Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 293(18): 6893-6904, 2018 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-29549124

RESUMEN

The voltage-dependent K+ (Kv) channel Kv2.1 is a major delayed rectifier in many secretory cells, including pancreatic ß cells. In addition, Kv2.1 has a direct role in exocytosis at an undefined step, involving SNARE proteins, that is independent of its ion-conducting pore function. Here, we elucidated the precise step in exocytosis. We previously reported that syntaxin-3 (Syn-3) is the key syntaxin that mediates exocytosis of newcomer secretory granules that spend minimal residence time on the plasma membrane before fusion. Using high-resolution total internal reflection fluorescence microscopy, we now show that Kv2.1 forms reservoir clusters on the ß-cell plasma membrane and binds Syn-3 via its C-terminal C1b domain, which recruits newcomer insulin secretory granules into this large reservoir. Upon glucose stimulation, secretory granules were released from this reservoir to replenish the pool of newcomer secretory granules for subsequent fusion, occurring just adjacent to the plasma membrane Kv2.1 clusters. C1b deletion blocked the aforementioned Kv2.1-Syn-3-mediated events and reduced fusion of newcomer secretory granules. These insights have therapeutic implications, as Kv2.1 overexpression in type-2 diabetes rat islets restored biphasic insulin secretion.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas Qa-SNARE/metabolismo , Vesículas Secretoras/metabolismo , Canales de Potasio Shab/metabolismo , Animales , Membrana Celular/metabolismo , Exocitosis/fisiología , Glucosa/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Masculino , Ratones , Microscopía Fluorescente , Unión Proteica , Dominios Proteicos , Proteínas Qa-SNARE/química , Ratas , Ratas Wistar , Proteínas SNARE/metabolismo , Canales de Potasio Shab/fisiología
2.
Gastroenterology ; 154(6): 1805-1821.e5, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29360461

RESUMEN

BACKGROUND & AIMS: Pancreatic acinar cells are polarized epithelial cells that store enzymes required for digestion as inactive zymogens, tightly packed at the cell apex. Stimulation of acinar cells causes the zymogen granules to fuse with the apical membrane, and the cells undergo exocytosis to release proteases into the intestinal lumen. Autophagy maintains homeostasis of pancreatic acini. Syntaxin 2 (STX2), an abundant soluble N-ethyl maleimide sensitive factor attachment protein receptor in pancreatic acini, has been reported to mediate apical exocytosis. Using human pancreatic tissues and STX2-knockout (KO) mice, we investigated the functions of STX2 in zymogen granule-mediated exocytosis and autophagy. METHODS: We obtained pancreatic tissues from 5 patients undergoing surgery for pancreatic cancer and prepared 80-µm slices; tissues were exposed to supramaximal cholecystokinin octapeptide (CCK-8) or ethanol and a low concentration of CCK-8 and analyzed by immunoblot and immunofluorescence analyses. STX2-KO mice and syntaxin 2+/+ C57BL6 mice (controls) were given intraperitoneal injections of supramaximal caerulein (a CCK-8 analogue) or fed ethanol and then given a low dose of caerulein to induce acute pancreatitis, or saline (controls); pancreata were isolated and analyzed by histology and immunohistochemistry. Acini were isolated from mice, incubated with CCK-8, and analyzed by immunofluorescence microscopy or used in immunoprecipitation experiments. Exocytosis was quantified using live-cell exocytosis and Ca2+ imaging analyses and based on formation of exocytotic soluble N-ethyl maleimide sensitive factor attachment protein receptor complexes. Dysregulations in autophagy were identified using markers, electron and immunofluorescence microscopy, and protease activation assays. RESULTS: Human pancreatic tissues and dispersed pancreatic acini from control mice exposed to CCK-8 or ethanol plus CCK-8 were depleted of STX2. STX2-KO developed more severe pancreatitis after administration of supramaximal caerulein or a 6-week ethanol diet compared with control. Acini from STX2-KO mice had increased apical exocytosis after exposure to CCK-8, as well as increased basolateral exocytosis, which led to ectopic release of proteases. These increases in apical and basolateral exocytosis required increased formation of fusogenic soluble N-ethyl maleimide sensitive factor attachment protein receptor complexes, mediated by STX3 and STX4. STX2 bound ATG16L1 and prevented it from binding clathrin. Deletion of STX2 from acini increased binding of AT16L1 to clathrin, increasing formation of pre-autophagosomes and inducing autophagy. Induction of autophagy promoted the CCK-8-induced increase in autolysosome formation and the activation of trypsinogen. CONCLUSIONS: In studies of human pancreatic tissues and pancreata from STX2-KO and control mice, we found STX2 to block STX3- and STX4-mediated fusion of zymogen granules with the plasma membrane and exocytosis and prevent binding of ATG16L1 to clathrin, which contributes to induction of autophagy. Exposure of pancreatic tissues to CCK-8 or ethanol depletes acinar cells of STX2, increasing basolateral exocytosis and promoting autophagy induction, leading to activation of trypsinogen.


Asunto(s)
Autofagia/genética , Exocitosis/genética , Páncreas/citología , Pancreatitis/genética , Sintaxina 1/metabolismo , Células Acinares/metabolismo , Animales , Membrana Celular/metabolismo , Ceruletida , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Páncreas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/cirugía , Pancreatitis/inducido químicamente , Vesículas Secretoras/fisiología , Tripsinógeno/metabolismo
3.
J Biol Chem ; 292(6): 2203-2216, 2017 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-28031464

RESUMEN

In type-2 diabetes (T2D), severely reduced islet syntaxin-1A (Syn-1A) levels contribute to insulin secretory deficiency. We generated ß-cell-specific Syn-1A-KO (Syn-1A-ßKO) mice to mimic ß-cell Syn-1A deficiency in T2D. Glucose tolerance tests showed that Syn-1A-ßKO mice exhibited blood glucose elevation corresponding to reduced blood insulin levels. Perifusion of Syn-1A-ßKO islets showed impaired first- and second-phase glucose-stimulated insulin secretion (GSIS) resulting from reduction in readily releasable pool and granule pool refilling. To unequivocally determine the ß-cell exocytotic defects caused by Syn-1A deletion, EM and total internal reflection fluorescence microscopy showed that Syn-1A-KO ß-cells had a severe reduction in the number of secretory granules (SGs) docked onto the plasma membrane (PM) at rest and reduced SG recruitment to the PM after glucose stimulation, the latter indicating defects in replenishment of releasable pools required to sustain second-phase GSIS. Whereas reduced predocked SG fusion accounted for reduced first-phase GSIS, selective reduction of exocytosis of short-dock (but not no-dock) newcomer SGs accounted for the reduced second-phase GSIS. These Syn-1A actions on newcomer SGs were partly mediated by Syn-1A interactions with newcomer SG VAMP8.


Asunto(s)
Exocitosis , Insulina/metabolismo , Vesículas Secretoras/metabolismo , Sintaxina 1/fisiología , Animales , Glucosa/metabolismo , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Sintaxina 1/genética
4.
Traffic ; 14(4): 428-39, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23346930

RESUMEN

RalA GTPase has been implicated in the regulated delivery of exocytotic vesicles to the plasma membrane (PM) in mammalian cells. We had reported that RalA regulates biphasic insulin secretion, which we have now determined to be contributed by RalA direct interaction with voltage-gated calcium (Cav ) channels. RalA knockdown (KD) in INS-1 cells and primary rat ß-cells resulted in a reduction in Ca(2+) currents arising specifically from L-(Cav 1.2 and Cav 1.3) and R-type (Cav 2.3) Ca(2+) channels. Restoration of RalA expression in RalA KD cells rescued these defects in Ca(2+) currents. RalA co-immunoprecipitated with the Cav α2 δ-1 auxiliary subunit known to bind the three Cav s. Moreover, the functional molecular interactions between Cav α2 δ-1 and RalA on the PM shown by total internal reflection fluorescent microscopy/FRET analysis could be induced by glucose stimulation. KD of RalA inhibited trafficking of α2 δ-1 to insulin granules without affecting the localization of the other Cav subunits. Furthermore, we confirmed that RalA and α2 δ-1 functionally interact since RalA KD-induced inhibition of Cav currents could not be recovered by RalA when α2 δ-1 was simultaneously knocked down. These data provide a mechanism for RalA function in insulin secretion, whereby RalA binds α2 δ-1 on insulin granules to tether these granules to PM Ca(2+) channels. This acts as a chaperoning step prior to and in preparation for sequential assembly of exocyst and excitosome complexes that mediate biphasic insulin secretion.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo R/metabolismo , Insulina/metabolismo , Subunidades de Proteína/metabolismo , Vesículas Secretoras/metabolismo , Proteínas de Unión al GTP ral/metabolismo , Animales , Calcio/metabolismo , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo R/genética , Membrana Celular/metabolismo , Exocitosis , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Humanos , Células Secretoras de Insulina/metabolismo , Unión Proteica , Subunidades de Proteína/genética , Transporte de Proteínas , ARN Interferente Pequeño , Ratas , Ratas Sprague-Dawley
5.
Diabetologia ; 58(6): 1250-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25762204

RESUMEN

AIMS/HYPOTHESIS: Of the four exocytotic syntaxins (Syns), much is now known about the role of Syn-1A (pre-docked secretory granules [SGs]) and Syn-3 (newcomer SGs) in insulin exocytosis. Some work was reported on Syn-4's role in biphasic glucose-stimulated insulin secretion (GSIS), but its precise role in insulin SG exocytosis remains unclear. In this paper we examine this role in human beta cells. METHODS: Endogenous function of Syn-4 in human islets was assessed by knocking down its expression with lentiviral single hairpin RNA (lenti-shRNA)-RFP. Biphasic GSIS was determined by islet perifusion assay. Single-cell analysis of exocytosis of red fluorescent protein (RFP)-positive beta cells (exhibiting near-total depletion of Syn-4) was by patch clamp capacitance measurements (Cm) and total internal reflection fluorescence microscopy (TIRFM), the latter to further assess single SG behaviour. Co-immunoprecipitations were conducted on INS-1 cells to assess exocytotic complexes. RESULTS: Syn-4 knockdown (KD) of 77% in human islets caused a concomitant reduction in cognate Munc18c expression (46%) without affecting expression of other exocytotic proteins; this resulted in reduction of GSIS in the first phase (by 42%) and the second phase (by 40%). Cm of RFP-tagged Syn-4-KD beta cells showed severe inhibition in the readily releasable pool (by 71%) and mobilisation from reserve pools (by 63%). TIRFM showed that Syn-4-KD-induced inhibition of first-phase GSIS was attributed to reduction in exocytosis of both pre-docked and newcomer SGs (which undergo minimal residence or docking time at the plasma membrane before fusion). Second-phase inhibition was attributed to reduction in newcomer SGs. Stx-4 co-immunoprecipitated Munc18c, VAMP2 and VAMP8, suggesting that these exocytotic complexes may be involved in exocytosis of pre-docked and newcomer SGs. CONCLUSIONS/INTERPRETATION: Syn-4 is involved in distinct molecular machineries that influence exocytosis of both pre-docked and newcomer SGs in a manner functionally redundant to Syn-1A and Syn-3, respectively; this underlies Syn-4's role in mediating portions of first-phase and second-phase GSIS.


Asunto(s)
Insulinas Bifásicas/sangre , Exocitosis , Células Secretoras de Insulina/citología , Insulina/metabolismo , Proteínas Qa-SNARE/metabolismo , Animales , Células Cultivadas , Técnicas de Silenciamiento del Gen , Glucosa/metabolismo , Humanos , Secreción de Insulina , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Proteínas Munc18/metabolismo , Técnicas de Placa-Clamp , Proteínas R-SNARE/metabolismo , ARN Interferente Pequeño/metabolismo , Análisis de la Célula Individual , Proteína 2 de Membrana Asociada a Vesículas/metabolismo , Proteína Fluorescente Roja
6.
J Biol Chem ; 289(9): 6028-40, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24429282

RESUMEN

In ß-cells, syntaxin (Syn)-1A interacts with SUR1 to inhibit ATP-sensitive potassium channels (KATP channels). PIP2 binds the Kir6.2 subunit to open KATP channels. PIP2 also modifies Syn-1A clustering in plasma membrane (PM) that may alter Syn-1A actions on PM proteins like SUR1. Here, we assessed whether the actions of PIP2 on activating KATP channels is contributed by sequestering Syn-1A from binding SUR1. In vitro binding showed that PIP2 dose-dependently disrupted Syn-1A·SUR1 complexes, corroborated by an in vivo Forster resonance energy transfer assay showing disruption of SUR1(-EGFP)/Syn-1A(-mCherry) interaction along with increased Syn-1A cluster formation. Electrophysiological studies of rat ß-cells, INS-1, and SUR1/Kir6.2-expressing HEK293 cells showed that PIP2 dose-dependent activation of KATP currents was uniformly reduced by Syn-1A. To unequivocally distinguish between PIP2 actions on Syn-1A and Kir6.2, we employed several strategies. First, we showed that PIP2-insensitive Syn-1A-5RK/A mutant complex with SUR1 could not be disrupted by PIP2, consequently reducing PIP2 activation of KATP channels. Next, Syn-1A·SUR1 complex modulation of KATP channels could be observed at a physiologically low PIP2 concentration that did not disrupt the Syn-1A·SUR1 complex, compared with higher PIP2 concentrations acting directly on Kir6.2. These effects were specific to PIP2 and not observed with physiologic concentrations of other phospholipids. Finally, depleting endogenous PIP2 with polyphosphoinositide phosphatase synaptojanin-1, known to disperse Syn-1A clusters, freed Syn-1A from Syn-1A clusters to bind SUR1, causing inhibition of KATP channels that could no longer be further inhibited by exogenous Syn-1A. These results taken together indicate that PIP2 affects islet ß-cell KATP channels not only by its actions on Kir6.2 but also by sequestering Syn-1A to modulate Syn-1A availability and its interactions with SUR1 on PM.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Receptores de Sulfonilureas/metabolismo , Sintaxina 1/metabolismo , Animales , Línea Celular Tumoral , Células HEK293 , Humanos , Células Secretoras de Insulina/citología , Masculino , Mutación , Fosfatidilinositol 4,5-Difosfato/genética , Canales de Potasio de Rectificación Interna/genética , Ratas , Ratas Wistar , Receptores de Sulfonilureas/genética , Sintaxina 1/genética
7.
J Mol Cell Cardiol ; 75: 100-10, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25073062

RESUMEN

Cardiac sarcolemmal syntaxin (Syn)-1A interacts with sulfonylurea receptor (SUR) 2A to inhibit ATP-sensitive potassium (KATP) channels. Phosphatidylinositol 4,5-bisphosphate (PIP2), a ubiquitous endogenous inositol phospholipid, known to bind Kir6.2 subunit to open KATP channels, has recently been shown to directly bind Syn-1A in plasma membrane to form Syn-1A clusters. Here, we sought to determine whether the interaction between Syn-1A and PIP2 interferes with the ability of Syn-1A to bind SUR2A and inhibit KATP channel activity. We found that PIP2 dose-dependently reduced SUR2A binding to GST-Syn-1A by in vitro pulldown assays. FRET studies in intact cells using TIRFM revealed that increasing endogenous PIP2 levels led to increased Syn-1A (-EGFP) cluster formation and a severe reduction in availability of Syn-1A molecules to interact with SUR2A (-mCherry) molecules outside the Syn-1A clusters. Correspondingly, electrophysiological studies employing SUR2A/Kir6.2-expressing HEK cells showed that increasing endogenous or exogenous PIP2 diminished the inhibitory effect of Syn-1A on KATP currents. The physiological relevance of these findings was confirmed by ability of exogenous PIP2 to block exogenous Syn-1A inhibition of cardiac KATP currents in inside-out patches of mouse ventricular myocytes. The effect of PIP2 on physical and functional interactions between Syn-1A and KATP channels is specific and not observed with physiologic concentrations of other phospholipids. To unequivocally demonstrate the specificity of PIP2 interaction with Syn-1A and its impact on KATP channel modulation by Syn-1A, we employed a PIP2-insensitive Syn-1A-5RK/A mutant. The Syn-1A-5RK/A mutant retains the ability to interact with SUR2A in both in vitro binding and in vivo FRET assays, although as expected the interaction is no longer disrupted by PIP2. Interestingly, at physiological PIP2 concentrations, Syn-1A-5RK/A inhibited KATP currents to a greater extent than Syn-1A-WT, indicating that the inhibitory effect of Syn-1A on KATP channels is not due to direct competition between Syn-1A and Kir6.2 for PIP2 binding. At high-dose PIP2, however, inhibition of KATP currents by Syn-1A-5RK/A was greatly reduced, likely overridden by the direct activating effect of PIP2 on KATP channels. Finally, depleting endogenous PIP2 with polyphosphoinositide phosphatase synaptojanin-1 known to disperse Syn-1A clusters, freed Syn-1A from Syn-1A clusters to bind SUR2A, causing optimal inhibition of KATP channels. These results taken together led us to conclude that PIP2 affects cardiac KATP channels not only by its actions on the channel directly but also by multi-modal effects of dynamically modulating Syn-1A mobility from Syn-1A clusters and thereby the availability of Syn-1A to inhibit KATP channels via interaction with SUR2A on the plasma membrane.


Asunto(s)
Canales KATP/metabolismo , Miocardio/metabolismo , Fosfatidilinositol 4,5-Difosfato/farmacología , Receptores de Sulfonilureas/metabolismo , Sintaxina 1/metabolismo , Animales , Células HEK293 , Humanos , Activación del Canal Iónico/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Canales de Potasio de Rectificación Interna/metabolismo , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Ratas , Sintaxina 1/química
8.
J Biol Chem ; 286(26): 23308-18, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21540180

RESUMEN

The ATP-sensitive potassium (K(ATP)) channel regulates pancreatic ß-cell function by linking metabolic status to electrical activity. Syntaxin-1A (Syn-1A), a SNARE protein mediating exocytotic fusion, binds and inhibits the K(ATP) channel via the nucleotide-binding folds (NBFs) of its sulfonylurea receptor-1 (SUR1) regulatory subunit. In this study, we elucidated the precise regions within the NBFs required for Syn-1A-mediated K(ATP) inhibition, using in vitro binding assays, whole cell patch clamp and FRET assay. Specifically, NBF1 and NBF2 were each divided into three subregions, Walker A (W(A)), signature sequence linker, and Walker B (W(B)), to make GST fusion proteins. In vitro binding assays revealed that Syn-1A associates with W(A) and W(B) regions of both NBFs. Patch clamp recordings on INS-1 and primary rat ß-cells showed that Syn-1A-mediated channel inhibition was reversed by co-addition of NBF1-W(B) (not NBF1-W(A)), NBF2-W(A), and NBF2-W(B). The findings were corroborated by FRET studies showing that these truncates disrupted Syn-1A interactions with full-length SUR1. To further identify the binding sites, series single-site mutations were made in the Walker motifs of the NBFs. Only NBF1-W(A) (K719M) or NBF2-W(A) (K1385M) mutant no longer bound to Syn-1A; K1385M failed to disrupt Syn-1A-mediated inhibition of K(ATP) channels. These data suggest that NBF1-W(A) (Lys-719) and NBF2-W(A) (Lys-1385) are critical for Syn-1A-K(ATP) channel interaction. Taken together, Syn-1A intimately and functionally associates with the SUR1-NBF1/2 dimer via direct interactions with W(A) motifs and sites adjacent to W(B) motifs of NBF1 and NBF2 but transduces its inhibitory actions on K(ATP) channel activity via some but not all of these NBF domains.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Células Secretoras de Insulina/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Pliegue de Proteína , Receptores de Droga/metabolismo , Sintaxina 1/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Secuencias de Aminoácidos , Animales , Sitios de Unión , Células HEK293 , Humanos , Células Secretoras de Insulina/citología , Masculino , Canales de Potasio de Rectificación Interna/genética , Estructura Terciaria de Proteína , Ratas , Ratas Wistar , Receptores de Droga/genética , Receptores de Sulfonilureas , Sintaxina 1/genética
9.
J Biol Chem ; 286(7): 5876-83, 2011 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-21173146

RESUMEN

ATP-sensitive potassium (K(ATP)) channels are regulated by a variety of cytosolic factors (adenine nucleotides, Mg(2+), phospholipids, and pH). We previously reported that K(ATP) channels are also regulated by endogenous membrane-bound SNARE protein syntaxin-1A (Syn-1A), which binds both nucleotide-binding folds of sulfonylurea receptor (SUR)1 and 2A, causing inhibition of K(ATP) channel activity in pancreatic islet ß-cells and cardiac myocytes, respectively. In this study, we show that ATP dose-dependently inhibits Syn-1A binding to SUR1 at physiological concentrations, with the addition of Mg(2+) causing a decrease in the ATP-induced inhibitory effect. This ATP disruption of Syn-1A binding to SUR1 was confirmed by FRET analysis in living HEK293 cells. Electrophysiological studies in pancreatic ß-cells demonstrated that reduced ATP concentrations increased K(ATP) channel sensitivity to Syn-1A inhibition. Depletion of endogenous Syn-1A in insulinoma cells by botulinum neurotoxin C1 proteolysis followed by rescue with exogenous Syn-1A showed that Syn-1A modulates K(ATP) channel sensitivity to ATP. Thus, our data indicate that although both ATP and Syn-1A independently inhibit ß-cell K(ATP) channel gating, they could also influence the sensitivity of K(ATP) channels to each other. These findings provide new insight into an alternate mechanism by which ATP regulates pancreatic ß-cell K(ATP) channel activity, not only by its direct actions on Kir6.2 pore subunit, but also via ATP modulation of Syn-1A binding to SUR1.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Adenosina Trifosfato/metabolismo , Células Secretoras de Insulina/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Receptores de Droga/metabolismo , Sintaxina 1/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Adenosina Trifosfato/genética , Animales , Células HEK293 , Humanos , Activación del Canal Iónico/fisiología , Ratones , Canales de Potasio de Rectificación Interna/genética , Unión Proteica , Ratas , Ratas Sprague-Dawley , Receptores de Droga/genética , Receptores de Sulfonilureas , Sintaxina 1/genética
10.
Nat Commun ; 13(1): 6512, 2022 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-36316316

RESUMEN

Enhancing pancreatic ß-cell secretion is a primary therapeutic target for type-2 diabetes (T2D). Syntaxin-2 (Stx2) has just been identified to be an inhibitory SNARE for insulin granule exocytosis, holding potential as a treatment for T2D, yet its molecular underpinnings remain unclear. We show that excessive Stx2 recruitment to raft-like granule docking sites at higher binding affinity than pro-fusion syntaxin-1A effectively competes for and inhibits fusogenic SNARE machineries. Depletion of Stx2 in human ß-cells improves insulin secretion by enhancing trans-SNARE complex assembly and cis-SNARE disassembly. Using a genetically-encoded reporter, glucose stimulation is shown to induce Stx2 flipping across the plasma membrane, which relieves its suppression of cytoplasmic fusogenic SNARE complexes to promote insulin secretion. Targeting the flipping efficiency of Stx2 profoundly modulates secretion, which could restore the impaired insulin secretion in diabetes. Here, we show that Stx2 acts to assist this precise tuning of insulin secretion in ß-cells, including in diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2 , Insulina , Humanos , Sintaxina 1/genética , Sintaxina 1/metabolismo , Insulina/metabolismo , Exocitosis/fisiología , Proteínas SNARE/metabolismo , Membrana Celular/metabolismo
11.
J Mol Cell Cardiol ; 51(5): 790-802, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21884702

RESUMEN

We previously demonstrated that syntaxin (Syn)-1A is present in the sarcolemma of rat cardiomyocytes and binds sulfonylurea receptor (SUR) 2A nucleotide binding folds (NBFs) to inhibit ATP-sensitive potassium (K(ATP)) channel. Here, we examined for the precise domains within the NBFs of SUR2A that may interact with Syn-1A. Specifically, we tested truncated NBF protein segments encompassing the conserved motifs Walker A (W(A)), signature/Linker (L), and Walker B (W(B)). In vitro binding results indicate that the domains encompassing W(A) and L of NBF-1 and all three conserved motifs of NBF-2 bound Syn-1A. Electrophysiological studies, employing inside-out patch-clamp recordings from SUR2A/Kir6.2 expressing HEK cells and mouse cardiomyocytes, show that W(B) and L of NBF-1 and all three NBF-2 truncated protein segments reduced Syn-1A inhibition of SUR2A/K(ATP) channels. Remarkably, these same NBF-1 and -2 truncated proteins could independently disrupt the intimate FRET interactions of full length SUR2A (-mCherry) and Syn-1A (-EGFP). These results taken together indicate that Syn-1A possibly maintains inhibition of cardiac ventricular K(ATP) channels by binding to large regions of NBF-1 and NBF-2 to stabilize the NBF-1-NBF-2 heterodimer formation and prevent ATP-binding and ATP hydrolysis. Since K(ATP) channels are closely coupled to metabolic states, we postulate that these very intimate Syn-1A-SUR2A interactions are critically important for myocardial protection during stress, in which profound changes in metabolic factors (pH, ATP) could modulate these Syn-1A-SUR2A interactions.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Canales KATP/metabolismo , Miocitos Cardíacos/metabolismo , Oligopéptidos/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Receptores de Droga/metabolismo , Proteínas Recombinantes/metabolismo , Transducción de Señal/fisiología , Sintaxina 1/metabolismo , Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/genética , Adenosina Trifosfato/biosíntesis , Secuencias de Aminoácidos , Animales , Sitios de Unión , Membrana Celular/metabolismo , Secuencia Conservada , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Humanos , Canales KATP/química , Canales KATP/genética , Masculino , Ratones , Microscopía Fluorescente , Miocitos Cardíacos/citología , Oligopéptidos/química , Oligopéptidos/genética , Técnicas de Placa-Clamp , Plásmidos , Potasio/metabolismo , Canales de Potasio de Rectificación Interna/química , Canales de Potasio de Rectificación Interna/genética , Unión Proteica , Estructura Terciaria de Proteína , Receptores de Droga/química , Receptores de Droga/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Receptores de Sulfonilureas , Sintaxina 1/química , Sintaxina 1/genética
12.
Endocr Rev ; 28(6): 653-63, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17878408

RESUMEN

The three SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) proteins, syntaxin, SNAP25 (synaptosome-associated protein of 25 kDa), and synaptobrevin, constitute the minimal machinery for exocytosis in secretory cells such as neurons and neuroendocrine cells by forming a series of complexes prior to and during vesicle fusion. It was subsequently found that these SNARE proteins not only participate in vesicle fusion, but also tether with voltage-dependent Ca(2+) channels to form an excitosome that precisely regulates calcium entry at the site of exocytosis. In pancreatic islet beta-cells, ATP-sensitive K(+) (K(ATP)) channel closure by high ATP concentration leads to membrane depolarization, voltage-dependent Ca(2+) channel opening, and insulin secretion, whereas subsequent opening of voltage-gated K(+) (Kv) channels repolarizes the cell to terminate exocytosis. We have obtained evidence that syntaxin-1A physically interacts with Kv2.1 (the predominant Kv in beta-cells) and the sulfonylurea receptor subunit of beta-cell K(ATP) channel to modify their gating behaviors. A model has proposed that the conformational changes of syntaxin-1A during exocytosis induce distinct functional modulations of K(ATP) and Kv2.1 channels in a manner that optimally regulates cell excitability and insulin secretion. Other proteins involved in exocytosis, such as Munc-13, tomosyn, rab3a-interacting molecule, and guanyl nucleotide exchange factor II, have also been implicated in direct or indirect regulation of beta-cell ion channel activities and excitability. This review discusses this interesting aspect that exocytotic proteins not only promote secretion per se, but also fine-tune beta-cell excitability via modulation of ion channel gating.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Activación del Canal Iónico/fisiología , Canales KATP/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Proteínas SNARE/fisiología , Sintaxina 1/fisiología , Animales , Exocitosis/fisiología , Humanos
13.
Autophagy ; 17(10): 3068-3081, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33213278

RESUMEN

Intrapancreatic trypsin activation by dysregulated macroautophagy/autophagy and pathological exocytosis of zymogen granules (ZGs), along with activation of inhibitor of NFKB/NF-κB kinase (IKK) are necessary early cellular events in pancreatitis. How these three pancreatitis events are linked is unclear. We investigated how SNAP23 orchestrates these events leading to pancreatic acinar injury. SNAP23 depletion was by knockdown (SNAP23-KD) effected by adenovirus-shRNA (Ad-SNAP23-shRNA/mCherry) treatment of rodent and human pancreatic slices and in vivo by infusion into rat pancreatic duct. In vitro pancreatitis induction by supraphysiological cholecystokinin (CCK) or ethanol plus low-dose CCK were used to assess SNAP23-KD effects on exocytosis and autophagy. Pancreatitis stimuli resulted in SNAP23 translocation from its native location at the plasma membrane to autophagosomes, where SNAP23 would bind and regulate STX17 (syntaxin17) SNARE complex-mediated autophagosome-lysosome fusion. This SNAP23 relocation was attributed to IKBKB/IKKß-mediated SNAP23 phosphorylation at Ser95 Ser120 in rat and Ser120 in human, which was blocked by IKBKB/IKKß inhibitors, and confirmed by the inability of IKBKB/IKKß phosphorylation-disabled SNAP23 mutant (Ser95A Ser120A) to bind STX17 SNARE complex. SNAP23-KD impaired the assembly of STX4-driven basolateral exocytotic SNARE complex and STX17-driven SNARE complex, causing respective reduction of basolateral exocytosis of ZGs and autolysosome formation, with consequent reduction in trypsinogen activation in both compartments. Consequently, pancreatic SNAP23-KD rats were protected from caerulein and alcoholic pancreatitis. This study revealed the roles of SNAP23 in mediating pathological basolateral exocytosis and IKBKB/IKKß's involvement in autolysosome formation, both where trypsinogen activation would occur to cause pancreatitis. SNAP23 is a strong candidate to target for pancreatitis therapy.Abbreviations: AL: autolysosome; AP: acute pancreatitis; AV: autophagic vacuole; CCK: cholecystokinin; IKBKB/IKKß: inhibitor of nuclear factor kappa B kinase subunit beta; SNAP23: synaptosome associated protein 23; SNARE: soluble NSF (N-ethylmaleimide-sensitive factor) attachment protein receptor; STX: syntaxin; TAP: trypsinogen activation peptide; VAMP: vesicle associated membrane protein; ZG: zymogen granule.


Asunto(s)
Pancreatitis , Proteínas Qb-SNARE , Proteínas Qc-SNARE , Enfermedad Aguda , Animales , Autofagia , Exocitosis , Humanos , Lisosomas , Páncreas , Pancreatitis/genética , Pancreatitis/prevención & control , Proteínas Qb-SNARE/genética , Proteínas Qc-SNARE/genética , Ratas , Tripsina/farmacología , Proteínas de Transporte Vesicular
14.
J Biol Chem ; 284(31): 20840-7, 2009 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-19487699

RESUMEN

Cab45b is a cytosolic Ca(2+)-binding protein reported to regulate zymogen secretion in pancreatic acini. We now show that Cab45b is also expressed in pancreatic islet beta-cells and interacts there with the Sec1-Munc18 protein Munc18b. We employed patch clamp cell capacitance measurements to show that antibodies against Cab45b inhibited depolarization-evoked membrane capacitance increments, suggesting an impact on beta-cell granule exocytosis, both the readily releasable granule pool and refilling of this pool. Site-specific mutants in the Cab45b EF-hands were used to dissect the molecular interactions involved in Cab45b function. Mutants in EF-hands 2 and 3 had no detectable effects on interaction of Cab45b with Munc18b and did not affect the depolarization-evoked calcium currents, but remarkably, they facilitated the complex formation of Munc18b with syntaxin-2 and -3. As a result, these two EF-hand mutants inhibited beta-cell membrane capacitance increments. This inhibition is mediated via Munc18b because Munc18b silencing with small interfering RNA abolished the effects of these two mutants. The results suggest a mechanism for Cab45b action that involves regulating the dynamic association of Munc18b with SNAREs to impact beta-cell granule exocytosis.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Exocitosis , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Proteínas Munc18/metabolismo , Animales , Anticuerpos/metabolismo , Señalización del Calcio , Proteínas de Unión al Calcio/química , Gránulos Citoplasmáticos/metabolismo , Células Secretoras de Glucagón/citología , Células Secretoras de Glucagón/metabolismo , Potenciales de la Membrana , Proteínas Mutantes/metabolismo , Células Neuroendocrinas/citología , Células Neuroendocrinas/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Qa-SNARE/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas
15.
JCI Insight ; 5(3)2020 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-32051343

RESUMEN

SNAP23 is the ubiquitous SNAP25 isoform that mediates secretion in non-neuronal cells, similar to SNAP25 in neurons. However, some secretory cells like pancreatic islet ß cells contain an abundance of both SNAP25 and SNAP23, where SNAP23 is believed to play a redundant role to SNAP25. We show that SNAP23, when depleted in mouse ß cells in vivo and human ß cells (normal and type 2 diabetes [T2D] patients) in vitro, paradoxically increased biphasic glucose-stimulated insulin secretion corresponding to increased exocytosis of predocked and newcomer insulin granules. Such effects on T2D Goto-Kakizaki rats improved glucose homeostasis that was superior to conventional treatment with sulfonylurea glybenclamide. SNAP23, although fusion competent in slower secretory cells, in the context of ß cells acts as a weak partial fusion agonist or inhibitory SNARE. Here, SNAP23 depletion promotes SNAP25 to bind calcium channels more quickly and longer where granule fusion occurs to increase exocytosis efficiency. ß Cell SNAP23 antagonism is a strategy to treat diabetes.


Asunto(s)
Canales de Calcio/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Exocitosis , Insulina/metabolismo , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Animales , Glucosa/metabolismo , Homeostasis , Humanos , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Noqueados , Técnicas de Placa-Clamp , Proteínas Qb-SNARE/genética , Proteínas Qc-SNARE/genética , Ratas
16.
Am J Physiol Endocrinol Metab ; 297(2): E452-61, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19509185

RESUMEN

Synaptosomal protein of 25 kDa (SNAP-25) is important for Ca(2+)-dependent fusion of large dense core vesicles (LDCVs) in insulin-secreting cells. Exocytosis is further enhanced by cAMP-increasing agents such as glucagon-like peptide-1 (GLP-1), and this augmentation includes interaction with both PKA and cAMP-GEFII. To investigate the coupling between SNAP-25- and cAMP-dependent stimulation of insulin exocytosis, we have used capacitance measurements, protein-binding assays, and Western blot analysis. In insulin-secreting INS-1 cells overexpressing wild-type SNAP-25 (SNAP-25(WT)), rapid exocytosis was stimulated more than threefold by cAMP, similar to the situation in nontransfected cells. However, cAMP failed to potentiate rapid exocytosis in INS-1 cells overexpressing a truncated form of SNAP-25 (SNAP-25(1-197)) or Botulinum neurotoxin A (BoNT/A). Close dissection of the exocytotic response revealed that the inability of cAMP to stimulate exocytosis in the presence of a truncated SNAP-25 was confined to the release of primed LDCVs within the readily releasable pool, especially from the immediately releasable pool, whereas cAMP enhanced mobilization of granules from the reserve pool in both SNAP-25(1-197) (P < 0.01) and SNAP-25(WT) (P < 0.05) cells. This was supported by hormone release measurements. Augmentation of the immediately releasable pool by cAMP has been suggested to act through the cAMP-GEFII-dependent, PKA-independent pathway. Indeed, we were able to verify an interaction between SNAP-25 with both cAMP-GEFII and RIM2, two proteins involved in the PKA-independent pathway. Thus we hypothesize that SNAP-25 is a necessary partner in the complex mediating cAMP-enhanced rapid exocytosis in insulin-secreting cells.


Asunto(s)
AMP Cíclico/farmacología , Exocitosis/efectos de los fármacos , Exocitosis/genética , Células Secretoras de Insulina/efectos de los fármacos , Proteína 25 Asociada a Sinaptosomas/genética , Animales , Toxinas Botulínicas Tipo A/farmacología , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Resistencia a Medicamentos/efectos de los fármacos , Resistencia a Medicamentos/genética , Eliminación de Gen , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica , Estructura Terciaria de Proteína/genética , Ratas , Proteína 25 Asociada a Sinaptosomas/química , Proteína 25 Asociada a Sinaptosomas/metabolismo , Proteína 25 Asociada a Sinaptosomas/fisiología , Factores de Tiempo
17.
Biochem J ; 396(2): 363-9, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16478442

RESUMEN

Distinct domains within the SNARE (soluble N-ethylmaleimide-sensitive fusion protein attachment protein receptor) proteins, STX1A (syntaxin 1A) and SNAP-25 (synaptosome-associated protein-25 kDa), regulate hormone secretion by their actions on the cell's exocytotic machinery, as well as voltage-gated Ca2+ and K+ channels. We examined the action of distinct domains within SNAP-25 on Kv2.1 (voltage gated K+ 2.1) channel gating. Dialysis of N-terminal SNAP-25 domains, S197 (SNAP-25(1-197)) and S180 (SNAP-25(1-180)), but not S206 (full-length SNAP-25(1-206)) increased the rate of Kv2.1 channel activation and slowed channel inactivation. Remarkably, these N-terminal SNAP-25 domains, acting on the Kv2.1 cytoplasmic N-terminus, potentiated the external TEA (tetraethylammonium)-mediated block of Kv2.1. To further examine whether these are effects of the channel pore domain, internal K+ was replaced with Na+ and external K+ was decreased from 4 to 1 mM, which decreased the IC50 of the TEA block from 6.8+/-0.9 mM to >100 mM. Under these conditions S180 completely restored TEA sensitivity (7.9+/-1.5 mM). SNAP-25 C-terminal domains, SNAP-25(198-206) and SNAP-25(181-197), had no effect on Kv2.1 gating kinetics. We conclude that different domains within SNAP-25 can form distinct complexes with Kv2.1 to execute a fine allosteric regulation of channel gating and the architecture of the outer pore structure in order to modulate cell excitability.


Asunto(s)
Activación del Canal Iónico , Estructura Terciaria de Proteína , Canales de Potasio Shab/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Tetraetilamonio/farmacología , Regulación Alostérica , Animales , Antidiscinéticos/metabolismo , Antidiscinéticos/farmacología , Toxinas Botulínicas/metabolismo , Toxinas Botulínicas/farmacología , Toxinas Botulínicas Tipo A/metabolismo , Toxinas Botulínicas Tipo A/farmacología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Electrofisiología , Humanos , Activación del Canal Iónico/efectos de los fármacos , Péptidos/metabolismo , Unión Proteica , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Sensibilidad y Especificidad , Canales de Potasio Shab/genética , Proteína 25 Asociada a Sinaptosomas/genética , Proteína 25 Asociada a Sinaptosomas/farmacología , Tetraetilamonio/metabolismo , Transfección
18.
EBioMedicine ; 16: 262-274, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28163042

RESUMEN

Reduced pancreatic islet levels of Munc18a/SNARE complex proteins have been postulated to contribute to the deficient glucose-stimulated insulin secretion (GSIS) in type-2 diabetes (T2D). Whereas much previous work has purported Munc18a/SNARE complex (Syntaxin-1A/VAMP-2/SNAP25) to be primarily involved in predocked secretory granule (SG) fusion, less is known about newcomer SGs that undergo minimal docking time at the plasma membrane before fusion. Newcomer SG fusion has been postulated to involve a distinct SM/SNARE complex (Munc18b/Syntaxin-3/VAMP8/SNAP25), whose levels we find also reduced in islets of T2D humans and T2D Goto-Kakizaki (GK) rats. Munc18b overexpression by adenovirus infection (Ad-Munc18b), by increasing assembly of Munc18b/SNARE complexes, mediated increased fusion of not only newcomer SGs but also predocked SGs in T2D human and GK rat islets, resulting in rescue of the deficient biphasic GSIS. Infusion of Ad-Munc18b into GK rat pancreas led to sustained improvement in glucose homeostasis. However, Munc18b overexpression in normal islets increased only newcomer SG fusion. Therefore, Munc18b could potentially be deployed in human T2D to rescue the deficient GSIS.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Glucosa/metabolismo , Homeostasis , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Proteínas Munc18/metabolismo , Anciano , Animales , Western Blotting , Diabetes Mellitus Tipo 2/genética , Femenino , Humanos , Secreción de Insulina , Masculino , Microscopía Confocal , Persona de Mediana Edad , Complejos Multiproteicos/metabolismo , Proteínas Munc18/genética , Proteínas Qa-SNARE/metabolismo , Proteínas R-SNARE/metabolismo , Ratas Endogámicas , Proteína 25 Asociada a Sinaptosomas/metabolismo
19.
Diabetes ; 66(4): 948-959, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28115395

RESUMEN

Of the four syntaxins specialized for exocytosis, syntaxin (Syn)-2 is the least understood. In this study, we used Syn-2/epimorphin knockout mice to examine the role of Syn-2 in insulin secretory granule (SG) exocytosis. Unexpectedly, Syn-2 knockout mice exhibited paradoxical superior glucose homeostasis resulting from an enhanced insulin secretion. This was confirmed in vitro by pancreatic islet perifusion showing an amplified biphasic glucose-stimulated insulin secretion arising from an increase in size of the readily releasable pool of insulin SGs and enhanced SG pool refilling. The increase in insulin exocytosis was attributed mainly to an enhanced recruitment of the larger pool of newcomer SGs that undergoes no residence time on plasma membrane before fusion and, to a lesser extent, also the predocked SGs. Consistently, Syn-2 depletion resulted in a stimulation-induced increase in abundance of exocytotic complexes we previously demonstrated as mediating the fusion of newcomer SGs (Syn-3/VAMP8/SNAP25/Munc18b) and predocked SGs (Syn-1A/VAMP2/SNAP25/Muncn18a). This work is the first to show in mammals that Syn-2 could function as an inhibitory SNARE protein that, when relieved, could promote exocytosis in pancreatic islet ß-cells. Thus, Syn-2 may serve as a potential target to treat diabetes.


Asunto(s)
Exocitosis/genética , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Proteínas SNARE/metabolismo , Sintaxina 1/genética , Animales , Western Blotting , Inmunoprecipitación , Secreción de Insulina , Ratones , Ratones Noqueados , Proteínas Munc18/metabolismo , Proteínas Qa-SNARE/metabolismo , Proteínas R-SNARE/metabolismo , Vesículas Secretoras/metabolismo , Fracciones Subcelulares , Proteína 25 Asociada a Sinaptosomas/metabolismo , Sintaxina 1/metabolismo , Proteína 2 de Membrana Asociada a Vesículas/metabolismo
20.
Endocrinology ; 147(8): 3727-36, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16644915

RESUMEN

Although proglucagon gene expression and the synthesis of proglucagon encoded peptide hormones could be activated by protein kinase A (PKA) activators such as forskolin/3-isobutyl-1-methylxanthine (IBMX) and cholera toxin, whether the activation is entirely attributed to PKA has not been previously examined. We found that forskolin/IBMX also activate ERK1/2 phosphorylation in intestinal and pancreatic proglucagon-producing cell lines. The MEK inhibitors PD98059 and U0126 were found to repress the expression of proglucagon promoter as well as endogenous proglucagon mRNA in two intestinal proglucagon-producing cell lines and to block the stimulatory effect of forskolin/IBMX on proglucagon mRNA expression. The repressive effect of the PKA-specific inhibitors H-89 and KT-5720, however, was either not observable or much less potent. Forskolin could activate ERK1/2 phosphorylation and proglucagon gene transcription on its own, whereas forskolin plus IBMX are required to effectively activate the PKA pathway in the proglucagon-producing cells. Exchange protein directly activated by cyclic AMP 2 (Epac2, or cAMP-binding guanine nucleotide exchange factor-2) was found to be expressed in gut and pancreatic proglucagon-producing cell lines, whereas the Epac-pathway-specific cAMP analog, 8-pMeOPT-2'O-Me-cAMP, effectively stimulated ERK1/2 phosphorylation as well as proglucagon mRNA expression. We therefore suggest that cAMP at least partially regulates proglucagon gene expression via the Epac-Ras/Rap-Raf-MEK-ERK signaling pathway.


Asunto(s)
Proteínas Portadoras/metabolismo , AMP Cíclico/metabolismo , Células Enteroendocrinas/fisiología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proglucagón/genética , 1-Metil-3-Isobutilxantina/farmacología , Animales , Proteínas Portadoras/genética , Línea Celular , Colforsina/farmacología , AMP Cíclico/análogos & derivados , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Enteroendocrinas/citología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Factores de Intercambio de Guanina Nucleótido/genética , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Inhibidores de Fosfodiesterasa/farmacología , Fosforilación , Regiones Promotoras Genéticas/genética , ARN Mensajero/metabolismo , Transcripción Genética/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA