Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 186(16): 3386-3399.e15, 2023 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-37541196

RESUMO

The gastrointestinal tract is in a state of constant motion. These movements are tightly regulated by the presence of food and help digestion by mechanically breaking down and propelling gut content. Mechanical sensing in the gut is thought to be essential for regulating motility; however, the identity of the neuronal populations, the molecules involved, and the functional consequences of this sensation are unknown. Here, we show that humans lacking PIEZO2 exhibit impaired bowel sensation and motility. Piezo2 in mouse dorsal root, but not nodose ganglia is required to sense gut content, and this activity slows down food transit rates in the stomach, small intestine, and colon. Indeed, Piezo2 is directly required to detect colon distension in vivo. Our study unveils the mechanosensory mechanisms that regulate the transit of luminal contents throughout the gut, which is a critical process to ensure proper digestion, nutrient absorption, and waste removal.


Assuntos
Trânsito Gastrointestinal , Canais Iônicos , Mecanotransdução Celular , Animais , Humanos , Camundongos , Digestão , Canais Iônicos/metabolismo , Neurônios/metabolismo
2.
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
3.
Int J Mol Sci ; 25(2)2024 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-38255906

RESUMO

Individuals with autism often experience gastrointestinal issues but the cause is unknown. Many gene mutations that modify neuronal synapse function are associated with autism and therefore may impact the enteric nervous system that regulates gastrointestinal function. A missense mutation in the Nlgn3 gene encoding the cell adhesion protein Neuroligin-3 was identified in two brothers with autism who both experienced severe gastrointestinal dysfunction. Mice expressing this mutation (Nlgn3R451C mice) are a well-studied preclinical model of autism and show autism-relevant characteristics, including impaired social interaction and communication, as well as repetitive behaviour. We previously showed colonic dysmotility in response to GABAergic inhibition and increased myenteric neuronal numbers in the small intestine in Nlgn3R451C mice bred on a mixed genetic background. Here, we show that gut dysfunction is a persistent phenotype of the Nlgn3 R451C mutation in mice backcrossed onto a C57BL/6 background. We report that Nlgn3R451C mice show a 30.9% faster gastrointestinal transit (p = 0.0004) in vivo and have 6% longer small intestines (p = 0.04) compared to wild-types due to a reduction in smooth muscle tone. In Nlgn3R451C mice, we observed a decrease in resting jejunal diameter (proximal jejunum: 10.6% decrease, p = 0.02; mid: 9.8%, p = 0.04; distal: 11.5%, p = 0.009) and neurally regulated dysmotility as well as shorter durations of contractile complexes (mid: 25.6% reduction in duration, p = 0.009; distal: 30.5%, p = 0.004) in the ileum. In Nlgn3R451C mouse colons, short contractions were inhibited to a greater extent (57.2% by the GABAA antagonist, gabazine, compared to 40.6% in wild-type mice (p = 0.007). The inhibition of nitric oxide synthesis decreased the frequency of contractile complexes in the jejunum (WT p = 0.0006, Nlgn3R451C p = 0.002), but not the ileum, in both wild-type and Nlgn3R451C mice. These findings demonstrate that changes in enteric nervous system function contribute to gastrointestinal dysmotility in mice expressing the autism-associated R451C missense mutation in the Neuroligin-3 protein.


Assuntos
Transtorno Autístico , Masculino , Animais , Camundongos , Camundongos Endogâmicos C57BL , Transtorno Autístico/genética , Trânsito Gastrointestinal , Intestino Delgado , Jejuno , Modelos Animais de Doenças , Cafeína , Antagonistas GABAérgicos
4.
Med Mol Morphol ; 56(3): 177-186, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36995439

RESUMO

Adenomatous polyposis coli (APC) is recognized as an antioncogene related to familial adenomatous polyposis and colorectal cancers. However, APC is a large protein with multiple binding partners, indicating APC has diverse roles besides as a tumor suppressor. We have ever studied the roles of APC by using APC1638T/1638T (APC1638T) mice. Through those studies, we have noticed stools of APC1638T mice were smaller than those of APC+/+ mice and hypothesized there be a disturbance in fecal formation processes in APC1638T mice. The gut motility was morphologically analyzed by immunohistochemical staining of the Auerbach's plexus. Gut microbiota was analyzed by terminal restriction fragment length polymorphism (T-RFLP). IgA concentration in stools was determined by enzyme-linked immunosorbent assay (ELISA). As results, macroscopic findings suggestive of large intestinal dysmotility and microscopic findings of disorganization and inflammation of the plexus were obtained in APC1638T mice. An alteration of microbiota composition, especially increased Bacteroidetes population was observed. Increases in IgA positive cells and dendritic cells in the ileum with high fecal IgA concentration were also confirmed, suggesting over-activation of gut immunity. Our findings will contribute to our understanding of APC's functions in the gastrointestinal motility, and lead to a development of novel therapies for gut dysmotility-related diseases.


Assuntos
Proteína da Polipose Adenomatosa do Colo , Polipose Adenomatosa do Colo , Camundongos , Animais , Proteína da Polipose Adenomatosa do Colo/metabolismo , Imunoglobulina A
5.
Pharmacol Res ; 181: 106291, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35690329

RESUMO

Saccharomyces boulardii (Sb) is a widely used fungal probiotic in treating various digestive diseases, including irritable bowel syndrome (IBS). However, the specific mechanisms of Sb relieving IBS remain unclear. The abnormal serotonin transporter (SERT) / 5-hydroxytryptamine (5-HT) system could cause disordered gastrointestinal sensation and motility, which closely related to IBS pathogenesis. The aim of this study was to explore the effects and mechanisms of Sb on regulating gut motility. Sb supernatant (SbS) was administered to intestinal epithelial cells and mice. SbS upregulated SERT expression via enhancing heparin-binding epidermal growth factor (HB-EGF) release to activate epidermal growth factor receptor (EGFR). EGFR kinase inhibitor treatment or HB-EGF siRNA transfection in cells blocked SbS upregulating SERT. Consistently, SbS-treated mice presented inhibited gut motility, and EGFR activation and SERT upregulation were found. Moreover, 16 S rDNA sequence presented an evident decrease in Firmicutes / Bacteroidetes ratio in SbS group. In genus level, SbS reduced Escherichia_Shigella, Alistipes, Clostridium XlVa, and Saccharibacteria_genera_incertae_sedis, meanwhile, increased Parasutterella. The abundance of Saccharibacteria_genera_incertae_sedis positively correlated with defecation parameters and intestinal 5-HT content. Fecal microbiota transplantation showed that SbS could modulate gut microbiota to influence gut motility. Interestingly, elimination of gut microbiota with antibiotic cocktail did not entirely block SbS regulating gut motility. Furthermore, SbS administration to IBS-D mice significantly upregulated SERT and inhibited gut motility. In conclusion, SbS could upregulate SERT by EGFR activation, and modulate gut microbiota to inhibit gut motility. This finding would provide more evidence for the application of this yeast probiotic in IBS and other diarrheal disorders.


Assuntos
Microbioma Gastrointestinal , Síndrome do Intestino Irritável , Probióticos , Saccharomyces boulardii , Animais , Bactérias/metabolismo , Receptores ErbB/metabolismo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/metabolismo , Camundongos , Probióticos/farmacologia , Saccharomyces boulardii/metabolismo , Serotonina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo
6.
Adv Exp Med Biol ; 1383: 271-294, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36587166

RESUMO

Over 150 years ago, methods for quantitative analysis of gastrointestinal motor patterns first appeared. Graphic representations of physiological variables were recorded with the kymograph after the mid-1800s. Changes in force or length of intestinal muscles could be quantified, however most recordings were limited to a single point along the digestive tract.In parallel, photography and cinematography with X-Rays visualised changes in intestinal shape, but were hard to quantify. More recently, the ability to record physiological events at many sites along the gut in combination with computer processing allowed construction of spatiotemporal maps. These included diameter maps (DMaps), constructed from video recordings of intestinal movements and pressure maps (PMaps), constructed using data from high-resolution manometry catheters. Combining different kinds of spatiotemporal maps revealed additional details about gut wall status, including compliance, which relates forces to changes in length. Plotting compliance values along the intestine enabled combined DPMaps to be constructed, which can distinguish active contractions and relaxations from passive changes. From combinations of spatiotemporal maps, it is possible to deduce the role of enteric circuits and pacemaker cells in the generation of complex motor patterns. Development and application of spatiotemporal methods to normal and abnormal motor patterns in animals and humans is ongoing, with further technical improvements arising from their combination with impedance manometry, magnetic resonance imaging, electrophysiology, and ultrasonography.


Assuntos
Motilidade Gastrointestinal , Intestino Delgado , Humanos , Animais , Motilidade Gastrointestinal/fisiologia , Manometria/métodos , Gravação em Vídeo , Músculos
7.
World J Microbiol Biotechnol ; 38(12): 222, 2022 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-36100774

RESUMO

Intestinal peristalsis is essential for gastrointestinal function, which could maintain the appropriate progression and digestion of food and reduce bacterial aggregation through mixing function. Even though certain ingredients of foodstuff are known to increase or decrease intestinal peristalsis, the role of environmental pollutants on intestinal peristalsis is relatively unknown. Therefore, the effects of four typical environmental pollutants (oxytetracycline, arsenic, polychlorinated biphenyls and chlorpyrifos) on intestinal peristalsis in the zebrafish model and then tested the recovery effect of the constipation-resistant probiotic. The results showed that 4-day environmental pollutants exposures on the zebrafish embryos at 1 day post fertilization clearly decreased the intestinal peristalsis through decreasing the serotonin (5-HT) production and down-regulating the expression of key genes involved in 5-HT synthesis. Pollutants-evoked change of gut motility could be normalized in the presence of Lactobacillus rhamnosus GG (LGG) via increasing 5-HT secretion. Exogenous 5-hydroxytryptophan (100 µg/L) could also rescue the dysfunction of gut motility in pollutants-treated zebrfish. The data identified that LGG normalized disorder of intestinal peristalsis induced by environmental pollutants through increasing 5-HT level. The stimulant effect of LGG on peristalsis may be associated with 5-HT system, which could provide references for the application of probiotics in regulation of gut dysmotility.


Assuntos
Poluentes Ambientais , Lacticaseibacillus rhamnosus , Animais , Poluentes Ambientais/metabolismo , Poluentes Ambientais/toxicidade , Lacticaseibacillus rhamnosus/genética , Lacticaseibacillus rhamnosus/metabolismo , Larva , Serotonina/metabolismo , Peixe-Zebra
8.
Dig Dis Sci ; 66(10): 3505-3515, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33063188

RESUMO

BACKGROUND AND AIMS: Limited means exist to assess gastrointestinal activity in a noninvasive, objective way that is highly predictive of underlying motility disorders. The aim of this paper is to demonstrate the feasibility of recording myoelectric gastrointestinal activity by cutaneous patches and to correlate myoelectric signals with gastrointestinal function in various clinical settings. METHODS: A novel wireless patch system (WPS) (G-Tech Medical) that acquires gastrointestinal myoelectrical signals was placed on the patients' anterior abdomens. Data were transmitted wirelessly to a mobile device with a user interface and forwarded to a cloud server where processing algorithms identified episodes of motor activity, quantified their parameters, and nominally assigned them to specific gastrointestinal organs based on their frequencies. RESULTS: The inherent reproducibility of the WPS measurement technique itself and from the underlying gut activity, coupled with source validation and sensitivity to changes in gut activity in several physiologic and pathologic states, demonstrates its feasibility, safety, and performance in clinical settings. CONCLUSIONS: The novel WPS technology, measuring myoelectric intestinal activity noninvasively and continuously over multiple days, is feasible in a wide range of clinical settings, highlighting its promise in the diagnosis and management of motility disorders. Further research is required for more extensive validation and to determine how best to employ this information to optimize patient care.


Assuntos
Fenômenos Eletrofisiológicos/fisiologia , Gastroenteropatias/diagnóstico , Motilidade Gastrointestinal/fisiologia , Trato Gastrointestinal/fisiologia , Monitorização Fisiológica/instrumentação , Tecnologia sem Fio/instrumentação , Gastroenteropatias/patologia , Humanos , Monitorização Fisiológica/métodos , Projetos Piloto
9.
BMC Pediatr ; 21(1): 536, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34852816

RESUMO

BACKGROUND: It has been proven that gut microbiota alterations are involved in the development of Henoch-Schönlein Purpura (HSP). However, the pathogenesis of HSP hasn't been eluciated. This study was to investigate the impact of gut microbiota from HSP on ASIC3 expression and interactions between microbiota and ASIC3 expression in the development of HSP. METHODS: Feces collected from HSP and healthy children at the First Affiliated Hospital of Anhui Medical University were made into fecal microbial solutions. Germ-free rats were randomly assigned to either the control or HSP groups. The HSP group of rats were administered the fecal microbiota solution of HSP children, while the control group rats were administered the fecal microbiota solution of healthy children. Abdominal withdrawal reflex (AWR) and intestinal propulsion rate of the rats were used to determine visceral sensitivity. Composition of the gut microbiota of HSP children was determined using 16S rRNA gene sequencing. ASIC3 expression in the colon was ascertained through qRT-PCR as well as western blotting analysis. RESULTS: The results showed a reduction in the number of species and abundance in the intestinal microbiota of children with HSP. Visceral sensitivity and intestinal propulsion rate of HSP group rats increased significantly, compared with the control group. Colon ASIC3 mRNA and protein levels in the HSP group were found to be upregulated. The microbiota dysbiosis of HSP patients could stimulate ASIC3 expression in the colon of Germ-free rats, which in turn affected intestinal motility. CONCLUSIONS: These results suggested that HSP children had intestinal microbiota disorder, which might affect gut motility by down-regulating colon ASIC3 expression in rats.


Assuntos
Microbioma Gastrointestinal , Vasculite por IgA , Animais , Motilidade Gastrointestinal , Humanos , Canais Iônicos , RNA Ribossômico 16S/genética , Ratos
10.
Gastroenterology ; 155(2): 514-528.e6, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29782847

RESUMO

BACKGROUND & AIMS: Strategies are needed to increase gastrointestinal transit without systemic pharmacologic agents. We investigated whether optogenetics, focal application of light to control enteric nervous system excitability, could be used to evoke propagating contractions and increase colonic transit in mice. METHODS: We generated transgenic mice with Cre-mediated expression of light-sensitive channelrhodopsin-2 (ChR2) in calretinin neurons (CAL-ChR2 Cre+ mice); Cre- littermates served as controls. Colonic myenteric neurons were analyzed by immunohistochemistry, patch-clamp, and calcium imaging studies. Motility was assessed by mechanical, electrophysiological, and video recording in vitro and by fecal output in vivo. RESULTS: In isolated colons, focal light stimulation of calretinin enteric neurons evoked classic polarized motor reflexes (50/58 stimulations), followed by premature anterograde propagating contractions (39/58 stimulations). Light stimulation could evoke motility from sites along the entire colon. These effects were prevented by neural blockade with tetrodotoxin (n = 2), and did not occur in control mice (n = 5). Light stimulation of proximal colon increased the proportion of natural fecal pellets expelled over 15 minutes in vitro (75% ± 17% vs 32% ± 8% for controls) (P < .05). In vivo, activation of wireless light-emitting diodes implanted onto the colon wall significantly increased hourly fecal pellet output in conscious, freely moving mice (4.2 ± 0.4 vs 1.3 ± 0.3 in controls) (P < .001). CONCLUSIONS: In studies of mice, we found that focal activation of a subset of enteric neurons can increase motility of the entire colon in vitro, and fecal output in vivo. Optogenetic control of enteric neurons might therefore be used to modify gut motility.


Assuntos
Colo/fisiologia , Sistema Nervoso Entérico/fisiologia , Trânsito Gastrointestinal/efeitos da radiação , Luz , Optogenética/métodos , Animais , Calbindina 2/genética , Calbindina 2/metabolismo , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Channelrhodopsins/efeitos da radiação , Colo/inervação , Colo/efeitos da radiação , Sistema Nervoso Entérico/citologia , Trânsito Gastrointestinal/genética , Camundongos , Camundongos Transgênicos , Modelos Animais , Neurônios/metabolismo , Neurônios/efeitos da radiação
11.
Dev Dyn ; 247(2): 268-278, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28975691

RESUMO

The enteric nervous system (ENS) is the largest part of the peripheral nervous system and is entirely neural crest-derived. It provides the intrinsic innervation of the gut, controlling different aspects of gut function, such as motility. In this review, we will discuss key points of Zebrafish ENS development, genes, and signaling pathways regulating ENS development, as well as contributions of the Zebrafish model system to better understand ENS disorders. During their migration, enteric progenitor cells (EPCs) display a gradient of developmental states based on their proliferative and migratory characteristics, and show spatiotemporal heterogeneity based on gene expression patterns. Many genes and signaling pathways that regulate the migration and proliferation of EPCs have been identified, but later stages of ENS development, especially steps of neuronal and glial differentiation, remain poorly understood. In recent years, Zebrafish have become increasingly important to test candidate genes for ENS disorders (e.g., from genome-wide association studies), to identify environmental influences on ENS development (e.g., through large-scale drug screens), and to investigate the role the gut microbiota play in ENS development and disease. With its unique advantages as a model organism, Zebrafish will continue to contribute to a better understanding of ENS development, function, and disease. Developmental Dynamics 247:268-278, 2018. © 2017 Wiley Periodicals, Inc.


Assuntos
Sistema Nervoso Entérico , Peixe-Zebra , Animais , Sistema Nervoso Entérico/fisiologia , Sistema Nervoso Entérico/fisiopatologia , Microbioma Gastrointestinal , Regulação da Expressão Gênica no Desenvolvimento , Estudo de Associação Genômica Ampla , Modelos Biológicos , Transdução de Sinais
12.
Am J Physiol Gastrointest Liver Physiol ; 314(3): G341-G348, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29167116

RESUMO

Colorectal motility is regulated by two defecation centers located in the brain and spinal cord. In previous studies, we have shown that administration of serotonin (5-HT) in the lumbosacral spinal cord causes enhancement of colorectal motility. Because spinal 5-HT is derived from neurons of the medullary raphe nuclei, including the raphe magnus, raphe obscurus, and raphe pallidus, we examined whether stimulation of the medullary raphe nuclei enhances colorectal motility via the lumbosacral defecation center. Colorectal pressure was recorded with a balloon in vivo in anesthetized rats. Electrical stimulation of the medullary raphe nuclei failed to enhance colorectal motility. Because GABAergic neurons can be simultaneously activated by the raphe stimulation and released GABA masks accelerating actions of the raphe nuclei on the lumbosacral defecation center, a GABAA receptor antagonist was preinjected intrathecally to manifest excitatory responses. When spinal GABAA receptors were blocked by the antagonist, electrical stimulation of the medullary raphe nuclei increased colorectal contractions. This effect of the raphe nuclei was inhibited by intrathecal injection of 5-hydroxytryptamine type 2 (5-HT2) and type 3 (5-HT3) receptor antagonists. In addition, injection of a selective 5-HT reuptake inhibitor in the lumbosacral spinal cord augmented the raphe stimulation-induced enhancement of colorectal motility. Transection of the pelvic nerves, but not transection of the colonic nerves, prevented the effect of the raphe nuclei on colorectal motility. These results demonstrate that activation of the medullary raphe nuclei causes augmented contractions of the colorectum via 5-HT2 and 5-HT3 receptors in the lumbosacral defecation center. NEW & NOTEWORTHY We have shown that electrical stimulation of the medullary raphe nuclei causes augmented contractions of the colorectum via pelvic nerves in rats. The effect of the medullary raphe nuclei on colorectal motility is exerted through activation of 5-hydroxytryptamine type 2 and type 3 receptors in the lumbosacral defecation center. The descending serotoninergic raphespinal tract represents new potential therapeutic targets against colorectal dysmotility such as irritable bowel syndrome.


Assuntos
Colo/inervação , Defecação , Motilidade Gastrointestinal , Plexo Lombossacral/fisiologia , Bulbo/fisiologia , Núcleos da Rafe/fisiologia , Neurônios Serotoninérgicos/fisiologia , Animais , Defecação/efeitos dos fármacos , Estimulação Elétrica , Neurônios GABAérgicos/metabolismo , Neurônios GABAérgicos/fisiologia , Motilidade Gastrointestinal/efeitos dos fármacos , Injeções Espinhais , Plexo Lombossacral/efeitos dos fármacos , Plexo Lombossacral/metabolismo , Masculino , Bulbo/metabolismo , Inibição Neural , Pressão , Núcleos da Rafe/metabolismo , Ratos Sprague-Dawley , Receptores de GABA-A/metabolismo , Receptores 5-HT2 de Serotonina/metabolismo , Receptores 5-HT3 de Serotonina/metabolismo , Neurônios Serotoninérgicos/efeitos dos fármacos , Neurônios Serotoninérgicos/metabolismo , Serotonina/administração & dosagem , Serotonina/metabolismo
13.
Nitric Oxide ; 73: 81-88, 2018 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-28602746

RESUMO

Periodontal disease (PD), a severe form of gum disease, is among the most prevalent chronic infection in humans and is associated with complex microbial synergistic dysbiosis in the subgingival cavity. The immune system of the body interacts with the microbes as the plaque extends and propagates below the gingival sulcus. Once bacteria reach the gingival sulcus, it can enter the blood stream and affect various areas of the human body. The polymicrobial nature of periodontal disease, if left untreated, promotes chronic inflammation, not only within the oral cavity, but also throughout the human body. Alterations seen in the concentrations of healthy gut microbiota may lead to systemic alterations, such as gut motility disorders, high blood pressure, and atherosclerosis. Although gut microbiome has been shown to play a vital role in intestinal motility functions, the role of oral bacteria in this setting remains to be investigated. It is unclear whether oral microbial DNA is present in the large intestine and, if so, whether it alters the gut microbiome. In addition, polybacterial infection induced PD reduced nitric oxide (NO) synthesis and antioxidant enzymes in rodent colon. In this review, we will discuss the interactions between oral and gut microbiome, specifics of how the oral microbiome may modulate the activities of the gut microbiome, and possible ramifications of these alterations.


Assuntos
Microbioma Gastrointestinal , Boca/microbiologia , Óxido Nítrico/fisiologia , Doenças Periodontais/microbiologia , Biofilmes , Gastroenteropatias/microbiologia , Motilidade Gastrointestinal , Humanos , Saliva/microbiologia
14.
J Clin Pharm Ther ; 43(5): 752-756, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30014556

RESUMO

WHAT IS KNOWN AND OBJECTIVE: Irritable bowel syndrome (IBS) is a complex and chronic, relapsing gastrointestinal condition that affects more than 10% of the population worldwide. There is a pressing need for new therapeutic strategies in the management of IBS. Increasing research has shed light on the modulatory functions of melatonin on pain, local inflammation and motility in the gastrointestinal tract. However, melatonin's effects are limited by its extensive first-pass metabolism and short half-life. COMMENT: Agomelatine, a naphthalene analog of melatonin, is a novel melatonergic drug with a longer half-life and a comparatively greater affinity for MT1 and MT2 melatonin receptors than melatonin itself. Agomelatine also shows serotonin 5-HT3 receptor antagonist activity, which is theoretically of benefit for patients with IBS with diarrhoea (IBS-D) as it regulates gastrointestinal motility and visceral sensory mechanisms. Although only one clinical study of agomelatine use in patients with IBS exists, we believe that agomelatine is a safe and efficacious multimodal agent with untapped potential in the management of IBS. WHAT IS NEW AND CONCLUSION: Numerous comorbidities are associated with IBS, including chronic pain syndromes and psychiatric disorders. Coupled with its antidepressant actions, agomelatine could serve as an effective adjunct therapeutic. Agomelatine should be considered in our therapeutic armamentarium for IBS management.


Assuntos
Acetamidas/uso terapêutico , Hipnóticos e Sedativos/uso terapêutico , Síndrome do Intestino Irritável/tratamento farmacológico , Adolescente , Adulto , Idoso , Feminino , Humanos , Síndrome do Intestino Irritável/metabolismo , Masculino , Melatonina/uso terapêutico , Pessoa de Meia-Idade , Receptores de Serotonina/metabolismo , Adulto Jovem
15.
Dev Biol ; 417(2): 168-81, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27235816

RESUMO

Co-ordinated gastrointestinal function is the result of integrated communication between the enteric nervous system (ENS) and "effector" cells in the gastrointestinal tract. Unlike smooth muscle cells, interstitial cells, and the vast majority of cell types residing in the mucosa, enteric neurons and glia are not generated within the gut. Instead, they arise from neural crest cells that migrate into and colonise the developing gastrointestinal tract. Although they are "later" arrivals into the developing gut, enteric neural crest-derived cells (ENCCs) respond to many of the same secreted signalling molecules as the "resident" epithelial and mesenchymal cells, and several factors that control the development of smooth muscle cells, interstitial cells and epithelial cells also regulate ENCCs. Much progress has been made towards understanding the migration of ENCCs along the gastrointestinal tract and their differentiation into neurons and glia. However, our understanding of how enteric neurons begin to communicate with each other and extend their neurites out of the developing plexus layers to innervate the various cell types lining the concentric layers of the gastrointestinal tract is only beginning. It is critical for postpartum survival that the gastrointestinal tract and its enteric circuitry are sufficiently mature to cope with the influx of nutrients and their absorption that occurs shortly after birth. Subsequently, colonisation of the gut by immune cells and microbiota during postnatal development has an important impact that determines the ultimate outline of the intrinsic neural networks of the gut. In this review, we describe the integrated development of the ENS and its target cells.


Assuntos
Sistema Nervoso Entérico/embriologia , Trato Gastrointestinal/inervação , Mesoderma/embriologia , Crista Neural/embriologia , Animais , Comunicação Celular/fisiologia , Diferenciação Celular , Movimento Celular/fisiologia , Trato Gastrointestinal/embriologia , Humanos , Crista Neural/citologia , Neurônios/citologia , Transdução de Sinais/fisiologia
16.
Am J Med Genet C Semin Med Genet ; 175(4): 496-506, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29082627

RESUMO

CHARGE syndrome is an autosomal dominant genetic condition that is primarily diagnosed based on clinical features, with genetic testing available for confirmation. The CHARGE mnemonic stands for some of the common characteristics: coloboma, heart defects, atresia/stenosis of the choanae, retardation of growth/development, genitourinary anomalies, and ear abnormalities (CHARGE). However, many of the common clinical features are not captured by this mnemonic, including cranial nerve dysfunction, considered by some to be one of the major diagnostic criteria. Over 90% of individuals experience feeding and gastrointestinal dysfunction, which carries great morbidity and mortality. The aim of this review is to examine the nature of gastrointestinal (GI) symptoms and feeding difficulties in CHARGE syndrome, focusing on their underlying pathology, associated investigations, and available treatment options. We also provide information on available tools (for parents, clinicians, and researchers) that are important additions to the lifelong healthcare management of every individual with CHARGE syndrome. We review how cranial nerve dysfunction is one of the most important characteristics underlying the pervasive GI and feeding dysfunction, and discuss the need for future research on gut innervation and motility in this genetic disorder.


Assuntos
Síndrome CHARGE/diagnóstico , Síndrome CHARGE/etiologia , Transtornos da Alimentação e da Ingestão de Alimentos/etiologia , Trato Gastrointestinal/anormalidades , Trato Gastrointestinal/fisiopatologia , Fenótipo , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/fisiopatologia , Animais , Síndrome CHARGE/terapia , Doenças dos Nervos Cranianos/genética , Doenças dos Nervos Cranianos/fisiopatologia , Feminino , Motilidade Gastrointestinal/genética
17.
Am J Med Genet C Semin Med Genet ; 175(1): 181-187, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28186368

RESUMO

Current evidence suggests that an association exists between non-inflammatory hereditary disorders of connective tissue such as the Ehlers-Danlos syndromes (EDS) and gastrointestinal (GI) symptoms. Patients with EDS can present with both structural problems such as hiatus hernias, visceroptosis, rectoceles, and rectal prolapse as well as functional problems such as disordered gut motility. It has recently been demonstrated that patients with hypermobile EDS (hEDS) present with GI symptoms related to the fore and hind-gut and these patients frequently meet the criteria for functional gastrointestinal disorders such as functional dyspepsia and irritable bowel syndrome. Presence of GI symptoms in EDS patients influences their quality of life. Specific evidence based management guidelines for the management of GI symptoms in EDS patients do not exist and these patients are often treated symptomatically. There is, however, recognition that certain precautions need to be taken for those patients undergoing surgical treatment. Future studies are required to identify the mechanisms that lead to GI symptoms in patients with EDS and more specific treatment guidelines are required. © 2017 Wiley Periodicals, Inc.


Assuntos
Síndrome de Ehlers-Danlos/complicações , Gastroenteropatias/etiologia , Gerenciamento Clínico , Gastroenteropatias/diagnóstico , Humanos , Guias de Prática Clínica como Assunto , Qualidade de Vida
18.
J Transl Med ; 15(1): 13, 2017 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-28086815

RESUMO

BACKGROUND: The gastrointestinal motility is affected by gut microbiota and the relationship between them has become a hot topic. However, mechanisms of microbiota in regulating motility have not been well defined. We thus investigated the effect of microbiota depletion by antibiotics on gastrointestinal motility, colonic serotonin levels, and bile acids metabolism. METHODS: After 4 weeks with antibiotics treatments, gastrointestinal and colon transit, defecation frequency, water content, and other fecal parameters were measured and analyzed in both wild-type and antibiotics-treated mice, respectively. Contractility of smooth muscle, serotonin levels, and bile acids levels in wild-type and antibiotics-treated mice were also analyzed. RESULTS: After antibiotics treatment, the richness and diversity of intestinal microbiota decreased significantly, and the fecal of mice had less output (P < 0.01), more water content (P < 0.01), and longer pellet length (P < 0.01). Antibiotics treatment in mice also resulted in delayed gastrointestinal and colonic motility (P < 0.05), and inhibition of phasic contractions of longitudinal muscle from isolated proximal colon (P < 0.01). In antibiotics-treated mice, serotonin, tryptophan hydroxylase 1, and secondary bile acids levels were decreased. CONCLUSION: Gut microbiota play an important role in the regulation of intestinal bile acids and serotonin metabolism, which could probably contribute to the association between gut microbiota and gastrointestinal motility as intermediates.


Assuntos
Antibacterianos/farmacologia , Motilidade Gastrointestinal/efeitos dos fármacos , Microbiota/efeitos dos fármacos , Serotonina/biossíntese , Animais , Ácidos e Sais Biliares/metabolismo , Ceco/efeitos dos fármacos , Ceco/patologia , Fezes , Microbioma Gastrointestinal/efeitos dos fármacos , Masculino , Metaboloma/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Contração Muscular , Tamanho do Órgão/efeitos dos fármacos
19.
Exp Physiol ; 102(9): 1118-1132, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28671737

RESUMO

NEW FINDINGS: What is the central question of this study? What are the dynamical rules governing interstitial cell of Cajal (ICC)-generated slow wave contractions in the small intestine, as reflected in their phase response curve and state space? What is the main finding and its importance? The phase response curve has a region of phase advance surrounding a phase delay peak. This pattern is important in generating a stable synchrony within the ICC network and is related to the state space of the ICC; in particular, the phase delay peak corresponds to the unstable equilibrium point that threads the ICC's limit cycle. Interstitial cells of Cajal (ICCs) generate electrical oscillations in the gut. Synchronization of the ICC population is required for generation of coherent electrical waves ('slow waves') that cause muscular contraction and thereby move gut content. The phase response curve (PRC) is an experimental measure of the dynamical rules governing a population of oscillators that determine their synchrony and gives an experimental window onto the state space of the oscillator, its dynamical landscape. We measured the PRC of slow wave contractions in the mouse small intestine by the novel combination of diameter mapping and single pulse electrical field stimulation. Phase change (τ) was measured as a function of old phase (ϕ) and distance from the stimulation electrode (d). Plots of τ(ϕ, d) showed an arrowhead-shaped region of phase advance enclosing at its base a phase delay peak. The phase change mirrored the perturbed pattern of contraction waves in response to a pulse. The (ϕ, d) plane is the surface of a displacement tube extending from the limit cycle through state space. To visualize the state space vector field on this tube, latent phase (ϕlat ) was calculated from τ. At the transition from advance to delay, isochrons made boomerang turns before tightening and winding around the phase delay peak corresponding to the unstable equilibrium point that threads the limit cycle. This isochron foliation had previously been observed in oscillator models such as the Fitzhugh-Nagumo but has not been demonstrated experimentally. The spatial extension of the PRC afforded by diameter mapping allows a better understanding of the dynamical properties of ICCs and how they synchronize as a population.


Assuntos
Intestino Delgado/fisiologia , Contração Muscular/fisiologia , Animais , Feminino , Células Intersticiais de Cajal/fisiologia , Camundongos , Músculo Liso/fisiologia
20.
FASEB J ; 29(4): 1395-403, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25550456

RESUMO

Gut microbiota alterations have been described in several diseases with altered gastrointestinal (GI) motility, and awareness is increasing regarding the role of the gut microbiome in modulating GI function. Serotonin [5-hydroxytryptamine (5-HT)] is a key regulator of GI motility and secretion. To determine the relationship among gut microbes, colonic contractility, and host serotonergic gene expression, we evaluated mice that were germ-free (GF) or humanized (HM; ex-GF colonized with human gut microbiota). 5-HT reduced contractile duration in both GF and HM colons. Microbiota from HM and conventionally raised (CR) mice significantly increased colonic mRNAs Tph1 [(tryptophan hydroxylase) 1, rate limiting for mucosal 5-HT synthesis; P < 0.01] and chromogranin A (neuroendocrine secretion; P < 0.01), with no effect on monoamine oxidase A (serotonin catabolism), serotonin receptor 5-HT4, or mouse serotonin transporter. HM and CR mice also had increased colonic Tph1 protein (P < 0.05) and 5-HT concentrations (GF, 17 ± 3 ng/mg; HM, 25 ± 2 ng/mg; and CR, 35 ± 3 ng/mg; P < 0.05). Enterochromaffin (EC) cell numbers (cells producing 5-HT) were unchanged. Short-chain fatty acids (SCFAs) promoted TPH1 transcription in BON cells (human EC cell model). Thus, gut microbiota acting through SCFAs are important determinants of enteric 5-HT production and homeostasis.


Assuntos
Colo/metabolismo , Sistema Digestório/microbiologia , Células Enterocromafins/metabolismo , Ácidos Graxos Voláteis/metabolismo , Serotonina/biossíntese , Animais , Contagem de Células , Linhagem Celular , Cromogranina A/genética , Colo/citologia , Colo/microbiologia , Células Enterocromafins/citologia , Células Enterocromafins/microbiologia , Feminino , Motilidade Gastrointestinal , Vida Livre de Germes , Humanos , Masculino , Camundongos , Microbiota , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Triptofano Hidroxilase/genética , Triptofano Hidroxilase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA