Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Physiol ; 601(7): 1183-1206, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36752210

RESUMO

The enteric nervous system (ENS) regulates the motor, secretory and defensive functions of the gastrointestinal tract. Enteric neurons integrate mechanical and chemical inputs from the gut lumen to generate complex motor outputs. How intact enteric neural circuits respond to changes in the gut lumen is not well understood. We recorded intracellular calcium in live-cell confocal recordings in neurons from intact segments of mouse intestine in order to investigate neuronal response to luminal mechanical and chemical stimuli. Wnt1-, ChAT- and Calb1-GCaMP6 mice were used to record neurons from the jejunum and colon. We measured neuronal calcium response to KCl (75 mM), veratridine (10 µM), 1,1-dimethyl-4-phenylpiperazinium (DMPP; 100 µM) or luminal nutrients (Ensure®), in the presence or absence of intraluminal distension. In the jejunum and colon, distension generated by the presence of luminal content (chyme and faecal pellets, respectively) renders the underlying enteric circuit unresponsive to depolarizing stimuli. In the distal colon, high levels of distension inhibit neuronal response to KCl, while intermediate levels of distension reorganize Ca2+ response in circumferentially propagating slow waves. Mechanosensitive channel inhibition suppresses distension-induced Ca2+ elevations, and calcium-activated potassium channel inhibition restores neuronal response to KCl, but not DMPP in the distended colon. In the jejunum, distension prevents a previously unknown tetrodotoxin-resistant neuronal response to luminal nutrient stimulation. Our results demonstrate that intestinal distension regulates the excitability of ENS circuits via mechanosensitive channels. Physiological levels of distension locally silence or synchronize neurons, dynamically regulating the excitability of enteric neural circuits based on the content of the intestinal lumen. KEY POINTS: How the enteric nervous system of the gastrointestinal tract responds to luminal distension remains to be fully elucidated. Here it is shown that intestinal distension modifies intracellular calcium levels in the underlying enteric neuronal network, locally and reversibly silencing neurons in the distended regions. In the distal colon, luminal distension is integrated by specific mechanosensitive channels and coordinates the dynamics of neuronal activation within the enteric network. In the jejunum, distension suppresses the neuronal calcium responses induced by luminal nutrients. Physiological levels of distension dynamically regulate the excitability of enteric neuronal circuits.


Assuntos
Cálcio , Sistema Nervoso Entérico , Camundongos , Animais , Sistema Nervoso Entérico/fisiologia , Neurônios/fisiologia , Intestino Delgado , Jejuno , Colo/fisiologia , Plexo Mientérico
2.
J Appl Microbiol ; 134(1)2023 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-36724218

RESUMO

AIMS: At conception, the infant gut barrier is immature, gradually developing with regular intake of maternal milk. This study addressed whether the barrier-strengthening effect of breast feeding might be attributable, at least in part, to autochthonous beneficial human milk bacteria. METHODS AND RESULTS: Twelve bacterial strains from the breast milk of Pakistani mothers who underwent cesarean delivery (NPL-88, NPL-157, NPL-179, NPL-181, NPL-388 (Limosilactobacillus reuteri), NPL-76, NPL-495, NPL-504 (Limosilactobacillus fermentum), NPL-415 (Lactobacillus pentosus), NPL-412, NPL-416 (Lactiplantibacilllus plantarum) and NPL-374 (Bifidobacterium longum) were shortlisted based on their tolerance to acidic pH (2.8-4.2) and bile (0.1-0.3%). The effect of these bacteria on gut barrier function in the presence and absence of pathogens was assessed as changes in transepithelial electrical resistance (TEER) in the human T84 colonic epithelial cell line and in murine enteroid-derived monolayers (EDMs). The TEER of T84 cells monolayers rose in the presence of most of the human milk strains, being most pronounced in case of L. reuteri NPL-88 (34% within five h), exceeding the effect of the well-known probiotic L. acidophilus (20%). qRT-PCR, western blot and immunofluorescent staining associated the increase in TEER with enhanced expression of tight junction proteins. Pretreatment of murine EDMs with NPL-88 also largely prevented the ability of the pathogen, Salmonella, to decrease TEER (87 ± 1.50%; P < 0.0001, n = 4). CONCLUSIONS: Human milk lactic acid bacteria are potential probiotics that can strengthen gut barrier function and protect breastfed neonates against enteric infections.


Assuntos
Limosilactobacillus fermentum , Limosilactobacillus reuteri , Probióticos , Lactente , Feminino , Recém-Nascido , Camundongos , Humanos , Animais , Leite Humano , Limosilactobacillus reuteri/genética , Bactérias , Probióticos/metabolismo
3.
J Pharmacol Sci ; 143(2): 65-73, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32229084

RESUMO

Glucagon-like peptide 1 (GLP-1) released from enteroendocrine (L) cells regulates insulin secretion. Intestinal inflammation and impaired GLP-1 release have been found in type 2 diabetes mellitus (T2DM) patients. Fructo-oligosaccharides (FOS), a known prebiotic, improve GLP-1 release and glucose homeostasis in T2DM models. This study aimed to investigate the effect of tumor necrosis factor-α (TNF-α), a proinflammatory cytokine associated with intestinal inflammation in T2DM, on L cell apoptosis and the effect of FOS on inflammation-associated impairment of GLP-1 secretion. Herein, using cell death assays, immunofluorescence staining, real time PCR and Western blot analyses, we found that TNF-α induced L cell apoptosis via nuclear factor kappa B (NF-κB)- inducible nitric oxide synthase (iNOS)-cleaved caspase-3-dependent pathways. Interestingly, FOS did not suppress TNF-α-induced NF-κB nuclear translocation, but inhibited expression of iNOS and cleaved caspase-3. In addition, FOS alleviated apoptosis and rescued impaired GLP-1 release in TNF-α-treated L cells. Altogether, our data indicate that TNF-α induces L cell apoptosis via an NF-κB-iNOS-caspase-3-dependent pathway. FOS may be useful in suppressing inflammation-associated L cell apoptosis and maintaining GLP-1 level in T2DM patients.


Assuntos
Apoptose/efeitos dos fármacos , Caspase 3/genética , Caspase 3/metabolismo , Células Enteroendócrinas/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Oligossacarídeos/farmacologia , Apoptose/genética , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Inflamação , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
4.
Pharmacol Res ; 100: 271-80, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26297981

RESUMO

Piperine is the principal alkaloid in black peppers (Piper nigrum L.), which is a commonly included spice in anti-diarrheal formulations. Piperine has antispasmodic activities, but its anti-secretory effect is not known. Therefore, this study investigated the anti-secretory effect of piperine and its underlying mechanism. Piperine inhibited cAMP-mediated Cl- secretion in human intestinal epithelial (T84) cells, similar to black pepper extract. Intraluminal administration of piperine (2 µg/loop) suppressed cholera toxin-induced intestinal fluid accumulation by ∼85% in mice. The anti-secretory mechanism of piperine was investigated by evaluating its effects on the activity of transport proteins involved in cAMP-mediated Cl- secretion. Notably, piperine inhibited CFTR Cl- channel activity (IC50#8'6#10 µM) without affecting intracellular cAMP levels. The mechanisms of piperine-induced CFTR inhibition did not involve MRP4-mediated cAMP efflux, AMPK or TRPV1. Piperine also inhibited cAMP-activated basolateral K+ channels, but it had no effect on Na+-K+-Cl- cotransporters or Na+-K+ ATPases. Piperine suppressed Ca2+-activated Cl- channels (CaCC) without affecting intracellular Ca2+ concentrations or Ca2+-activated basolateral K+ channels. Collectively, this study indicates that the anti-secretory effect of piperine involves the inhibition of CFTR, CaCC and cAMP-activated basolateral K+ channels. Piperine represents a novel class of drug candidates for the treatment of diarrheal diseases caused by the intestinal hypersecretion of Cl-.


Assuntos
Alcaloides/farmacologia , Benzodioxóis/farmacologia , Cloretos/metabolismo , Intestinos/efeitos dos fármacos , Piper nigrum/química , Piperidinas/farmacologia , Alcamidas Poli-Insaturadas/farmacologia , Animais , Transporte Biológico/efeitos dos fármacos , Células CACO-2 , Linhagem Celular Tumoral , Toxina da Cólera/farmacologia , AMP Cíclico/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos CFTR , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio Cálcio-Ativados/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo
5.
Methods Mol Biol ; 2367: 273-290, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33861461

RESUMO

Intestinal barrier function relies primarily on the assembly and integrity of tight junctions, which forms a size-selective barrier. This barrier restricts paracellular movement of solutes in various types of epithelia. Of note, extracellular Ca2+ concentration affects tight junction assembly. Therefore, the removal and re-addition of Ca2+ into cell culture medium of cultured intestinal epithelial cells causes destabilization and reassembly of tight junction to membrane periphery near apical surface, respectively. Based on this principle, the Ca2+-switch assay was established to investigate tight junction assembly in fully differentiated intestinal epithelial cells. This chapter provides a stepwise protocol for culture of intestinal epithelial cell monolayers using T84 cell line as an in vitro model and the Ca2+-switch assay for evaluating tight junction assembly.


Assuntos
Junções Íntimas , Cálcio , Células Epiteliais , Mucosa Intestinal , Intestinos
6.
Biomed Pharmacother ; 129: 110415, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32603892

RESUMO

Tight junctions play an important role in maintaining barrier integrity of intestinal epithelia. Activation of AMP-activated protein kinase (AMPK) promotes tight junction assembly in intestinal epithelial cells (IEC). Fructo-oligosaccharides (FOS), well-known prebiotics, have previously been shown to alleviate inflammation-associated intestinal epithelial disruption although the mechanisms were unclear. This study aimed to investigate any effect of FOS on AMPK activity and tight junction assembly under non-inflammatory and inflammatory conditions using T84 cells as an IEC model. As analyzed by western blot, FOS induced AMPK activation through a calcium sensing receptor (CaSR)-phospholipase C (PLC)- Ca2+/calmodulin-dependent protein kinase kinase-ß (CaMKKß) pathway. Calcium switch assays and immunofluorescence staining of zonula occludens-1 (ZO-1) revealed that FOS induced tight junction assembly via an CaMKKß-AMPK-dependent mechanism in IEC. Interestingly, FOS reversed the suppressive effect of lipopolysaccharide (LPS) on AMPK activity and tight junction assembly via a CaMKKß pathway. Taken together, these findings uncover a prebiotic-independent effect of FOS in promoting intestinal epithelial tight junction assembly through AMPK activation, which may have implications for the treatment of diseases whose pathogenesis involves impaired intestinal barrier function.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Oligossacarídeos/farmacologia , Prebióticos , Junções Íntimas/efeitos dos fármacos , Sinalização do Cálcio , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Linhagem Celular , Claudina-1/metabolismo , Humanos , Mucosa Intestinal/enzimologia , Lipopolissacarídeos/farmacologia , Ocludina/metabolismo , Fosforilação , Receptores de Detecção de Cálcio/metabolismo , Junções Íntimas/enzimologia , Fosfolipases Tipo C/metabolismo , Proteína da Zônula de Oclusão-1/metabolismo
7.
Biomedicines ; 7(4)2019 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-31614968

RESUMO

Mannan oligosaccharide (MOS) is well-known as an effective fed supplement for livestock to increase their nutrients absorption and health status. Pentasaccharide of mannan (MOS5) was reported as a molecule that possesses the ability to increase tight junction of epithelial tissue, but the structure and mechanism of action remains undetermined. In this study, the mechanism of action and structure of MOS5 were investigated. T84 cells were cultured and treated with MOS5 compared with vehicle and compound C, a 5'-adenosine monophosphate-activated protein kinase (AMPK) inhibitor. The results demonstrated that the ability of MOS5 to increase tight junction integration was inhibited in the presence of dorsomorphine (compound C). Phosphorylation level of AMPK was elevated in MOS5 treated group as determined by Western blot analysis. Determination of MOS5 structure was performed using enzymatic mapping together with 1H, 13C NMR, and 2D-NMR analysis. The results demonstrated that the structure of MOS5 is a ß-(1,4)-mannotetraose with α-(1,6)-galactose attached at the second mannose unit from non-reducing end.

8.
Eur J Pharmacol ; 798: 94-104, 2017 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-28119077

RESUMO

Nuclear factor kappa B (NF-κB)-mediated inflammatory responses play crucial roles in the pathogenesis of diarrhea caused by the Vibrio cholerae El Tor variant (EL), which is a major bacterial strain causing recent cholera outbreaks. Flufenamic acid (FFA) has previously been demonstrated to be a potent activator of AMP-activated protein kinase (AMPK), which is a negative regulator of NF-κB signaling. This study aimed to investigate the anti-diarrheal efficacy of FFA in a mouse model of EL infection and to investigate the mechanisms by which FFA activates AMPK in intestinal epithelial cells (IEC). In a mouse closed loop model of EL infection, FFA treatment (20mg/kg) significantly abrogated EL-induced intestinal fluid secretion and barrier disruption. In addition, FFA suppressed NF-κB nuclear translocation and expression of proinflammatory mediators and promoted AMPK phosphorylation in the EL-infected mouse intestine. In T84 cells, FFA induced AMPK activation. Furthermore, FFA promoted tight junction assembly and prevented interferon gamma (IFN-γ)-induced barrier disruption in an AMPK-dependent manner. Biochemical and molecular docking analyses indicated that FFA activates AMPK via a direct stimulation of calcium/calmodulin-dependent protein kinase kinase beta (CaMKKß) activity. Collectively, our data indicate that FFA represents a class of existing drugs that may be of potential utility in the treatment of cholera caused by EL infection via AMPK-mediated suppression of NF-κB signaling in IEC.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Líquidos Corporais/metabolismo , Cólera/tratamento farmacológico , Ácido Flufenâmico/farmacologia , Intestinos/efeitos dos fármacos , NF-kappa B/antagonistas & inibidores , Vibrio cholerae/fisiologia , Animais , Líquidos Corporais/efeitos dos fármacos , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/química , Quinase da Proteína Quinase Dependente de Cálcio-Calmodulina/metabolismo , Domínio Catalítico , Linhagem Celular , Cólera/enzimologia , Cólera/metabolismo , Diarreia/tratamento farmacológico , Diarreia/virologia , Modelos Animais de Doenças , Ácido Flufenâmico/metabolismo , Ácido Flufenâmico/uso terapêutico , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Camundongos , Simulação de Acoplamento Molecular
9.
Carbohydr Polym ; 145: 30-6, 2016 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-27106148

RESUMO

Novel, effective and safe agents are needed for the chemoprevention of colorectal cancer (CRC). This study investigated the effects of chitosan oligosaccharides (COS) on CRC progression and their underlying mechanisms and safety profiles in mice. Using a mouse model of colitis-associated CRC, we found that oral administration of COS (500mg/kg/day) resulted in a ∼60% reduction of tumor size and tumor numbers/sectioning. In addition, COS treatment increased AMPK activity, suppressed the NF-κB-mediated inflammatory response and reduced the expressions of cyclin D1, phosphorylated ribosomal protein S6, and MMP-9 in the colon tissues of these mice. Importantly, administration of COS (500mg/kg/day; 50 days) had no adverse effects on renal or liver functions. Our results indicate that COS suppressed CRC progression via AMPK activation and the suppression of NF-κB and mTOR signaling. COS may be of potential utility in the chemoprevention of CRC.


Assuntos
Anticarcinógenos/farmacologia , Anticarcinógenos/uso terapêutico , Quitosana/química , Neoplasias Colorretais/tratamento farmacológico , Oligossacarídeos/farmacologia , Oligossacarídeos/uso terapêutico , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Colite/complicações , Colite/metabolismo , Colite/patologia , Colo/efeitos dos fármacos , Colo/metabolismo , Colo/patologia , Neoplasias Colorretais/etiologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Ciclina D1/metabolismo , Modelos Animais de Doenças , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Carga Tumoral/efeitos dos fármacos
10.
Biochem Pharmacol ; 96(3): 225-36, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26047848

RESUMO

Chitosan oligosaccharide (COS), a biomaterial derived from chitin, is absorbed by intestinal epithelia without degradation and has diverse biological activities including intestinal epithelial function. However, the mode of action is still unclear. This study aimed to investigate the effect of COS on AMP-activated protein kinase (AMPK) in intestinal epithelial cells (IEC) and its potential applications in the intestinal diseases benefited from AMPK activation. COS with molecular weights (MW) from 5,000Da to 14,000Da induced AMPK activation in T84 cells. That with MW of 5,000-Da was the most potent polymer and was used in the subsequent experiments. COS also activated AMPK in other IEC including HT-29 and Caco-2 cells. Mechanism of COS-induced AMPK activation in T84 cells involves calcium-sensing receptor (CaSR)-phospholipase C (PLC)-IP3 receptor channel-mediated calcium release from endoplasmic reticulum (ER). In addition, COS promoted tight junction assembly in T84 cells in an AMPK-dependent manner. COS also inhibited NF-κB transcriptional activity and NF-κB-mediated inflammatory response and barrier disruption via AMPK-independent mechanisms. Interestingly, luminal exposure to COS suppressed cholera toxin-induced intestinal fluid secretion by ∼30% concurrent with AMPK activation in a mouse closed loop model. Importantly, oral administration of COS prevented the development of aberrant crypt foci in a mouse model of colitis-associated colorectal cancer (CRC) via a mechanism involving AMPK activation-induced ß-catenin suppression and caspase-3 activation. Collectively, this study reveals a novel action of COS in activating AMPK via CaSR-PLC-IP3 receptor channel-mediated calcium release from ER. COS may be beneficial in the treatment of secretory diarrheas and CRC chemoprevention.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Quitosana/farmacologia , Colite/tratamento farmacológico , Neoplasias Colorretais/tratamento farmacológico , Células Epiteliais/efeitos dos fármacos , Fármacos Gastrointestinais/farmacologia , Proteínas Quinases Ativadas por AMP/genética , Animais , Azoximetano , Cálcio/metabolismo , Caspase 3/genética , Caspase 3/metabolismo , Linhagem Celular Tumoral , Toxina da Cólera/toxicidade , Colite/induzido quimicamente , Colite/genética , Colite/patologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Sulfato de Dextrana , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Regulação da Expressão Gênica , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Intestinos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , NF-kappa B/metabolismo , Receptores de Detecção de Cálcio , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Fosfolipases Tipo C/genética , Fosfolipases Tipo C/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa