormones was studied using bioluminescence resonance energy transfer biosensors. RESULTS: The GPR39 KO mice displayed a significantly increased food intake without corresponding increases in respiratory exchange ratios or energy expenditure. Oral administration of a GPR39 agonist induced an acute decrease in food intake and subsequent weight loss in high-fat diet (HFD)-fed mice without affecting their energy expenditure. The tool compound, Cpd1324, increased GLP-1 secretion in the mice as well as in mouse and human intestinal organoids, but not in GPR39 KO mouse organoids. In contrast, the GPR39 agonist had no effect on PYY or GIP secretion. Transepithelial ion transport was acutely affected by GPR39 agonism in a GLP-1- and calcitonin gene-related peptide (CGRP)-dependent manner. Analysis of Cpd1324 signaling properties showed activation of Gαq and Gαi/o signaling pathways in L cells, but not Gαs signaling. CONCLUSIONS: The GPR39 agonist described in this study can potentially be used by oral administration as a weight-lowering agent due to its stimulatory effect on GLP-1 secretion, which is most likely mediated through a unique activation of Gα subunits. Thus, GPR39 agonism may represent a novel approach to effectively treat obesity through selective modulation of gastrointestinal hormonal axes.


Subject(s)
Gastrointestinal Hormones/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Animals , Appetite Regulation , Bariatric Surgery , Body Weight , Eating , Enteroendocrine Cells , Gastric Inhibitory Polypeptide/pharmacology , Ghrelin/metabolism , Glucagon-Like Peptide 1/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Peptide YY/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Gastrointestinal Hormone , Weight Loss
5.
J Med Chem ; 64(5): 2801-2814, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33595306

ABSTRACT

Human neuropeptide Y receptors (Y1R, Y2R, Y4R, and Y5R) belong to the superfamily of G protein-coupled receptors and play an important role in the regulation of food intake and energy metabolism. We identified and characterized the first selective Y4R allosteric antagonist (S)-VU0637120, an important step toward validating Y receptors as therapeutic targets for metabolic diseases. To obtain insight into the antagonistic mechanism of (S)-VU0637120, we conducted a variety of in vitro, ex vivo, and in silico studies. These studies revealed that (S)-VU0637120 selectively inhibits native Y4R function and binds in an allosteric site located below the binding pocket of the endogenous ligand pancreatic polypeptide in the core of the Y4R transmembrane domains. Taken together, our studies provide a first-of-its-kind tool for probing Y4R function and improve the general understanding of allosteric modulation, ultimately contributing to the rational development of allosteric modulators for peptide-activated G protein-coupled receptors (GPCRs).


Subject(s)
Benzothiazoles/pharmacology , Receptors, Neuropeptide Y/antagonists & inhibitors , Sulfonamides/pharmacology , Allosteric Site , Animals , Benzothiazoles/chemical synthesis , Benzothiazoles/metabolism , Chlorocebus aethiops , HEK293 Cells , Humans , Molecular Docking Simulation , Mutagenesis , Mutation , Protein Binding , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism , Stereoisomerism , Sulfonamides/chemical synthesis , Sulfonamides/metabolism
6.
NPJ Parkinsons Dis ; 7(1): 9, 2021 Jan 21.
Article in English | MEDLINE | ID: mdl-33479243

ABSTRACT

The pathological changes underlying gastrointestinal (GI) dysfunction in Parkinson's disease (PD) are poorly understood and the symptoms remain inadequately treated. In this study we compared the functional and neurochemical changes in the enteric nervous system in the colon of adult, L-DOPA-responsive, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated common marmoset, with naïve controls. Measurement of mucosal vectorial ion transport, spontaneous longitudinal smooth muscle activity and immunohistochemical assessment of intrinsic innervation were each performed in discrete colonic regions of naïve and MPTP-treated marmosets. The basal short circuit current (Isc) was lower in MPTP-treated colonic mucosa while mucosal resistance was unchanged. There was no difference in basal cholinergic tone, however, there was an increased excitatory cholinergic response in MPTP-treated tissues when NOS was blocked with L-Nω-nitroarginine. The amplitude and frequency of spontaneous contractions in longitudinal smooth muscle as well as carbachol-evoked post-junctional contractile responses were unaltered, despite a decrease in choline acetyltransferase and an increase in the vasoactive intestinal polypeptide neuron numbers per ganglion in the proximal colon. There was a low-level inflammation in the proximal but not the distal colon accompanied by a change in α-synuclein immunoreactivity. This study suggests that MPTP treatment produces long-term alterations in colonic mucosal function associated with amplified muscarinic mucosal activity but decreased cholinergic innervation in myenteric plexi and increased nitrergic enteric neurotransmission. This suggests that long-term changes in either central or peripheral dopaminergic neurotransmission may lead to adaptive changes in colonic function resulting in alterations in ion transport across mucosal epithelia that may result in GI dysfunction in PD.

7.
Neurogastroenterol Motil ; 32(12): e13943, 2020 12.
Article in English | MEDLINE | ID: mdl-32656959

ABSTRACT

BACKGROUND: The G protein-coupled bile acid (BA) receptor, GPBA (previously named TGR5), mediates BA gastrointestinal (GI) activities. Our aim was to elucidate the mucosal and motility responses to selective GPBA agonists compared with conjugated BA (eg, taurodeoxycholate, TDCA) in mouse and human colon. METHODS: Ion transport responses to GPBA agonists or BAs were measured in mucosal preparations with intact submucous innervation, from C57Bl/6, PYY-/-, or GPBA-/- mice and compared with GPBA signaling in human colon. We also investigated the mechanisms underlying GPBA agonism in mucosae and on natural fecal pellet propulsion. KEY RESULTS: GPBA agonist Merck V stimulated basolateral responses involving peptide YY (PYY), cholinergic, and 5-HT mechanisms in colonic mucosa. The PYY-mediated GPBA signal was glucose-sensitive. Luminal TDCA crossed the epithelial lining via the apical sodium-dependent BA transporter (ASBT) and its inhibitor, GSK2330672 significantly reduced luminal, but not basolateral TDCA activity. Merck V also slowed natural fecal pellet progression in wild-type and PYY-/- colons but not in GPBA-/- colon, while TDCA increased motility in wild-type colon. The antimotile GPBA effect was reversed by blockade of glucagon-like peptide 1 (GLP-1) receptors or nitric oxide synthase, indicating involvement of GLP-1 and nitric oxide. CONCLUSIONS & INFERENCES: We conclude that several different targets within the lamina propria express GPBA, including L cells (that release PYY and GLP-1), enterochromaffin cells and neurons (that release 5-HT), and other enteric neurons. Furthermore, luminal-conjugated BAs require transport across the epithelium via ASBT in order to activate basolateral GPBA.


Subject(s)
Bile Acids and Salts/administration & dosage , Colon/metabolism , Ileum/metabolism , Intestinal Mucosa/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Aged , Animals , Colon/drug effects , Female , Humans , Ileum/drug effects , Intestinal Mucosa/drug effects , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Organ Culture Techniques , Receptors, G-Protein-Coupled/agonists , Receptors, Gastrointestinal Hormone/agonists
8.
Neurogastroenterol Motil ; 30(12): e13454, 2018 12.
Article in English | MEDLINE | ID: mdl-30136343

ABSTRACT

BACKGROUND: Propionate exhibits affinity for free fatty acid receptor 2 (FFA2, formerly GPR43) and FFA3 (GPR41). These two G protein-coupled receptors (GPCRs) are expressed by enteroendocrine L cells that contain anorectic peptide YY (PYY) and glucagon-like peptide 1 (GLP-1), while FFA3 is also expressed by enteric neurons. Few studies have investigated the individual roles of FFA2 and FFA3 in propionate's gastrointestinal (GI) effects. Here, we compared FFA2, FFA3, and propionate mucosal responses utilizing selective ligands including an FFA3 antagonist, in mouse and human colonic mucosa. METHODS: Vectorial ion transport was measured in native colonic preparations from normal mouse and human colon with intact submucosal innervation. Endogenous fecal pellet propulsion was monitored in colons isolated from wild-type (WT) and PYY-/- mice. KEY RESULTS: FFA2 and FFA3 signaling differed significantly. FFA2 agonism involved endogenous L cell-derived PYY and was glucose dependent, while FFA3 agonism was independent of PYY and glucose, but required submucosal enteric neurons for activity. Tonic FFA3 activity was observed in mouse and human colon mucosa. Apical propionate responses were a combination of FFA2-PYY mediation and FFA3 neuronal GLP-1- and CGRP-dependent signaling in mouse ascending colon mucosa. Propionate also slowed WT and PYY-/- colonic transit, and this effect was blocked by a GLP-1 receptor antagonist. CONCLUSIONS & INFERENCES: We conclude that luminal propionate costimulates FFA2 and FFA3 pathways, reducing anion secretion and slowing colonic motility; FFA2 via PYY mediation and FFA3 signaling by activation of enteric sensory neurons.


Subject(s)
Colon/metabolism , Intestinal Mucosa/metabolism , Propionates/pharmacology , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Colon/drug effects , Female , Humans , Intestinal Mucosa/drug effects , Male , Mice , Mice, Inbred C57BL , Middle Aged , Propionates/metabolism , Receptors, Cell Surface/drug effects , Receptors, G-Protein-Coupled/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology
9.
J Physiol ; 596(17): 4237-4251, 2018 09.
Article in English | MEDLINE | ID: mdl-29917237

ABSTRACT

KEY POINTS: Tenascin-X (TNX) is an extracellular matrix glycoprotein with anti-adhesive properties in skin and joints. Here we report the novel finding that TNX is expressed in human and mouse gut tissue where it is exclusive to specific subpopulations of neurones. Our studies with TNX-deficient mice show impaired defecation and neural control of distal colonic motility that can be rescued with a 5-HT4 receptor agonist. However, colonic secretion is unchanged. They are also susceptible to internal rectal intussusception. Colonic afferent sensitivity is increased in TNX-deficient mice. Correspondingly, there is increased density of and sensitivity of putative nociceptive fibres in TNX-deficient mucosa. A group of TNX-deficient patients report symptoms highly consistent with those in the mouse model. These findings suggest TNX plays entirely different roles in gut to non-visceral tissues - firstly a role in enteric motor neurones and secondly a role influencing nociceptive sensory neurones Studying further the mechanisms by which TNX influences neuronal function will lead to new targets for future treatment. ABSTRACT: The extracellular matrix (ECM) is not only an integral structural molecule, but is also critical for a wide range of cellular functions. The glycoprotein tenascin-X (TNX) predominates in the ECM of tissues like skin and regulates tissue structure through anti-adhesive interactions with collagen. Monogenic TNX deficiency causes painful joint hypermobility and skin hyperelasticity, symptoms characteristic of hypermobility Ehlers Danlos syndrome (hEDS). hEDS patients also report consistently increased visceral pain and gastrointestinal (GI) dysfunction. We investigated whether there is a direct link between TNX deficiency and GI pain or motor dysfunction. We set out first to learn where TNX is expressed in human and mouse, then determine how GI function, specifically in the colon, is disordered in TNX-deficient mice and humans of either sex. In human and mouse tissue, TNX was predominantly associated with cholinergic colonic enteric neurones, which are involved in motor control. TNX was absent from extrinsic nociceptive peptidergic neurones. TNX-deficient mice had internal rectal prolapse and a loss of distal colonic contractility which could be rescued by prokinetic drug treatment. TNX-deficient patients reported increased sensory and motor GI symptoms including abdominal pain and constipation compared to controls. Despite absence of TNX from nociceptive colonic neurones, neuronal sprouting and hyper-responsiveness to colonic distension was observed in the TNX-deficient mice. We conclude that ECM molecules are not merely support structures but an integral part of the microenvironment particularly for specific populations of colonic motor neurones where TNX exerts functional influences.


Subject(s)
Colon/pathology , Extracellular Matrix/metabolism , Gastrointestinal Diseases/pathology , Motor Neurons/pathology , Sensory Receptor Cells/pathology , Tenascin/metabolism , Animals , Cell Movement , Colon/metabolism , Female , Gastrointestinal Diseases/metabolism , Humans , Male , Mice , Mice, Knockout , Motor Neurons/metabolism , Sensory Receptor Cells/metabolism , Tenascin/genetics
10.
Endocrinology ; 159(4): 1704-1717, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29471473

ABSTRACT

The lipid sensor G protein-coupled receptor 119 (GPR119) is highly expressed by enteroendocrine L-cells and pancreatic ß-cells that release the hormones, peptide YY (PYY) and glucagonlike peptide 1, and insulin, respectively. Endogenous oleoylethanolamide (OEA) and the dietary metabolite, 2-monoacylglycerol (2-OG), can each activate GPR119. Here, we compared mucosal responses with selective, synthetic GPR119 agonists (AR440006 and AR231453) and the lipids, OEA, 2-OG, and N-oleoyldopamine (OLDA), monitoring epithelial ion transport as a readout for L-cell activity in native mouse and human gastrointestinal (GI) mucosae. We also assessed GPR119 modulation of colonic motility in wild-type (WT), GPR119-deficient (GPR119-/-), and PYY-deficient (PYY-/-) mice. The water-soluble GPR119 agonist, AR440006 (that cannot traverse epithelial tight junctions), elicited responses, when added apically or basolaterally in mouse and human colonic mucosae. In both species, GPR119 responses were PYY, Y1 receptor mediated, and glucose dependent. AR440006 efficacy matched the GI distribution of L-cells in WT tissues but was absent from GPR119-/- tissue. OEA and 2-OG responses were significantly reduced in the GPR119-/- colon, but OLDA responses were unchanged. Alternative L-cell activation via free fatty acid receptors 1, 3, and 4 and the G protein-coupled bile acid receptor TGR5 or by the melanocortin 4 receptor, was unchanged in GPR119-/- tissues. The GPR119 agonist slowed transit in WT but not the PYY-/- colon in vitro. AR440006 (intraperitoneally) slowed WT colonic and upper-GI transit significantly in vivo. These data indicate that luminal or blood-borne GPR119 agonism can stimulate L-cell PYY release with paracrine consequences and slower motility. We suggest that this glucose-dependent L-cell response to a gut-restricted GPR119 stimulus has potential therapeutic advantage in modulating insulinotropic signaling with reduced risk of hypoglycemia.


Subject(s)
Colon/metabolism , Glucose/pharmacology , Intestinal Mucosa/metabolism , Peptide YY/metabolism , Receptors, G-Protein-Coupled/agonists , Animals , Colon/drug effects , Dopamine/analogs & derivatives , Dopamine/pharmacology , Endocannabinoids/pharmacology , Gastrointestinal Motility/drug effects , Humans , Intestinal Mucosa/drug effects , Ion Transport/drug effects , Mice , Mice, Knockout , Monoglycerides/pharmacology , Oleic Acids/pharmacology , Oxadiazoles/pharmacology , Peptide YY/genetics , Pyrimidines/pharmacology , Signal Transduction/drug effects
11.
Br J Pharmacol ; 174(23): 4508-4522, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28971469

ABSTRACT

BACKGROUND AND PURPOSE: Free fatty acid receptors FFA1 and FFA4 are located on enteroendocrine L cells with the highest gastrointestinal (GI) expression in descending colon. Their activation causes the release of glucagon-like peptide 1 and peptide YY (PYY) from L cells. Additionally, FFA1 agonism releases insulin from pancreatic ß cells. As these receptors are modulators of nutrient-stimulated glucose regulation, the aim of this study was to compare the pharmacology of commercially available agonists (TUG424, TUG891, GW9508) with proven selective agonists (JTT, TAK-875, AZ423, Metabolex-36) in mice. EXPERIMENTAL APPROACH: Mouse mucosa was mounted in Ussing chambers, voltage-clamped and the resultant short-circuit current (Isc ) was recorded continuously. Pretreatments included antagonists of FFA1, Y1  or Y2 receptors. Glucose sensitivity was investigated by mannitol replacement apically, and colonic and upper GI transit was assessed in vitro and in vivo. KEY RESULTS: FFA1 and FFA4 agonism required glucose and reduced Isc in a PYY-Y1 receptor-dependent manner. The novel compounds were more potent than GW9508. The FFA1 antagonists (GW1100 and ANT825) blocked FFA1 activity only and revealed FFA1 tonic activity. The FFA4 agonist, Metabolex-36, slowed colonic transit in vitro but increased small intestinal transit in vivo. CONCLUSIONS AND IMPLICATIONS: The selective FFA1 and FFA4 agonists were more potent at reducing Isc than GW9508, a dual FFA1 and FFA4 agonist. A paracrine epithelial mechanism involving PYY-stimulated Y1 receptors mediated their responses, which were glucose sensitive, potentially limiting hypoglycaemia. ANT825 revealed tonic activity and the possibility of endogenous FFA1 ligands causing PYY release. Finally, FFA4 agonism induced regional differences in transit.


Subject(s)
Colon/metabolism , Peptide YY/metabolism , Receptors, G-Protein-Coupled/agonists , Animals , Biphenyl Compounds/pharmacology , Colon/drug effects , Female , Glucose/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Methylamines/pharmacology , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Phenylpropionates/pharmacology , Propionates/pharmacology , Receptors, G-Protein-Coupled/metabolism
12.
J Med Chem ; 60(17): 7605-7612, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28795803

ABSTRACT

The human Y4 receptor (Y4R) and its cognate ligand, pancreatic polypeptide (PP), are involved in the regulation of energy expenditure, satiety, and food intake. This system represents a potential target for the treatment of metabolic diseases and has been extensively investigated and validated in vivo. Here, we present the compound tBPC (tert-butylphenoxycyclohexanol), a novel and selective Y4R positive allosteric modulator that potentiates Y4R activation in G-protein signaling and arrestin3 recruitment experiments. The compound has no effect on the binding of the orthosteric ligands, implying its allosteric mode of action at the Y4R and evidence for a purely efficacy-driven positive allosteric modulation. Finally, the ability of tBPC to selectively potentiate Y4R agonism initiated by PP was confirmed in mouse descending colon mucosa preparations expressing native Y4R, demonstrating Y4R positive allosteric modulation in vitro.


Subject(s)
Allosteric Regulation/drug effects , Cyclohexanols/chemistry , Cyclohexanols/pharmacology , GTP-Binding Proteins/metabolism , Receptors, Neuropeptide Y/agonists , Receptors, Neuropeptide Y/metabolism , Signal Transduction/drug effects , Animals , Arrestins/metabolism , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans , Models, Molecular
13.
Nature ; 520(7545): 94-8, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25600267

ABSTRACT

The regulated release of anorexigenic α-melanocyte stimulating hormone (α-MSH) and orexigenic Agouti-related protein (AgRP) from discrete hypothalamic arcuate neurons onto common target sites in the central nervous system has a fundamental role in the regulation of energy homeostasis. Both peptides bind with high affinity to the melanocortin-4 receptor (MC4R); existing data show that α-MSH is an agonist that couples the receptor to the Gαs signalling pathway, while AgRP binds competitively to block α-MSH binding and blocks the constitutive activity mediated by the ligand-mimetic amino-terminal domain of the receptor. Here we show that, in mice, regulation of firing activity of neurons from the paraventricular nucleus of the hypothalamus (PVN) by α-MSH and AgRP can be mediated independently of Gαs signalling by ligand-induced coupling of MC4R to closure of inwardly rectifying potassium channel, Kir7.1. Furthermore, AgRP is a biased agonist that hyperpolarizes neurons by binding to MC4R and opening Kir7.1, independently of its inhibition of α-MSH binding. Consequently, Kir7.1 signalling appears to be central to melanocortin-mediated regulation of energy homeostasis within the PVN. Coupling of MC4R to Kir7.1 may explain unusual aspects of the control of energy homeostasis by melanocortin signalling, including the gene dosage effect of MC4R and the sustained effects of AgRP on food intake.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Potassium Channels, Inwardly Rectifying/metabolism , Receptor, Melanocortin, Type 4/metabolism , Action Potentials , Agouti-Related Protein/metabolism , Animals , Eating/genetics , Energy Metabolism , Female , HEK293 Cells , Homeostasis/genetics , Humans , Ligands , Male , Melanocortins/metabolism , Mice , Receptor, Melanocortin, Type 4/genetics , Signal Transduction/genetics , alpha-MSH/metabolism
14.
Cell Metab ; 20(6): 1018-29, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25453189

ABSTRACT

The melanocortin-4 receptor (MC4R) is expressed in the brainstem and vagal afferent nerves and regulates a number of aspects of gastrointestinal function. Here we show that the receptor is also diffusely expressed in cells of the gastrointestinal system, from stomach to descending colon. Furthermore, MC4R is the second most highly enriched GPCR in peptide YY (PYY) and glucagon-like peptide 1 (GLP-1) expressing enteroendocrine L cells. When vectorial ion transport is measured across mouse or human intestinal mucosa, administration of α-MSH induces a MC4R-specific PYY-dependent antisecretory response consistent with a role for the MC4R in paracrine inhibition of electrolyte secretion. Finally, MC4R-dependent acute PYY and GLP-1 release from L cells can be stimulated in vivo by intraperitoneal (i.p.) administration of melanocortin peptides to mice. This suggests physiological significance for MC4R in L cells and indicates a previously unrecognized peripheral role for the MC4R, complementing vagal and central receptor functions.


Subject(s)
Enteroendocrine Cells/metabolism , Glucagon-Like Peptide 1/metabolism , Peptide YY/metabolism , Receptor, Melanocortin, Type 4/metabolism , Acids, Heterocyclic/pharmacology , Animals , Colon/cytology , Colon/drug effects , Colon/metabolism , Enteroendocrine Cells/drug effects , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxadiazoles/pharmacology , Peptides, Cyclic/pharmacology , Receptor, Melanocortin, Type 4/agonists
15.
J Physiol ; 592(4): 777-93, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24344165

ABSTRACT

Submucosal neurons are vital regulators of water and electrolyte secretion and local blood flow in the gut. Due to the availability of transgenic models for enteric neuropathies, the mouse has emerged as the research model of choice, but much is still unknown about the murine submucosal plexus. The progeny of choline acetyltransferase (ChAT)-Cre × ROSA26(YFP) reporter mice, ChAT-Cre;R26R-yellow fluorescent protein (YFP) mice, express YFP in every neuron that has ever expressed ChAT. With the aid of the robust YFP staining in these mice, we correlated the neurochemistry, morphology and electrophysiology of submucosal neurons in distal colon. We also examined whether there are differences in neurochemistry along the colon and in neurally mediated vectorial ion transport between the proximal and distal colon. All YFP(+) submucosal neurons also contained ChAT. Two main neurochemical but not electrophysiological groups of neurons were identified: cholinergic (containing ChAT) or non-cholinergic. The vast majority of neurons in the middle and distal colon were non-cholinergic but contained vasoactive intestinal peptide. In the distal colon, non-cholinergic neurons had one or two axons, whereas the cholinergic neurons examined had only one axon. All submucosal neurons exhibited S-type electrophysiology, shown by the lack of long after-hyperpolarizing potentials following their action potentials and fast excitatory postsynaptic potentials (EPSPs). Fast EPSPs were predominantly nicotinic, and somatic action potentials were mediated by tetrodotoxin-resistant voltage-gated channels. The size of submucosal ganglia decreased but the proportion of cholinergic neurons increased distally along the colon. The distal colon had a significantly larger nicotinic ion transport response than the proximal colon. This work shows that the properties of murine submucosal neurons and their control of epithelial ion transport differ between colonic regions. There are several key differences between the murine submucous plexus and that of other animals, including a lack of conventional intrinsic sensory neurons, which suggests there is an incomplete neuronal circuitry within the murine submucous plexus.


Subject(s)
Action Potentials , Cholinergic Neurons/physiology , Colon/innervation , Submucous Plexus/cytology , Animals , Axons/metabolism , Axons/physiology , Choline O-Acetyltransferase/genetics , Choline O-Acetyltransferase/metabolism , Cholinergic Neurons/cytology , Cholinergic Neurons/metabolism , Colon/cytology , Excitatory Postsynaptic Potentials , Inhibitory Postsynaptic Potentials , Mice , Mice, Inbred C57BL , Submucous Plexus/metabolism , Submucous Plexus/physiology , Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/metabolism
16.
Cell Metab ; 11(6): 532-42, 2010 Jun 09.
Article in English | MEDLINE | ID: mdl-20519124

ABSTRACT

Peptide YY (PYY) is released following food intake and regulates intestinal function and glucose homeostasis, but the mechanisms underpinning these processes are unclear. Enteroendocrine L cells contain PYY and express the acylethanolamine receptor, Gpr119. Here, we show that Gpr119 activation inhibited epithelial electrolyte secretion in human and mouse colon in a glucose-sensitive manner. Endogenous PYY selectively mediated these effects, since PYY(-/-) mice showed no Gpr119 response, but responses were observed in NPY(-/-) mice. Importantly, Gpr119 responses in wild-type (WT) mouse tissue and human colon were abolished by Y(1) receptor antagonism, but were not enhanced by dipeptidylpeptidase IV blockade, indicating that PYY processing to PYY(3-36) was not important. In addition, Gpr119 agonism reduced glycemic excursions after oral glucose delivery to WT mice but not PYY(-/-) mice. Taken together, these data demonstrate a previously unrecognized role of PYY in mediating intestinal Gpr119 activity and an associated function in controlling glucose tolerance.


Subject(s)
Gastric Mucosa/metabolism , Peptide YY/metabolism , Receptors, G-Protein-Coupled/metabolism , Acids, Heterocyclic/pharmacology , Animals , Colon/drug effects , Colon/metabolism , Electrolytes/metabolism , Enteroendocrine Cells/metabolism , Glucagon-Like Peptide 1/metabolism , Glucose/metabolism , Humans , Mice , Mice, Knockout , Oxadiazoles/pharmacology , Peptide YY/genetics , Receptors, G-Protein-Coupled/agonists , Receptors, Neuropeptide Y/antagonists & inhibitors , Receptors, Neuropeptide Y/metabolism
17.
J Pharmacol Exp Ther ; 319(1): 20-30, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16807358

ABSTRACT

The antisecretory effects of several Y agonists, including pancreatic polypeptide (PP), indicate the presence of Y(1), Y(2), and Y(4) receptors in mouse and human (h) colon mucosae. Here, we used preparations from human and from wild-type (WT), Y(4), and Y(1) receptor knockout ((-/-)) mice, alongside Y(4) receptor-transfected cells to define the relative functional contribution of the Y(4) receptor. First, rat (r) PP antisecretory responses were lost in murine Y(4)(-/-) preparations, but hPP and Pro(34) peptide YY (PYY) costimulated Y(4) and Y(1) receptors in WT mucosa. The Y(1) antagonist/Y(4) agonist GR231118 [(Ile,Glu,Pro,Dpr,Tyr,Arg,Leu,Arg,Try-NH(2))-2-cyclic(2,4'),(2',4)-diamide] elicited small Y(4)-mediated antisecretory responses in human tissues pretreated with the Y(1) antagonist, BIBO3304 [(R)-N-[[4-(aminocarbonylaminomethyl)-phenyl]methyl]-N(2)-(diphenylacetyl)-argininamide trifluoroacetate)], and attenuated Y(4)-mediated hPP responses in mouse and human mucosa. GR231118 and rPP were also antisecretory in hY(4)-transfected epithelial monolayers but were partial agonists compared with hPP at this receptor. In Y(4)-transfected human embryonic kidney (HEK) 293 cells, Y(4) ligands displaced [(125)I]hPP binding with orders of affinity (pK(i)) at human (hPP = rPP > GR231118 > Pro(34)PYY = PYY) and mouse (rPP = hPP > GR231118 > Pro(34)PYY > PYY) Y(4) receptors. GR231118- and rPP-stimulated guanosine 5'-3-O-(thio)triphosphate binding through hY(4) receptors with significantly lower efficacy than hPP. GR231118 marginally increased basal but abolished further PP-induced hY(4) internalization to recycling (transferrin-labeled) pathways in HEK293 cells. Taken together, these findings show that Y(4) receptors play a definitive role in attenuating colonic anion transport and may be useful targets for novel antidiarrheal agents due to their limited peripheral expression.


Subject(s)
Colon/drug effects , Pancreatic Polypeptide/pharmacology , Receptors, Neuropeptide Y/physiology , Amino Acid Sequence , Animals , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Intestinal Mucosa/drug effects , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Peptides, Cyclic/pharmacology , Rats , Species Specificity
18.
Mol Pharmacol ; 67(3): 655-64, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15576634

ABSTRACT

We have studied truncation mutants of the rat neuropeptide Y (NPY) Y1 receptor lacking four (Thr361stop, Y1T361*) or eight (Ser352stop, Y1S352*) potential serine/threonine C-terminal phosphorylation sites. NPY-stimulated hemagglutinin-tagged Y1, Y1T361*, and Y1S352* receptors all efficiently activated G proteins in Chinese hamster ovary (CHO) cell membranes, but desensitization after NPY pretreatment was only prevented in the HAY1S352* clone. In transfected colonic carcinoma epithelial layers, functional Y1 and Y1T361* peptide YY responses became more transient as the agonist concentration increased, whereas those mediated by the Y1S352* receptor remained sustained. NPY-stimulated HAY1 receptor phosphorylation was increased by transient overexpression of G protein-coupled receptor kinase 2, and only Ser352stop truncation abolished this response in CHO or human embryonic kidney (HEK) 293 cells. Rapid internalization of cell-surface HAY1 receptors in HEK293 cells was observed in response to agonist, resulting in partial colocalization with transferrin, a marker for clathrin-mediated endocytosis and recycling. It is surprising that both truncated receptors were constitutively internalized, predominantly in transferrin-positive compartments. NPY increased cell-surface localization of HAY1S352* receptors, whereas the distribution of both mutants was unaltered by BIBO3304. Recruitment of green fluorescent protein-tagged beta-arrestin2 to punctate endosomes was observed only for HAY1 and HAY1T361* receptors and solely under NPY-stimulated conditions. Thus, the key C-terminal sequence between Ser352 and Lys360 is a major site for Y1 receptor phosphorylation, is critical for its desensitization, and contributes to the association between the receptor and beta-arrestin proteins. However, additional beta-arrestin-independent mechanisms control Y1 receptor trafficking under basal conditions.


Subject(s)
Receptors, Neuropeptide Y/chemistry , Receptors, Neuropeptide Y/metabolism , Amino Acid Sequence , Animals , Base Sequence , Biological Transport , CHO Cells , Cell Line , Cricetinae , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Neuropeptide Y/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphorylation , Protein Conformation , Rats , Receptors, Gastrointestinal Hormone/metabolism , Transfection
19.
Br J Pharmacol ; 139(4): 863-71, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12813010

ABSTRACT

1 Neuropeptide Y (NPY), peptide YY (PYY) and pancreatic polypeptide (PP) differentially activate three Y receptors (Y(1), Y(2) and Y(4)) in mouse and human isolated colon. 2 The aim of this study was to characterise Y(2) receptor-mediated responses in colon mucosa and longitudinal smooth muscle preparations from wild type (Y(2)+/+) and knockout (Y(2)-/-) mice and to compare the former with human mucosal Y agonist responses. Inhibition of mucosal short-circuit current and increases in muscle tone were monitored in colonic tissues from Y(2)+/+ and Y(2)-/- mice+/-Y(1) ((R)-N-[[4-(aminocarbonylaminomethyl)phenyl)methyl]-N(2)-(diphenylacetyl)-argininamide-trifluoroacetate (BIBO3304) or Y(2) (S)-N(2)-[[1-[2-[4-[(R,S)-5,11-dihydro-6(6H)-oxodibenz[b,e]azepin-11-yl]-1-piperazinyl]-2-oxoethyl]cyclopentyl]acetyl]-N-[2-[1,2-dihydro-3,5(4H)-dioxo-1,2-diphenyl-3H-1,2,4-triazol-4-yl]ethyl]-argininamide (BIIE0246) antagonists. 3 Predictably, Y(2)-/- tissues were insensitive to Y(2)-preferred agonist PYY(3-36) (

Subject(s)
Arginine/analogs & derivatives , Colon/drug effects , Intestinal Mucosa/drug effects , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism , Animals , Arginine/pharmacology , Benzazepines/pharmacology , Colon/metabolism , Electric Stimulation , Female , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Pancreatic Polypeptide/pharmacokinetics , Peptide Fragments , Peptide YY/antagonists & inhibitors , Peptide YY/pharmacokinetics , Receptors, Neuropeptide Y/antagonists & inhibitors , Sex Characteristics , Tetrodotoxin/pharmacology
20.
Naunyn Schmiedebergs Arch Pharmacol ; 367(2): 104-8, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12595950

ABSTRACT

The neurokinin (NK) receptors, NK(1) and NK(2), which are activated by substance P (SP) and NKA, have been identified as potential therapeutic targets in irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD). Here we have investigated the effects of a novel dual NK(1) and NK(2) receptor antagonist, namely DNK333 upon responses elicited by [Sar(9), Met(O(2))(11)]-SP (SMSP) and [betaAla(8)]-NKA(4-10) in isolated human colon mucosa mounted in Ussing chambers. A selective NK(1) receptor antagonist, SR140333 and NK(2) receptor antagonist, SR48968 have been tested for comparison. Additions of SMSP (100 nM) or [betaAla(8)]-NKA(4-10) (100 nM) increased basal short-circuit current and responses to both peptides were inhibited by DNK333, while SR140333 only inhibited SMSP and SR48968 blocked only [betaAla(8)]-NKA(4-10) responses. SR140333 did not attenuate [betaAla(8)]-NKA(4-10) effects and SR48968 had no effect upon SMSP responses. Carbachol (1 micro M) responses were not altered by any of the three NK antagonists. We conclude that activation of either NK(1) or NK(2) receptors can stimulate epithelial ion transport in human colon mucosa and that the novel dual antagonist, DNK333 may be of potential therapeutic interest in the treatment of IBD and IBS.


Subject(s)
Intestinal Mucosa/drug effects , Neurokinin-1 Receptor Antagonists , Receptors, Neurokinin-2/antagonists & inhibitors , Aged , Aza Compounds/pharmacology , Benzamides/pharmacology , Colon/drug effects , Colon/physiology , Female , Humans , In Vitro Techniques , Intestinal Mucosa/metabolism , Ion Transport/drug effects , Male , Patch-Clamp Techniques , Piperidines/pharmacology , Quinuclidines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...