Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34810264

RESUMEN

Calcium (Ca2+) homeostasis is maintained through coordination between intestinal absorption, renal reabsorption, and bone remodeling. Intestinal and renal (re)absorption occurs via transcellular and paracellular pathways. The latter contributes the bulk of (re)absorption under conditions of adequate intake. Epithelial paracellular permeability is conferred by tight-junction proteins called claudins. However, the molecular identity of the paracellular Ca2+ pore remains to be delineated. Claudins (Cldn)-2 and -12 confer Ca2+ permeability, but deletion of either claudin does not result in a negative Ca2+ balance or increased calciotropic hormone levels, suggesting the existence of additional transport pathways or parallel roles for the two claudins. To test this, we generated a Cldn2/12 double knockout mouse (DKO). These animals have reduced intestinal Ca2+ absorption. Colonic Ca2+ permeability is also reduced in DKO mice and significantly lower than single-null animals, while small intestine Ca2+ permeability is unaltered. The DKO mice display significantly greater urinary Ca2+ wasting than Cldn2 null animals. These perturbations lead to hypocalcemia and reduced bone mineral density, which was not observed in single-KO animals. Both claudins were localized to colonic epithelial crypts and renal proximal tubule cells, but they do not physically interact in vitro. Overexpression of either claudin increased Ca2+ permeability in cell models with endogenous expression of the other claudin. We find claudin-2 and claudin-12 form partially redundant, independent Ca2+ permeable pores in renal and colonic epithelia that enable paracellular Ca2+ (re)absorption in these segments, with either one sufficient to maintain Ca2+ balance.


Asunto(s)
Calcio/metabolismo , Claudinas/genética , Hipocalcemia/metabolismo , Animales , Calcificación Fisiológica , Cationes , Genotipo , Células HEK293 , Homeostasis , Humanos , Técnicas In Vitro , Ratones , Ratones Noqueados , Permeabilidad
2.
J Physiol ; 594(17): 4865-78, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27037495

RESUMEN

KEY POINTS: Arginine vasopressin (AVP) stimulates the release of enteroendocrine L-cell derived hormones glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) in vitro from mouse and human colons. This is mediated by the AVP receptor 1B, which is highly enriched in colonic L-cells and linked to the elevation of L-cell calcium and cAMP concentrations. By means of Ussing chambers, we show that AVP reduced colonic anion secretion, although this was blocked by a specific neuropeptide Y receptor Y1 receptor antagonist, suggesting that L-cell-released PYY acts locally on the epithelium to modulate fluid balance. In human serum samples, PYY concentrations were higher in samples with raised osmolality and copeptin (surrogate marker for AVP). These findings describe, for the first time, the role of L-cells in AVP regulated intestinal fluid secretion, potentially linking together hormonal control of blood volume and blood glucose levels, and thus adding to our understanding of the complex pathways involved in the gut hormonal response to different stimuli. ABSTRACT: Arginine vasopressin (AVP) regulates fluid balance and blood pressure via AVP receptor (AVPR)2 in the kidney and AVP receptor 1A in vascular smooth muscle. Its role in intestinal function has received less attention. We hypothesized that enteroendocrine L-cells producing glucagon-like peptide 1 (GLP-1) and peptide YY (PYY) may be a target of AVP and contribute to the control of fluid balance. Avpr1b expression was assessed by quantitative RT-PCR on flourescence-activated cell sorting-isolated L- and control cells and was enriched in colonic L-cells. AVP stimulated GLP-1 and PYY release from primary cultured murine and human colonic cells and was associated with elevated calcium and cAMP concentrations in L-cells as measured in cultures from GLU-Cre/ROSA26-GCaMP3 and GLU-Epac2camps mice. An antagonist of AVPR1B reduced AVP-triggered hormone secretion from murine and human cells. In Ussing chambers, basolaterally applied AVP reduced colonic anion secretion and this effect was blocked by a specific neuropeptide Y receptor Y1 (NPY1R) antagonist. In human serum, PYY concentrations were higher in samples with raised osmolality or copeptin (a surrogate marker for AVP). In conclusion, we propose that AVP activates L-cell AVPR1B, causing GLP-1 and PYY secretion. PYY in turn reduces colonic anion secretion via epithelial NPY1R. Our data suggest L-cells are active players in the hypothalamic control of intestinal fluid homeostasis, providing a potential link between the regulation of blood volume/pressure/osmolality and blood glucose.


Asunto(s)
Arginina Vasopresina/farmacología , Colon/metabolismo , Células Enteroendocrinas/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Péptido YY/metabolismo , Anciano , Animales , Calcio/metabolismo , Células Cultivadas , Colon/citología , AMP Cíclico/metabolismo , Femenino , Expresión Génica , Humanos , Intestino Delgado/citología , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Péptido YY/sangre , Receptores de Vasopresinas/genética
3.
Biochem Cell Biol ; 92(6): 467-80, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25386841

RESUMEN

Calcium (Ca(2+)) is a key constituent in a myriad of physiological processes from intracellular signalling to the mineralization of bone. As a consequence, Ca(2+) is maintained within narrow limits when circulating in plasma. This is accomplished via regulated interplay between intestinal absorption, renal tubular reabsorption, and exchange with bone. Many studies have focused on the highly regulated active transcellular transport pathways for Ca(2+) from the duodenum of the intestine and the distal nephron of the kidney. However, comparatively little work has examined the molecular constituents creating the paracellular shunt across intestinal and renal epithelium, the transport pathway responsible for the majority of transepithelial Ca(2+) flux. More specifically, passive paracellular Ca(2+) absorption occurs across the majority of the intestine in addition to the renal proximal tubule and thick ascending limb of Henle's loop. Importantly, recent studies demonstrated that Ca(2+) transport through the paracellular shunt is significantly regulated. Therefore, we have summarized the evidence for different modes of paracellular Ca(2+) flux across renal and intestinal epithelia and highlighted recent molecular insights into both the mechanism of secondarily active paracellular Ca(2+) movement and the identity of claudins that permit the passage of Ca(2+) through the tight junction of these epithelia.


Asunto(s)
Calcio/metabolismo , Duodeno/metabolismo , Mucosa Intestinal/metabolismo , Nefronas/metabolismo , Animales , Humanos , Transporte Iónico/fisiología
4.
Am J Physiol Gastrointest Liver Physiol ; 305(4): G303-13, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23764894

RESUMEN

Intestinal calcium (Ca²âº) absorption occurs via paracellular and transcellular pathways. Although the transcellular route has been extensively studied, mechanisms mediating paracellular absorption are largely unexplored. Unlike passive diffusion, secondarily active paracellular Ca²âº uptake occurs against an electrochemical gradient with water flux providing the driving force. Water movement is dictated by concentration differences that are largely determined by Na⁺ fluxes. Consequently, we hypothesized that Na⁺ absorption mediates Ca²âº flux. NHE3 is central to intestinal Na⁺ absorption. NHE3 knockout mice (NHE3-/-) display impaired intestinal Na⁺, water, and Ca²âº absorption. However, the mechanism mediating this latter abnormality is not clear. To investigate this, we used Ussing chambers to measure net Ca²âº absorption across different segments of wild-type mouse intestine. The cecum was the only segment with net Ca²âº absorption. Quantitative RT-PCR measurements revealed cecal expression of all genes implicated in intestinal Ca²âº absorption, including NHE3. We therefore employed this segment for further studies. Inhibition of NHE3 with 100 µM 5-(N-ethyl-N-isopropyl) amiloride decreased luminal-to-serosal and increased serosal-to-luminal Ca²âº flux. NHE3-/- mice had a >60% decrease in luminal-to-serosal Ca²âº flux. Ussing chambers experiments under altered voltage clamps (-25, 0, +25 mV) showed decreased transcellular and secondarily active paracellular Ca²âº absorption in NHE3-/- mice relative to wild-type animals. Consistent with this, cecal Trpv6 expression was diminished in NHE3-/- mice. Together these results implicate NHE3 in intestinal Ca(2+) absorption and support the theory that this is, at least partially, due to the role of NHE3 in Na⁺ and water absorption.


Asunto(s)
Calcio/metabolismo , Ciego/metabolismo , Absorción Intestinal , Mucosa Intestinal/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Sodio/metabolismo , Amilorida/análogos & derivados , Amilorida/farmacología , Animales , Canales de Calcio/metabolismo , Ciego/efectos de los fármacos , Femenino , Regulación de la Expresión Génica , Absorción Intestinal/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Transporte Iónico , Masculino , Potenciales de la Membrana , Moduladores del Transporte de Membrana/farmacología , Ratones , Ratones Noqueados , Técnicas de Placa-Clamp , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rojo de Rutenio/farmacología , Intercambiador 3 de Sodio-Hidrógeno , Intercambiadores de Sodio-Hidrógeno/antagonistas & inhibidores , Intercambiadores de Sodio-Hidrógeno/genética , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/metabolismo , Agua/metabolismo
5.
Am J Physiol Cell Physiol ; 303(12): C1278-91, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23076790

RESUMEN

The effect of claudins on paracellular fluxes has been predominantly studied in either Madin-Darby canine kidney (MDCK) or LLCPK cells. Neither model system has a very low transepithelial resistance (TER) as observed in leaky epithelia. Moreover, results from one model system are not always consistent with another. Opossum kidney (OK) cells form tight junctions yet have a very low TER. We therefore set out to characterize the paracellular transport properties of this cell culture model. Ussing chamber dilution potential measurements revealed that OK cells exhibit a very low TER (11.7 ± 1.4 Ω·cm(2)), slight cation selectivity (P(Na)/P(Cl) = 1.10 ± 0.01), and the Eisenman permeability sequence IV; the permeability of monovalent cations ranking K(+) > Cs(+) > Rb(+) > Na(+) > Li(+). Quantitative real-time PCR studies found that OK cells endogenously express claudin-4 > -1 > -6 > -20 > -9 > -12 > -11 > -15. Overexpression of claudin-4 significantly increased TER, decreased Na(+) and Cl(-) permeability, and increased levels of claudin-1, -6, and -9 mRNA. Knockdown of claudin-4 in the overexpressing cells significantly decreased TER without altering claudin expression; thus claudin-4 forms a barrier in OK cells. Knockdown of endogenous claudin-4 decreased claudin-1, -9, and -12 expression without altering TER. Claudin-2 overexpression decreased TER, significantly increased Na(+) and Cl(-) permeability, and decreased claudin-12 and -6 expression. Together these results demonstrate that claudin expression is tightly coupled in OK cells.


Asunto(s)
Claudina-4/biosíntesis , Células Epiteliales/metabolismo , Riñón/metabolismo , Animales , Cationes Monovalentes/metabolismo , Células Cultivadas , Claudina-4/genética , Claudinas/biosíntesis , Perros , Silenciador del Gen , Zarigüeyas , Permeabilidad , Uniones Estrechas/metabolismo
6.
Pflugers Arch ; 463(3): 497-509, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22170096

RESUMEN

Protease-activated receptor 2 (PAR-2) is a G protein-coupled receptor possibly involved in the pathogenesis of asthma. PAR-2 also modulates ion transport in cultured epithelial cells, but these effects in native airways are controversial. The influence of allergic inflammation on PAR-2-induced changes in ion transport has received little attention. Here, we studied immediate changes in transepithelial short circuit current (I (sc)) induced by PAR-2 activation in the tracheas of naive and allergic mice. Activation of PAR-2 with an apically added activation peptide (AP) induced a small increase in I (sc), while a much larger increase was observed following basolateral AP addition. In ovalbumin-sensitized and -challenged animals used as a model of allergic airway inflammation, the effect of basolateral AP addition was enhanced. Responses to basolateral AP in both naive and allergic mice were not decreased by blocking sodium absorption with amiloride or CFTR function with CFTR(inh)172 but were reduced by the cyclooxygenase inhibitor indomethacin and largely blocked (>80%) by niflumic acid, a calcium-activated chloride channels' (CaCC) blocker. Allergic mice also showed an enhanced response to ATP and thapsigargin. There was no change in mRNA expression of Par-2 or of the chloride channels Ano1 (Tmem16a) and Bestrophin 2 in tracheas from allergic mice, while mRNA levels of Bestrophin 1 were increased. In conclusion, basolateral PAR-2 activation in the mouse airways led to increased anion secretion through apical CaCC, which was more pronounced in allergic animals. This could be a protective mechanism aimed at clearing allergens and defending against mucus plugging.


Asunto(s)
Canales de Cloruro/fisiología , Hipersensibilidad/fisiopatología , Receptor PAR-2/fisiología , Traqueítis/fisiopatología , Amilorida/farmacología , Animales , Asma/fisiopatología , Benzoatos/farmacología , Bestrofinas , Canales de Cloruro/efectos de los fármacos , Proteínas del Ojo/biosíntesis , Indometacina/farmacología , Canales Iónicos/biosíntesis , Masculino , Ratones , Ratones Endogámicos BALB C , Ácido Niflúmico/farmacología , Oligopéptidos/farmacología , Ovalbúmina , Receptor PAR-2/efectos de los fármacos , Tiazolidinas/farmacología
7.
EBioMedicine ; 55: 102759, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32344198

RESUMEN

BACKGROUND: The bile acid (BA) pathway plays a role in regulation of food intake and glucose metabolism, based mainly on findings in animal models. Our aim was to determine whether the BA pathway is altered and correctable in human obesity and diabetes. METHODS: We conducted 3 investigations: 1) BA receptor pathways were studied in NCI-H716 enteroendocrine cell (EEC) line, whole human colonic mucosal tissue and in human colonic EEC isolated by Fluorescence-activated Cell Sorting (ex vivo) from endoscopically-obtained biopsies colon mucosa; 2) We characterized the BA pathway in 307 participants by measuring during fasting and postprandial levels of FGF19, 7αC4 and serum BA; 3) In a placebo-controlled, double-blind, randomised, 28-day trial, we studied the effect of ileo-colonic delivery of conjugated BAs (IC-CBAS) on glucose metabolism, incretins, and lipids, in participants with obesity and diabetes. FINDINGS: Human colonic GLP-1-producing EECs express TGR5, and upon treatment with bile acids in vitro, human EEC differentially expressed GLP-1 at the protein and mRNA level. In Ussing Chamber, GLP-1 release was stimulated by Taurocholic acid in either the apical or basolateral compartment. FGF19 was decreased in obesity and diabetes compared to controls. When compared to placebo, IC-CBAS significantly decreased postprandial glucose, fructosamine, fasting insulin, fasting LDL, and postprandial FGF19 and increased postprandial GLP-1 and C-peptide. Increase in faecal BA was associated with weight loss and with decreased fructosamine. INTERPRETATIONS: In humans, BA signalling machinery is expressed in colonic EECs, deficient in obesity and diabetes, and when stimulated with IC-CBAS, improved glucose homeostasis. ClinicalTrials.gov number, NCT02871882, NCT02033876. FUNDING: Research support and drug was provided by Satiogen Pharmaceuticals (San Diego, CA). AA, MC, and NFL report grants (AA- C-Sig P30DK84567, K23 DK114460; MC- NIH R01 DK67071; NFL- R01 DK057993) from the NIH. JR was supported by an Early Career Grant from Society for Endocrinology.


Asunto(s)
Ácidos y Sales Biliares/administración & dosificación , Glucemia/metabolismo , Colon/efectos de los fármacos , Diabetes Mellitus Tipo 2/terapia , Íleon/efectos de los fármacos , Obesidad/terapia , Administración Oral , Ácidos y Sales Biliares/química , Ácidos y Sales Biliares/metabolismo , Transporte Biológico , Cápsulas , Línea Celular , Colestenonas/sangre , Colon/metabolismo , Colon/patología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Cámaras de Difusión de Cultivos , Células Enteroendocrinas/citología , Células Enteroendocrinas/efectos de los fármacos , Células Enteroendocrinas/metabolismo , Ayuno/fisiología , Factores de Crecimiento de Fibroblastos/sangre , Fructosamina/sangre , Expresión Génica , Péptido 1 Similar al Glucagón/sangre , Péptido 1 Similar al Glucagón/genética , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Humanos , Íleon/metabolismo , Íleon/patología , Insulina/sangre , Obesidad/genética , Obesidad/metabolismo , Obesidad/patología , Periodo Posprandial , Cultivo Primario de Células , Receptores Acoplados a Proteínas G/sangre , Receptores Acoplados a Proteínas G/genética
8.
Nat Commun ; 10(1): 1029, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30833673

RESUMEN

Enteroendocrine cells are specialised sensory cells located in the intestinal epithelium and generate signals in response to food ingestion. Whilst traditionally considered hormone-producing cells, there is evidence that they also initiate activity in the afferent vagus nerve and thereby signal directly to the brainstem. We investigate whether enteroendocrine L-cells, well known for their production of the incretin hormone glucagon-like peptide-1 (GLP-1), also release other neuro-transmitters/modulators. We demonstrate regulated ATP release by ATP measurements in cell supernatants and by using sniffer patches that generate electrical currents upon ATP exposure. Employing purinergic receptor antagonists, we demonstrate that evoked ATP release from L-cells triggers electrical responses in neighbouring enterocytes through P2Y2 and nodose ganglion neurones in co-cultures through P2X2/3-receptors. We conclude that L-cells co-secrete ATP together with GLP-1 and PYY, and that ATP acts as an additional signal triggering vagal activation and potentially synergising with the actions of locally elevated peptide hormone concentrations.


Asunto(s)
Adenosina Trifosfato/metabolismo , Enterocitos/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Intestinos , Neuronas Aferentes/metabolismo , Vías Aferentes , Animales , Línea Celular , Ingestión de Alimentos , Células Enteroendocrinas/metabolismo , Femenino , Ganglión/metabolismo , Ganglión/patología , Incretinas/metabolismo , Mucosa Intestinal/inervación , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/patología , Ganglio Nudoso/metabolismo , Ganglio Nudoso/patología , Péptido YY/metabolismo , Receptores Purinérgicos P2X2/metabolismo , Receptores Purinérgicos P2X3/metabolismo , Nervio Vago/metabolismo
9.
Sci Rep ; 9(1): 15574, 2019 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-31666564

RESUMEN

Guanylin, a peptide implicated in regulation of intestinal fluid secretion, is expressed in the mucosa, but the exact cellular origin remains controversial. In a new transgenic mouse model fluorescent reporter protein expression driven by the proguanylin promoter was observed throughout the small intestine and colon in goblet and Paneth(-like) cells and, except in duodenum, in mature enterocytes. In Ussing chamber experiments employing both human and mouse intestinal tissue, proguanylin was released predominantly in the luminal direction. Measurements of proguanylin expression and secretion in cell lines and organoids indicated that secretion is largely constitutive and requires ER to Golgi transport but was not acutely regulated by salt or other stimuli. Using a newly-developed proguanylin assay, we found plasma levels to be raised in humans after total gastrectomy or intestinal transplantation, but largely unresponsive to nutrient ingestion. By LC-MS/MS we identified processed forms in tissue and luminal extracts, but in plasma we only detected full-length proguanylin. Our transgenic approach provides information about the cellular origins of proguanylin, complementing previous immunohistochemical and in-situ hybridisation results. The identification of processed forms of proguanylin in the intestinal lumen but not in plasma supports the notion that the primary site of action is the gut itself.


Asunto(s)
Hormonas Gastrointestinales/metabolismo , Regulación de la Expresión Génica , Mucosa Intestinal/metabolismo , Precursores de Proteínas/metabolismo , Hormonas Gastrointestinales/sangre , Humanos , Péptidos Natriuréticos/metabolismo , Precursores de Proteínas/sangre
10.
Chem Senses ; 33(4): 397-404, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18303030

RESUMEN

Organotypic cultures of the mouse olfactory epithelium connected to the olfactory bulb were obtained with the roller tube technique from postnatal mice aged between 13 and 66 days. To test the functionality of the cultures, we measured electroolfactograms (EOGs) at different days in vitro (DIV), up to 7 DIV, and we compared them with EOGs from identical acute preparations (0 DIV). Average amplitudes of EOG responses to 2 mixtures of various odorants at concentrations of 1 mM or 100 microM decreased in cultures between 2 and 5 DIV compared with 0 DIV. The percentage of responsive cultures was 57%. We also used the phosphodiesterase inhibitor 3-isobutyl-1-methylxanthine (IBMX) to trigger the olfactory transduction cascade bypassing odorant receptor activation. Average amplitudes of EOG responses to 500 microM IBMX were not significantly different in cultures up to 6 DIV or 0 DIV, and the average percentage of responsive cultures between 2 and 5 DIV was 72%. The dose-response curve to IBMX measured in cultures up to 7 DIV was similar to that at 0 DIV. Moreover, the percentage of EOG response to IBMX blocked by niflumic acid, a blocker of Ca-activated Cl channels, was not significantly different in cultured or acute preparations.


Asunto(s)
Odorantes , Mucosa Olfatoria/fisiología , Olfato/fisiología , Animales , Animales Recién Nacidos , Canales de Cloruro/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Electrofisiología , Ratones , Ratones Endogámicos BALB C , Odorantes/análisis , Mucosa Olfatoria/enzimología , Mucosa Olfatoria/metabolismo , Técnicas de Cultivo de Órganos , Inhibidores de Fosfodiesterasa/farmacología , Hidrolasas Diéster Fosfóricas/metabolismo , Factores de Tiempo
11.
Endocrinology ; 157(10): 3821-3831, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27447725

RESUMEN

Angiotensin II (Ang II) is the key hormone mediator of the renin angiotensin system, which regulates blood pressure and fluid and electrolyte balance in the body. Here we report that in the colonic epithelium, the Ang II type 1 receptor is highly and exclusively expressed in enteroendocrine L cells, which produce the gut hormones glucagon-like peptide-1 and peptide YY (PYY). Ang II stimulated glucagon-like peptide-1 and PYY release from primary cultures of mouse and human colon, which was antagonized by the specific Ang II type 1 receptor blocker candesartan. Ang II raised intracellular calcium levels in L cells in primary cultures, recorded by live-cell imaging of L cells specifically expressing the fluorescent calcium sensor GCaMP3. In Ussing chamber recordings, Ang II reduced short circuit currents in mouse distal colon preparations, which was antagonized by candesartan or a specific neuropeptide Y1 receptor inhibitor but insensitive to amiloride. We conclude that Ang II stimulates PYY secretion, in turn inhibiting epithelial anion fluxes, thereby reducing net fluid secretion into the colonic lumen. Our findings highlight an important role of colonic L cells in whole-body fluid homeostasis by controlling water loss through the intestine.


Asunto(s)
Angiotensina II/fisiología , Colon/fisiología , Células Enteroendocrinas/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Péptido YY/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Animales , Señalización del Calcio , Femenino , Humanos , Mucosa Intestinal/fisiología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Sistema Renina-Angiotensina , Equilibrio Hidroelectrolítico
12.
Biochim Biophys Acta ; 1567(1-2): 176-82, 2002 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-12488051

RESUMEN

Several factors, including the exposure of the negatively charged PL and transmembrane potential (TMP), may affect the binding of merocyanine 540 dye (MC540) to membrane lipids. Our aim was to quantify the significance of each of these two determinants in MC540 interactions with phosphatidylserine:phosphatidylcholine (PS/PC) vesicles. The effects of the altered PS content (PS/PC molar ratio: 5:95, 10:90 and 20:80) and TMP on MC540 binding were monitored using flow cytometry. Rapid [K(+)] flux across the vesicle membrane lipid bilayer was generated using valinomycin. We showed that the increased PS content leads to attenuated MC540 binding, while having no influence on the dynamic parameters of PS/PC vesicle membranes (electron spin resonance (ESR) spectrometry). Higher [K(+)](out) makes PS/PC liposomes bind more MC540, which implies that TMP-which becomes more positive inside the vesicles-favours the interactions of MC540 with the PL bilayer. Overall, the variability attributed to MC540-PL interactions is explained only to a minor extent by the generated TMP (7%) and largely by the variations in PS content (by up to 60%). In conclusion, the content of negatively charged PL is more important than TMP in determining the interactions of MC540 with PS/PC membranes.


Asunto(s)
Membrana Dobles de Lípidos/metabolismo , Potenciales de la Membrana , Fosfatidilserinas/metabolismo , Pirimidinonas/metabolismo , Espectroscopía de Resonancia por Spin del Electrón , Citometría de Flujo , Liposomas , Potasio/metabolismo
13.
Endocrinology ; 156(11): 3961-70, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26280129

RESUMEN

Bile acids are well-recognized stimuli of glucagon-like peptide-1 (GLP-1) secretion. This action has been attributed to activation of the G protein-coupled bile acid receptor GPBAR1 (TGR5), although other potential bile acid sensors include the nuclear farnesoid receptor and the apical sodium-coupled bile acid transporter ASBT. The aim of this study was to identify pathways important for GLP-1 release and to determine whether bile acids target their receptors on GLP-1-secreting L-cells from the apical or basolateral compartment. Using transgenic mice expressing fluorescent sensors specifically in L-cells, we observed that taurodeoxycholate (TDCA) and taurolithocholate (TLCA) increased intracellular cAMP and Ca(2+). In primary intestinal cultures, TDCA was a more potent GLP-1 secretagogue than taurocholate (TCA) and TLCA, correlating with a stronger Ca(2+) response to TDCA. Using small-volume Ussing chambers optimized for measuring GLP-1 secretion, we found that both a GPBAR1 agonist and TDCA stimulated GLP-1 release better when applied from the basolateral than from the luminal direction and that luminal TDCA was ineffective when intestinal tissue was pretreated with an ASBT inhibitor. ASBT inhibition had no significant effect in nonpolarized primary cultures. Studies in the perfused rat gut confirmed that vascularly administered TDCA was more effective than luminal TDCA. Intestinal primary cultures and Ussing chamber-mounted tissues from GPBAR1-knockout mice did not secrete GLP-1 in response to either TLCA or TDCA. We conclude that the action of bile acids on GLP-1 secretion is predominantly mediated by GPBAR1 located on the basolateral L-cell membrane, suggesting that stimulation of gut hormone secretion may include postabsorptive mechanisms.


Asunto(s)
Ácidos y Sales Biliares/farmacología , Células Enteroendocrinas/efectos de los fármacos , Péptido 1 Similar al Glucagón/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , AMP Cíclico/metabolismo , Células Enteroendocrinas/metabolismo , Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Masculino , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente , Ratas Wistar , Receptores Acoplados a Proteínas G/genética , Ácido Taurodesoxicólico/farmacología , Ácido Taurolitocólico/farmacología , Técnicas de Cultivo de Tejidos
14.
PLoS One ; 4(3): e4672, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19262692

RESUMEN

Bestrophins are a recently discovered family of Cl(-) channels, for which no structural information is available. Some family members are activated by increased intracellular Ca2+ concentration. Bestrophins feature a well conserved Asp-rich tract in their COOH terminus (Asp-rich domain), which is homologous to Ca2+-binding motifs in human thrombospondins and in human big-conductance Ca2+- and voltage-gated K+ channels (BK(Ca)). Consequently, the Asp-rich domain is also a candidate for Ca2+ binding in bestrophins. Based on these considerations, we constructed homology models of human bestrophin-1 (Best1) Asp-rich domain using human thrombospondin-1 X-ray structure as a template. Molecular dynamics simulations were used to identify Asp and Glu residues binding Ca2+ and to predict the effects of their mutations to alanine. We then proceeded to test selected mutations in the Asp-rich domain of the highly homologous mouse bestrophin-2. The mutants expressed in HEK-293 cells were investigated by electrophysiological experiments using the whole-cell voltage-clamp technique. Based on our molecular modeling results, we predicted that Asp-rich domain has two defined binding sites and that D301A and D304A mutations may impact the binding of the metal ions. The experiments confirmed that these mutations do actually affect the function of the protein causing a large decrease in the Ca2+-activated Cl(-) current, fully consistent with our predictions. In addition, other studied mutations (E306A, D312A) did not decrease Ca2+-activated Cl(-) current in agreement with modeling results.


Asunto(s)
Calcio/metabolismo , Canales de Cloruro/fisiología , Simulación por Computador , Fenómenos Electrofisiológicos , Proteínas del Ojo/fisiología , Animales , Bestrofinas , Sitios de Unión , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Humanos , Ratones , Modelos Moleculares , Mutación , Técnicas de Placa-Clamp , Homología de Secuencia de Aminoácido , Trombospondina 1/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA