Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 146
Filtrar
1.
Int J Mol Sci ; 22(17)2021 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-34502396

RESUMO

The monoamine serotonin, 5-hydroxytryptamine (5-HT), is a remarkable molecule with conserved production in prokaryotes and eukaryotes and a wide range of functions. In the gastrointestinal tract, enterochromaffin cells are the most important source for 5-HT production. Some intestinal bacterial species are also able to produce 5-HT. Besides its role as a neurotransmitter, 5-HT acts on immune cells to regulate their activation. Several lines of evidence indicate that intestinal 5-HT signaling is altered in patients with inflammatory bowel disease. In this review, we discuss the current knowledge on the production, secretion, and signaling of 5-HT in the intestine. We present an inventory of intestinal immune and epithelial cells that respond to 5-HT and describe the effects of these signaling processes on intestinal homeostasis. Further, we detail the mechanisms by which 5-HT could affect inflammatory bowel disease course and describe the effects of interventions that target intestinal 5-HT signaling.


Assuntos
Trato Gastrointestinal/metabolismo , Serotonina/metabolismo , Serotonina/fisiologia , Animais , Colite , Células Enterocromafins/metabolismo , Células Enterocromafins/fisiologia , Células Epiteliais/metabolismo , Trato Gastrointestinal/fisiologia , Homeostase/fisiologia , Humanos , Inflamação , Doenças Inflamatórias Intestinais , Mucosa Intestinal/metabolismo , Intestinos , Transdução de Sinais/efeitos dos fármacos
2.
Gastroenterology ; 160(7): 2451-2466.e19, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33662386

RESUMO

BACKGROUND & AIMS: Gastrointestinal (GI) motility is regulated by serotonin (5-hydroxytryptamine [5-HT]), which is primarily produced by enterochromaffin (EC) cells in the GI tract. However, the precise roles of EC cell-derived 5-HT in regulating gastric motility remain a major point of conjecture. Using a novel transgenic mouse line, we investigated the distribution of EC cells and the pathophysiologic roles of 5-HT deficiency in gastric motility in mice and humans. METHODS: We developed an inducible, EC cell-specific Tph1CreERT2/+ mouse, which was used to generate a reporter mouse line, Tph1-tdTom, and an EC cell-depleted line, Tph1-DTA. We examined EC cell distribution, morphology, and subpopulations in reporter mice. GI motility was measured in vivo and ex vivo in EC cell-depleted mice. Additionally, we evaluated 5-HT content in biopsy and plasma specimens from patients with idiopathic gastroparesis (IG). RESULTS: Tph1-tdTom mice showed EC cells that were heterogeneously distributed throughout the GI tract with the greatest abundance in the antrum and proximal colon. Two subpopulations of EC cells were identified in the gut: self-renewal cells located at the base of the crypt and mature cells observed in the villi. Tph1-DTA mice displayed delayed gastric emptying, total GI transit, and colonic transit. These gut motility alterations were reversed by exogenous provision of 5-HT. Patients with IG had a significant reduction of antral EC cell numbers and 5-HT content, which negatively correlated with gastric emptying rate. CONCLUSIONS: The Tph1CreERT2/+ mouse provides a powerful tool to study the functional roles of EC cells in the GI tract. Our findings suggest a new pathophysiologic mechanism of 5-HT deficiency in IG.


Assuntos
Esvaziamento Gástrico/genética , Trânsito Gastrointestinal/genética , Serotonina/deficiência , Animais , Linhagem Celular , Células Enterocromafins/fisiologia , Humanos , Camundongos , Camundongos Transgênicos , Triptofano Hidroxilase/metabolismo
3.
Immunity ; 54(1): 151-163.e6, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33220232

RESUMO

The gastrointestinal tract is known as the largest endocrine organ that encounters and integrates various immune stimulations and neuronal responses due to constant environmental challenges. Enterochromaffin (EC) cells, which function as chemosensors on the gut epithelium, are known to translate environmental cues into serotonin (5-HT) production, contributing to intestinal physiology. However, how immune signals participate in gut sensation and neuroendocrine response remains unclear. Interleukin-33 (IL-33) acts as an alarmin cytokine by alerting the system of potential environmental stresses. We here demonstrate that IL-33 induced instantaneous peristaltic movement and facilitated Trichuris muris expulsion. We found that IL-33 could be sensed by EC cells, inducing release of 5-HT. IL-33-mediated 5-HT release activated enteric neurons, subsequently promoting gut motility. Mechanistically, IL-33 triggered calcium influx via a non-canonical signaling pathway specifically in EC cells to induce 5-HT secretion. Our data establish an immune-neuroendocrine axis in calibrating rapid 5-HT release for intestinal homeostasis.


Assuntos
Células Enterocromafins/fisiologia , Interleucina-33/metabolismo , Intestinos/fisiologia , Neurônios/fisiologia , Serotonina/metabolismo , Tricuríase/imunologia , Trichuris/fisiologia , Animais , Sinalização do Cálcio , Homeostase , Interleucina-33/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuroimunomodulação , Peristaltismo
4.
Am J Physiol Gastrointest Liver Physiol ; 319(4): G494-G501, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32845170

RESUMO

Small intestinal neuroendocrine tumors (SI-NET) are serotonin-secreting well-differentiated neuroendocrine tumors of putative enterochromaffin (EC) cell origin. Recent studies recognize a subset of EC cells that is label-retaining at the +4 position in the crypt and functions as a reserve intestinal stem cell. Importantly, this +4 reserve EC cell subset not only contributes to regeneration of the intestinal epithelium during injury and inflammation but also to basal crypt homeostasis at a constant rate. The latter function suggests that the +4 EC cell subset serves as an active reserve stem cell via a constant rate of dedifferentiation. Characterization of early tumor formation of SI-NET, observed as crypt-based EC cell clusters in many cases of familial SI-NETs, suggests that the +4 active reserve EC cell subset is the cell of origin. This newly discovered active reserve stem cell property of EC cells can account for unique biological mechanisms and processes associated with the genesis and development of SI-NETs. The recognition of this property of the +4 active reserve EC cell subset may provide novel opportunities to explore NETs in the gastrointestinal tract and other organs.


Assuntos
Células Enterocromafins/patologia , Neoplasias Intestinais/patologia , Intestino Delgado/patologia , Tumores Neuroendócrinos/patologia , Células-Tronco/patologia , Células-Tronco/fisiologia , Animais , Carcinogênese/patologia , Desdiferenciação Celular , Células Enterocromafins/fisiologia , Humanos , Camundongos , Tumores Neuroendócrinos/metabolismo , Serotonina/metabolismo
5.
Life Sci ; 239: 116886, 2019 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-31678286

RESUMO

Enterochromaffin (EC) cell is the main cell type that responsible for 5-hydroxytryptamine (5-HT) synthesis, storage and release of the gut. Intestinal 5-HT play a key role in visceral sensation, intestinal motility and permeability, EC cell hyperplasia and increased 5-HT bioavailability in the gut have been found to be involved in the symptoms generation of irritable bowel syndrome and inflammatory bowel disease. EC cells originate from intestinal stem cells, the interaction between proliferation and differentiation signals on intestinal stem cells enable EC cell number to be regulated in a normal level. This review focuses on the impact factors, pathogenesis mechanisms, and therapeutic clues for intestinal EC cells hyperplasia, and showed that EC cell hyperplasia was observed under the condition of physiological stress, intestinal infection or intestinal inflammation, the disordered proliferation and/or differentiation of intestinal stem cells as well as their progenitor cells all contribute to the pathogenesis of intestinal EC cell hyperplasia. The altered intestinal niche, i.e. increased corticotrophin releasing factor (CRF) signal, elevated nerve growth factor (NGF) signal, and Th2-dominant cytokines production, has been found to have close correlation with intestinal EC cell hyperplasia. Currently, CRF receptor antagonist, nuclear factor-κB inhibitor, and NGF receptor neutralizing antibody have been proved useful to attenuate intestinal EC cell hyperplasia, which may provide a promising clue for the therapeutic strategy in EC cell hyperplasia related diseases.


Assuntos
Células Enterocromafins/metabolismo , Células Enterocromafins/fisiologia , Hiperplasia/patologia , Animais , Colo/metabolismo , Humanos , Hiperplasia/metabolismo , Infecções/metabolismo , Inflamação/metabolismo , Intestinos/patologia , Síndrome do Intestino Irritável/metabolismo , Serotonina/metabolismo , Estresse Fisiológico/fisiologia
6.
J Vis Exp ; (139)2018 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-30320764

RESUMO

Enterochromaffin (EC) cells in the gastrointestinal (GI) epithelium constitute the largest subpopulation of enteroendocrine cells. As specialized sensory cells, EC cells sense luminal stimuli and convert them into serotonin (5-hyroxytryptamine, 5-HT) release events. However, the electrophysiology of these cells is poorly understood because they are difficult to culture and to identify. The method presented in this paper outlines primary EC cell cultures optimized for single cell electrophysiology. This protocol utilizes a transgenic cyan fluorescent protein (CFP) reporter to identify mouse EC cells in mixed primary cultures, advancing the approach to obtaining high-quality recordings of whole cell electrophysiology in voltage- and current-clamp modes.


Assuntos
Fenômenos Eletrofisiológicos , Células Enterocromafins/fisiologia , Animais , Células Cultivadas , Camundongos
7.
Proc Natl Acad Sci U S A ; 115(32): E7632-E7641, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-30037999

RESUMO

Enterochromaffin (EC) cells constitute the largest population of intestinal epithelial enteroendocrine (EE) cells. EC cells are proposed to be specialized mechanosensory cells that release serotonin in response to epithelial forces, and thereby regulate intestinal fluid secretion. However, it is unknown whether EE and EC cells are directly mechanosensitive, and if so, what the molecular mechanism of their mechanosensitivity is. Consequently, the role of EE and EC cells in gastrointestinal mechanobiology is unclear. Piezo2 mechanosensitive ion channels are important for some specialized epithelial mechanosensors, and they are expressed in mouse and human EC cells. Here, we use EC and EE cell lineage tracing in multiple mouse models to show that Piezo2 is expressed in a subset of murine EE and EC cells, and it is distributed near serotonin vesicles by superresolution microscopy. Mechanical stimulation of a subset of isolated EE cells leads to a rapid inward ionic current, which is diminished by Piezo2 knockdown and channel inhibitors. In these mechanosensitive EE cells force leads to Piezo2-dependent intracellular Ca2+ increase in isolated cells as well as in EE cells within intestinal organoids, and Piezo2-dependent mechanosensitive serotonin release in EC cells. Conditional knockout of intestinal epithelial Piezo2 results in a significant decrease in mechanically stimulated epithelial secretion. This study shows that a subset of primary EE and EC cells is mechanosensitive, uncovers Piezo2 as their primary mechanotransducer, defines the molecular mechanism of their mechanotransduction and mechanosensitive serotonin release, and establishes the role of epithelial Piezo2 mechanosensitive ion channels in regulation of intestinal physiology.


Assuntos
Células Enterocromafins/fisiologia , Canais Iônicos/metabolismo , Jejuno/fisiologia , Mecanotransdução Celular/fisiologia , Serotonina/metabolismo , Animais , Células Cultivadas , Canais Iônicos/genética , Jejuno/citologia , Camundongos , Camundongos Transgênicos , Organoides/fisiologia , Cultura Primária de Células , RNA Interferente Pequeno/metabolismo , Análise de Célula Única
9.
Artigo em Inglês | MEDLINE | ID: mdl-28485065

RESUMO

Enterochromaffin cells were the first endocrine cells of the gastrointestinal tract to be chemically distinguished, almost 150 years ago. It is now known that the chromaffin reaction of these cells was due to their content of the reactive aromatic amine, 5-hydroxytryptamine (5-HT, also known as serotonin). They have commonly been thought to be a special class of gut endocrine cells (enteroendocrine cells) that are distinct from the enteroendocrine cells that contain peptide hormones. The study by Martin et al. in the current issue of this journal reveals that the patterns of expression of nutrient receptors and transporters differ considerably between chromaffin cells of the mouse duodenum and colon. However, even within regions, chromaffin cells differ; in the duodenum there are chromaffin cells that contain both secretin and 5-HT, cholecystokinin and 5-HT, and all three of secretin, cholecystokinin, and 5-HT. Moreover, the ratios of these different cell types differ substantially between species. And, in terms of function, 5-HT has many roles, including in appetite, motility, fluid secretion, release of digestive enzymes and bone metabolism. The paper thus emphasizes the need to define the many different classes of enterochromaffin cells and relate this to their roles.


Assuntos
Células Enterocromafins/fisiologia , Trato Gastrointestinal/fisiologia , Animais , Doença Celíaca/fisiopatologia , Trato Gastrointestinal/citologia , Humanos , Síndrome do Intestino Irritável/fisiopatologia
10.
J Physiol ; 595(1): 79-91, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27392819

RESUMO

KEY POINTS: The gastrointestinal epithelial enterochromaffin (EC) cell synthesizes the vast majority of the body's serotonin. As a specialized mechanosensor, the EC cell releases this serotonin in response to mechanical forces. However, the molecular mechanism of EC cell mechanotransduction is unknown. In the present study, we show, for the first time, that the mechanosensitive ion channel Piezo2 is specifically expressed by the human and mouse EC cells. Activation of Piezo2 by mechanical forces results in a characteristic ionic current, the release of serotonin and stimulation of gastrointestinal secretion. Piezo2 inhibition by drugs or molecular knockdown decreases mechanosensitive currents, serotonin release and downstream physiological effects. The results of the present study suggest that the mechanosensitive ion channel Piezo2 is specifically expressed by the EC cells of the human and mouse small bowel and that it is important for EC cell mechanotransduction. ABSTRACT: The enterochromaffin (EC) cell in the gastrointestinal (GI) epithelium is the source of nearly all systemic serotonin (5-hydroxytryptamine; 5-HT), which is an important neurotransmitter and endocrine, autocrine and paracrine hormone. The EC cell is a specialized mechanosensor, and it is well known that it releases 5-HT in response to mechanical forces. However, the EC cell mechanotransduction mechanism is unknown. The present study aimed to determine whether Piezo2 is involved in EC cell mechanosensation. Piezo2 mRNA was expressed in human jejunum and mouse mucosa from all segments of the small bowel. Piezo2 immunoreactivity localized specifically within EC cells of human and mouse small bowel epithelium. The EC cell model released 5-HT in response to stretch, and had Piezo2 mRNA and protein, as well as a mechanically-sensitive inward non-selective cation current characteristic of Piezo2. Both inward currents and 5-HT release were inhibited by Piezo2 small interfering RNA and antagonists (Gd3+ and D-GsMTx4). Jejunum mucosal pressure increased 5-HT release and short-circuit current via submucosal 5-HT3 and 5-HT4 receptors. Pressure-induced secretion was inhibited by the mechanosensitive ion channel antagonists gadolinium, ruthenium red and D-GsMTx4. We conclude that the EC cells in the human and mouse small bowel GI epithelium selectively express the mechanosensitive ion channel Piezo2, and also that activation of Piezo2 by force leads to inward currents, 5-HT release and an increase in mucosal secretion. Therefore, Piezo2 is critical to EC cell mechanosensitivity and downstream physiological effects.


Assuntos
Células Enterocromafins/fisiologia , Canais Iônicos/fisiologia , Mecanotransdução Celular/fisiologia , Animais , Linhagem Celular , Humanos , Mucosa Intestinal/fisiologia , Intestino Delgado/fisiologia , Canais Iônicos/genética , Camundongos , Estimulação Física , Pressão , RNA Mensageiro/metabolismo , Serotonina/metabolismo
11.
Neurogastroenterol Motil ; 28(10): 1465-79, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27206689

RESUMO

BACKGROUND: Advanced age is associated with a reduction in clinical visceral pain perception. However, the underlying mechanisms remain largely unknown. Previous studies have suggested that an abnormal interplay between mast cells, enterochromaffin (EC) cells, and afferent nerves contribute to nociception in gastrointestinal disorders. The aim of this study was to investigate how aging affects afferent sensitivity and neuro-immune association in the human bowel. METHODS: Mechanical and chemical sensitivity of human bowel afferents were examined by ex vivo afferent nerve recordings. Age-related changes in the density of mast cells, EC cells, sensory nerve terminals, and mast cell-nerve micro-anatomical association were investigated by histological and immune staining. KEY RESULTS: Human afferents could be broadly classified into subpopulations displaying mechanical and chemical sensitivity, adaptation, chemo-sensitization, and recruitment. Interestingly human bowel afferent nerve sensitivity was attenuated with age. The density of substance P-immunoreactive (SP-IR) nerve varicosities was also reduced with age. In contrast, the density of ileal and colonic mucosal mast cells was increased with age, as was ileal EC cell number. An increased proportion of mast cells was found in close apposition to SP-IR nerves. CONCLUSIONS & INFERENCES: Afferent sensitivity in human bowel was reduced with advancing age. Augmentation of mast cells and EC cell numbers and the mast cell-nerve association suggest a compensatory mechanism for sensory neurodegeneration.


Assuntos
Envelhecimento/fisiologia , Colo Sigmoide/fisiologia , Células Enterocromafins/fisiologia , Íleo/fisiologia , Mastócitos/fisiologia , Neurônios Aferentes/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/patologia , Colo Sigmoide/inervação , Colo Sigmoide/patologia , Células Enterocromafins/patologia , Feminino , Humanos , Íleo/inervação , Íleo/patologia , Mucosa Intestinal/inervação , Mucosa Intestinal/patologia , Mucosa Intestinal/fisiologia , Masculino , Mastócitos/patologia , Pessoa de Meia-Idade , Técnicas de Cultura de Órgãos , Células Receptoras Sensoriais/fisiologia , Transdução de Sinais/fisiologia
12.
J Physiol ; 591(23): 5959-75, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-24099799

RESUMO

The major source of serotonin (5-HT) in the body is the enterochromaffin (EC) cells lining the intestinal mucosa of the gastrointestinal tract. Despite the fact that EC cells synthesise ∼95% of total body 5-HT, and that this 5-HT has important paracrine and endocrine roles, no studies have investigated the mechanisms of 5-HT release from single primary EC cells. We have developed a rapid primary culture of guinea-pig and human EC cells, allowing analysis of single EC cell function using electrophysiology, electrochemistry, Ca(2+) imaging, immunocytochemistry and 3D modelling. Ca(2+) enters EC cells upon stimulation and triggers quantal 5-HT release via L-type Ca(2+) channels. Real time amperometric techniques reveal that EC cells release 5-HT at rest and this release increases upon stimulation. Surprisingly for an endocrine cell storing 5-HT in large dense core vesicles (LDCVs), EC cells release 70 times less 5-HT per fusion event than catecholamine released from similarly sized LDCVs in endocrine chromaffin cells, and the vesicle release kinetics instead resembles that observed in mammalian synapses. Furthermore, we measured EC cell density along the gastrointestinal tract to create three-dimensional (3D) simulations of 5-HT diffusion using the minimal number of variables required to understand the physiological relevance of single cell 5-HT release in the whole-tissue milieu. These models indicate that local 5-HT levels are likely to be maintained around the activation threshold for mucosal 5-HT receptors and that this is dependent upon stimulation and location within the gastrointestinal tract. This is the first study demonstrating single cell 5-HT release in primary EC cells. The mode of 5-HT release may represent a unique mode of exocytosis amongst endocrine cells and is functionally relevant to gastrointestinal sensory and motor function.


Assuntos
Cálcio/fisiologia , Células Enterocromafins/fisiologia , Serotonina/fisiologia , Animais , Canais de Cálcio Tipo L/fisiologia , Células Cultivadas , Trato Gastrointestinal/citologia , Cobaias , Humanos , Cinética , Modelos Biológicos
13.
ACS Chem Neurosci ; 4(5): 879-87, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23631514

RESUMO

Constipation and fecal impaction are conditions of the bowel whose prevalence increases with age. Limited information is known about how these conditions manifest; however, functional deficits are likely to be due to changes in signaling within the bowel. This study investigated the effects of age on colonic mucosal melatonin (MEL) release and the consequences this had on colonic motility. Electrochemical measurements of MEL overflow demonstrated that both basal and mechanically stimulated MEL release decreased with age. The MEL/serotonin also decreased with increasing age, and the trend was similar to that of MEL overflow, suggestive that age-related changes were primarily due to a reduction in MEL levels. Levels of N-acetylserotonin and the N-acetylserotonin/serotonin ratio were reduced with age, providing an explanation for the reduction in MEL release. Decreases in colonic motility were observed in animals between 3 and 24 months old. Exogenous application of MEL could reverse this deficit in aged colon. In summary, we propose that the age-related decline in MEL release may be due to either decreases or alterations in mechanosensory channels and/or a loss in levels/activity of the N-acetyltransferase enzyme responsible for the synthesis of N-acetylserotonin. Decreases in MEL release may explain the decreases in colonic motility observed in 24 month old animals and could offer a new potential therapeutic treatment for age-related constipation.


Assuntos
Envelhecimento/fisiologia , Colo/metabolismo , Mucosa Intestinal/metabolismo , Melatonina/metabolismo , Animais , Depressores do Sistema Nervoso Central/farmacologia , Técnicas Eletroquímicas , Células Enterocromafins/metabolismo , Células Enterocromafins/fisiologia , Motilidade Gastrointestinal/efeitos dos fármacos , Motilidade Gastrointestinal/fisiologia , Masculino , Melatonina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Acetiltransferases N-Terminal/fisiologia , Serotonina/análogos & derivados , Serotonina/metabolismo
14.
Am J Physiol Gastrointest Liver Physiol ; 303(10): G1164-73, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22936271

RESUMO

The chemomechanosensory function of the gut enterochromaffin (EC) cell enables it to respond to dietary agents and mechanical stretch. We hypothesized that the EC cell, which also sensed alterations in luminal or mucosal oxygen level, was physiologically sensitive to fluctuations in O(2). Given that low oxygen levels induce 5-HT production and secretion through a hypoxia inducible factor 1α (HIF-1α)-dependent pathway, we also hypothesized that increasing O(2) would reduce 5-HT production and secretion. Isolated normal EC cells as well as the well-characterized EC cell model KRJ-I were used to examine HIF signaling (luciferase-assays), hypoxia transcriptional response element (HRE)-mediated transcription (PCR), signaling pathways (Western blot), and 5-HT release (ELISA) during exposure to different oxygen levels. Normal EC cells and KRJ-I cells express HIF-1α, and transient transfection with Renilla luciferase under HRE control identified a hypoxia-mediated pathway in these cells. PCR confirmed activation of HIF-downstream targets, GLUT1, IGF2, and VEGF under reduced O(2) levels (0.5%). Reducing O(2) also elevated 5-HT secretion (2-3.2-fold) as well as protein levels of HIF-1α (1.7-3-fold). Increasing O(2) to 100% inhibited HRE-mediated signaling, transcription, reduced 5-HT secretion, and significantly lowered HIF-1α levels (∼75% of control). NF-κB signaling was also elevated during hypoxia (1.2-1.6-fold), but no significant changes were noted in PKA/cAMP. We concluded that gut EC cells are oxygen responsive, and alterations in O(2) levels differentially activate HIF-1α and tryptophan hydroxylase 1, as well as NF-κB signaling. This results in alterations in 5-HT production and secretion and identifies that the chemomechanosensory role of EC cells extends to oxygen sensing.


Assuntos
Células Enterocromafins/fisiologia , Oxigênio/farmacologia , Serotonina/fisiologia , Transdução de Sinais/fisiologia , Humanos , Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , NF-kappa B/fisiologia , Oxigênio/administração & dosagem , Serotonina/metabolismo , Triptofano Hidroxilase/metabolismo
15.
Trans Am Clin Climatol Assoc ; 123: 268-80; discussion 280, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23303993

RESUMO

The gut contains the bulk of the body's serotonin (5-hydroxytryptamine, 5-HT); nevertheless, the physiological role that enteric 5-HT plays has not been determined. 5-HT is linked to gastrointestinal (GI) motility; increased intraluminal pressure causes enterochromaffin (EC) cells to secrete 5-HT, which stimulates intrinsic primary afferent neurons that initiate peristaltic reflexes. 5-HT is also an enteric neurotransmitter. Surprisingly, deletion of tryptophan hydroxylase-1 (TPH1), upon which 5-HT biosynthesis in EC cells depends, does not alter constitutive GI motility, whereas deletion of TPH2, upon which biosynthesis of neuronal 5-HT depends, slows intestinal transit and accelerates gastric emptying. TPH1 deletion, however, protects mice from experimental inflammation; 5-HT potentiation and TPH2 deletion each make inflammation more severe. Neuronal 5-HT is neuroprotective and recruits stem cells to give rise to new enteric neurons in adult mice. Mucosal 5-HT, therefore, may mobilize inflammatory effectors, which protect the gut from invasion, whereas neuronal 5-HT shields enteric neurons from inflammatory damage.


Assuntos
Sistema Nervoso Entérico/fisiologia , Gastrite/fisiopatologia , Trato Gastrointestinal/inervação , Trato Gastrointestinal/fisiologia , Neurogênese/fisiologia , Serotonina/fisiologia , Animais , Sistema Nervoso Entérico/citologia , Células Enterocromafins/citologia , Células Enterocromafins/fisiologia , Motilidade Gastrointestinal/fisiologia , Trato Gastrointestinal/citologia , Camundongos , Camundongos Knockout , Modelos Animais , Neurônios Serotoninérgicos/citologia , Neurônios Serotoninérgicos/fisiologia , Transmissão Sináptica/fisiologia , Triptofano Hidroxilase/deficiência , Triptofano Hidroxilase/genética , Triptofano Hidroxilase/fisiologia
16.
Am J Physiol Gastrointest Liver Physiol ; 301(3): G519-27, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21700904

RESUMO

The mechanisms underlying distension-evoked peristalsis in the colon are incompletely understood. It is well known that, following colonic distension, 5-hydroxytryptamine (5-HT) is released from enterochromaffin (EC) cells in the intestinal mucosa. It is also known that exogenous 5-HT can stimulate peristalsis. These observations have led some investigators to propose that endogenous 5-HT release from EC cells might be involved in the initiation of colonic peristalsis, following distension. However, because no direct evidence exists to support this hypothesis, the aim of this study was to determine directly whether release of 5-HT from EC cells was required for distension-evoked colonic peristalsis. Real-time amperometric recordings of 5-HT release and video imaging of colonic wall movements were performed on isolated segments of guinea pig distal colon, during distension-evoked peristalsis. Amperometric recordings revealed basal and transient release of 5-HT from EC cells before and during the initiation of peristalsis, respectively. However, removal of mucosa (and submucosal plexus) abolished 5-HT release but did not inhibit the initiation of peristalsis nor prevent the propagation of fecal pellets or intraluminal fluid. Maintained colonic distension by fecal pellets induced repetitive peristaltic waves, whose intrinsic frequency was also unaffected by removal of the submucosal plexus and mucosa, although their propagation velocities were slower. In conclusion, the mechanoreceptors and sensory neurons activated by radial distension to initiate peristalsis lie in the myenteric plexus and/or muscularis externa, and their activation does not require the submucosal plexus, release of 5-HT from EC cells, nor the presence of the mucosa. The propagation of peristalsis and propulsion of liquid or solid content along the colon is entrained by activity within the myenteric plexus and/or muscularis externa and does not require sensory feedback from the mucosa, nor neural inputs arising from submucosal ganglia.


Assuntos
Células Enterocromafins/fisiologia , Peristaltismo/fisiologia , Serotonina/metabolismo , Animais , Colo/fisiologia , Dilatação Patológica , Células Enterocromafins/metabolismo , Feminino , Cobaias , Técnicas In Vitro , Mucosa Intestinal/fisiologia , Masculino , Plexo Submucoso/fisiologia
17.
World J Surg ; 35(8): 1879-86, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21559999

RESUMO

Gastric carcinoid tumors comprise 7% of all gastrointestinal carcinoids and have significantly increased in incidence over the past few decades. Seventy to 80% of gastric carcinoids are type I, which usually are clinically asymptomatic and found incidentally at endoscopic evaluation for abdominal pain or anemia. In this review, advances in understanding the pathophysiology of type I gastric carcinoid are highlighted. In addition, various current diagnostic and treatment options are discussed. Although type I carcinoids generally hold a benign course, rigorous investigation is needed to ensure accurate diagnosis and optimal treatment. This includes appropriate diagnostic procedures and imaging and accurate staging of tumor. Tumor size, depth of invasion, presence of metastasis, and the tumor's gastrin dependency dictate treatment options. Appropriate treatments can consist of endoscopic resection, antrectomy, medical management, or frequent follow-up. This article provides a systematic method of evaluating and treating type I gastric carcinoid.


Assuntos
Tumor Carcinoide/diagnóstico , Tumor Carcinoide/fisiopatologia , Gastrectomia , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/fisiopatologia , Tumor Carcinoide/patologia , Tumor Carcinoide/cirurgia , Células Enterocromafins/patologia , Células Enterocromafins/fisiologia , Determinação da Acidez Gástrica , Fundo Gástrico/patologia , Fundo Gástrico/fisiopatologia , Fundo Gástrico/cirurgia , Mucosa Gástrica/patologia , Mucosa Gástrica/fisiopatologia , Gastrinas/sangue , Gastrite Atrófica/complicações , Gastrite Atrófica/patologia , Gastrite Atrófica/fisiopatologia , Gastrite Atrófica/cirurgia , Gastroscopia , Humanos , Estadiamento de Neoplasias , Pólipos/diagnóstico , Pólipos/patologia , Pólipos/fisiopatologia , Pólipos/cirurgia , Prognóstico , Antro Pilórico/patologia , Antro Pilórico/fisiopatologia , Antro Pilórico/cirurgia , Cintilografia , Fatores de Risco , Neoplasias Gástricas/patologia , Neoplasias Gástricas/cirurgia
18.
Neurogastroenterol Motil ; 23(7): e282-93, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21303432

RESUMO

BACKGROUND: Lumenal glucose initiates changes in gastrointestinal (GI) function, including inhibition of gastric emptying, stimulation of pancreatic exocrine and endocrine secretion, and intestinal fluid secretion. Glucose stimulates the release of GI hormones and 5-hydroxytryptamine (5-HT), and activates intrinsic and extrinsic neuronal pathways to initiate changes in GI function. The precise mechanisms involved in luminal glucose-sensing are not clear; studying gut endocrine cells is difficult due to their sparse and irregular localization within the epithelium. METHODS: Here we show a technique to determine activation of gut epithelial cells and the gut-brain pathway in vivo in rats using immunohistochemical detection of the activated, phosphorylated, form of calcium-calmodulin kinase II (pCaMKII). KEY RESULTS: Perfusion of the gut with glucose (60 mg) increased pCaMKII immunoreactivity in 5-HT-expressing enterochromaffin (EC) cells, cytokeratin-18 immunopositive brush cells, but not in enterocytes or cholecystokinin-expressing cells. Lumenal glucose increased pCaMKII in neurons in the myenteric plexus and nodose ganglion, nucleus of the solitary tract, dorsal motor nucleus of the vagus and the arcuate nucleus. pCaMKII expression in neurons, but not in EC cells, was significantly attenuated by pretreatment with the 5-HT(3) R antagonist ondansetron. Deoxynojirimycin, a selective agonist for the putative glucose sensor, sodium-glucose cotransporter-3 (SGLT-3), mimicked the effects of glucose with increased pCaMKII in ECs and neurons; galactose had no effect. CONCLUSIONS & INFERENCES: The data suggest that native EC cells in situ respond to glucose, possibly via SGLT-3, to activate intrinsic and extrinsic neurons and thereby regulate GI function.


Assuntos
Encéfalo/fisiologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/fisiologia , Estado de Consciência/fisiologia , Trato Gastrointestinal/fisiologia , Glucose/farmacologia , Mucosa Intestinal/fisiologia , Transdução de Sinais/efeitos dos fármacos , 1-Desoxinojirimicina/farmacologia , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/efeitos dos fármacos , Células Enterocromafins/citologia , Células Enterocromafins/fisiologia , Glucosamina/análogos & derivados , Glucosamina/farmacologia , Mucosa Intestinal/citologia , Masculino , Modelos Animais , Plexo Mientérico/fisiologia , Ondansetron/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores 5-HT3 de Serotonina/efeitos dos fármacos , Receptores 5-HT3 de Serotonina/fisiologia , Antagonistas da Serotonina/farmacologia , Transdução de Sinais/fisiologia , Proteínas de Transporte de Sódio-Glucose/efeitos dos fármacos , Proteínas de Transporte de Sódio-Glucose/fisiologia
19.
J Endocrinol ; 207(3): 329-41, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20876237

RESUMO

Although vgf gene knockout mice are hypermetabolic, administration of the VGF peptide TLQP-21 itself increased energy consumption. Agonist-antagonist roles are thus suggested for different VGF peptides, and the definition of their tissue heterogeneity is mandatory. We studied the rat stomach using antisera to C- or N-terminal sequences of known or predicted VGF peptides in immunohistochemistry and ELISA. TLQP (rat VGF(556-565)) peptide/s were most abundant (162±11 pmol/g, mean±s.e.m.) and were brightly immunostained in enterochromaffin-like (ECL) cells and somatostatin cells. A peptide co-eluting with TLQP-21 was revealed in HPLC of gastric and hypothalamic extracts, while the extended TLQP-62 form was restricted to the hypothalamus. Novel PGH (rat VGF(422-430)) peptide/s were revealed in ghrelin cells, mostly corresponding to low MW forms (0.8-1.5  kDa), while VGF C-terminus peptides were confined to neurons. VGF mRNA was present in the above gastric endocrine cell types, and was prominent in chief cells, in parallel with low-intensity staining for further cleaved products from the C-terminal region of VGF (HVLL peptides: VGF(605-614)). In swine stomach, a comparable profile of VGF peptides was revealed by immunohistochemistry. When fed and fasted rats were studied, a clear-cut, selective decrease on fasting was observed for TLQP peptides only (162±11 vs 74±5.3  pmol/g, fed versus fasted rats, mean±s.e.m., P<0.00001). In conclusion, specific VGF peptides appear to be widely represented in different gastric endocrine and other mucosal cell populations. The selective modulation of TLQP peptides suggests their involvement in peripheral neuro-endocrine mechanisms related to feeding responses and/or ECL cell regulation.


Assuntos
Ingestão de Alimentos/fisiologia , Mucosa Gástrica/metabolismo , Células Neuroendócrinas/metabolismo , Neuropeptídeos/biossíntese , Fragmentos de Peptídeos/biossíntese , Animais , Celulas Principais Gástricas/química , Células Enterocromafins/química , Células Enterocromafins/fisiologia , Jejum/fisiologia , Feminino , Grelina/análise , Hipotálamo/química , Masculino , Neuropeptídeos/análise , Fragmentos de Peptídeos/análise , Ratos , Ratos Sprague-Dawley , Células Secretoras de Somatostatina/química , Células Secretoras de Somatostatina/fisiologia , Estômago/citologia , Suínos
20.
Gastroenterology ; 139(6): 2018-2027.e2, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20708616

RESUMO

BACKGROUND & AIMS: Gastric stem cells are located in the isthmus of the gastric glands and give rise to epithelial progenitors that undergo bipolar migration and differentiation into pit and oxyntic lineages. Although gastric mucus neck cells located below the isthmus express trefoil factor family 2 (TFF2) protein, TFF2 messenger RNA transcripts are concentrated in cells above the neck region in normal corpus mucosa, suggesting that TFF2 transcription is a marker of gastric progenitor cells. METHODS: Using a BAC strategy, we generated a transgenic mouse with a tamoxifen-inducible Cre under the control of the TFF2 promoter (TFF2-BAC-Cre(ERT2)) and analyzed the lineage derivation from TFF2 mRNA transcript-expressing (TTE) cells. RESULTS: TTE cells were localized to the isthmus, above and distinct from TFF2 protein-expressing mucus neck cells. Lineage tracing revealed that these cells migrated toward the bottom of the gland within 20 days, giving rise to parietal, mucous neck, and chief cells, but not to enterochromaffin-like-cell. Surface mucus cells were not derived from TTE cells and the progeny of the TTE lineage did not survive beyond 200 days. TTE cells were localized in the isthmus adjacent to doublecortin CaM kinase-like-1(+) putative progenitor cells. Induction of spasmolytic polypeptide-expressing metaplasia with DMP-777-induced acute parietal cell loss revealed that this metaplastic phenotype might arise in part through transdifferentiation of chief cells as opposed to expansion of mucus neck or progenitor cells. CONCLUSIONS: TFF2 transcript-expressing cells are progenitors for mucus neck, parietal and zymogenic, but not for pit or enterochromaffin-like cell lineages in the oxyntic gastric mucosa.


Assuntos
Mucosa Gástrica/citologia , Mucosa Gástrica/fisiologia , Mucinas/genética , Proteínas Musculares/genética , Peptídeos/genética , Células-Tronco/citologia , Células-Tronco/fisiologia , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Celulas Principais Gástricas/citologia , Celulas Principais Gástricas/fisiologia , Duodeno/citologia , Duodeno/fisiologia , Células Enterocromafins/citologia , Células Enterocromafins/fisiologia , Integrases/genética , Rim/citologia , Rim/fisiologia , Pulmão/citologia , Pulmão/fisiologia , Camundongos , Camundongos Transgênicos , Células Parietais Gástricas/citologia , Células Parietais Gástricas/fisiologia , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Transcrição Gênica/fisiologia , Fator Trefoil-2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...