Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 116(37): 18684-18690, 2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31451647

RESUMEN

Given the global epidemic in type 2 diabetes, novel antidiabetic drugs with increased efficacy and reduced side effects are urgently needed. Previous work has shown that M3 muscarinic acetylcholine (ACh) receptors (M3Rs) expressed by pancreatic ß cells play key roles in stimulating insulin secretion and maintaining physiological blood glucose levels. In the present study, we tested the hypothesis that a positive allosteric modulator (PAM) of M3R function can improve glucose homeostasis in mice by promoting insulin release. One major advantage of this approach is that allosteric agents respect the ACh-dependent spatiotemporal control of M3R activity. In this study, we first demonstrated that VU0119498, a drug known to act as a PAM at M3Rs, significantly augmented ACh-induced insulin release from cultured ß cells and mouse and human pancreatic islets. This stimulatory effect was absent in islets prepared from mice lacking M3Rs, indicative of the involvement of M3Rs. VU0119498 treatment of wild-type mice caused a significant increase in plasma insulin levels, accompanied by a striking improvement in glucose tolerance. These effects were mediated by ß-cell M3Rs, since they were absent in mutant mice selectively lacking M3Rs in ß cells. Moreover, acute VU0119498 treatment of obese, glucose-intolerant mice triggered enhanced insulin release and restored normal glucose tolerance. Interestingly, doses of VU0119498 that led to pronounced improvements in glucose homeostasis did not cause any significant side effects due to activation of M3Rs expressed by other peripheral cell types. Taken together, the data from this proof-of-concept study strongly suggest that M3R PAMs may become clinically useful as novel antidiabetic agents.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/farmacología , Islotes Pancreáticos/efectos de los fármacos , Agonistas Muscarínicos/farmacología , Receptor Muscarínico M3/efectos de los fármacos , Acetilcolina/metabolismo , Adulto , Regulación Alostérica/efectos de los fármacos , Animales , Glucemia/análisis , Glucemia/metabolismo , Línea Celular Tumoral , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Femenino , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/tratamiento farmacológico , Intolerancia a la Glucosa/metabolismo , Humanos , Hipoglucemiantes/uso terapéutico , Secreción de Insulina/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Obesos , Ratones Transgénicos , Persona de Mediana Edad , Agonistas Muscarínicos/uso terapéutico , Obesidad/sangre , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Cultivo Primario de Células , Prueba de Estudio Conceptual , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Adulto Joven
2.
Proc Natl Acad Sci U S A ; 112(49): E6818-24, 2015 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-26598688

RESUMEN

G protein-coupled receptors (GPCRs) regulate virtually all physiological functions including the release of insulin from pancreatic ß-cells. ß-Cell M3 muscarinic receptors (M3Rs) are known to play an essential role in facilitating insulin release and maintaining proper whole-body glucose homeostasis. As is the case with other GPCRs, M3R activity is regulated by phosphorylation by various kinases, including GPCR kinases and casein kinase 2 (CK2). At present, it remains unknown which of these various kinases are physiologically relevant for the regulation of ß-cell activity. In the present study, we demonstrate that inhibition of CK2 in pancreatic ß-cells, knockdown of CK2α expression, or genetic deletion of CK2α in ß-cells of mutant mice selectively augmented M3R-stimulated insulin release in vitro and in vivo. In vitro studies showed that this effect was associated with an M3R-mediated increase in intracellular calcium levels. Treatment of mouse pancreatic islets with CX4945, a highly selective CK2 inhibitor, greatly reduced agonist-induced phosphorylation of ß-cell M3Rs, indicative of CK2-mediated M3R phosphorylation. We also showed that inhibition of CK2 greatly enhanced M3R-stimulated insulin secretion in human islets. Finally, CX4945 treatment protected mice against diet-induced hyperglycemia and glucose intolerance in an M3R-dependent fashion. Our data demonstrate, for the first time to our knowledge, the physiological relevance of CK2 phosphorylation of a GPCR and suggest the novel concept that kinases acting on ß-cell GPCRs may represent novel therapeutic targets.


Asunto(s)
Quinasa de la Caseína II/fisiología , Insulina/metabolismo , Receptor Muscarínico M3/fisiología , Animales , Células COS , Chlorocebus aethiops , Femenino , Células HEK293 , Humanos , Secreción de Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Naftiridinas/farmacología , Fenazinas
3.
Cell Biochem Funct ; 33(2): 67-72, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25663655

RESUMEN

Adenylate kinase plays an important role in cellular energy homeostasis by catalysing the interconversion of adenine nucleotides. The goal of present study was to evaluate the contribution of the adenylate kinase reaction to oxidative ATP synthesis by direct measurements of ATP using (31) P NMR spectroscopy. Results show that AMP can stimulate ATP synthesis in the presence or absence of ADP. In particular, addition of 1 mM AMP to the 0.6 mM ADP superfusion system of isolated superfused mitochondria (contained and maintained in agarose beads) led to a 25% increase in ATP synthesis as measured by the increase in ßATP signal. More importantly, we show that AMP can support ATP synthesis in the absence of ADP, demonstrated as follows. Superfusion of mitochondria without ADP led to the disappearance of ATP γ, α and ß signals and the increase of Pi . Addition of AMP to the medium restored the production of ATP, as demonstrated by the reappearance of γ, α and ß ATP signals, in conjunction with a decrease in Pi , which is being used for ATP synthesis. Polarographic studies showed Mg(2+) dependence of this process, confirming the specificity of the adenylate kinase reaction. Furthermore, data obtained from this study demonstrate, for the first time, that different aspects of the adenylate kinase reaction can be evaluated with (31) P NMR spectroscopy. SIGNIFICANCE OF RESEARCH PARAGRAPH: The data generated in the present study indicate that (31) P NMR spectroscopy can effectively be used to study the adenylate kinase reaction under a variety of conditions. This is important because understanding of adenylate kinase function and/or malfunction is essential to understanding its role in health and disease. The data obtained with (31) P NMR were confirmed by polarographic studies, which further strengthens the robustness of the NMR findings. In summary, (31) P NMR spectroscopy provides a sensitive tool to study adenylate kinase activity in different physiological and pathophysiological conditions, including but not exclusive of, cancer, ischemic injury, hemolytic anemia and neurological problems such as sensorineural deafness.


Asunto(s)
Adenosina Monofosfato/metabolismo , Adenosina Trifosfato/biosíntesis , Adenilato Quinasa/metabolismo , Mitocondrias/metabolismo , Miocardio/metabolismo , Consumo de Oxígeno , Animales , Espectroscopía de Resonancia Magnética/métodos , Polarografía/métodos , Ratas
4.
J Biol Chem ; 288(6): 3938-51, 2013 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-23266825

RESUMEN

Paracrine signaling between pancreatic islet ß-cells and α-cells has been proposed to play a role in regulating glucagon responses to elevated glucose and hypoglycemia. To examine this possibility in human islets, we used a metabolomic approach to trace the responses of amino acids and other potential neurotransmitters to stimulation with [U-(13)C]glucose in both normal individuals and type 2 diabetics. Islets from type 2 diabetics uniformly showed decreased glucose stimulation of insulin secretion and respiratory rate but demonstrated two different patterns of glucagon responses to glucose: one group responded normally to suppression of glucagon by glucose, but the second group was non-responsive. The non-responsive group showed evidence of suppressed islet GABA levels and of GABA shunt activity. In further studies with normal human islets, we found that γ-hydroxybutyrate (GHB), a potent inhibitory neurotransmitter, is generated in ß-cells by an extension of the GABA shunt during glucose stimulation and interacts with α-cell GHB receptors, thus mediating the suppressive effect of glucose on glucagon release. We also identified glycine, acting via α-cell glycine receptors, as the predominant amino acid stimulator of glucagon release. The results suggest that glycine and GHB provide a counterbalancing receptor-based mechanism for controlling α-cell secretory responses to metabolic fuels.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Glucagón/metabolismo , Glucagón/metabolismo , Glucosa/metabolismo , Glicina/metabolismo , Células Secretoras de Insulina/metabolismo , Oxibato de Sodio/metabolismo , Adulto , Diabetes Mellitus Tipo 2/patología , Femenino , Células Secretoras de Glucagón/patología , Humanos , Células Secretoras de Insulina/patología , Masculino , Persona de Mediana Edad , Receptores de GABA/metabolismo , Receptores de Glicina/metabolismo , Ácido gamma-Aminobutírico/metabolismo
5.
Nat Commun ; 15(1): 5129, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38879678

RESUMEN

Glucagon, a hormone released from pancreatic α-cells, is critical for maintaining euglycemia and plays a key role in the pathophysiology of diabetes. To stimulate the development of new classes of therapeutic agents targeting glucagon release, key α-cell signaling pathways that regulate glucagon secretion need to be identified. Here, we focused on the potential importance of α-cell Gs signaling on modulating α-cell function. Studies with α-cell-specific mouse models showed that activation of α-cell Gs signaling causes a marked increase in glucagon secretion. We also found that intra-islet adenosine plays an unexpected autocrine/paracrine role in promoting glucagon release via activation of α-cell Gs-coupled A2A adenosine receptors. Studies with α-cell-specific Gαs knockout mice showed that α-cell Gs also plays an essential role in stimulating the activity of the Gcg gene, thus ensuring proper islet glucagon content. Our data suggest that α-cell enriched Gs-coupled receptors represent potential targets for modulating α-cell function for therapeutic purposes.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs , Células Secretoras de Glucagón , Glucagón , Ratones Noqueados , Transducción de Señal , Glucagón/metabolismo , Animales , Células Secretoras de Glucagón/metabolismo , Ratones , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Adenosina/metabolismo , Receptor de Adenosina A2A/metabolismo , Receptor de Adenosina A2A/genética , Masculino , Ratones Endogámicos C57BL , Islotes Pancreáticos/metabolismo
6.
J Clin Invest ; 134(12)2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38950317

RESUMEN

Glucose plays a key role in shaping pancreatic ß cell function. Thus, deciphering the mechanisms by which this nutrient stimulates ß cells holds therapeutic promise for combating ß cell failure in type 2 diabetes (T2D). ß Cells respond to hyperglycemia in part by rewiring their mRNA metabolism, yet the mechanisms governing these changes remain poorly understood. Here, we identify a requirement for the RNA-binding protein PCBP2 in maintaining ß cell function basally and during sustained hyperglycemic challenge. PCBP2 was induced in primary mouse islets incubated with elevated glucose and was required to adapt insulin secretion. Transcriptomic analysis of primary Pcbp2-deficient ß cells revealed impacts on basal and glucose-regulated mRNAs encoding core components of the insulin secretory pathway. Accordingly, Pcbp2-deficient ß cells exhibited defects in calcium flux, insulin granule ultrastructure and exocytosis, and the amplification pathway of insulin secretion. Further, PCBP2 was induced by glucose in primary human islets, was downregulated in islets from T2D donors, and impacted genes commonly altered in islets from donors with T2D and linked to single-nucleotide polymorphisms associated with T2D. Thus, these findings establish a paradigm for PCBP2 in governing basal and glucose-adaptive gene programs critical for shaping the functional state of ß cells.


Asunto(s)
Diabetes Mellitus Tipo 2 , Glucosa , Células Secretoras de Insulina , Insulina , Proteínas de Unión al ARN , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Animales , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Ratones , Humanos , Glucosa/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Insulina/metabolismo , Secreción de Insulina , Ratones Noqueados , Masculino , Adaptación Fisiológica
7.
Diabetes ; 73(4): 554-564, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38266068

RESUMEN

Assessment of pancreas cell type composition is crucial to the understanding of the genesis of diabetes. Current approaches use immunodetection of protein markers, for example, insulin as a marker of ß-cells. A major limitation of these methods is that protein content varies in physiological and pathological conditions, complicating the extrapolation to actual cell number. Here, we demonstrate the use of cell type-specific DNA methylation markers for determining the fraction of specific cell types in human islet and pancreas specimens. We identified genomic loci that are uniquely demethylated in specific pancreatic cell types and applied targeted PCR to assess the methylation status of these loci in tissue samples, enabling inference of cell type composition. In islet preparations, normalization of insulin secretion to ß-cell DNA revealed similar ß-cell function in pre-type 1 diabetes (T1D), T1D, and type 2 diabetes (T2D), which was significantly lower than in donors without diabetes. In histological pancreas specimens from recent-onset T1D, this assay showed ß-cell fraction within the normal range, suggesting a significant contribution of ß-cell dysfunction. In T2D pancreata, we observed increased α-cell fraction and normal ß-cell fraction. Methylation-based analysis provides an accurate molecular alternative to immune detection of cell types in the human pancreas, with utility in the interpretation of insulin secretion assays and the assessment of pancreas cell composition in health and disease.


Asunto(s)
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Células Secretoras de Glucagón , Células Secretoras de Insulina , Islotes Pancreáticos , Humanos , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Islotes Pancreáticos/metabolismo , Metilación de ADN , Páncreas/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Glucagón/metabolismo
8.
Dis Model Mech ; 16(5)2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36107856

RESUMEN

Friedreich ataxia, the most common hereditary ataxia, is a neuro- and cardio-degenerative disorder caused, in most cases, by decreased expression of the mitochondrial protein frataxin. Cardiomyopathy is the leading cause of premature death. Frataxin functions in the biogenesis of iron-sulfur clusters, which are prosthetic groups that are found in proteins involved in many biological processes. To study the changes associated with decreased frataxin in human cardiomyocytes, we developed a novel isogenic model by acutely knocking down frataxin, post-differentiation, in cardiomyocytes derived from induced pluripotent stem cells (iPSCs). Transcriptome analysis of four biological replicates identified severe mitochondrial dysfunction and a type I interferon response as the pathways most affected by frataxin knockdown. We confirmed that, in iPSC-derived cardiomyocytes, loss of frataxin leads to mitochondrial dysfunction. The type I interferon response was activated in multiple cell types following acute frataxin knockdown and was caused, at least in part, by release of mitochondrial DNA into the cytosol, activating the cGAS-STING sensor pathway.


Asunto(s)
Ataxia de Friedreich , Células Madre Pluripotentes Inducidas , Interferón Tipo I , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Interferón Tipo I/metabolismo , Proteínas de Unión a Hierro/genética , Proteínas de Unión a Hierro/metabolismo , Ataxia de Friedreich/genética , Ataxia de Friedreich/metabolismo , Proteínas Mitocondriales/metabolismo , Hierro/metabolismo , ADN Mitocondrial/metabolismo , Nucleotidiltransferasas/metabolismo , Azufre/metabolismo , Frataxina
9.
Diabetes ; 72(12): 1809-1819, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37725835

RESUMEN

The importance of glucokinase (GK) in the regulation of insulin secretion has been highlighted by the phenotypes of individuals with activating and inactivating mutations in the glucokinase gene (GCK). Here we report 10 individuals with congenital hyperinsulinism (HI) caused by eight unique activating mutations of GCK. Six are novel and located near previously identified activating mutations sites. The first recognized episode of hypoglycemia in these patients occurred between birth and 24 years, and the severity of the phenotype was also variable. Mutant enzymes were expressed and purified for enzyme kinetics in vitro. Mutant enzymes had low glucose half-saturation concentration values and an increased enzyme activity index compared with wild-type GK. We performed functional evaluation of islets from the pancreata of three children with GCK-HI who required pancreatectomy. Basal insulin secretion in perifused GCK-HI islets was normal, and the response to glyburide was preserved. However, the threshold for glucose-stimulated insulin secretion in perifused glucokinase hyperinsulinism (GCK-HI) islets was decreased, and glucagon secretion was greatly suppressed. Our evaluation of novel GCK disease-associated mutations revealed that the detrimental effects of these mutations on glucose homeostasis can be attributed not only to a lowering of the glucose threshold of insulin secretion but also to a decreased counterregulatory glucagon secretory response. ARTICLE HIGHLIGHTS: Our evaluation of six novel and two previously published activating GCK mutations revealed that the detrimental effects of these mutations on glucose homeostasis can be attributed not only to a lowering of the glucose threshold of insulin secretion but also to a decreased counterregulatory glucagon secretory response. These studies provide insights into the pathophysiology of GCK-hyperinsulinism and the dual role of glucokinase in ß-cells and α-cells to regulate glucose homeostasis.


Asunto(s)
Hiperinsulinismo Congénito , Hiperinsulinismo , Niño , Humanos , Glucoquinasa/genética , Glucagón , Hiperinsulinismo Congénito/genética , Hiperinsulinismo/genética , Glucosa , Mutación , Fenotipo
10.
Cell Metab ; 6(4): 267-79, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17908556

RESUMEN

The winged-helix transcription factor Foxa2 regulates Pdx1 gene expression and fetal endocrine pancreas development. We show here by inducible gene ablation that Foxa2 inactivation in mature beta cells induces hyperinsulinemic hypoglycemia in Foxa2(loxP/loxP),Pdx1-CreERT2 adult mice. Mutant beta cells exhibited a markedly increased pool of docked insulin granules, some of which were engaged in sequential or compound exocytosis, consistent with increased first-phase glucose-stimulated insulin secretion. Expression of multiple genes involved in vesicular trafficking, membrane targeting, and fuel-secretion pathways is dependent on Foxa2. In addition, impaired cytosolic Ca(2+) oscillations and elevated intracellular cyclic AMP production accompanied this secretory defect and were likely contributors to the sensitization of the exocytotic machinery. Thus, in the absence of Foxa2, alterations in intracellular second-messenger signaling redistribute the insulin granules into the readily releasable pool. We conclude that Foxa2 is required for both fetal pancreas development and the function of mature beta cells.


Asunto(s)
Factor Nuclear 3-beta del Hepatocito/fisiología , Hiperinsulinismo/genética , Hipoglucemia/genética , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Animales , Señalización del Calcio/genética , Eliminación de Gen , Factor Nuclear 3-beta del Hepatocito/deficiencia , Factor Nuclear 3-beta del Hepatocito/genética , Secreción de Insulina , Células Secretoras de Insulina/ultraestructura , Ratones , Vesículas Secretoras/metabolismo , Vesículas Secretoras/ultraestructura
11.
Am J Physiol Endocrinol Metab ; 302(1): E87-E102, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21952036

RESUMEN

It was reported previously that isolated human islets from individuals with type 2 diabetes mellitus (T2DM) show reduced glucose-stimulated insulin release. To assess the possibility that impaired bioenergetics may contribute to this defect, glucose-stimulated respiration (Vo(2)), glucose usage and oxidation, intracellular Ca(2+), and insulin secretion (IS) were measured in pancreatic islets isolated from three healthy and three type 2 diabetic organ donors. Isolated mouse and rat islets were studied for comparison. Islets were exposed to a "staircase" glucose stimulus, whereas IR and Vo(2) were measured. Vo(2) of human islets from normals and diabetics increased sigmoidally from equal baselines of 0.25 nmol/100 islets/min as a function of glucose concentration. Maximal Vo(2) of normal islets at 24 mM glucose was 0.40 ± 0.02 nmol·min(-1)·100 islets(-1), and the glucose S(0.5) was 4.39 ± 0.10 mM. The glucose stimulation of respiration of islets from diabetics was lower, V(max) of 0.32 ± 0.01 nmol·min(-1)·100 islets(-1), and the S(0.5) shifted to 5.43 ± 0.13 mM. Glucose-stimulated IS and the rise of intracellular Ca(2+) were also reduced in diabetic islets. A clinically effective glucokinase activator normalized the defective Vo(2), IR, and free calcium responses during glucose stimulation in islets from type 2 diabetics. The body of data shows that there is a clear relationship between the pancreatic islet energy (ATP) production rate and IS. This relationship was similar for normal human, mouse, and rat islets and the data for all species fitted a single sigmoidal curve. The shared threshold rate for IS was ∼13 pmol·min(-1)·islet(-1). Exendin-4, a GLP-1 analog, shifted the ATP production-IS curve to the left and greatly potentiated IS with an ATP production rate threshold of ∼10 pmol·min(-1)·islet(-1). Our data suggest that impaired ß-cell bioenergetics resulting in greatly reduced ATP production is critical in the molecular pathogenesis of type 2 diabetes mellitus.


Asunto(s)
Bencenoacetamidas/farmacología , Diabetes Mellitus Tipo 2/metabolismo , Activadores de Enzimas/farmacología , Glucoquinasa/metabolismo , Glucosa/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Adulto , Animales , Señalización del Calcio/efectos de los fármacos , Respiración de la Célula/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Exenatida , Femenino , Péptido 1 Similar al Glucagón/análogos & derivados , Glucoquinasa/química , Glucólisis/efectos de los fármacos , Humanos , Hipoglucemiantes/farmacología , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Fosforilación Oxidativa/efectos de los fármacos , Péptidos/farmacología , Ratas , Especificidad de la Especie , Técnicas de Cultivo de Tejidos , Ponzoñas/farmacología
12.
Diabetes Obes Metab ; 14 Suppl 3: 109-19, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22928571

RESUMEN

Glucokinase activators (GKAs) are being developed and clinically tested for potential antidiabetic therapy. The potential benefits and limitations of this approach continue to be intensively debated. To contribute to the understanding of experimental pharmacology and therapeutics of GKAs, we have tested the efficacy of one of these agents (Piragliatin) in isolated islets from humans with type 2 diabetes mellitus (T2DM), from mice with glucokinase (GK) mutations induced by ethyl-nitroso-urea (ENU) as models of Maturity Onset Diabetes of the Young linked to GK and Permanent Neonatal Diabetes Mellitus linked to GK (PNDM-GK) and finally of islets rendered glucose insensitive by treatment with the sulphonyl urea compound glyburide in organ culture. We found that the GKA repaired the defect in all three instances as manifest in increased glucose-induced insulin release and elevated intracellular calcium responses. The results show the remarkable fact that acute pharmacological activation of GK reverses secretion defects of ß-cells caused by molecular mechanism that differ vastly in nature, including the little understood multifactorial lesion of ß-cells in T2DM of man, the complex GK mutations in mice resembling GK disease and acute sulphonylurea failure of mouse ß-cells in tissue culture. The implications of these results are to be discussed on the theoretical basis underpinning the strategy of developing these drugs and in light of recent results of clinical trials with GKAs that failed for little understood reasons.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Activadores de Enzimas/farmacología , Glucoquinasa/metabolismo , Hipoglucemiantes/farmacología , Resistencia a la Insulina , Células Secretoras de Insulina/metabolismo , Animales , Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/genética , Modelos Animales de Enfermedad , Activación Enzimática , Prueba de Tolerancia a la Glucosa , Humanos , Resistencia a la Insulina/genética , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Ratones Mutantes , Fenotipo
13.
Life Sci ; 309: 120952, 2022 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-36100080

RESUMEN

AIMS: Glucokinase (GK) is expressed in the glucose-sensing cells of the islets of Langerhans and plays a critical role in glucose homeostasis. Here, we tested the hypothesis that genetic activation of GK in a small subset of ß-cells is sufficient to change the glucose set-point of the whole islet. MATERIAL AND METHODS: Mouse models of cell-type specific GK deficiency (GKKO) and genetic enzyme activation (GKKI) in a subset of ß-cells were obtained by crossing the αGSU (gonadotropin alpha subunit)-Cre transgene with the appropriate GK mutant alleles. Metabolic analyses consisted of glucose tolerance tests, perifusion of isolated islets and intracellular calcium measurements. KEY FINDINGS: The αGSU-Cre transgene produced genetically mosaic islets, as Cre was active in 15 ± 1.2 % of ß-cells. While mice deficient for GK in a subset of islet cells were normal, unexpectedly, GKKI mice were chronically hypoglycemic, glucose intolerant, and had a lower threshold for glucose stimulated insulin secretion. GKKI mice exhibited an average fasting blood glucose level of 3.5 mM. GKKI islets responded with intracellular calcium signals that spread through the whole islets at 1 mM and secreted insulin at 3 mM glucose. SIGNIFICANCE: Genetic activation of GK in a minority of ß-cells is sufficient to change the glucose threshold for insulin secretion in the entire islet and thereby glucose homeostasis in the whole animal. These data support the model in which ß-cells with higher GK activity function as 'hub' or 'trigger' cells and thus control insulin secretion by the ß-cell collective within the islet.


Asunto(s)
Hipoglucemia , Células Secretoras de Insulina , Islotes Pancreáticos , Ratones , Animales , Glucoquinasa/genética , Glucoquinasa/metabolismo , Células Secretoras de Insulina/metabolismo , Glucemia/metabolismo , Calcio/metabolismo , Islotes Pancreáticos/metabolismo , Insulina/metabolismo , Glucosa/metabolismo , Hipoglucemia/metabolismo , Hipoglucemiantes/metabolismo
14.
Redox Biol ; 56: 102457, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36063729

RESUMEN

We previously reported a depletion of murine regenerating islet-derived protein 2 (REG2) in pancreatic islets of glutathione peroxidase-1 (Gpx1) overexpressing (OE) mice. The present study was to explore if and how the REG2 depletion contributed to an augmented glucose stimulated insulin secretion (GSIS) in OE islets. After we verified a consistent depletion (90%, p < 0.05) of REG2 mRNA, transcript, and protein in OE islets compared with wild-type (WT) controls, we treated cultured and perifused OE islets (70 islets/sample) with REG2 (1 µg/ml or ml · min) and observed 30-40% (p < 0.05) inhibitions of GSIS by REG2. Subsequently, we obtained evidences of co-immunoprecipitation, cell surface ligand binding, and co-immunofluorescence for a ligand-receptor binding between REG2 and transmembrane, L-type voltage-dependent Ca2+ channel (CaV1.2) in beta TC3 cells. Mutating the C-type lectin binding domain of REG2 or deglycosylating CaV1.2 removed the inhibition of REG2 on GSIS and(or) the putative binding between the two proteins. Treating cultured OE and perifused WT islets with REG2 (1 µg/ml or ml · min) decreased (p < 0.05) Ca2+ influx triggered by glucose or KCl. An intraperitoneal (ip) injection of REG2 (2 µg/g) to OE mice (6-month old, n = 10) decreased their plasma insulin concentration (46%, p < 0.05) and elevated their plasma glucose concentration (25%, p < 0.05) over a 60 min period after glucose challenge (ip, 1 g/kg). In conclusion, our study identifies REG2 as a novel regulator of Ca2+ influx and insulin secretion, and reveals a new cascade of GPX1/REG2/CaV1.2 to explain how REG2 depletion in OE islets could decrease its binding to CaV1.2, resulting in uninhibited Ca2+ influx and augmented GSIS. These findings create new links to bridge redox biology, tissue regeneration, and insulin secretion.


Asunto(s)
Células Secretoras de Insulina , Islotes Pancreáticos , Animales , Glucemia/metabolismo , Glucosa/metabolismo , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ligandos , Ratones , Proteínas Asociadas a Pancreatitis/metabolismo , ARN Mensajero/metabolismo , Glutatión Peroxidasa GPX1
15.
J Clin Invest ; 132(11)2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35642629

RESUMEN

BACKGROUNDMultiple islet autoantibodies (AAbs) predict the development of type 1 diabetes (T1D) and hyperglycemia within 10 years. By contrast, T1D develops in only approximately 15% of individuals who are positive for single AAbs (generally against glutamic acid decarboxylase [GADA]); hence, the single GADA+ state may represent an early stage of T1D.METHODSHere, we functionally, histologically, and molecularly phenotyped human islets from nondiabetic GADA+ and T1D donors.RESULTSSimilar to the few remaining ß cells in the T1D islets, GADA+ donor islets demonstrated a preserved insulin secretory response. By contrast, α cell glucagon secretion was dysregulated in both GADA+ and T1D islets, with impaired glucose suppression of glucagon secretion. Single-cell RNA-Seq of GADA+ α cells revealed distinct abnormalities in glycolysis and oxidative phosphorylation pathways and a marked downregulation of cAMP-dependent protein kinase inhibitor ß (PKIB), providing a molecular basis for the loss of glucose suppression and the increased effect of 3-isobutyl-1-methylxanthine (IBMX) observed in GADA+ donor islets.CONCLUSIONWe found that α cell dysfunction was present during the early stages of islet autoimmunity at a time when ß cell mass was still normal, raising important questions about the role of early α cell dysfunction in the progression of T1D.FUNDINGThis work was supported by grants from the NIH (3UC4DK112217-01S1, U01DK123594-02, UC4DK112217, UC4DK112232, U01DK123716, and P30 DK019525) and the Vanderbilt Diabetes Research and Training Center (DK20593).


Asunto(s)
Diabetes Mellitus Tipo 1 , Glutamato Descarboxilasa , Autoanticuerpos , Glucagón , Glucosa , Humanos
16.
Handb Exp Pharmacol ; (203): 357-401, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21484579

RESUMEN

Glucokinase Glucokinase (GK GK ; EC 2.7.1.1.) phosphorylates and regulates glucose metabolism in insulin-producing pancreatic beta-cells, hepatocytes, and certain cells of the endocrine and nervous systems allowing it to play a central role in glucose homeostasis glucose homeostasis . Most importantly, it serves as glucose sensor glucose sensor in pancreatic beta-cells mediating glucose-stimulated insulin biosynthesis and release and it governs the capacity of the liver to convert glucose to glycogen. Activating and inactivating mutations of the glucokinase gene cause autosomal dominant hyperinsulinemic hypoglycemia and hypoinsulinemic hyperglycemia in humans, respectively, illustrating the preeminent role of glucokinase in the regulation of blood glucose and also identifying the enzyme as a potential target for developing antidiabetic drugs antidiabetic drugs . Small molecules called glucokinase activators (GKAs) glucokinase activators (GKAs) which bind to an allosteric activator allosteric activator site of the enzyme have indeed been discovered and hold great promise as new antidiabetic agents. GKAs increase the enzyme's affinity for glucose and also its maximal catalytic rate. Consequently, they stimulate insulin biosynthesis and secretion, enhance hepatic glucose uptake, and augment glucose metabolism and related processes in other glucokinase-expressing cells. Manifestations of these effects, most prominently a lowering of blood glucose, are observed in normal laboratory animals and man but also in animal models of diabetes and patients with type 2 diabetes mellitus (T2DM T2DM ) type 2 diabetes mellitus (T2DM) . These compelling concepts and results sustain a strong R&D effort by many pharmaceutical companies to generate GKAs with characteristics allowing for a novel drug treatment of T2DM.


Asunto(s)
Diabetes Mellitus/tratamiento farmacológico , Activadores de Enzimas/farmacología , Glucoquinasa/metabolismo , Hipoglucemiantes/uso terapéutico , Animales , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Activadores de Enzimas/uso terapéutico , Homeostasis/efectos de los fármacos , Humanos , Hiperinsulinismo/tratamiento farmacológico , Hipoglucemiantes/farmacología , Receptores de Droga/efectos de los fármacos
17.
JCI Insight ; 6(24)2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34752420

RESUMEN

Glucagon, a hormone released from pancreatic α cells, plays a key role in maintaining euglycemia. New insights into the signaling pathways that control glucagon secretion may stimulate the development of novel therapeutic agents. In this study, we investigated the potential regulation of α cell function by G proteins of the Gq family. The use of a chemogenetic strategy allowed us to selectively activate Gq signaling in mouse α cells in vitro and in vivo. Acute stimulation of α cell Gq signaling led to elevated plasma glucagon levels, accompanied by increased insulin release and improved glucose tolerance. Moreover, chronic activation of this pathway greatly improved glucose tolerance in obese mice. We also identified an endogenous Gq-coupled receptor (vasopressin 1b receptor; V1bR) that was enriched in mouse and human α cells. Agonist-induced activation of the V1bR strongly stimulated glucagon release in a Gq-dependent fashion. In vivo studies indicated that V1bR-mediated glucagon release played a key role in the counterregulatory hyperglucagonemia under hypoglycemic and glucopenic conditions. These data indicate that α cell Gq signaling represents an important regulator of glucagon secretion, resulting in multiple beneficial metabolic effects. Thus, drugs that target α cell-enriched Gq-coupled receptors may prove useful to restore euglycemia in various pathophysiological conditions.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Hipoglucemiantes/metabolismo , Transducción de Señal/inmunología , Animales , Humanos , Masculino , Ratones
18.
Am J Physiol Endocrinol Metab ; 299(3): E475-85, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20606076

RESUMEN

Fatty acids, acetylcholine, and GLP-1 enhance insulin secretion in a glucose-dependent manner. However, the interplay between glucose, fatty acids, and the neuroendocrine regulators of insulin secretion is not well understood. Therefore, we studied the acute effects of PA (alone or in combination with glucose, acetylcholine, or GLP-1) on isolated cultured mouse islets. Two different sets of experiments were designed. In one, a fixed concentration of 0.5 mM of PA bound to 0.15 mM BSA was used; in the other, a PA ramp from 0 to 0.5 mM was applied at a fixed albumin concentration of 0.15 mM so that the molar PA/BSA ratio changed within the physiological range. At a fixed concentration of 0.5 mM, PA markedly inhibited acetylcholine-stimulated insulin release, the rise of intracellular Ca(2+), and enhancement of cAMP production but did not influence the effects of GLP-1 on these parameters of islet cell function. 2-ADB, an IP(3) receptor inhibitor, reduced the effect of acetylcholine on insulin secretion and reversed the effect of PA on acetylcholine-stimulated insulin release. Islet perfusion for 35-40 min with 0.5 mM PA significantly reduced the calcium storage capacity of ER measured by the thapsigargin-induced Ca(2+) release. Oxygen consumption due to low but not high glucose was reduced by PA. When a PA ramp from 0 to 0.5 mM was applied in the presence of 8 mM glucose, PA at concentrations as low as 50 microM significantly augmented glucose-stimulated insulin release and markedly reduced acetylcholine's effects on hormone secretion. We thus demonstrate that PA acutely reduces the total oxygen consumption response to glucose, glucose-dependent acetylcholine stimulation of insulin release, Ca(2+), and cAMP metabolism, whereas GLP-1's actions on these parameters remain unaffected or potentiated. We speculate that acute emptying of the ER calcium by PA results in decreased glucose stimulation of respiration and acetylcholine potentiation of insulin secretion.


Asunto(s)
Acetilcolina/antagonistas & inhibidores , Péptido 1 Similar al Glucagón/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/fisiología , Ácido Palmítico/farmacología , Acetilcolina/metabolismo , Animales , Calcio/metabolismo , AMP Cíclico/metabolismo , Técnicas In Vitro , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Consumo de Oxígeno/fisiología
19.
Int Immunopharmacol ; 81: 106267, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32044662

RESUMEN

A key feature of type 2 diabetes (T2D) is that beta-cells of the pancreatic islets fail to release sufficient amounts of insulin to overcome peripheral insulin resistance. Glucose-stimulated insulin secretion (GSIS) is regulated by the activity of numerous neurotransmitters, hormones and paracrine factors that act by stimulating specific G protein-coupled receptors expressed by pancreatic beta-cells. Studies with both mouse and human islets suggest that acetylcholine (ACh) acts on beta-cell M3 muscarinic receptors (M3Rs) to promote GSIS. In mouse islets, beta-cell M3Rs are thought to be activated by ACh released from parasympathetic nerve endings. Interestingly, studies with human pancreatic islets suggest that ACh is synthesized, stored and released by alpha-cells, which, in human pancreatic islets, are intermingled with beta-cells. Independent of the source of pancreatic islet ACh, recent studies indicate that beta-cell M3Rs represent a potential target for drugs capable of promoting insulin release for therapeutic purposes. In this review, we will provide an overview about signaling pathways and molecules that regulate the activity of beta-cell M3Rs. We will also discuss a novel pharmacological strategy to stimulate the activity of these receptors to reduce the metabolic impairments associated with T2D.


Asunto(s)
Acetilcolina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Receptor Muscarínico M3/metabolismo , Receptores Colinérgicos/metabolismo , Regulación Alostérica , Animales , Diabetes Mellitus Tipo 2/terapia , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/patología , Terapia Molecular Dirigida
20.
Mol Metab ; 26: 45-56, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31178390

RESUMEN

OBJECTIVE: Pancreatic ß cell failure plays a central role in the development of type 2 diabetes (T2D). While the transcription factors shaping the ß cell gene expression program have received much attention, the post-transcriptional controls that are activated in ß cells during stress are largely unknown. We recently identified JUND as a pro-oxidant transcription factor that is post-transcriptionally upregulated in ß cells during metabolic stress. Here we seek to uncover the mechanisms underlying this maladaptive response to metabolic stress. METHODS: RNA-protein and protein-protein interactions were measured using RNA immunoprecipitation and co-immunoprecipitation, respectively, in Min6 cells and mouse islets. Phos-tag analyses were used to assess hnRNPK phosphorylation in primary mouse and human islets and Min6 cells. Translating ribosome affinity purification (TRAP) followed by RT-qPCR was used to identify changes in the ribosome occupancy of mRNAs in Min6 cells. Gene depletion studies used lentiviral delivery of CRISPR-Cas9 to Min6 cells. Apoptosis was measured in primary islets using a cell-permeable dye with a fluorescence readout of activated cleaved caspase-3 and-7. RESULTS: A de novo motif analysis was performed on a subset of genes previously found to be regulated at the level of ribosome binding during PDX1-deficiency, which identified a poly-cytosine (polyC) motif in the 3'UTR of the transcript encoding JUND. The polyC-binding protein hnRNPK bound to the mRNA encoding JUND, leading us to hypothesize that hnRNPK regulates JUND expression during glucolipotoxicity. Indeed, loss of hnRNPK blocked the post-transcriptional upregulation of JUND during metabolic stress. hnRNPK was phosphorylated in mouse and human islets during glucolipotoxicity and in islets of diabetic db/db mice. The MEK/ERK signaling pathway was both necessary and sufficient for the phosphorylation of hnRNPK, upregulation of JUND levels, and induction of pro-oxidant and pro-inflammatory genes. Further, we identified the RNA helicase DDX3X as a new binding partner for hnRNPK that is required for efficient translation of JUND mRNA. Loss of hnRNPK reduced DDX3X binding to translation machinery, suggesting that these factors cooperate to regulate translation in ß cells. CONCLUSIONS: Our results identify a novel ERK/hnRNPK/DDX3X pathway that influences ß cell survival and is activated under conditions associated with T2D.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo K/metabolismo , Células Secretoras de Insulina/metabolismo , Estrés Fisiológico , Animales , ARN Helicasas DEAD-box/genética , Diabetes Mellitus Tipo 2/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Células HEK293 , Ribonucleoproteína Heterogénea-Nuclear Grupo K/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA