Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Metab ; 35(7): 1242-1260.e9, 2023 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-37339634

RESUMEN

Type 1 (T1D) or type 2 diabetes (T2D) are caused by a deficit of functional insulin-producing ß cells. Thus, the identification of ß cell trophic agents could allow the development of therapeutic strategies to counteract diabetes. The discovery of SerpinB1, an elastase inhibitor that promotes human ß cell growth, prompted us to hypothesize that pancreatic elastase (PE) regulates ß cell viability. Here, we report that PE is up-regulated in acinar cells and in islets from T2D patients, and negatively impacts ß cell viability. Using high-throughput screening assays, we identified telaprevir as a potent PE inhibitor that can increase human and rodent ß cell viability in vitro and in vivo and improve glucose tolerance in insulin-resistant mice. Phospho-antibody microarrays and single-cell RNA sequencing analysis identified PAR2 and mechano-signaling pathways as potential mediators of PE. Taken together, our work highlights PE as a potential regulator of acinar-ß cell crosstalk that acts to limit ß cell viability, leading to T2D.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Humanos , Ratones , Animales , Células Acinares/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Elastasa Pancreática/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Comunicación Celular
2.
Diabetes Obes Metab ; 24(7): 1255-1266, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35293666

RESUMEN

AIM: To establish if alpha-7 nicotinic acetylcholine receptor (α7nAChR) agonist GTS-21 exerts a blood glucose-lowering action in db/db mice, and to test if this action requires coordinate α7nAChR and GLP-1 receptor (GLP-1R) stimulation by GTS-21 and endogenous GLP-1, respectively. MATERIALS AND METHODS: Blood glucose levels were measured during an oral glucose tolerance test (OGTT) using db/db mice administered intraperitoneal GTS-21. Plasma GLP-1, peptide tyrosine tyrosine 1-36 (PYY1-36), glucose-dependent insulinotropic peptide (GIP), glucagon, and insulin levels were measured by ELISA. A GLP-1R-mediated action of GTS-21 that is secondary to α7nAChR stimulation was evaluated using α7nAChR and GLP-1R knockout (KO) mice, or by co-administration of GTS-21 with the dipeptidyl peptidase-4 inhibitor, sitagliptin, or the GLP-1R antagonist, exendin (9-39). Insulin sensitivity was assessed in an insulin tolerance test. RESULTS: Single or multiple dose GTS-21 (0.5-8.0 mg/kg) acted in a dose-dependent manner to lower levels of blood glucose in the OGTT using 10-14 week-old male and female db/db mice. This action of GTS-21 was reproduced by the α7nAChR agonist, PNU-282987, was enhanced by sitagliptin, was counteracted by exendin (9-39), and was absent in α7nAChR and GLP-1R KO mice. Plasma GLP-1, PYY1-36, GIP, glucagon, and insulin levels increased in response to GTS-21, but insulin sensitivity, body weight, and food intake were unchanged. CONCLUSIONS: α7nAChR agonists improve oral glucose tolerance in db/db mice. This action is contingent to coordinate α7nAChR and GLP-1R stimulation. Thus α7nAChR agonists administered in combination with sitagliptin might serve as a new treatment for type 2 diabetes.


Asunto(s)
Compuestos de Bencilideno , Glucemia , Resistencia a la Insulina , Agonistas Nicotínicos , Piridinas , Receptor Nicotínico de Acetilcolina alfa 7 , Animales , Compuestos de Bencilideno/farmacología , Glucemia/análisis , Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Femenino , Polipéptido Inhibidor Gástrico/metabolismo , Glucagón/metabolismo , Péptido 1 Similar al Glucagón , Receptor del Péptido 1 Similar al Glucagón/genética , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Prueba de Tolerancia a la Glucosa , Humanos , Incretinas/uso terapéutico , Insulina/uso terapéutico , Masculino , Ratones , Ratones Noqueados , Agonistas Nicotínicos/farmacología , Piridinas/farmacología , Fosfato de Sitagliptina/uso terapéutico , Tirosina/uso terapéutico , Receptor Nicotínico de Acetilcolina alfa 7/agonistas , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
3.
J Biol Chem ; 298(2): 101484, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34896391

RESUMEN

We report that intra-islet glucagon secreted from α-cells signals through ß-cell glucagon and GLP-1 receptors (GcgR and GLP-1R), thereby conferring to rat islets their competence to exhibit first-phase glucose-stimulated insulin secretion (GSIS). Thus, in islets not treated with exogenous glucagon or GLP-1, first-phase GSIS is abolished by a GcgR antagonist (LY2786890) or a GLP-1R antagonist (Ex[9-39]). Mechanistically, glucose competence in response to intra-islet glucagon is conditional on ß-cell cAMP signaling because it is blocked by the cAMP antagonist prodrug Rp-8-Br-cAMPS-pAB. In its role as a paracrine hormone, intra-islet glucagon binds with high affinity to the GcgR, while also exerting a "spillover" effect to bind with low affinity to the GLP-1R. This produces a right shift of the concentration-response relationship for the potentiation of GSIS by exogenous glucagon. Thus, 0.3 nM glucagon fails to potentiate GSIS, as expected if similar concentrations of intra-islet glucagon already occupy the GcgR. However, 10 to 30 nM glucagon effectively engages the ß-cell GLP-1R to potentiate GSIS, an action blocked by Ex[9-39] but not LY2786890. Finally, we report that the action of intra-islet glucagon to support insulin secretion requires a step-wise increase of glucose concentration to trigger first-phase GSIS. It is not measurable when GSIS is stimulated by a gradient of increasing glucose concentrations, as occurs during an oral glucose tolerance test in vivo. Collectively, such findings are understandable if defective intra-islet glucagon action contributes to the characteristic loss of first-phase GSIS in an intravenous glucose tolerance test that is diagnostic of type 2 diabetes in the clinical setting.


Asunto(s)
Diabetes Mellitus Tipo 2 , Receptor del Péptido 1 Similar al Glucagón , Glucagón , Glucosa , Secreción de Insulina , Islotes Pancreáticos , Animales , Diabetes Mellitus Tipo 2/metabolismo , Glucagón/metabolismo , Glucagón/farmacología , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Glucosa/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratas
4.
Rev Endocr Metab Disord ; 21(4): 431-447, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32851581

RESUMEN

The cholinergic anti-inflammatory reflex (CAIR) represents an important homeostatic regulatory mechanism for sensing and controlling the body's response to inflammatory stimuli. Vagovagal reflexes are an integral component of CAIR whose anti-inflammatory effects are mediated by acetylcholine (ACh) acting at α7 nicotinic acetylcholine receptors (α7nAChR) located on cells of the immune system. Recently, it is appreciated that CAIR and α7nAChR also participate in the control of metabolic homeostasis. This has led to the understanding that defective vagovagal reflex circuitry underlying CAIR might explain the coexistence of obesity, diabetes, and inflammation in the metabolic syndrome. Thus, there is renewed interest in the α7nAChR that mediates CAIR, particularly from the standpoint of therapeutics. Of special note is the recent finding that α7nAChR agonist GTS-21 acts at L-cells of the distal intestine to stimulate the release of two glucoregulatory and anorexigenic hormones: glucagon-like peptide-1 (GLP-1) and peptide YY (PYY). Furthermore, α7nAChR agonist PNU 282987 exerts trophic factor-like actions to support pancreatic ß-cell survival under conditions of stress resembling diabetes. This review provides an overview of α7nAChR function as it pertains to CAIR, vagovagal reflexes, and metabolic homeostasis. We also consider the possible usefulness of α7nAChR agonists for treatment of obesity, diabetes, and inflammation.


Asunto(s)
Sistema Nervioso Autónomo/fisiología , Diabetes Mellitus/tratamiento farmacológico , Homeostasis/fisiología , Inflamación/tratamiento farmacológico , Agonistas Nicotínicos/farmacología , Obesidad/tratamiento farmacológico , Receptor Nicotínico de Acetilcolina alfa 7/agonistas , Animales , Diabetes Mellitus/metabolismo , Humanos , Inflamación/metabolismo , Obesidad/metabolismo
6.
J Biol Chem ; 294(7): 2247-2248, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30765510

RESUMEN

The activity of the archetypal protein kinase A (PKA) is typically thought of in regards to the catalytic subunit, which is inhibited by the regulatory subunits in the absence of cAMP. However, it is now reported that one of the regulatory subunit isoforms (PKA-RIα) takes on a function of its own upon binding to cAMP, acting independently of this canonical cAMP signaling mechanism. PKA-RIα instead binds to and stimulates the catalytic activity of a guanine nucleotide exchange factor (P-REX1) that itself promotes Rac1 GTPase activation. This newly discovered function of PKA-RIα adds an additional layer of complexity to our understanding of cAMP signal transduction.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , AMP Cíclico/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Sistemas de Mensajero Secundario , Proteínas Quinasas Activadas por AMP/genética , Animales , AMP Cíclico/genética , Factores de Intercambio de Guanina Nucleótido/genética , Humanos
7.
J Biol Chem ; 294(10): 3514-3531, 2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30622136

RESUMEN

G protein-coupled receptors (GPCRs) for glucagon (GluR) and glucagon-like peptide-1 (GLP-1R) are normally considered to be highly selective for glucagon and GLP-1, respectively. However, glucagon secreted from pancreatic α-cells may accumulate at high concentrations to exert promiscuous effects at the ß-cell GLP-1R, as may occur in the volume-restricted microenvironment of the islets of Langerhans. Furthermore, systemic administration of GluR or GLP-1R agonists and antagonists at high doses may lead to off-target effects at other receptors. Here, we used molecular modeling to evaluate data derived from FRET assays that detect cAMP as a read-out for GluR and GLP-1R activation. This analysis established that glucagon is a nonconventional GLP-1R agonist, an effect inhibited by the GLP-1R orthosteric antagonist exendin(9-39) (Ex(9-39)). The GluR allosteric inhibitors LY2409021 and MK 0893 antagonized glucagon and GLP-1 action at the GLP-1R, whereas des-His1-[Glu9]glucagon antagonized glucagon action at the GluR, while having minimal inhibitory action versus glucagon or GLP-1 at the GLP-1R. When testing Ex(9-39) in combination with des-His1-[Glu9]glucagon in INS-1 832/13 cells, we validated a dual agonist action of glucagon at the GluR and GLP-1R. Hybrid peptide GGP817 containing glucagon fused to a fragment of peptide YY (PYY) acted as a triagonist at the GluR, GLP-1R, and neuropeptide Y2 receptor (NPY2R). Collectively, these findings provide a new triagonist strategy with which to target the GluR, GLP-1R, and NPY2R. They also provide an impetus to reevaluate prior studies in which GluR and GLP-1R agonists and antagonists were assumed not to exert promiscuous actions at other GPCRs.


Asunto(s)
AMP Cíclico/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Receptores de Glucagón/agonistas , Receptores de Glucagón/antagonistas & inhibidores , Secuencia de Aminoácidos , Descubrimiento de Drogas , Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/química , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Células HEK293 , Humanos , Simulación del Acoplamiento Molecular , Péptidos/química , Péptidos/metabolismo , Péptidos/farmacología , Conformación Proteica , Receptores de Glucagón/química , Receptores de Glucagón/metabolismo
8.
Diabetes ; 67(10): 1999-2011, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29986926

RESUMEN

Glucose metabolism stimulates cell division control protein 42 homolog (Cdc42)-p21-activated kinase (Pak1) activity and initiates filamentous actin (F-actin) cytoskeleton remodeling in pancreatic ß-cells so that cytoplasmic secretory granules can translocate to the plasma membrane where insulin exocytosis occurs. Since glucose metabolism also generates cAMP in ß-cells, the cross talk of cAMP signaling with Cdc42-Pak1 activation might be of fundamental importance to glucose-stimulated insulin secretion (GSIS). Previously, the type-2 isoform of cAMP-regulated guanine nucleotide exchange factor 2 (Epac2) was established to mediate a potentiation of GSIS by cAMP-elevating agents. Here we report that nondiabetic human islets and INS-1 832/13 ß-cells treated with the selective Epac activator 8-pCPT-2'-O-Me-cAMP-AM exhibited Cdc42-Pak1 activation at 1 mmol/L glucose and that the magnitude of this effect was equivalent to that which was measured during stimulation with 20 mmol/L glucose in the absence of 8-pCPT-2'-O-Me-cAMP-AM. Conversely, the cAMP antagonist Rp-8-Br-cAMPS-pAB prevented glucose-stimulated Cdc42-Pak1 activation, thereby blocking GSIS while also increasing cellular F-actin content. Although islets from donors with type 2 diabetes had profound defects in glucose-stimulated Cdc42-Pak1 activation and insulin secretion, these defects were rescued by the Epac activator so that GSIS was restored. Collectively, these findings indicate an unexpected role for cAMP as a permissive or direct metabolic coupling factor in support of GSIS that is Epac2 and Cdc42-Pak1 regulated.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Glucosa/farmacología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Quinasas p21 Activadas/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/análogos & derivados , 8-Bromo Monofosfato de Adenosina Cíclica/química , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Animales , Línea Celular , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Ratas , Tionucleótidos/química , Tionucleótidos/farmacología
9.
Endocrinology ; 159(9): 3132-3142, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29992246

RESUMEN

Enteroendocrine L cells secrete the incretin hormone glucagon-like peptide-1 (GLP-1), and they also express the α7 nicotinic acetylcholine receptor (α7nAChR), which may regulate GLP-1 secretion. Here, GTS-21, a selective α7nAChR agonist, was used to examine the effect of α7nAChR activation in L-cell lines, mouse intestinal primary cell cultures, and C57BL/6 mice. GTS-21 stimulated GLP-1 secretion in vitro, and this effect was attenuated by an α7nAChR antagonist or by α7nAChR-specific small interfering RNA. Under in vitro cell culture conditions of glucotoxicity, GTS-21 restored GLP-1 secretion and improved L-cell viability while also acting in vivo to raise levels of circulating GLP-1 in mice. To assess potential signaling mechanisms underlying these actions of GTS-21, we first monitored Ca2+, cAMP, and phosphatidylinositol 3-kinase (PI3K) activity. As expected for a GLP-1 secretagogue promoting Ca2+ influx through α7nAChR cation channels, [Ca2+]i increased in response to GTS-21, but [cAMP]i was unchanged. Surprisingly, pharmacological inhibition of growth factor signaling pathways revealed that GTS-21 also acts on the PI3K-protein kinase B-mammalian target of rapamycin pathway to promote L-cell viability. Moreover, the Ca2+ chelator BAPTA-AM counteracted GTS-21‒stimulated PI3K activity, thereby indicating unexpected crosstalk of L-cell Ca2+ and growth factor signaling pathways. Collectively, these data demonstrate that α7nAChR activation enhances GLP-1 secretion by increasing levels of cytosolic Ca2+ while also revealing Ca2+- and PI3K-dependent processes of α7nAChR activation that promote L-cell survival.


Asunto(s)
Calcio/metabolismo , Supervivencia Celular/fisiología , Células Enteroendocrinas/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Animales , Compuestos de Bencilideno/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Quelantes/farmacología , AMP Cíclico/metabolismo , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Células Enteroendocrinas/efectos de los fármacos , Péptido 1 Similar al Glucagón/efectos de los fármacos , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasa/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piridinas/farmacología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/agonistas
10.
Int J Mol Sci ; 19(5)2018 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-29783744

RESUMEN

Store-operated calcium entry (SOCE), a fundamentally important homeostatic and Ca2+ signaling pathway in many types of cells, is activated by the direct interaction of stromal interaction molecule 1 (STIM1), an endoplasmic reticulum (ER) Ca2+-binding protein, with Ca2+-selective Orai1 channels localized in the plasma membrane. While much is known about the regulation of SOCE by STIM1, the role of stromal interaction molecule 2 (STIM2) in SOCE remains incompletely understood. Here, using clustered regularly interspaced short palindromic repeats -CRISPR associated protein 9 (CRISPR-Cas9) genomic editing and molecular imaging, we investigated the function of STIM2 in NIH 3T3 fibroblast and αT3 cell SOCE. We found that deletion of Stim2 expression reduced SOCE by more than 90% in NIH 3T3 cells. STIM1 expression levels were unaffected in the Stim2 null cells. However, quantitative confocal fluorescence imaging demonstrated that in the absence of Stim2 expression, STIM1 did not translocate or form punctae in plasma membrane-associated ER membrane (PAM) junctions following ER Ca2+ store depletion. Fluorescence resonance energy transfer (FRET) imaging of intact, living cells revealed that the formation of STIM1 and Orai1 complexes in PAM nanodomains was significantly reduced in the Stim2 knockout cells. Our findings indicate that STIM2 plays an essential role in regulating SOCE in NIH 3T3 and αT3 cells and suggests that dynamic interplay between STIM1 and STIM2 induced by ER Ca2+ store discharge is necessary for STIM1 translocation, its interaction with Orai1, and activation of SOCE.


Asunto(s)
Señalización del Calcio , Proteínas de Neoplasias/metabolismo , Molécula de Interacción Estromal 1/metabolismo , Molécula de Interacción Estromal 2/metabolismo , Células 3T3 , Animales , Calcio/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Microdominios de Membrana/metabolismo , Ratones , Proteínas de Neoplasias/genética , Proteína ORAI1/metabolismo , Unión Proteica , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 2/genética
11.
Sci Rep ; 8(1): 6192, 2018 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-29654321

RESUMEN

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

12.
Sci Rep ; 8(1): 3749, 2018 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-29491394

RESUMEN

We report the design and target validation of chimeric peptide EP45, a novel 45 amino acid monomeric dual agonist peptide that contains amino acid sequence motifs present within the blood glucose-lowering agent exendin-4 (Ex-4) and the appetite-suppressing agent PYY(3-36). In a new high-throughput FRET assay that provides real-time kinetic information concerning levels of cAMP in living cells, EP45 recapitulates the action of Ex-4 to stimulate cAMP production via the glucagon-like peptide-1 receptor (GLP-1R), while also recapitulating the action of PYY(3-36) to inhibit cAMP production via the neuropeptide Y2 receptor (NPY2R). EP45 fails to activate glucagon or GIP receptors, whereas for cells that co-express NPY2R and adenosine A2B receptors, EP45 acts in an NPY2R-mediated manner to suppress stimulatory effects of adenosine on cAMP production. Collectively, such findings are remarkable in that they suggest a new strategy in which the co-existing metabolic disorders of type 2 diabetes and obesity will be treatable using a single peptide such as EP45 that lowers levels of blood glucose by virtue of its GLP-1R-mediated effect, while simultaneously suppressing appetite by virtue of its NPY2R-mediated effect.


Asunto(s)
Péptido 1 Similar al Glucagón/agonistas , Péptidos/farmacología , Receptores de Neuropéptido Y/agonistas , Secuencia de Aminoácidos , Células HEK293 , Humanos , Péptidos/química
13.
J Biol Chem ; 292(6): 2266-2277, 2017 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-28003364

RESUMEN

Stromal interaction molecule 1 (STIM1) regulates store-operated Ca2+ entry (SOCE) and other ion channels either as an endoplasmic reticulum Ca2+-sensing protein or when present in the plasma membrane. However, the role of STIM1 in insulin-secreting ß-cells is unresolved. We report that lowering expression of STIM1, the gene that encodes STIM1, in insulin-secreting MIN6 ß-cells with RNA interference inhibits SOCE and ATP-sensitive K+ (KATP) channel activation. The effects of STIM1 knockdown were reversed by transduction of MIN6 cells with an adenovirus gene shuttle vector that expressed human STIM1 Immunoprecipitation studies revealed that STIM1 binds to nucleotide binding fold-1 (NBF1) of the sulfonylurea receptor 1 (SUR1) subunit of the KATP channel. Binding of STIM1 to SUR1 was enhanced by poly-lysine. Our data indicate that SOCE and KATP channel activity are regulated by STIM1. This suggests that STIM1 is a multifunctional signaling effector that participates in the control of membrane excitability and Ca2+ signaling events in ß-cells.


Asunto(s)
Canales de Calcio/fisiología , Islotes Pancreáticos/metabolismo , Canales KATP/fisiología , Proteínas de Neoplasias/fisiología , Molécula de Interacción Estromal 1/fisiología , Animales , Señalización del Calcio , Línea Celular , Técnicas de Silenciamiento del Gen , Humanos , Transporte Iónico , Ratones , Proteínas de Neoplasias/genética , Molécula de Interacción Estromal 1/genética
15.
Sci Rep ; 6: 28177, 2016 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-27378176

RESUMEN

Endoplasmic reticulum (ER) stress is among several pathological features that underlie ß-cell failure in the development of type 1 and type 2 diabetes. Adaptor proteins in the insulin/insulin-like-growth factor-1 signaling pathways, such as insulin receptor substrate-1 (IRS1) and IRS2, differentially impact ß-cell survival but the underlying mechanisms remain unclear. Here we report that ß-cells deficient in IRS1 (IRS1KO) are resistant, while IRS2 deficiency (IRS2KO) makes them susceptible to ER stress-mediated apoptosis. IRS1KOs exhibited low nuclear accumulation of spliced XBP-1 due to its poor stability, in contrast to elevated accumulation in IRS2KO. The reduced nuclear accumulation in IRS1KO was due to protein instability of Xbp1 secondary to proteasomal degradation. IRS1KO also demonstrated an attenuation in their general translation status in response to ER stress revealed by polyribosomal profiling. Phosphorylation of eEF2 was dramatically increased in IRS1KO enabling the ß-cells to adapt to ER stress by blocking translation. Furthermore, significantly high ER calcium (Ca(2+)) was detected in IRS1KO ß-cells even upon induction of ER stress. These observations suggest that IRS1 could be a therapeutic target for ß-cell protection against ER stress-mediated cell death by modulating XBP-1 stability, protein synthesis, and Ca(2+) storage in the ER.


Asunto(s)
Calcio/metabolismo , Proteínas Sustrato del Receptor de Insulina/deficiencia , Células Secretoras de Insulina/citología , Proteína 1 de Unión a la X-Box/química , Proteína 1 de Unión a la X-Box/metabolismo , Animales , Apoptosis , Núcleo Celular/metabolismo , Células Cultivadas , Estrés del Retículo Endoplásmico , Técnicas de Inactivación de Genes , Proteínas Sustrato del Receptor de Insulina/genética , Células Secretoras de Insulina/metabolismo , Ratones , Biosíntesis de Proteínas , Estabilidad Proteica
16.
Sci Rep ; 6: 28934, 2016 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-27352904

RESUMEN

Rational assembly of small molecule libraries for purposes of drug discovery requires an efficient approach in which the synthesis of bioactive compounds is enabled so that numerous structurally related compounds of a similar basic formulation can be derived. Here, we describe (4 + 3) and (3 + 2) indole annulation strategies that quickly generate complex indole heterocycle libraries that contain novel cyclohepta- and cyclopenta[b]indoles, respectively. Screening of one such library comprised of these indoles identifies JWU-A021 to be an especially potent stimulator of glucagon-like peptide-1 (GLP-1) secretion in vitro. Surprisingly, JWU-A021 is also a potent stimulator of Ca(2+) influx through TRPA1 cation channels (EC50 ca. 200 nM), thereby explaining its ability to stimulate GLP-1 release. Of additional importance, the available evidence indicates that JWU-A021 is one of the most potent non-electrophilic TRPA-1 channel agonists yet to be reported in the literature.


Asunto(s)
Péptido 1 Similar al Glucagón/metabolismo , Indoles/síntesis química , Animales , Señalización del Calcio/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Células Enteroendocrinas/efectos de los fármacos , Células Enteroendocrinas/metabolismo , Células HEK293 , Humanos , Indoles/farmacología , Ratones , Estereoisomerismo , Canal Catiónico TRPA1/agonistas
17.
Mol Endocrinol ; 30(6): 614-29, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27082897

RESUMEN

GPR119 is a G protein-coupled receptor expressed on intestinal L cells that synthesize and secrete the blood glucose-lowering hormone glucagon-like peptide-1 (GLP-1). GPR119 agonists stimulate the release of GLP-1 from L cells, and for this reason there is interest in their potential use as a new treatment for type 2 diabetes mellitus. AS1269574 is one such GPR119 agonist, and it is the prototype of a series of 2,4,6 trisubstituted pyrimidines that exert positive glucoregulatory actions in mice. Here we report the unexpected finding that AS1269574 stimulates GLP-1 release from the STC-1 intestinal cell line by directly promoting Ca(2+) influx through transient receptor potential ankyrin 1 (TRPA1) cation channels. These GPR119-independent actions of AS1269574 are inhibited by TRPA1 channel blockers (AP-18, A967079, HC030031) and are not secondary to intracellular Ca(2+) release or cAMP production. Patch clamp studies reveal that AS1269574 activates an outwardly rectifying membrane current with properties expected of TRPA1 channels. However, the TRPA1 channel-mediated action of AS1269574 to increase intracellular free calcium concentration is not replicated by GPR119 agonists (AR231453, oleoylethanolamide) unrelated in structure to AS1269574. Using human embryonic kidney-293 cells expressing recombinant rat TRPA1 channels but not GPR119, direct TRPA1 channel activating properties of AS1269574 are validated. Because we find that AS1269574 also acts in a conventional GPR119-mediated manner to stimulate proglucagon gene promoter activity in the GLUTag intestinal L cell line, new findings reported here reveal the surprising capacity of AS1269574 to act as a dual agonist at two molecular targets (GPR119/TRPA1) important to the control of L-cell function and type 2 diabetes mellitus drug discovery research.


Asunto(s)
Etanolaminas/farmacología , Péptido 1 Similar al Glucagón/metabolismo , Activación del Canal Iónico/efectos de los fármacos , Pirimidinas/farmacología , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Canal Catiónico TRPA1/metabolismo , Animales , Calcio/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Cimenos , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glucagón/metabolismo , Células HEK293 , Humanos , Isotiocianatos/farmacología , Luciferasas/metabolismo , Monoterpenos/farmacología , Proteínas Mutantes/metabolismo , Proglucagón/genética , Proglucagón/metabolismo , Ratas , Proteínas Recombinantes/farmacología , Transfección
18.
Mol Endocrinol ; 29(7): 988-1005, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26061564

RESUMEN

cAMP-elevating agents such as the incretin hormone glucagon-like peptide-1 potentiate glucose-stimulated insulin secretion (GSIS) from pancreatic ß-cells. However, a debate has existed since the 1970s concerning whether or not cAMP signaling is essential for glucose alone to stimulate insulin secretion. Here, we report that the first-phase kinetic component of GSIS is cAMP-dependent, as revealed through the use of a novel highly membrane permeable para-acetoxybenzyl (pAB) ester prodrug that is a bioactivatable derivative of the cAMP antagonist adenosine-3',5'-cyclic monophosphorothioate, Rp-isomer (Rp-cAMPS). In dynamic perifusion assays of human or rat islets, a step-wise increase of glucose concentration leads to biphasic insulin secretion, and under these conditions, 8-bromoadenosine-3',5'-cyclic monophosphorothioate, Rp-isomer, 4-acetoxybenzyl ester (Rp-8-Br-cAMPS-pAB) inhibits first-phase GSIS by up to 80%. Surprisingly, second-phase GSIS is inhibited to a much smaller extent (≤20%). Using luciferase, fluorescence resonance energy transfer, and bioluminescence resonance energy transfer assays performed in living cells, we validate that Rp-8-Br-cAMPS-pAB does in fact block cAMP-dependent protein kinase activation. Novel effects of Rp-8-Br-cAMPS-pAB to block the activation of cAMP-regulated guanine nucleotide exchange factors (Epac1, Epac2) are also validated using genetically encoded Epac biosensors, and are independently confirmed in an in vitro Rap1 activation assay using Rp-cAMPS and Rp-8-Br-cAMPS. Thus, in addition to revealing the cAMP dependence of first-phase GSIS from human and rat islets, these findings establish a pAB-based chemistry for the synthesis of highly membrane permeable prodrug derivatives of Rp-cAMPS that act with micromolar or even nanomolar potency to inhibit cAMP signaling in living cells.


Asunto(s)
8-Bromo Monofosfato de Adenosina Cíclica/análogos & derivados , AMP Cíclico/farmacología , Glucosa/farmacología , Insulina/metabolismo , Profármacos/farmacología , Tionucleótidos/farmacología , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Animales , Alcohol Bencilo/farmacología , Línea Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citosol/metabolismo , Activación Enzimática/efectos de los fármacos , Esterasas/metabolismo , Femenino , Transferencia Resonante de Energía de Fluorescencia , Regulación de la Expresión Génica/efectos de los fármacos , Factores de Intercambio de Guanina Nucleótido/metabolismo , Holoenzimas/metabolismo , Humanos , Secreción de Insulina , Integrasas/metabolismo , Luciferasas/metabolismo , Masculino , Persona de Mediana Edad , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
19.
Biochim Biophys Acta ; 1842(12 Pt B): 2593-600, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24980705

RESUMEN

Recently published findings indicate that a knockout (KO) of soluble adenylyl cyclase (sAC, also known as AC-10) gene expression in mice leads to defective glucoregulation that is characterized by reduced pancreatic insulin secretion and reduced intraperitoneal glucose tolerance. Summarized here are current concepts regarding the molecular basis for this phenotype, with special emphasis on the potential role of sAC as a determinant of glucose-stimulated insulin secretion. Highlighted is new evidence that in pancreatic beta cells, oxidative glucose metabolism stimulates mitochondrial CO2production that in turn generates bicarbonate ion (HCO(3)(-)). Since HCO(3)(-) binds to and directly stimulates the activity of sAC, we propose that glucose-stimulated cAMP production in beta cells is mediated not simply by transmembrane adenylyl cyclases (TMACs), but also by sAC. Based on evidence that sAC is expressed in mitochondria, there exists the possibility that beta-cell glucose metabolism is linked to mitochondrial cAMP production with consequent facilitation of oxidative phosphorylation. Since sAC is also expressed in the cytoplasm, sAC catalyzed cAMP production may activate cAMP sensors such as PKA and Epac2 to control ion channel function, intracellular Ca²âº handling, and Ca²âº-dependent exocytosis. Thus, we propose that the existence of sAC in beta cells provides a new and unexpected explanation for previously reported actions of glucose metabolism to stimulate cAMP production. It seems possible that alterations of sAC activity might be of importance when evaluating new strategies for the treatment of type 2 diabetes (T2DM), or when evaluating why glucose metabolism fails to stimulate insulin secretion in patients diagnosed with T2DM. This article is part of a Special Issue entitled: The role of soluble adenylyl cyclase in health and disease.


Asunto(s)
Adenilil Ciclasas/metabolismo , Glucosa/metabolismo , Adenilil Ciclasas/genética , Animales , Diabetes Mellitus Tipo 2/metabolismo , Insulina/metabolismo , Secreción de Insulina , Ratones , Ratones Noqueados
20.
J Fluoresc ; 24(2): 279-84, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24151033

RESUMEN

Resveratrol, a naturally occurring polyphenol found in some fruits and especially in grapes, has been reported to provide diverse health benefits. Resveratrol's mechanism of action is the subject of many investigations, and some studies using the ratiometric calcium indicator Fura-2 suggest that it modulates cellular calcium responses. In the current study, contradictory cellular calcium responses to resveratrol applied at concentrations exceeding 10 µM were observed during in vitro imaging studies depending on the calcium indicator used, with Fura-2 indicating an increase in intracellular calcium while Fluo-4 and the calcium biosensor YC3.60 indicated no response. When cells loaded with Fura-2 were treated with 100 µM resveratrol, excitation at 340 nm resulted in a large intensity increase at 510 nm, but the expected concurrent decline with 380 nm excitation was not observed. Pre-treatment of cells with the calcium chelator BAPTA-AM did not prevent a rise in the 340/380 ratio when resveratrol was present, but it did prevent an increase in 340/380 when ATP was applied, suggesting that the resveratrol response was an artifact. Cautious data interpretation is recommended from imaging experiments using Fura-2 concurrently with resveratrol in calcium imaging experiments.


Asunto(s)
Calcio/metabolismo , Fura-2/química , Estilbenos/química , Línea Celular Tumoral , Humanos , Resveratrol , Espectrometría de Fluorescencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...