Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 298(5): 101847, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35314195

RESUMEN

Although capsaicin has been studied extensively as an activator of the transient receptor potential vanilloid cation channel subtype 1 (TRPV1) channels in sensory neurons, little is known about its TRPV1-independent actions in gastrointestinal health and disease. Here, we aimed to investigate the pharmacological actions of capsaicin as a food additive and medication on intestinal ion transporters in mouse models of ulcerative colitis (UC). The short-circuit current (Isc) of the intestine from WT, TRPV1-, and TRPV4-KO mice were measured in Ussing chambers, and Ca2+ imaging was performed on small intestinal epithelial cells. We also performed Western blots, immunohistochemistry, and immunofluorescence on intestinal epithelial cells and on intestinal tissues following UC induction with dextran sodium sulfate. We found that capsaicin did not affect basal intestinal Isc but significantly inhibited carbachol- and caffeine-induced intestinal Isc in WT mice. Capsaicin similarly inhibited the intestinal Isc in TRPV1 KO mice, but this inhibition was absent in TRPV4 KO mice. We also determined that Ca2+ influx via TRPV4 was required for cholinergic signaling-mediated intestinal anion secretion, which was inhibited by capsaicin. Moreover, the glucose-induced jejunal Iscvia Na+/glucose cotransporter was suppressed by TRPV4 activation, which could be relieved by capsaicin. Capsaicin also stimulated ouabain- and amiloride-sensitive colonic Isc. Finally, we found that dietary capsaicin ameliorated the UC phenotype, suppressed hyperaction of TRPV4 channels, and rescued the reduced ouabain- and amiloride-sensitive Isc. We therefore conclude that capsaicin inhibits intestinal Cl- secretion and promotes Na+ absorption predominantly by blocking TRPV4 channels to exert its beneficial anti-colitic action.


Asunto(s)
Capsaicina , Colitis , Canales Catiónicos TRPV , Amilorida , Animales , Capsaicina/farmacología , Cloruros/metabolismo , Colitis/tratamiento farmacológico , Colon/metabolismo , Glucosa , Ratones , Ratones Noqueados , Ouabaína , Sodio/metabolismo , Canales Catiónicos TRPV/antagonistas & inhibidores
2.
Front Pharmacol ; 13: 816133, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35281933

RESUMEN

Oral glutamine (Gln) has been widely used in gastrointestinal (GI) clinical practice, but it is unclear if Ca2+ regulates intestinal Gln transport, although both of them are essential nutrients for mammals. Chambers were used to determine Gln (25 mM)-induced I sc through Na+/Gln co-transporters in the small intestine in the absence or the presence of selective activators or blockers of ion channels and transporters. Luminal but not serosal application of Gln induced marked intestinal I sc , especially in the distal ileum. Lowering luminal Na+ almost abolished the Gln-induced ileal I sc , in which the calcium-sensitive receptor (CaSR) activation were not involved. Ca2+ removal from both luminal and serosal sides of the ileum significantly reduced Gln- I sc . Blocking either luminal Ca2+ entry via the voltage-gated calcium channels (VGCC) or endoplasmic reticulum (ER) release via inositol 1,4,5-triphosphate receptor (IP3R) and ryanodine receptor (RyR) attenuated the Gln-induced ileal I sc , Likewise, blocking serosal Ca2+ entry via the store-operated Ca2+ entry (SOCE), TRPV1/2 channels, and Na+/Ca2+ exchangers (NCX) attenuated the Gln-induced ileal I sc . In contrast, activating TRPV1/2 channels enhanced the Gln-induced ileal I sc . We concluded that Ca2+ signaling is critical for intestinal Gln transport, and multiple plasma membrane Ca2+-permeable channels and transporters play roles in this process. The Ca2+ regulation of ileal Na+/Gln transport expands our understanding of intestinal nutrient uptake and may be significant in GI health and disease.

3.
Front Pharmacol ; 12: 684538, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34335254

RESUMEN

As little is known about the role of calcium (Ca2+) signaling mediating the small intestinal epithelial anion secretion, we aimed to study its regulatory role in secretagogue-stimulated duodenal anion secretion and the underlying molecular mechanisms. Therefore, intestinal anion secretion from native mouse duodenal epithelia was examined with Ussing chambers to monitor PGE2-, 5-HT-, and CCh-induced short-circuit currents (I sc ). PGE2 (10 µM) and 5-HT (10 µM) induced mouse duodenal I sc , markedly attenuated by serosal Ca2+-free solution and selective blockers of store-operated Ca2+ channels on the serosal side of the duodenum. Furthermore, PGE2- and 5-HT-induced duodenal I sc was also inhibited by ER Ca2+ chelator TPEN. However, dantrolene, a selective blocker of ryanodine receptors, inhibited PGE2-induced duodenal I sc , while LiCl, an inhibitor of IP3 production, inhibited 5-HT-induced I sc . Moreover, duodenal I sc response to the serosal applications of both PGE2 and 5-HT was significantly attenuated in transient receptor potential vanilloid 4 (TRPV4) knockout mice. Finally, mucosal application of carbachol (100 µM) also induced duodenal I sc via selective activation of muscarinic receptors, which was significantly inhibited in serosal Ca2+-free solution but neither in mucosal Ca2+-free solution nor by nifedipine. Therefore, the serosal TRPV4-constituted SOCE mechanism is likely universal for the most common and important secretagogues-induced and Ca2+-dependent intestinal anion secretion. These findings will enhance our knowledge about gastrointestinal (G.I.) epithelial physiology and the associated G.I. diseases, such as diarrhea and constipation.

4.
Oncol Rep ; 45(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33760177

RESUMEN

Metabolism is defined as the biochemical processes that produce or consume energy in living organisms. Otto Warburg suggested that cancer is a metabolic disease, thus metabolic reprogramming is widely considered as an emerging hallmark of cancer cells. Long non­coding RNAs (lncRNAs), which are defined as transcripts >200 nucleotides with limited protein coding potential, are involved in cancer metabolism. lncRNAs can control pathophysiological processes of cancer by regulating gene expression at epigenetic, transcriptional and post­transcriptional levels. The process of tumorigenesis is usually accompanied by alterations in metabolic patterns, involving glycolysis, the tricarboxylic acid cycle, mitochondrial oxidative phosphorylation, the pentose phosphate signaling pathway, glutamine metabolism and lipid metabolism, which is also known as metabolic reprogramming. The present review summarized the functions of lncRNAs in cancer metabolism and discussed how the dysregulation of lncRNAs contributed to metabolic reprogramming and tumorigenesis, which may provide novel therapeutic targets for cancer.


Asunto(s)
Carcinogénesis/genética , Neoplasias/genética , ARN Largo no Codificante/metabolismo , Carcinogénesis/patología , Ciclo del Ácido Cítrico/genética , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Humanos , Metabolismo de los Lípidos/genética , Neoplasias/patología , Fosforilación Oxidativa , Vía de Pentosa Fosfato/genética , Efecto Warburg en Oncología
5.
Br J Pharmacol ; 178(2): 346-362, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33080043

RESUMEN

BACKGROUND AND PURPOSE: Luminal glucose enhances intestinal Ca2+ absorption through apical Cav 1.3 channels necessary for GLUT2-mediated glucose absorption. As these reciprocal mechanisms are not well understood, we investigated the regulatory mechanisms of intestinal [Ca2+ ]cyt and SGLT1-mediated Na+ -glucose co-transports. EXPERIMENTAL APPROACH: Glucose absorption and channel expression were examined in mouse upper jejunal epithelium using an Ussing chamber, immunocytochemistry and Ca2+ and Na+ imaging in single intestinal epithelial cells. KEY RESULTS: Glucose induced jejunal Isc via Na+ -glucose cotransporter 1 (SGLT1) operated more efficiently in the presence of extracellular Ca2+ . A crosstalk between luminal Ca2+ entry via plasma Cav 1.3 channels and the ER Ca2+ release through ryanodine receptor (RYR) activation in small intestinal epithelial cell (IEC) or Ca2+ -induced Ca2+ release (CICR) mechanism was involve in Ca2+ -mediated jejunal glucose absorption. The ER Ca2+ release through RyR triggered basolateral Ca2+ entry or store-operated Ca2+ entry (SOCE) mechanism and the subsequent Ca2+ entry via Na+ /Ca2+ exchanger 1 (NCX1) were found to be critical in Na+ -glucose cotransporter-mediated glucose absorption. Blocking RyR, SOCE and NCX1 inhibited glucose induced [Na+ ]cyt and [Ca2+ ]cyt in single IEC and protein expression and co-localization of STIM1/Orai1, RyR1 and NCX1 were detected in IEC and jejunal mucosa. CONCLUSION AND IMPLICATIONS: Luminal Ca2+ influx through Cav 1.3 triggers the CICR through RyR1 to deplete the ER Ca2+ , which induces the basolateral STIM1/Orai1-mediated SOCE mechanism and the subsequent Ca2+ entry via NCX1 to regulate intestinal glucose uptake via Ca2+ signalling. Targeting these mechanisms in IEC may help to modulate blood glucose and sodium in the metabolic disease.


Asunto(s)
Calcio , Sodio , Animales , Calcio/metabolismo , Señalización del Calcio , Glucosa , Ratones , Proteína ORAI1/metabolismo , Molécula de Interacción Estromal 1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...