Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Physiol ; 594(17): 4865-78, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27037495

RESUMEN

KEY POINTS: Arginine vasopressin (AVP) stimulates the release of enteroendocrine L-cell derived hormones glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) in vitro from mouse and human colons. This is mediated by the AVP receptor 1B, which is highly enriched in colonic L-cells and linked to the elevation of L-cell calcium and cAMP concentrations. By means of Ussing chambers, we show that AVP reduced colonic anion secretion, although this was blocked by a specific neuropeptide Y receptor Y1 receptor antagonist, suggesting that L-cell-released PYY acts locally on the epithelium to modulate fluid balance. In human serum samples, PYY concentrations were higher in samples with raised osmolality and copeptin (surrogate marker for AVP). These findings describe, for the first time, the role of L-cells in AVP regulated intestinal fluid secretion, potentially linking together hormonal control of blood volume and blood glucose levels, and thus adding to our understanding of the complex pathways involved in the gut hormonal response to different stimuli. ABSTRACT: Arginine vasopressin (AVP) regulates fluid balance and blood pressure via AVP receptor (AVPR)2 in the kidney and AVP receptor 1A in vascular smooth muscle. Its role in intestinal function has received less attention. We hypothesized that enteroendocrine L-cells producing glucagon-like peptide 1 (GLP-1) and peptide YY (PYY) may be a target of AVP and contribute to the control of fluid balance. Avpr1b expression was assessed by quantitative RT-PCR on flourescence-activated cell sorting-isolated L- and control cells and was enriched in colonic L-cells. AVP stimulated GLP-1 and PYY release from primary cultured murine and human colonic cells and was associated with elevated calcium and cAMP concentrations in L-cells as measured in cultures from GLU-Cre/ROSA26-GCaMP3 and GLU-Epac2camps mice. An antagonist of AVPR1B reduced AVP-triggered hormone secretion from murine and human cells. In Ussing chambers, basolaterally applied AVP reduced colonic anion secretion and this effect was blocked by a specific neuropeptide Y receptor Y1 (NPY1R) antagonist. In human serum, PYY concentrations were higher in samples with raised osmolality or copeptin (a surrogate marker for AVP). In conclusion, we propose that AVP activates L-cell AVPR1B, causing GLP-1 and PYY secretion. PYY in turn reduces colonic anion secretion via epithelial NPY1R. Our data suggest L-cells are active players in the hypothalamic control of intestinal fluid homeostasis, providing a potential link between the regulation of blood volume/pressure/osmolality and blood glucose.


Asunto(s)
Arginina Vasopresina/farmacología , Colon/metabolismo , Células Enteroendocrinas/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Péptido YY/metabolismo , Anciano , Animales , Calcio/metabolismo , Células Cultivadas , Colon/citología , AMP Cíclico/metabolismo , Femenino , Expresión Génica , Humanos , Intestino Delgado/citología , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Péptido YY/sangre , Receptores de Vasopresinas/genética
2.
J Biol Chem ; 288(7): 4513-21, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23269670

RESUMEN

Although amino acids are dietary nutrients that evoke the secretion of glucagon-like peptide 1 (GLP-1) from intestinal L cells, the precise molecular mechanism(s) by which amino acids regulate GLP-1 secretion from intestinal L cells remains unknown. Here, we show that the G protein-coupled receptor (GPCR), family C group 6 subtype A (GPRC6A), is involved in amino acid-induced GLP-1 secretion from the intestinal L cell line GLUTag. Application of l-ornithine caused an increase in intracellular Ca(2+) concentration ([Ca(2+)](i)) in GLUTag cells. Application of a GPRC6A receptor antagonist, a phospholipase C inhibitor, or an IP(3) receptor antagonist significantly suppressed the l-ornithine-induced [Ca(2+)](i) increase. We found that the increase in [Ca(2+)](i) stimulated by l-ornithine correlated with GLP-1 secretion and that l-ornithine stimulation increased exocytosis in a dose-dependent manner. Furthermore, depletion of endogenous GPRC6A by a specific small interfering RNA (siRNA) inhibited the l-ornithine-induced [Ca(2+)](i) increase and GLP-1 secretion. Taken together, these findings suggest that the GPRC6A receptor functions as an amino acid sensor in GLUTag cells that promotes GLP-1 secretion.


Asunto(s)
Células Enteroendocrinas/metabolismo , Proteínas de Unión al GTP/metabolismo , Regulación de la Expresión Génica , Péptido 1 Similar al Glucagón/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Aminoácidos/metabolismo , Animales , Calcio/metabolismo , Vectores Genéticos , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica/métodos , Ligandos , Ratones , Ratones Transgénicos , Modelos Biológicos , Ornitina/farmacología , ARN Interferente Pequeño/metabolismo , Espectroscopía Infrarroja por Transformada de Fourier , Factores de Tiempo , Transfección
3.
Am J Physiol Gastrointest Liver Physiol ; 307(3): G330-7, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24875103

RESUMEN

Type 2 diabetes is associated with elevated circulating levels of the chemokine RANTES and with decreased plasma levels of the incretin hormone glucagon-like peptide 1 (GLP-1). GLP-1 is a peptide secreted from intestinal L-cells upon nutrient ingestion. It enhances insulin secretion from pancreatic ß-cells and protects from ß-cell loss but also promotes satiety and weight loss. In search of chemokines that may reduce GLP-1 secretion we identified RANTES and show that it reduces glucose-stimulated GLP-1 secretion in the human enteroendocrine cell line NCI-H716, blocked by the antagonist Met-RANTES, and in vivo in mice. RANTES exposure to mouse intestinal tissues lowers transport function of the intestinal glucose transporter SGLT1, and administration in mice reduces plasma GLP-1 and GLP-2 levels after an oral glucose load and thereby impairs insulin secretion. These data show that RANTES is involved in altered secretion of glucagon-like peptide hormones most probably acting through SGLT1, and our study identifies the RANTES-receptor CCR1 as a potential target in diabetes therapy.


Asunto(s)
Glucemia/metabolismo , Quimiocina CCL5/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Péptido 2 Similar al Glucagón/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Intestino Delgado/metabolismo , Animales , Línea Celular , Quimiocina CCL5/administración & dosificación , AMP Cíclico/metabolismo , Péptido 1 Similar al Glucagón/sangre , Péptido 2 Similar al Glucagón/sangre , Humanos , Inyecciones Intraperitoneales , Insulina/sangre , Secreción de Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Interferencia de ARN , Receptores CCR1/genética , Receptores CCR1/metabolismo , Transportador 1 de Sodio-Glucosa/metabolismo , Factores de Tiempo , Transfección
4.
Diabetologia ; 56(12): 2688-96, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24045836

RESUMEN

AIMS/HYPOTHESIS: Ingested protein is a well-recognised stimulus for glucagon-like peptide-1 (GLP-1) release from intestinal L cells. This study aimed to characterise the molecular mechanisms employed by L cells to detect oligopeptides. METHODS: GLP-1 secretion from murine primary colonic cultures and Ca(2+) dynamics in L cells were monitored in response to peptones and dipeptides. L cells were identified and purified based on their cell-specific expression of the fluorescent protein Venus, using GLU-Venus transgenic mice. Pharmacological tools and knockout mice were used to characterise candidate sensory pathways identified by expression analysis. RESULTS: GLP-1 secretion was triggered by peptones and di-/tripeptides, including the non-metabolisable glycine-sarcosine (Gly-Sar). Two sensory mechanisms involving peptide transporter-1 (PEPT1) and the calcium-sensing receptor (CaSR) were distinguishable. Responses to Gly-Sar (10 mmol/l) were abolished in the absence of extracellular Ca(2+) or by the L-type calcium-channel blocker nifedipine (10 µmol/l) and were PEPT1-dependent, as demonstrated by their sensitivity to pH and 4-aminomethylbenzoic acid and the finding of impaired responses in tissue from Pept1 (also known as Slc15a1) knockout mice. Peptone (5 mg/ml)-stimulated Ca(2+) responses were insensitive to nifedipine but were blocked by antagonists of CaSR. Peptone-stimulated GLP-1 secretion was not impaired in mice lacking the putative peptide-responsive receptor lysophosphatidic acid receptor 5 (LPAR5; also known as GPR92/93). CONCLUSIONS/INTERPRETATION: Oligopeptides stimulate GLP-1 secretion through PEPT1-dependent electrogenic uptake and activation of CaSR. Both pathways are highly expressed in native L cells, and likely contribute to the ability of ingested protein to elevate plasma GLP-1 levels. Targeting nutrient-sensing pathways in L cells could be used to mobilise endogenous GLP-1 stores in humans, and could mimic some of the metabolic benefits of bariatric surgery.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Calcio/metabolismo , Células Enteroendocrinas/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Oligopéptidos/metabolismo , Estado Prediabético/metabolismo , Receptores Sensibles al Calcio/metabolismo , Animales , Línea Celular , AMP Cíclico/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Peptonas/metabolismo , Protones , Receptores de Glucagón/metabolismo , Transducción de Señal
5.
Am J Physiol Gastrointest Liver Physiol ; 304(10): G897-907, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23494121

RESUMEN

The intestinal transporter PEPT1 mediates the absorption of di- and tripeptides originating from breakdown of dietary proteins. Whereas mice lacking PEPT1 did not display any obvious changes in phenotype on a high-carbohydrate control diet (HCD), Pept1(-/-) mice fed a high-fat diet (HFD) showed a markedly reduced weight gain and reduced body fat stores. They were additionally protected from hyperglycemia and hyperinsulinemia. Energy balance studies revealed that Pept1(-/-) mice on HFD have a reduced caloric intake, no changes in energy expenditure, but increased energy content in feces. Cecal biomass in Pept1(-/-) mice was as well increased twofold on both diets, suggesting a limited capacity in digesting and/or absorbing the dietary constituents in the small intestine. GC-MS-based metabolite profiling of cecal contents revealed high levels and a broad spectrum of sugars in PEPT1-deficient mice on HCD, whereas animals fed HFD were characterized by high levels of free fatty acids and absence of sugars. In search of the origin of the impaired digestion/absorption, we observed that Pept1(-/-) mice lack the adaptation of the upper small intestinal mucosa to the trophic effects of the diet. Whereas wild-type mice on HFD adapt to diet with increased villus length and surface area, Pept1(-/-) mice failed to show this response. In search for the origin of this, we recorded markedly reduced systemic IL-6 levels in all Pept1(-/-) mice, suggesting that IL-6 could contribute to the lack of adaptation of the mucosal architecture to the diets.


Asunto(s)
Digestión/genética , Ingestión de Energía/genética , Síndromes de Malabsorción/genética , Obesidad/genética , Simportadores/fisiología , Animales , Composición Corporal/genética , Temperatura Corporal/fisiología , Peso Corporal/genética , Peso Corporal/fisiología , Dieta , Ingestión de Líquidos/genética , Ingestión de Alimentos/genética , Ingestión de Alimentos/psicología , Heces/química , Cromatografía de Gases y Espectrometría de Masas , Tránsito Gastrointestinal/genética , Tránsito Gastrointestinal/fisiología , Metabolismo de los Lípidos/genética , Espectroscopía de Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/genética , Tamaño de los Órganos/genética , Tamaño de los Órganos/fisiología , Transportador de Péptidos 1 , ARN/biosíntesis , ARN/aislamiento & purificación , Reacción en Cadena en Tiempo Real de la Polimerasa , Recto/fisiología , Simportadores/genética
6.
Nat Commun ; 10(1): 1029, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30833673

RESUMEN

Enteroendocrine cells are specialised sensory cells located in the intestinal epithelium and generate signals in response to food ingestion. Whilst traditionally considered hormone-producing cells, there is evidence that they also initiate activity in the afferent vagus nerve and thereby signal directly to the brainstem. We investigate whether enteroendocrine L-cells, well known for their production of the incretin hormone glucagon-like peptide-1 (GLP-1), also release other neuro-transmitters/modulators. We demonstrate regulated ATP release by ATP measurements in cell supernatants and by using sniffer patches that generate electrical currents upon ATP exposure. Employing purinergic receptor antagonists, we demonstrate that evoked ATP release from L-cells triggers electrical responses in neighbouring enterocytes through P2Y2 and nodose ganglion neurones in co-cultures through P2X2/3-receptors. We conclude that L-cells co-secrete ATP together with GLP-1 and PYY, and that ATP acts as an additional signal triggering vagal activation and potentially synergising with the actions of locally elevated peptide hormone concentrations.


Asunto(s)
Adenosina Trifosfato/metabolismo , Enterocitos/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Intestinos , Neuronas Aferentes/metabolismo , Vías Aferentes , Animales , Línea Celular , Ingestión de Alimentos , Células Enteroendocrinas/metabolismo , Femenino , Ganglión/metabolismo , Ganglión/patología , Incretinas/metabolismo , Mucosa Intestinal/inervación , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/patología , Ganglio Nudoso/metabolismo , Ganglio Nudoso/patología , Péptido YY/metabolismo , Receptores Purinérgicos P2X2/metabolismo , Receptores Purinérgicos P2X3/metabolismo , Nervio Vago/metabolismo
7.
Sci Rep ; 8(1): 1207, 2018 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-29352262

RESUMEN

L cells are an important class of enteroendocrine cells secreting hormones such as glucagon like peptide-1 and peptide YY that have several metabolic and physiological effects. The gut is home to trillions of bacteria affecting host physiology, but there has been limited understanding about how the microbiota affects gene expression in L cells. Thus, we rederived the reporter mouse strain, GLU-Venus expressing yellow fluorescent protein under the control of the proglucagon gene, as germ-free (GF). Lpos cells from ileum and colon of GF and conventionally raised (CONV-R) GLU-Venus mice were isolated and subjected to transcriptomic profiling. We observed that the microbiota exerted major effects on ileal L cells. Gene Ontology enrichment analysis revealed that microbiota suppressed biological processes related to vesicle localization and synaptic vesicle cycling in Lpos cells from ileum. This finding was corroborated by electron microscopy of Lpos cells showing reduced numbers of vesicles as well as by demonstrating decreased intracellular GLP-1 content in primary cultures from ileum of CONV-R compared with GF GLU-Venus mice. By analysing Lpos cells following colonization of GF mice we observed that the greatest transcriptional regulation was evident within 1 day of colonization. Thus, the microbiota has a rapid and pronounced effect on the L cell transcriptome, predominantly in the ileum.


Asunto(s)
Células Enteroendocrinas/metabolismo , Células Enteroendocrinas/microbiología , Interacciones Huésped-Patógeno/genética , Microbiota , Transcriptoma , Animales , Biología Computacional/métodos , Células Enteroendocrinas/ultraestructura , Microbioma Gastrointestinal , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ontología de Genes , Péptido 1 Similar al Glucagón/genética , Péptido 1 Similar al Glucagón/metabolismo , Ratones , Ratones Transgénicos
8.
Mol Metab ; 16: 65-75, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30104166

RESUMEN

OBJECTIVE: Insulin-like peptide-5 (INSL5) is an orexigenic gut hormone found in a subset of colonic and rectal enteroendocrine L-cells together with the anorexigenic hormones glucagon-like peptide-1 (GLP-1) and peptideYY (PYY). Unlike GLP-1 and PYY, INSL5 levels are elevated by calorie restriction, raising questions about how these hormones respond to different stimuli when they arise from the same cell type. The aim of the current study was to identify whether and how INSL5, GLP-1 and PYY are co-secreted or differentially secreted from colonic L-cells. METHODS: An inducible reporter mouse (Insl5-rtTA) was created to enable selective characterisation of Insl5-expressing cells. Expression profiling and Ca2+-dynamics were assessed using TET-reporter mice. Secretion of INSL5, PYY, and GLP-1 from murine and human colonic crypt cultures was quantified by tandem mass spectrometry. Vesicular co-localisation of the three hormones was analysed in 3D-SIM images of immunofluorescently-labelled murine colonic primary cultures and tissue sections. RESULTS: INSL5-producing cells expressed a range of G-protein coupled receptors previously identified in GLP-1 expressing L-cells, including Ffar1, Gpbar1, and Agtr1a. Pharmacological or physiological agonists for these receptors triggered Ca2+ transients in INSL5-producing cells and stimulated INSL5 secretion. INSL5 secretory responses strongly correlated with those of PYY and GLP-1 across a range of stimuli. The majority (>80%) of secretory vesicles co-labelled for INSL5, PYY and GLP-1. CONCLUSIONS: INSL5 is largely co-stored with PYY and GLP-1 and all three hormones are co-secreted when INSL5-positive cells are stimulated. Opposing hormonal profiles observed in vivo likely reflect differential stimulation of L-cells in the proximal and distal gut.


Asunto(s)
Péptido 1 Similar al Glucagón/metabolismo , Insulina/metabolismo , Péptido YY/metabolismo , Proteínas/metabolismo , Animales , Células Cultivadas , Cromatografía Liquida , Colon/citología , Células Enteroendocrinas/metabolismo , Hormonas Gastrointestinales/metabolismo , Humanos , Secreciones Intestinales/metabolismo , Espectrometría de Masas , Ratones , Hormonas Peptídicas/metabolismo , Cultivo Primario de Células , Receptores Acoplados a Proteínas G/metabolismo
9.
Ther Adv Endocrinol Metab ; 7(1): 24-42, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26885360

RESUMEN

The incretin hormones glucose-dependent insulinotropic polypeptide (GIP) and glucagon like peptide-1 (GLP-1) are secreted from enteroendocrine cells in the gut and regulate physiological and homeostatic functions related to glucose control, metabolism and food intake. This review provides a systematic summary of the molecular mechanisms underlying secretion from incretin cells, and an understanding of how they sense and interact with lumen and vascular factors and the enteric nervous system through transporters and G-protein coupled receptors (GPCRs) present on their surface to ultimately culminate in hormone release. Some of the molecules described below such as sodium coupled glucose transporter 1 (SGLT1), G-protein coupled receptor (GPR) 119 and GPR40 are targets of novel therapeutics designed to enhance endogenous gut hormone release. Synthetic ligands at these receptors aimed at treating obesity and type 2 diabetes are currently under investigation.

10.
Peptides ; 77: 9-15, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26215048

RESUMEN

Glucagon like peptide-1 is an insulinotropic hormone released from intestinal L-cells in response to food ingestion. Here, we investigated mechanisms underlying the sensing of peptones by primary small intestinal L-cells. Meat, casein and vegetable-derived peptones (5 mg/ml), the L-amino acids Phe, Trp, Gln and Ala (20 mM each), and the dipeptide glycine-sarcosine (20 mM) stimulated GLP-1 secretion from primary cultures prepared from the small intestine. Further mechanistic studies were performed with meat peptone, and revealed the elevation of intracellular calcium in L-cells. Inhibition of the calcium sensing receptor (CaSR), transient receptor potential (TRP) channels and Q-type voltage gated calcium channels (VGCC) significantly attenuated peptone-stimulated GLP-1 release and reduced intracellular Ca(2+) responses. CaSR inhibition also attenuated the GLP-1 secretory response to Gln. Targeting these pathways in L-cells could be used to increase endogenous production of GLP-1 and offer exploitable avenues for the development of therapeutics to treat diabetes and obesity.


Asunto(s)
Señalización del Calcio , Células Enteroendocrinas/fisiología , Peptonas/fisiología , Animales , Canales de Calcio Tipo L/metabolismo , Células Enteroendocrinas/efectos de los fármacos , Péptido 1 Similar al Glucagón/metabolismo , Ratones Transgénicos , Peptonas/farmacología , Técnicas de Cultivo de Tejidos , Canales de Potencial de Receptor Transitorio/metabolismo
11.
Endocrinology ; 157(10): 3821-3831, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27447725

RESUMEN

Angiotensin II (Ang II) is the key hormone mediator of the renin angiotensin system, which regulates blood pressure and fluid and electrolyte balance in the body. Here we report that in the colonic epithelium, the Ang II type 1 receptor is highly and exclusively expressed in enteroendocrine L cells, which produce the gut hormones glucagon-like peptide-1 and peptide YY (PYY). Ang II stimulated glucagon-like peptide-1 and PYY release from primary cultures of mouse and human colon, which was antagonized by the specific Ang II type 1 receptor blocker candesartan. Ang II raised intracellular calcium levels in L cells in primary cultures, recorded by live-cell imaging of L cells specifically expressing the fluorescent calcium sensor GCaMP3. In Ussing chamber recordings, Ang II reduced short circuit currents in mouse distal colon preparations, which was antagonized by candesartan or a specific neuropeptide Y1 receptor inhibitor but insensitive to amiloride. We conclude that Ang II stimulates PYY secretion, in turn inhibiting epithelial anion fluxes, thereby reducing net fluid secretion into the colonic lumen. Our findings highlight an important role of colonic L cells in whole-body fluid homeostasis by controlling water loss through the intestine.


Asunto(s)
Angiotensina II/fisiología , Colon/fisiología , Células Enteroendocrinas/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Péptido YY/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Animales , Señalización del Calcio , Femenino , Humanos , Mucosa Intestinal/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Sistema Renina-Angiotensina , Equilibrio Hidroelectrolítico
12.
Peptides ; 77: 16-20, 2016 03.
Artículo en Inglés | MEDLINE | ID: mdl-26144594

RESUMEN

AIMS/HYPOTHESIS: Glucagon-like peptide-1 (GLP-1) is an incretin hormone derived from proglucagon, which is released from intestinal L-cells and increases insulin secretion in a glucose dependent manner. GPR119 is a lipid derivative receptor present in L-cells, believed to play a role in the detection of dietary fat. This study aimed to characterize the responses of primary murine L-cells to GPR119 agonism and assess the importance of GPR119 for the detection of ingested lipid. METHODS: GLP-1 secretion was measured from murine primary cell cultures stimulated with a panel of GPR119 ligands. Plasma GLP-1 levels were measured in mice lacking GPR119 in proglucagon-expressing cells and controls after lipid gavage. Intracellular cAMP responses to GPR119 agonists were measured in single primary L-cells using transgenic mice expressing a cAMP FRET sensor driven by the proglucagon promoter. RESULTS: L-cell specific knockout of GPR119 dramatically decreased plasma GLP-1 levels after a lipid gavage. GPR119 ligands triggered GLP-1 secretion in a GPR119 dependent manner in primary epithelial cultures from the colon, but were less effective in the upper small intestine. GPR119 agonists elevated cAMP in ∼70% of colonic L-cells and 50% of small intestinal L-cells. CONCLUSIONS/INTERPRETATION: GPR119 ligands strongly enhanced GLP-1 release from colonic cultures, reflecting the high proportion of colonic L-cells that exhibited cAMP responses to GPR119 agonists. Less GPR119-dependence could be demonstrated in the upper small intestine. In vivo, GPR119 in L-cells plays a key role in oral lipid-triggered GLP-1 secretion.


Asunto(s)
Aceite de Maíz/farmacología , Células Enteroendocrinas/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Aceite de Oliva/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Células Cultivadas , AMP Cíclico/metabolismo , Células Enteroendocrinas/efectos de los fármacos , Femenino , Masculino , Ratones Transgénicos , Aceite de Oliva/administración & dosificación , Cultivo Primario de Células , Sistemas de Mensajero Secundario
13.
Peptides ; 77: 21-7, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26145551

RESUMEN

UNLABELLED: Glucagon-like peptide-1 (GLP-1) acts as a satiety signal and enhances insulin release. This study examined how GLP-1 production from intestinal L-cells is modified by dietary changes. METHODS: Transgenic mouse models were utilized in which L-cells could be purified by cell specific expression of a yellow fluorescent protein, Venus. Mice were fed on chow or 60% high fat diet (HFD) for 2 or 16 weeks. L-cells were purified by flow cytometry and analysed by microarray and quantitative RT-PCR. Enteroendocrine cell populations were examined by FACS analysis, and GLP-1 secretion was assessed in primary intestinal cultures. RESULTS: Two weeks HFD reduced the numbers of GLP-1 positive cells in the colon, and of GIP positive cells in the small intestine. Purified small intestinal L-cells showed major shifts in their gene expression profiles. In mice on HFD for 16 weeks, significant reductions were observed in the expression of L-cell specific genes, including those encoding gut hormones (Gip, Cck, Sct, Nts), prohormone processing enzymes (Pcsk1, Cpe), granins (Chgb, Scg2), nutrient sensing machinery (Slc5a1, Slc15a1, Abcc8, Gpr120) and enteroendocrine-specific transcription factors (Etv1, Isl1, Mlxipl, Nkx2.2 and Rfx6). A corresponding reduction in the GLP-1 secretory responsiveness to nutrient stimuli was observed in primary small intestinal cultures. CONCLUSION: Mice fed on HFD exhibited reduced expression in L-cells of many L-cell specific genes, suggesting an impairment of enteroendocrine cell function. Our results suggest that a western style diet may detrimentally affect the secretion of gut hormones and normal post-prandial signaling, which could impact on insulin secretion and satiety.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Células Enteroendocrinas/fisiología , Péptido 1 Similar al Glucagón/metabolismo , Animales , Células Cultivadas , Expresión Génica , Péptido 1 Similar al Glucagón/genética , Proteína Homeobox Nkx-2.2 , Masculino , Ratones Endogámicos C57BL , Cultivo Primario de Células
14.
J Endocrinol ; 228(1): 39-48, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26483393

RESUMEN

The incretin hormones glucagon-like peptide-1 (GLP1) and glucose-dependent insulinotropic polypeptide (GIP) are secreted from intestinal endocrine cells, the so-called L- and K-cells. The cells are derived from a common precursor and are highly related, and co-expression of the two hormones in so-called L/K-cells has been reported. To investigate the relationship between the GLP1- and GIP-producing cells more closely, we generated a transgenic mouse model expressing a fluorescent marker in GIP-positive cells. In combination with a mouse strain with fluorescent GLP1 cells, we were able to estimate the overlap between the two cell types. Furthermore, we used primary cultured intestinal cells and isolated perfused mouse intestine to measure the secretion of GIP and GLP1 in response to different stimuli. Overlapping GLP1 and GIP cells were rare (∼5%). KCl, glucose and forskolin+IBMX increased the secretion of both GLP1 and GIP, whereas bombesin/neuromedin C only stimulated GLP1 secretion. Expression analysis showed high expression of the bombesin 2 receptor in GLP1 positive cells, but no expression in GIP-positive cells. These data indicate both expressional and functional differences between the GLP1-producing 'L-cell' and the GIP-producing 'K-cell'.


Asunto(s)
Células Enteroendocrinas/clasificación , Células Enteroendocrinas/metabolismo , Polipéptido Inhibidor Gástrico/biosíntesis , Péptido 1 Similar al Glucagón/biosíntesis , Receptores de Bombesina/análisis , Animales , Calcio/análisis , Separación Celular , Células Cultivadas , Células Enteroendocrinas/química , Femenino , Citometría de Flujo , Colorantes Fluorescentes , Polipéptido Inhibidor Gástrico/análisis , Polipéptido Inhibidor Gástrico/metabolismo , Péptido 1 Similar al Glucagón/análisis , Péptido 1 Similar al Glucagón/metabolismo , Integrasas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Bombesina/genética
15.
J Clin Invest ; 125(1): 379-85, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25500886

RESUMEN

Glucagon-like peptide-1-based (GLP-1-based) therapies improve glycemic control in patients with type 2 diabetes. While these agents augment insulin secretion, they do not mimic the physiological meal-related rise and fall of GLP-1 concentrations. Here, we tested the hypothesis that increasing the number of intestinal L cells, which produce GLP-1, is an alternative strategy to augment insulin responses and improve glucose tolerance. Blocking the NOTCH signaling pathway with the γ-secretase inhibitor dibenzazepine increased the number of L cells in intestinal organoid-based mouse and human culture systems and augmented glucose-stimulated GLP-1 secretion. In a high-fat diet-fed mouse model of impaired glucose tolerance and type 2 diabetes, dibenzazepine administration increased L cell numbers in the intestine, improved the early insulin response to glucose, and restored glucose tolerance. Dibenzazepine also increased K cell numbers, resulting in increased gastric inhibitory polypeptide (GIP) secretion. Using a GLP-1 receptor antagonist, we determined that the insulinotropic effect of dibenzazepine was mediated through an increase in GLP-1 signaling. Together, our data indicate that modulation of the development of incretin-producing cells in the intestine has potential as a therapeutic strategy to improve glycemic control.


Asunto(s)
Células Enteroendocrinas/fisiología , Incretinas/metabolismo , Insulina/metabolismo , Animales , Células Cultivadas , Dieta Alta en Grasa , Péptido 1 Similar al Glucagón/metabolismo , Intolerancia a la Glucosa , Humanos , Íleon/citología , Secreción de Insulina , Masculino , Ratones Endogámicos C57BL , Receptores Notch/antagonistas & inhibidores , Receptores Notch/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA