Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39229244

RESUMO

Ca2+-driven oscillations in insulin secretion are crucial for glycemic control. Prior studies, performed with single-plane imaging, suggest that subpopulations of electrically coupled ß-cells have privileged roles in leading and coordinating the propagation of Ca2+ waves. Here, we used 3D light-sheet imaging to analyze the location and Ca2+ activity of single ß-cells within the entire islet at >2 Hz. Network analysis indicates that the most highly synchronized ß-cells are located at the islet center and remain geographically stable between oscillations. In contrast, ß-cells that initiate the Ca2+ wave ('leaders') are located at the islet periphery and change their identity over time via rotations in the wave axis. Activators of glucokinase and pyruvate kinase were used to further test the influence of glycolysis on these ß-cell subpopulations. Glucokinase activation, which increased oscillation period, reinforced leader cells and stabilized the wave axis. Pyruvate kinase activation, despite increasing oscillation frequency, had no effect on leader cells, indicating the wave origin is patterned by fuel input. The 3D islet ß-cell network, which did not respond to either treatment, is insensitive to changes in fuel metabolism. These findings emphasize the stochastic nature of Ca2+ oscillations and identify a role for glucokinase in spatially patterning 'leader' ß-cells.

3.
Diabetes ; 73(9): 1426-1439, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38870025

RESUMO

Glucagon is critical for the maintenance of blood glucose, however nutrient regulation of pancreatic α-cells remains poorly understood. Here, we identified a role of leucine, a well-known ß-cell fuel, in the α-cell-intrinsic regulation of glucagon release. In islet perifusion assays, physiologic concentrations of leucine strongly inhibited alanine- and arginine-stimulated glucagon secretion from human and mouse islets under hypoglycemic conditions. Mechanistically, leucine dose-dependently reduced α-cell cAMP, independently of Ca2+, ATP/ADP, or fatty acid oxidation. Leucine also reduced α-cell cAMP in islets treated with somatostatin receptor 2 antagonists or diazoxide, compounds that limit paracrine signaling from ß/δ-cells. Studies in dispersed mouse islets confirmed an α-cell-intrinsic effect. The inhibitory effect of leucine on cAMP was mimicked by glucose, α-ketoisocaproate, succinate, and the glutamate dehydrogenase activator BCH and blocked by cyanide, indicating a mechanism dependent on mitochondrial metabolism. Glucose dose-dependently reduced the impact of leucine on α-cell cAMP, indicating an overlap in function; however, leucine was still effective at suppressing glucagon secretion in the presence of elevated glucose, amino acids, and the incretin GIP. Taken together, these findings show that leucine plays an intrinsic role in limiting the α-cell secretory tone across the physiologic range of glucose levels, complementing the inhibitory paracrine actions of ß/δ-cells.


Assuntos
AMP Cíclico , Células Secretoras de Glucagon , Glucagon , Leucina , Comunicação Parácrina , Animais , Glucagon/metabolismo , AMP Cíclico/metabolismo , Células Secretoras de Glucagon/metabolismo , Células Secretoras de Glucagon/efeitos dos fármacos , Camundongos , Humanos , Leucina/farmacologia , Comunicação Parácrina/efeitos dos fármacos , Glucose/metabolismo , Cetoácidos/farmacologia , Masculino , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo
4.
Nat Commun ; 15(1): 5129, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38879678

RESUMO

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.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP , Células Secretoras de Glucagon , Glucagon , Camundongos Knockout , Transdução de Sinais , Glucagon/metabolismo , Animais , Células Secretoras de Glucagon/metabolismo , Camundongos , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Adenosina/metabolismo , Receptor A2A de Adenosina/metabolismo , Receptor A2A de Adenosina/genética , Masculino , Camundongos Endogâmicos C57BL , Ilhotas Pancreáticas/metabolismo
5.
bioRxiv ; 2023 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-37577685

RESUMO

Objective: Pancreatic islets are nutrient sensors that regulate organismal blood glucose homeostasis. Glucagon release from the pancreatic α-cell is important under fasted, fed, and hypoglycemic conditions, yet metabolic regulation of α-cells remains poorly understood. Here, we identified a previously unexplored role for physiological levels of leucine, which is classically regarded as a ß-cell fuel, in the intrinsic regulation of α-cell glucagon release. Methods: GcgCreERT:CAMPER and GcgCreERT:GCaMP6s mice were generated to perform dynamic, high-throughput functional measurements of α-cell cAMP and Ca2+ within the intact islet. Islet perifusion assays were used for simultaneous, time-resolved measurements of glucagon and insulin release from mouse and human islets. The effects of leucine were compared with glucose and the mitochondrial fuels 2-aminobicyclo(2,2,1)heptane-2-carboxylic acid (BCH, non-metabolized leucine analog that activates glutamate dehydrogenase), α-ketoisocaproate (KIC, leucine metabolite), and methyl-succinate (complex II fuel). CYN154806 (Sstr2 antagonist), diazoxide (KATP activator, which prevents Ca2+-dependent exocytosis from α, ß, and δ-cells), and dispersed α-cells were used to inhibit islet paracrine signaling and identify α-cell intrinsic effects. Results: Mimicking the effect of glucose, leucine strongly suppressed amino acid-stimulated glucagon secretion. Mechanistically, leucine dose-dependently reduced α-cell cAMP at physiological concentrations, with an IC50 of 57, 440, and 1162 µM at 2, 6, and 10 mM glucose, without affecting α-cell Ca2+. Leucine also reduced α-cell cAMP in islets treated with Sstr2 antagonist or diazoxide, as well as dispersed α-cells, indicating an α-cell intrinsic effect. The effect of leucine was matched by KIC and the glutamate dehydrogenase activator BCH, but not methyl-succinate, indicating a dependence on mitochondrial anaplerosis. Glucose, which stimulates anaplerosis via pyruvate carboxylase, had the same suppressive effect on α-cell cAMP but with lower potency. Similarly to mouse islets, leucine suppressed glucagon secretion from human islets under hypoglycemic conditions. Conclusions: These findings highlight an important role for physiological levels of leucine in the metabolic regulation of α-cell cAMP and glucagon secretion. Leucine functions primarily through an α-cell intrinsic effect that is dependent on glutamate dehydrogenase, in addition to the well-established α-cell regulation by ß/δ-cell paracrine signaling. Our results suggest that mitochondrial anaplerosis-cataplerosis facilitates the glucagonostatic effect of both leucine and glucose, which cooperatively suppress α-cell tone by reducing cAMP.

6.
Elife ; 102021 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-34231467

RESUMO

The spatial architecture of the islets of Langerhans is hypothesized to facilitate synchronized insulin secretion among ß cells, yet testing this in vivo in the intact pancreas is challenging. Robo ßKO mice, in which the genes Robo1 and Robo2 are deleted selectively in ß cells, provide a unique model of altered islet spatial architecture without loss of ß cell differentiation or islet damage from diabetes. Combining Robo ßKO mice with intravital microscopy, we show here that Robo ßKO islets have reduced synchronized intra-islet Ca2+ oscillations among ß cells in vivo. We provide evidence that this loss is not due to a ß cell-intrinsic function of Robo, mis-expression or mis-localization of Cx36 gap junctions, or changes in islet vascularization or innervation, suggesting that the islet architecture itself is required for synchronized Ca2+ oscillations. These results have implications for understanding structure-function relationships in the islets during progression to diabetes as well as engineering islets from stem cells.


Assuntos
Secreção de Insulina/fisiologia , Células Secretoras de Insulina/fisiologia , Proteínas do Tecido Nervoso/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Receptores Imunológicos/deficiência , Receptores Imunológicos/metabolismo , Animais , Conexinas/genética , Conexinas/metabolismo , Junções Comunicantes/metabolismo , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Receptores Imunológicos/genética , Proteína delta-2 de Junções Comunicantes , Proteínas Roundabout
7.
Nature ; 543(7645): 443-446, 2017 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-28241148

RESUMO

Loss of proteostasis underlies ageing and neurodegeneration characterized by the accumulation of protein aggregates and mitochondrial dysfunction. Although many neurodegenerative-disease-associated proteins can be found in mitochondria, it remains unclear how mitochondrial dysfunction and protein aggregation could be related. In dividing yeast cells, protein aggregates that form under stress or during ageing are preferentially retained by the mother cell, in part through tethering to mitochondria, while the disaggregase Hsp104 helps to dissociate aggregates and thereby enables refolding or degradation of misfolded proteins. Here we show that, in yeast, cytosolic proteins prone to aggregation are imported into mitochondria for degradation. Protein aggregates that form under heat shock contain both cytosolic and mitochondrial proteins and interact with the mitochondrial import complex. Many aggregation-prone proteins enter the mitochondrial intermembrane space and matrix after heat shock, and some do so even without stress. Timely dissolution of cytosolic aggregates requires the mitochondrial import machinery and proteases. Blocking mitochondrial import but not proteasome activity causes a marked delay in the degradation of aggregated proteins. Defects in cytosolic Hsp70s leads to enhanced entry of misfolded proteins into mitochondria and elevated mitochondrial stress. We term this mitochondria-mediated proteostasis mechanism MAGIC (mitochondria as guardian in cytosol) and provide evidence that it may exist in human cells.


Assuntos
Citosol/metabolismo , Homeostase , Mitocôndrias/metabolismo , Agregados Proteicos/fisiologia , Dobramento de Proteína , Proteínas/química , Proteínas/metabolismo , Saccharomyces cerevisiae , Linhagem Celular , Citosol/efeitos dos fármacos , Proteínas de Choque Térmico HSP70/metabolismo , Resposta ao Choque Térmico , Humanos , Mitocôndrias/efeitos dos fármacos , Proteínas Mitocondriais/química , Proteínas Mitocondriais/metabolismo , Peptídeo Hidrolases/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Redobramento de Proteína , Estabilidade Proteica , Transporte Proteico/efeitos dos fármacos , Proteólise , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA