Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Gastroenterology ; 162(2): 535-547.e13, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34688712

RESUMO

BACKGROUND AND AIMS: The gastrointestinal (GI) tract extracts nutrients from ingested meals while protecting the organism from infectious agents frequently present in meals. Consequently, most animals conduct the entire digestive process within the GI tract while keeping the luminal contents entirely outside the body, separated by the tightly sealed GI epithelium. Therefore, like the skin and oral cavity, the GI tract must sense the chemical and physical properties of the its external interface to optimize its function. Specialized sensory enteroendocrine cells (EECs) in GI epithelium interact intimately with luminal contents. A subpopulation of EECs express the mechanically gated ion channel Piezo2 and are developmentally and functionally like the skin's touch sensor- the Merkel cell. We hypothesized that Piezo2+ EECs endow the gut with intrinsic tactile sensitivity. METHODS: We generated transgenic mouse models with optogenetic activators in EECs and Piezo2 conditional knockouts. We used a range of reference standard and novel techniques from single cells to living animals, including single-cell RNA sequencing and opto-electrophysiology, opto-organ baths with luminal shear forces, and in vivo studies that assayed GI transit while manipulating the physical properties of luminal contents. RESULTS: Piezo2+ EECs have transcriptomic features of synaptically connected, mechanosensory epithelial cells. EEC activation by optogenetics and forces led to Piezo2-dependent alterations in colonic propagating contractions driven by intrinsic circuitry, with Piezo2+ EECs detecting the small luminal forces and physical properties of the luminal contents to regulate transit times in the small and large bowel. CONCLUSIONS: The GI tract has intrinsic tactile sensitivity that depends on Piezo2+ EECs and allows it to detect luminal forces and physical properties of luminal contents to modulate physiology.


Assuntos
Células Enteroendócrinas/metabolismo , Mucosa Intestinal/metabolismo , Canais Iônicos/genética , Tato/fisiologia , Animais , Células Enteroendócrinas/fisiologia , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Técnicas de Inativação de Genes , Mucosa Intestinal/citologia , Mucosa Intestinal/fisiologia , Canais Iônicos/metabolismo , Mecanorreceptores , Camundongos , Camundongos Transgênicos , Optogenética , Peristaltismo/fisiologia
2.
Cell Host Microbe ; 29(2): 179-196.e9, 2021 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33352109

RESUMO

The intestinal epithelium senses nutritional and microbial stimuli using epithelial sensory enteroendocrine cells (EEC). EECs communicate nutritional information to the nervous system, but whether they also relay signals from intestinal microbes remains unknown. Using in vivo real-time measurements of EEC and nervous system activity in zebrafish, we discovered that the bacteria Edwardsiella tarda activate EECs through the receptor transient receptor potential ankyrin A1 (Trpa1) and increase intestinal motility. Microbial, pharmacological, or optogenetic activation of Trpa1+EECs directly stimulates vagal sensory ganglia and activates cholinergic enteric neurons by secreting the neurotransmitter 5-hydroxytryptamine (5-HT). A subset of indole derivatives of tryptophan catabolism produced by E. tarda and other gut microbes activates zebrafish EEC Trpa1 signaling. These catabolites also directly stimulate human and mouse Trpa1 and intestinal 5-HT secretion. These results establish a molecular pathway by which EECs regulate enteric and vagal neuronal pathways in response to microbial signals.


Assuntos
Edwardsiella tarda/metabolismo , Sistema Nervoso Entérico/metabolismo , Células Enteroendócrinas/fisiologia , Mucosa Intestinal/metabolismo , Canal de Cátion TRPA1/metabolismo , Animais , Animais Geneticamente Modificados , Neurônios Colinérgicos/metabolismo , Sistema Nervoso Entérico/citologia , Motilidade Gastrointestinal/fisiologia , Mucosa Intestinal/citologia , Mucosa Intestinal/inervação , Proteínas Proto-Oncogênicas c-ret/genética , Serotonina/metabolismo , Transdução de Sinais , Triptofano/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
3.
Int J Obes (Lond) ; 44(9): 1859-1871, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32015474

RESUMO

OBJECTIVES: Gastrointestinal hormones contribute to the beneficial effects of Roux-en-Y gastric bypass surgery (RYGB) on glycemic control. Secretin is secreted from duodenal S cells in response to low luminal pH, but it is unknown whether its secretion is altered after RYGB and if secretin contributes to the postoperative improvement in glycemic control. We hypothesized that secretin secretion increases after RYGB as a result of the diversion of nutrients to more distal parts of the small intestine, and thereby affects islet hormone release. METHODS: A specific secretin radioimmunoassay was developed, evaluated biochemically, and used to quantify plasma concentrations of secretin in 13 obese individuals before, 1 week after, and 3 months after RYGB. Distribution of secretin and its receptor was assessed by RNA sequencing, mass-spectrometry and in situ hybridization in human and rat tissues. Isolated, perfused rat intestine and pancreas were used to explore the molecular mechanism underlying glucose-induced secretin secretion and to study direct effects of secretin on glucagon, insulin, and somatostatin secretion. Secretin was administered alone or in combination with GLP-1 to non-sedated rats to evaluate effects on glucose regulation. RESULTS: Plasma postprandial secretin was more than doubled in humans after RYGB (P < 0.001). The distal small intestine harbored secretin expressing cells in both rats and humans. Glucose increased the secretion of secretin in a sodium-glucose cotransporter dependent manner when administered to the distal part but not into the proximal part of the rat small intestine. Secretin stimulated somatostatin secretion (fold change: 1.59, P < 0.05) from the perfused rat pancreas but affected neither insulin (P = 0.2) nor glucagon (P = 0.97) secretion. When administered to rats in vivo, insulin secretion was attenuated and glucagon secretion increased (P = 0.04), while blood glucose peak time was delayed (from 15 to 45 min) and gastric emptying time prolonged (P = 0.004). CONCLUSIONS: Glucose-sensing secretin cells located in the distal part of the small intestine may contribute to increased plasma concentrations observed after RYGB. The metabolic role of the distal S cells warrants further studies.


Assuntos
Células Enteroendócrinas , Derivação Gástrica , Glucose/metabolismo , Intestino Delgado/citologia , Animais , Células Enteroendócrinas/metabolismo , Células Enteroendócrinas/fisiologia , Masculino , Período Pós-Prandial/fisiologia , Ratos , Ratos Wistar
4.
Cell Mol Gastroenterol Hepatol ; 9(3): 447-464, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31756561

RESUMO

BACKGROUND & AIMS: The enteroendocrine cell (EEC) lineage is important for intestinal homeostasis. It was recently shown that EEC progenitors contribute to intestinal epithelial growth and renewal, but the underlying mechanisms remain poorly understood. MicroRNAs are under-explored along the entire EEC lineage trajectory, and comparatively little is known about their contributions to intestinal homeostasis. METHODS: We leverage unbiased sequencing and eight different mouse models and sorting methods to identify microRNAs enriched along the EEC lineage trajectory. We further characterize the functional role of EEC progenitor-enriched miRNA, miR-7, by in vivo dietary study as well as ex vivo enteroid in mice. RESULTS: First, we demonstrate that miR-7 is highly enriched across the entire EEC lineage trajectory and is the most enriched miRNA in EEC progenitors relative to Lgr5+ intestinal stem cells. Next, we show in vivo that in EEC progenitors miR-7 is dramatically suppressed under dietary conditions that favor crypt division and suppress EEC abundance. We then demonstrate by functional assays in mouse enteroids that miR-7 exerts robust control of growth, as determined by budding (proxy for crypt division), EdU and PH3 staining, and likely regulates EEC abundance also. Finally, we show by single-cell RNA sequencing analysis that miR-7 regulates Xiap in progenitor/stem cells and we demonstrate in enteroids that the effects of miR-7 on mouse enteroid growth depend in part on Xiap and Egfr signaling. CONCLUSIONS: This study demonstrates for the first time that EEC progenitor cell-enriched miR-7 is altered by dietary perturbations and that it regulates growth in enteroids via intact Xiap and Egfr signaling.


Assuntos
Células Enteroendócrinas/fisiologia , Proteínas Inibidoras de Apoptose/genética , Mucosa Intestinal/fisiologia , MicroRNAs/metabolismo , Células-Tronco/fisiologia , Animais , Linhagem da Célula/genética , Proliferação de Células/genética , Células Cultivadas , Biologia Computacional , Receptores ErbB/metabolismo , Comportamento Alimentar/fisiologia , Feminino , Proteínas Inibidoras de Apoptose/metabolismo , Mucosa Intestinal/citologia , Masculino , Camundongos , Camundongos Transgênicos , Modelos Animais , Organoides , Cultura Primária de Células , RNA-Seq , Transdução de Sinais/genética , Análise de Célula Única
6.
Dev Cell ; 47(1): 98-111.e5, 2018 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-30220569

RESUMO

Tissue homeostasis involves a complex balance of developmental signals and environmental cues that dictate stem cell function. We found that dietary lipids control enteroendocrine cell production from Drosophila posterior midgut stem cells. Dietary cholesterol influences new intestinal cell differentiation in an Hr96-dependent manner by altering the level and duration of Notch signaling. Exogenous lipids modulate Delta ligand and Notch extracellular domain stability and alter their trafficking in endosomal vesicles. Lipid-modulated Notch signaling occurs in other nutrient-dependent tissues, suggesting that Delta trafficking in many cells is sensitive to cellular sterol levels. These diet-mediated alterations in young animals contribute to a metabolic program that persists after the diet changes. A low-sterol diet also slows the proliferation of enteroendocrine tumors initiated by Notch pathway disruption. Thus, a specific dietary nutrient can modify a key intercellular signaling pathway to shift stem cell differentiation and cause lasting changes in tissue structure and physiology.


Assuntos
Colesterol na Dieta/efeitos adversos , Lipídeos/fisiologia , Receptores Notch/efeitos dos fármacos , Animais , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Colesterol/metabolismo , Colesterol na Dieta/metabolismo , Proteínas de Drosophila/efeitos dos fármacos , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Enterócitos/metabolismo , Células Enteroendócrinas/efeitos dos fármacos , Células Enteroendócrinas/fisiologia , Intestinos/citologia , Peptídeos e Proteínas de Sinalização Intracelular , Metabolismo dos Lipídeos/genética , Metabolismo dos Lipídeos/fisiologia , Proteínas de Membrana , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Esteróis/metabolismo
7.
Mol Nutr Food Res ; 62(3)2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28961371

RESUMO

SCOPE: Enteroendocrine cell lines are routinely assayed in simple buffers at ≈20% oxygen to screen foods for bioactives that boost satiety hormone levels. However, in vivo, enteroendocrine cells are exposed to different phases of food digestion and function at low oxygen concentration, ranging from 7.5% in the stomach to 0.5% in the colon-rectal junction. METHODS AND RESULTS: The objective of this study is to investigate the effect of physiologically relevant O2 concentrations of the gut on the production and secretion of the satiety hormone, glucagon-like peptide 1 (GLP-1), from the murine enteroendocrine cell line, secretin tumor cell line (STC-1), in response to dairy macronutrients as they transit the gut. GLP-1 exocytosis from STC-1 cells is influenced by both oxygen concentration and by individual macronutrients. At low oxygen, STC-1 cell viability is significantly improved for all macronutrient stimulations and cyclic adenosine monophosphate levels are dampened. GLP-1 secretion from STC-1 cells is influenced by both the phase of yogurt digestion and corresponding O2 concentration. Atmospheric oxygen at 4.5% combined with upper gastric digesta, which simulates ileum conditions, yields the highest GLP-1 response. CONCLUSION: This demonstrates the importance of considering physiological oxygen levels and food digestion along gastrointestinal tract for reliable in vitro analysis of gut hormone secretion.


Assuntos
Células Enteroendócrinas/fisiologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Nutrientes/farmacologia , Oxigênio/metabolismo , Iogurte , Animais , Sinalização do Cálcio , Hipóxia Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Digestão , Células Enteroendócrinas/citologia , Células Enteroendócrinas/efeitos dos fármacos , Hormônios Gastrointestinais/metabolismo , Camundongos , Oxigênio/farmacologia
8.
Curr Opin Endocrinol Diabetes Obes ; 25(1): 12-21, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29194046

RESUMO

PURPOSE OF REVIEW: To summarize the most recent findings relevant to the biology of serotonin (5-hydroxytryptamine; 5-HT) and the enzyme tryptophan hydroxylase (TPH) in human gastrointestinal disease. RECENT FINDINGS: Serotonin is synthesized in the central nervous system (CNS) and the gastrointestinal tract where it is secreted from enteroendocrine cells. Its biosynthesis is regulated by two isoforms of the enzyme TPH of which TPH1 is localized predominantly in gastrointestinal enteroendocrine cells. Serotonin activates the peristaltic reflexes, regulates gastrointestinal motility, and has a role in intestinal inflammation. Inhibition of TPH with novel molecules represents a new pharmacological tool in the successful management of carcinoid syndrome in patients with gastrointestinal neuroendocrine tumors (GI-NETs). Certain 5-HT receptor subtype agonists and antagonists are useful in the treatment of functional gastrointestinal disorders. SUMMARY: The gastrointestinal tract is the largest storage organ for serotonin where its biosynthesis is regulated by TPH1. It has several important functions in gastrointestinal motility, secretion, and inflammation. Furthermore, TPH represents a target for inhibitory pharmacological therapy of serotonin access states such as the carcinoid syndrome.


Assuntos
Serotonina/fisiologia , Triptofano Hidroxilase/fisiologia , Animais , Células Enteroendócrinas/fisiologia , Gastroenteropatias/etiologia , Gastroenteropatias/metabolismo , Motilidade Gastrointestinal/fisiologia , Humanos
9.
PLoS One ; 12(11): e0186507, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29091949

RESUMO

INTRODUCTION: Intestinal atresia, a rare congenital condition, is often associated with intestinal motility disorders despite adequate neonatal surgery. Previous studies have focused on changes in the enteric nervous system (ENS). We hypothesized that other components of the digestive tract could be involved in this condition. MATERIAL AND METHODS: In a rat model of surgically-induced intestinal obstruction, a transcriptome analysis was performed to measure the global gene expression. Then, analyzes were focused on genes expressed in ENS and neuroendocrine cells. Rat fetus small intestines at different developmental stages (ED15, ED17, ED19 and ED21, (n = 22)) were studied as controls and compared to the upper and lower segments of small intestines from rat fetuses with surgically-induced obstruction (n = 14; ligature at ED18). The gene expression pattern was confirmed by immunohistochemistry, electron microscopy and RT-qPCR. RESULTS: From ED15 to ED21, there was a physiological decrease in the gene expression of ENS markers and an increase in that of neuroendocrine genes. Regarding operated embryos, the changes in global gene expression were significantly higher in the proximal segment compared to the distal segment (18% vs. 9%). More precisely, a decrease in ENS gene expression and an increase in neuroendocrine gene expression were observed in the proximal segment compared to controls, indicating an accelerated maturation pattern. Immunohistochemistry and electron microscopy confirmed these findings. CONCLUSION: Fetal intestinal obstruction seems to induce an accelerated maturation in the proximal segment. Moreover, neuroendocrine cells undergo significant unexpected changes, suggesting that ENS changes could be associated with other changes to induce intestinal motility disorders.


Assuntos
Células Enteroendócrinas/fisiologia , Obstrução Intestinal/fisiopatologia , Animais , Células Enteroendócrinas/metabolismo , Feminino , Perfilação da Expressão Gênica , Atresia Intestinal/fisiopatologia , Gravidez , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real
10.
Endocr Dev ; 32: 8-19, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28873381

RESUMO

Gastrointestinal hormones are released from enteroendocrine cells in the digestive tract. More than 30 hormone genes are expressed, which make the gut the largest endocrine organ in the body. At present, it is feasible to conceive the hormones under 5 headings: the structural homology groups most hormones into 9 families, each of which is assumed to originate from a single gene. Today's hormone gene often has multiple phenotypes due to alternative splicing, tandem organization or differentiated maturation of the prohormone. By these mechanisms, more than 100 different hormonal peptides are released from the gut. Gut hormones are also widely expressed in extraintestinal cells. These cells may release different fragments of the same prohormone due to cell-specific processing pathways. Moreover, endocrine cells, immune cells, neurons, myocytes, kidney cells, sperm cells and cancer cells secrete gut peptides in different ways, so the same peptide may act for instance as a hormone, a neurotransmitter, a cytokine, a growth factor or a fertility factor. The targets of gastrointestinal hormones are specific G-protein coupled receptors that are expressed in the cell membrane all over the body. Thus, each gut hormone constitutes a regulatory system operating in the whole organism.


Assuntos
Endocrinologia/tendências , Hormônios Gastrointestinais/fisiologia , Trato Gastrointestinal/fisiologia , Animais , Glândulas Endócrinas , Células Enteroendócrinas/fisiologia , Hormônios Gastrointestinais/genética , Humanos , Fenótipo , Receptores Acoplados a Proteínas G/fisiologia
11.
Curr Opin Gastroenterol ; 33(6): 439-445, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28806271

RESUMO

PURPOSE OF REVIEW: We report recently published knowledge regarding gut chemosensory mechanisms focusing on nutrient-sensing G protein-coupled receptors (GPCRs) expressed on gut enteroendocrine cells (EECs), tuft cells, and in afferent nerves in the gastroduodenal mucosa and submucosa. RECENT FINDINGS: Gene profiling of EECs and tuft cells have revealed expression of a variety of nutrient-sensing GPCRs. The density of EEC and tuft cells is altered by luminal environmental changes that may occur following bypass surgery or in the presence of mucosal inflammation. Some EECs and tuft cells are directly linked to sensory nerves in the subepithelial space. Vagal afferent neurons that innervate the intestinal villi express nutrient receptors, contributing to the regulation of duodenal anion secretion in response to luminal nutrients. Nutrients are also absorbed via specific epithelial transporters. SUMMARY: Gastric and duodenal epithelial cells are continually exposed to submolar concentrations of nutrients that activate GPCRs expressed on EECs, tuft cells, and submucosal afferent nerves and are also absorbed through specific transporters, regulating epithelial cell proliferation, gastrointestinal physiological function, and metabolism. The chemical coding and distribution of EECs and tuft cells are keys to the development of GPCR-targeted therapies.


Assuntos
Células Quimiorreceptoras/fisiologia , Duodeno/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Intestinal/metabolismo , Vias Aferentes/fisiologia , Cirurgia Bariátrica , Duodeno/inervação , Células Enteroendócrinas/fisiologia , Mucosa Gástrica/inervação , Humanos , Mucosa Intestinal/inervação , Receptores Acoplados a Proteínas G/metabolismo
12.
J Diabetes Investig ; 7 Suppl 1: 8-12, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27186349

RESUMO

Glucose-dependent insulinotropic polypeptide (GIP) was established as a gut hormone more than 40 years ago, and there is good experimental support for its role as an incretin hormone although deletion of the GIP receptor or the GIP cells or GIP receptor mutations have only minor effects on glucose metabolism. Unlike the related hormone, GLP-1, GIP stimulates the secretion of glucagon, which in healthy individuals may help to stabilize glucose levels, but in people with type 2 diabetes may contribute to glucose intolerance. A role in lipid metabolism is supported by numerous indirect observations and by resistance to diet-induced obesity after deletion of the GIP receptor. However, a clear effect on lipid clearance could not be identified in humans, raising doubt about its importance. The GIP receptor is widely expressed in the body and also appears to be expressed on bone cells, and experimental studies in rodent point to effects on bone metabolism. Recent studies revealed pronounced inhibitory effects of GIP on bone resorption markers in humans and suggest that GIP may be (one of the) gastrointestinal regulators of bone turn-over. In support of this, a loss-of-function GIP receptor mutation in humans is associated with a marked increase in fracture risk. The lack of a reliable GIP receptor antagonist contributes to the uncertainty regarding the physiological role of GIP.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Polipeptídeo Inibidor Gástrico/metabolismo , Receptores dos Hormônios Gastrointestinais/metabolismo , Animais , Osso e Ossos/metabolismo , Células Enteroendócrinas/fisiologia , Glucagon/metabolismo , Humanos , Metabolismo dos Lipídeos
13.
J Diabetes Investig ; 7 Suppl 1: 87-93, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27186362

RESUMO

The gut epithelium's large surface area, its direct exposure to ingested nutrients, its vast stem cell population and its immunotolerogenic environment make it an excellent candidate for therapeutic cells to treat diabetes. Thus, several attempts have been made to coax immature gut cells to differentiate into insulin-producing cells by altering the expression patterns of specific transcription factors. Furthermore, because of similarities in enteroendocrine and pancreatic endocrine cell differentiation pathways, other approaches have used genetically engineered enteroendocrine cells to produce insulin in addition to their endogenous secreted hormones. Several studies support the utility of both of these approaches for the treatment of diabetes. Converting a patient's own gut cells into meal-regulated insulin factories in a safe and immunotolerogenic environment is an attractive approach to treat and potentially cure diabetes. Here, we review work on these approaches and indicate where we feel further advancements are required.


Assuntos
Engenharia Celular , Diabetes Mellitus/terapia , Células Enteroendócrinas/fisiologia , Células Secretoras de Insulina/fisiologia , Insulina/uso terapêutico , Células-Tronco/fisiologia , Animais , Diferenciação Celular , Células Enteroendócrinas/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina , Transplante das Ilhotas Pancreáticas , Camundongos , Ratos , Transplante de Células-Tronco
14.
Curr Opin Gastroenterol ; 32(2): 61-6, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26839962

RESUMO

PURPOSE OF REVIEW: Alterations in small intestinal physiology are proposed to play a causative role in the beneficial impact of Roux-en-Y gastric bypass on type 2 diabetes mellitus. The present article describes the key proposed mechanisms implicated with an emphasis on some of the newer findings in the field. RECENT FINDINGS: Augmented incretin and diminished anti-incretin effects postsurgery are explored and a model proposed that reconciles the hindgut and foregut hypotheses of improved glycaemic control as being complementary rather than mutually exclusive. Synthesis of recent findings on postbypass changes in intestinal glucose handling then follows. Finally an updated view of the role of distal bile diversion and changes in the microbiota on enteroendocrine signalling is presented. SUMMARY: A series of nonmutually exclusive changes in small intestinal physiology likely make a significant contribution to improved glycaemic control postgastric bypass. Longitudinal data indicate that these effects do not translate into a long-term cure. A number of surgery-induced changes, however, are amenable to device-based and pharmacology-based mimicry, and this is an area for prioritization of future research focus.


Assuntos
Bile/metabolismo , Diabetes Mellitus Tipo 2/cirurgia , Células Enteroendócrinas/metabolismo , Derivação Gástrica , Trato Gastrointestinal/metabolismo , Intestino Delgado/cirurgia , Obesidade Mórbida/cirurgia , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/metabolismo , Células Enteroendócrinas/fisiologia , Trato Gastrointestinal/fisiopatologia , Trato Gastrointestinal/cirurgia , Humanos , Intestino Delgado/metabolismo , Obesidade Mórbida/complicações , Obesidade Mórbida/metabolismo , Indução de Remissão , Transdução de Sinais , Resultado do Tratamento
15.
J Clin Endocrinol Metab ; 101(3): 778-86, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26649620

RESUMO

CONTEXT: Classical enteroenteroendocrine cell (EEC) biology evolved historically from identification of scattered hormone-producing endocrine cells within the epithelial mucosa of the stomach, small and large intestine. Purification of functional EEC hormones from intestinal extracts, coupled with molecular cloning of cDNAs and genes expressed within EECs has greatly expanded the complexity of EEC endocrinology, with implications for understanding the contribution of EECs to disease pathophysiology. EVIDENCE ACQUISITION: Pubmed searches identified manuscripts highlighting new concepts illuminating the molecular biology, classification and functional role(s) of EECs and their hormonal products. EVIDENCE SYNTHESIS: Molecular interrogation of EECs has been transformed over the past decade, raising multiple new questions that challenge historical concepts of EEC biology. Evidence for evolution of the EEC from a unihormonal cell type with classical endocrine actions, to a complex plurihormonal dynamic cell with pleiotropic interactive functional networks within the gastrointestinal mucosa is critically assessed. We discuss gaps in understanding how EECs sense and respond to nutrients, cytokines, toxins, pathogens, the microbiota, and the microbial metabolome, and highlight the expanding translational relevance of EECs in the pathophysiology and therapy of metabolic and inflammatory disorders. CONCLUSIONS: The EEC system represents the largest specialized endocrine network in human physiology, integrating environmental and nutrient cues, enabling neural and hormonal control of metabolic homeostasis. Updating EEC classification systems will enable more accurate comparative analyses of EEC subpopulations and endocrine networks in multiple regions of the gastrointestinal tract.


Assuntos
Células Enteroendócrinas/fisiologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Homeostase , Humanos , Neoplasias Intestinais/etiologia , Tumores Neuroendócrinos/etiologia , Terminologia como Assunto
16.
Horm Res Paediatr ; 83(1): 11-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25592084

RESUMO

Obesity is an epidemic and medical issue. Investigating the pathways regulating appetite, food intake, and body weight is crucial to find strategies for the prevention and treatment of obesity. In the context of therapeutic strategies, we focus here on the potential of enteroendocrine cells (EECs) and their secreted hormones in the regulation of body weight. We review the role of the enteroendocrine system during weight loss after lifestyle intervention or after bariatric surgery. We discuss the therapeutic potential of EECs and their hormones as targets for new treatment strategies. In fact, targeting nutrient receptors of EECs with a nutritional approach, pharmaceutical agents or prebiotics delivered to the lumen may provide a promising new approach.


Assuntos
Apetite/fisiologia , Peso Corporal/fisiologia , Células Enteroendócrinas/fisiologia , Obesidade/fisiopatologia , Saciação/fisiologia , Redução de Peso/fisiologia , Animais , Ingestão de Alimentos/fisiologia , Humanos , Obesidade/tratamento farmacológico
17.
J Pediatr Gastroenterol Nutr ; 60(2): 192-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25171319

RESUMO

OBJECTIVES: Severe congenital diarrhea occurs in approximately half of patients with Aristaless-Related Homeobox (ARX) null mutations. The cause of this diarrhea is unknown. In a mouse model of intestinal Arx deficiency, the prevalence of a subset of enteroendocrine cells is altered, leading to diarrhea. Because polyalanine expansions within the ARX protein are the most common mutations found in ARX-related disorders, we sought to characterize the enteroendocrine population in human tissue of an ARX mutation and in a mouse model of the corresponding polyalanine expansion (Arx). METHODS: Immunohistochemistry and quantitative real-time polymerase chain reaction were the primary modalities used to characterize the enteroendocrine populations. Daily weights were determined for the growth curves, and Oil-Red-O staining on stool and tissue identified neutral fats. RESULTS: An expansion of 7 alanines in the first polyalanine tract of both human ARX and mouse Arx altered enteroendocrine differentiation. In human tissue, cholecystokinin, glucagon-like peptide 1, and somatostatin populations were reduced, whereas the chromogranin A population was unchanged. In the mouse model, cholecystokinin and glucagon-like peptide 1 populations were also lost, although the somatostatin-expressing population was increased. The ARX protein was present in human tissue, whereas the Arx protein was degraded in the mouse intestine. CONCLUSIONS: ARX/Arx is required for the specification of a subset of enteroendocrine cells in both humans and mice. Owing to protein degradation, the Arx mouse recapitulates findings of the intestinal Arx null model, but is not able to further the study of the differential effects of the ARX protein on its transcriptional targets in the intestine.


Assuntos
Diarreia/genética , Duodenopatias/genética , Células Enteroendócrinas/fisiologia , Proteínas de Homeodomínio/genética , Pseudo-Obstrução Intestinal/genética , Peptídeos/metabolismo , Fatores de Transcrição/genética , Adolescente , Animais , Diferenciação Celular/genética , Colecistocinina/análise , Cromogranina A/análise , Diarreia/patologia , Modelos Animais de Doenças , Duodenopatias/patologia , Duodeno/patologia , Células Enteroendócrinas/química , Células Enteroendócrinas/patologia , Insuficiência de Crescimento/genética , Feminino , Peptídeo 1 Semelhante ao Glucagon/análise , Proteínas de Homeodomínio/análise , Humanos , Pseudo-Obstrução Intestinal/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese Insercional , Somatostatina/análise , Esteatorreia/genética , Fatores de Transcrição/análise
18.
J Clin Invest ; 125(1): 379-85, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25500886

RESUMO

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.


Assuntos
Células Enteroendócrinas/fisiologia , Incretinas/metabolismo , Insulina/metabolismo , Animais , Células Cultivadas , Dieta Hiperlipídica , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Intolerância à Glucose , Humanos , Íleo/citologia , Secreção de Insulina , Masculino , Camundongos Endogâmicos C57BL , Receptores Notch/antagonistas & inibidores , Receptores Notch/metabolismo
19.
Gut ; 64(4): 618-26, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25015642

RESUMO

OBJECTIVE: Inhibition of food intake and glucose homeostasis are both promoted when nutrients stimulate enteroendocrine cells (EEC) to release gut hormones. Several specific nutrient receptors may be located on EEC that respond to dietary sugars, amino acids and fatty acids. Bypass surgery for obesity and type II diabetes works by shunting nutrients to the distal gut, where it increases activation of nutrient receptors and mediator release, but cellular mechanisms of activation are largely unknown. We determined which nutrient receptors are expressed in which gut regions and in which cells in mouse and human, how they are associated with different types of EEC, how they are activated leading to hormone and 5-HT release. DESIGN AND RESULTS: mRNA expression of 17 nutrient receptors and EEC mediators was assessed by quantitative PCR and found throughout mouse and human gut epithelium. Many species similarities emerged, in particular the dense expression of several receptors in the distal gut. Immunolabelling showed specific colocalisation of receptors with EEC mediators PYY and GLP-1 (L-cells) or 5-HT (enterochromaffin cells). We exposed isolated proximal colonic mucosa to specific nutrients, which recruited signalling pathways within specific EEC extracellular receptor-regulated kinase (p-ERK) and calmodulin kinase II (pCAMKII), as shown by subsequent immunolabelling, and activated release of these mediators. Aromatic amino acids activated both pathways in mouse, but in humans they induced only pCAMKII, which was colocalised mainly with 5-HT expression. Activation was pertussis toxin-sensitive. Fatty acid (C12) potently activated p-ERK in human in all EEC types and evoked potent release of all three mediators. CONCLUSIONS: Specific nutrient receptors associate with distinct activation pathways within EEC. These may provide discrete, complementary pharmacological targets for intervention in obesity and type II diabetes.


Assuntos
Células Enteroendócrinas/fisiologia , Alimentos , Receptores de Superfície Celular/fisiologia , Animais , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Tecidos
20.
J Clin Endocrinol Metab ; 99(9): E1691-5, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24878048

RESUMO

BACKGROUND: The incretin effect is reduced in type 2 diabetes mellitus (T2DM) patients. Whether the impaired function of the enteropancreatic axis in these patients is due to defective GLP-1 receptor (GLP-1R) expression in extrapancreatic target organs is not known. AIMS AND METHODS: To compare the GLP-1R expression and distribution in gastric mucosa biopsies of patients with (n =22) and without (n =22) T2DM referred for routine esophagogastroduodenoscopies. GLP-1R mRNA levels were estimated by real-time PCR. The intensity of GLP-1R immunostaining, frequency, and types of glandular cells bearing GLP-1R and their glandular distribution in different stomach mucosa regions were evaluated by immunohistochemical morphological semiquantitative and quantitative analysis. RESULTS: Mean mRNA GLP-1R levels were significantly reduced in patients with T2DM compared with nondiabetic patients (P < .02). Immunohistochemical analysis revealed that the reduced GLP-1R expression in T2DM patients was due to a decreased intensity of immunostaining (P < .01). The number of glandular GLP-1R-bearing cells in both body and antrum mucosa was decreased in T2DM patients. Most notably, the frequency of GLP-1R immunoreactive acid-secreting parietal cells was reduced in the neck area of the gastric principal glands of T2DM patients (P < .01). No correlation was found between the reduced GLP-1R expression and clinical parameters including body mass index, age, glycosylated hemoglobin, and disease duration. CONCLUSION: This is the first evidence of reduced GLP-1R expression in gastric glands of T2DM patients. These data demonstrate that the defective function of the incretin axis in T2DM may also result from decreased GLP-1R expression in its extrapancreatic target organs.


Assuntos
Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Mucosa Gástrica/fisiologia , Receptores de Glucagon/genética , Receptores de Glucagon/metabolismo , Adulto , Idoso , Biópsia , Endoscopia do Sistema Digestório , Células Enteroendócrinas/citologia , Células Enteroendócrinas/fisiologia , Feminino , Mucosa Gástrica/citologia , Regulação da Expressão Gênica , Receptor do Peptídeo Semelhante ao Glucagon 1 , Humanos , Masculino , Pessoa de Meia-Idade , Células Parietais Gástricas/citologia , Células Parietais Gástricas/fisiologia , RNA Mensageiro/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA