Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 44
Filtrer
1.
Nat Commun ; 15(1): 5129, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38879678

RÉSUMÉ

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.


Sujet(s)
Sous-unités alpha Gs des protéines G , Cellules à glucagon , Glucagon , Souris knockout , Transduction du signal , Glucagon/métabolisme , Animaux , Cellules à glucagon/métabolisme , Souris , Sous-unités alpha Gs des protéines G/métabolisme , Adénosine/métabolisme , Récepteur A2A à l'adénosine/métabolisme , Récepteur A2A à l'adénosine/génétique , Mâle , Souris de lignée C57BL , Ilots pancréatiques/métabolisme
2.
Diabetes ; 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38870025

RÉSUMÉ

Glucagon is critical for the maintenance of blood glucose, however nutrient regulation of pancreatic α-cells remains poorly understood. Here, we identified a role for leucine, a well-known ß-cell fuel, in the α-cell intrinsic regulation of glucagon release. In islet perifusion assays, physiological 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 Sstr2 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 α-cell secretory tone across the physiological range of glucose levels, complementing the inhibitory paracrine actions of ß/δ-cells.

3.
Diabetes ; 73(6): 856-863, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38768366

RÉSUMÉ

An agreed-upon consensus model of glucose-stimulated insulin secretion from healthy ß-cells is essential for understanding diabetes pathophysiology. Since the discovery of the KATP channel in 1984, an oxidative phosphorylation (OxPhos)-driven rise in ATP has been assumed to close KATP channels to initiate insulin secretion. This model lacks any evidence, genetic or otherwise, that mitochondria possess the bioenergetics to raise the ATP/ADP ratio to the triggering threshold, and conflicts with genetic evidence demonstrating that OxPhos is dispensable for insulin secretion. It also conflates the stoichiometric yield of OxPhos with thermodynamics, and overestimates OxPhos by failing to account for established features of ß-cell metabolism, such as leak, anaplerosis, cataplerosis, and NADPH production that subtract from the efficiency of mitochondrial ATP production. We have proposed an alternative model, based on the spatial and bioenergetic specializations of ß-cell metabolism, in which glycolysis initiates insulin secretion. The evidence for this model includes that 1) glycolysis has high control strength over insulin secretion; 2) glycolysis is active at the correct time to explain KATP channel closure; 3) plasma membrane-associated glycolytic enzymes control KATP channels; 4) pyruvate kinase has favorable bioenergetics, relative to OxPhos, for raising ATP/ADP; and 5) OxPhos stalls before membrane depolarization and increases after. Although several key experiments remain to evaluate this model, the 1984 model is based purely on circumstantial evidence and must be rescued by causal, mechanistic experiments if it is to endure.


Sujet(s)
Glucose , Sécrétion d'insuline , Cellules à insuline , Insuline , Canaux KATP , Phosphorylation oxydative , Cellules à insuline/métabolisme , Humains , Glucose/métabolisme , Canaux KATP/métabolisme , Canaux KATP/génétique , Sécrétion d'insuline/physiologie , Animaux , Insuline/métabolisme , Glycolyse/physiologie , Modèles biologiques , Adénosine triphosphate/métabolisme
4.
Cell Rep ; 43(4): 114047, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38607916

RÉSUMÉ

Using 13C6 glucose labeling coupled to gas chromatography-mass spectrometry and 2D 1H-13C heteronuclear single quantum coherence NMR spectroscopy, we have obtained a comparative high-resolution map of glucose fate underpinning ß cell function. In both mouse and human islets, the contribution of glucose to the tricarboxylic acid (TCA) cycle is similar. Pyruvate fueling of the TCA cycle is primarily mediated by the activity of pyruvate dehydrogenase, with lower flux through pyruvate carboxylase. While the conversion of pyruvate to lactate by lactate dehydrogenase (LDH) can be detected in islets of both species, lactate accumulation is 6-fold higher in human islets. Human islets express LDH, with low-moderate LDHA expression and ß cell-specific LDHB expression. LDHB inhibition amplifies LDHA-dependent lactate generation in mouse and human ß cells and increases basal insulin release. Lastly, cis-instrument Mendelian randomization shows that low LDHB expression levels correlate with elevated fasting insulin in humans. Thus, LDHB limits lactate generation in ß cells to maintain appropriate insulin release.


Sujet(s)
Sécrétion d'insuline , Cellules à insuline , L-Lactate dehydrogenase , Acide lactique , Humains , Cellules à insuline/métabolisme , Animaux , L-Lactate dehydrogenase/métabolisme , Souris , Acide lactique/métabolisme , Glucose/métabolisme , Insuline/métabolisme , Isoenzymes/métabolisme , Cycle citrique , Souris de lignée C57BL , Mâle
5.
Elife ; 122023 10 03.
Article de Anglais | MEDLINE | ID: mdl-37787501

RÉSUMÉ

Insufficient insulin secretion to meet metabolic demand results in diabetes. The intracellular flux of Ca2+ into ß-cells triggers insulin release. Since genetics strongly influences variation in islet secretory responses, we surveyed islet Ca2+ dynamics in eight genetically diverse mouse strains. We found high strain variation in response to four conditions: (1) 8 mM glucose; (2) 8 mM glucose plus amino acids; (3) 8 mM glucose, amino acids, plus 10 nM glucose-dependent insulinotropic polypeptide (GIP); and (4) 2 mM glucose. These stimuli interrogate ß-cell function, α- to ß-cell signaling, and incretin responses. We then correlated components of the Ca2+ waveforms to islet protein abundances in the same strains used for the Ca2+ measurements. To focus on proteins relevant to human islet function, we identified human orthologues of correlated mouse proteins that are proximal to glycemic-associated single-nucleotide polymorphisms in human genome-wide association studies. Several orthologues have previously been shown to regulate insulin secretion (e.g. ABCC8, PCSK1, and GCK), supporting our mouse-to-human integration as a discovery platform. By integrating these data, we nominate novel regulators of islet Ca2+ oscillations and insulin secretion with potential relevance for human islet function. We also provide a resource for identifying appropriate mouse strains in which to study these regulators.


Sujet(s)
Ilots pancréatiques , Souris , Humains , Animaux , Ilots pancréatiques/métabolisme , Étude d'association pangénomique , Insuline/métabolisme , Glucose/métabolisme , Variation génétique , Acides aminés/métabolisme
6.
bioRxiv ; 2023 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-37577685

RÉSUMÉ

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.

7.
Islets ; 15(1): 2223327, 2023 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-37415404

RÉSUMÉ

Of the ß-cell signaling pathways altered by obesity and insulin resistance, some are adaptive while others contribute to ß-cell failure. Two critical second messengers are Ca2+ and cAMP, which control the timing and amplitude of insulin secretion. Previous work has shown the importance of the cAMP-inhibitory Prostaglandin EP3 receptor (EP3) in mediating the ß-cell dysfunction of type 2 diabetes (T2D). Here, we used three groups of C57BL/6J mice as a model of the progression from metabolic health to T2D: wildtype, normoglycemic LeptinOb (NGOB), and hyperglycemic LeptinOb (HGOB). Robust increases in ß-cell cAMP and insulin secretion were observed in NGOB islets as compared to wildtype controls; an effect lost in HGOB islets, which exhibited reduced ß-cell cAMP and insulin secretion despite increased glucose-dependent Ca2+ influx. An EP3 antagonist had no effect on ß-cell cAMP or Ca2+ oscillations, demonstrating agonist-independent EP3 signaling. Finally, using sulprostone to hyperactivate EP3 signaling, we found EP3-dependent suppression of ß-cell cAMP and Ca2+ duty cycle effectively reduces insulin secretion in HGOB islets, while having no impact insulin secretion on NGOB islets, despite similar and robust effects on cAMP levels and Ca2+ duty cycle. Finally, increased cAMP levels in NGOB islets are consistent with increased recruitment of the small G protein, Rap1GAP, to the plasma membrane, sequestering the EP3 effector, Gɑz, from inhibition of adenylyl cyclase. Taken together, these results suggest that rewiring of EP3 receptor-dependent cAMP signaling contributes to the progressive changes in ß cell function observed in the LeptinOb model of diabetes.


Sujet(s)
Diabète de type 2 , Insulinorésistance , Ilots pancréatiques , Souris , Animaux , Sécrétion d'insuline , Glucose/pharmacologie , Glucose/métabolisme , Ilots pancréatiques/métabolisme , Leptine/métabolisme , Leptine/pharmacologie , Insuline/métabolisme , Diabète de type 2/métabolisme , Souris de lignée C57BL , Transduction du signal , Obésité
8.
iScience ; 26(7): 107036, 2023 Jul 21.
Article de Anglais | MEDLINE | ID: mdl-37360692

RÉSUMÉ

Human and mouse genetics have delivered numerous diabetogenic loci, but it is mainly through the use of animal models that the pathophysiological basis for their contribution to diabetes has been investigated. More than 20 years ago, we serendipidously identified a mouse strain that could serve as a model of obesity-prone type 2 diabetes, the BTBR (Black and Tan Brachyury) mouse (BTBR T+ Itpr3tf/J, 2018) carrying the Lepob mutation. We went on to discover that the BTBR-Lepob mouse is an excellent model of diabetic nephropathy and is now widely used by nephrologists in academia and the pharmaceutical industry. In this review, we describe the motivation for developing this animal model, the many genes identified and the insights about diabetes and diabetes complications derived from >100 studies conducted in this remarkable animal model.

9.
Cell Rep ; 42(5): 112529, 2023 05 30.
Article de Anglais | MEDLINE | ID: mdl-37200193

RÉSUMÉ

Male mice lacking the androgen receptor (AR) in pancreatic ß cells exhibit blunted glucose-stimulated insulin secretion (GSIS), leading to hyperglycemia. Testosterone activates an extranuclear AR in ß cells to amplify glucagon-like peptide-1 (GLP-1) insulinotropic action. Here, we examined the architecture of AR targets that regulate GLP-1 insulinotropic action in male ß cells. Testosterone cooperates with GLP-1 to enhance cAMP production at the plasma membrane and endosomes via: (1) increased mitochondrial production of CO2, activating the HCO3--sensitive soluble adenylate cyclase; and (2) increased Gαs recruitment to GLP-1 receptor and AR complexes, activating transmembrane adenylate cyclase. Additionally, testosterone enhances GSIS in human islets via a focal adhesion kinase/SRC/phosphatidylinositol 3-kinase/mammalian target of rapamycin complex 2 actin remodeling cascade. We describe the testosterone-stimulated AR interactome, transcriptome, proteome, and metabolome that contribute to these effects. This study identifies AR genomic and non-genomic actions that enhance GLP-1-stimulated insulin exocytosis in male ß cells.


Sujet(s)
Cellules à insuline , Ilots pancréatiques , Mâle , Souris , Humains , Animaux , Glucagon-like peptide 1/métabolisme , Cellules à insuline/métabolisme , Adenylate Cyclase/métabolisme , Récepteurs aux androgènes/métabolisme , Insuline/métabolisme , Glucose/pharmacologie , Glucose/métabolisme , Testostérone , Ilots pancréatiques/métabolisme , Fragments peptidiques/métabolisme , Mammifères/métabolisme
10.
Cell Rep ; 42(4): 112394, 2023 04 25.
Article de Anglais | MEDLINE | ID: mdl-37058408

RÉSUMÉ

The ATP-sensitive K+ (KATP) channel is a key regulator of hormone secretion from pancreatic islet endocrine cells. Using direct measurements of KATP channel activity in pancreatic ß cells and the lesser-studied α cells, from both humans and mice, we provide evidence that a glycolytic metabolon locally controls KATP channels on the plasma membrane. The two ATP-consuming enzymes of upper glycolysis, glucokinase and phosphofructokinase, generate ADP that activates KATP. Substrate channeling of fructose 1,6-bisphosphate through the enzymes of lower glycolysis fuels pyruvate kinase, which directly consumes the ADP made by phosphofructokinase to raise ATP/ADP and close the channel. We further show the presence of a plasma membrane-associated NAD+/NADH cycle whereby lactate dehydrogenase is functionally coupled to glyceraldehyde-3-phosphate dehydrogenase. These studies provide direct electrophysiological evidence of a KATP-controlling glycolytic signaling complex and demonstrate its relevance to islet glucose sensing and excitability.


Sujet(s)
Membrane cellulaire , Cellules à glucagon , Glycolyse , Cellules à insuline , Humains , Animaux , Souris , Cellules à glucagon/métabolisme , Cellules à insuline/métabolisme , Canaux KATP/métabolisme , Techniques de patch-clamp , Électrophysiologie , Membrane cellulaire/enzymologie , Membrane cellulaire/métabolisme , Lactate dehydrogenases/métabolisme , Glyceraldehyde 3-phosphate dehydrogenases/métabolisme , ADP/métabolisme , Phosphofructokinases/métabolisme
11.
Elife ; 112022 08 23.
Article de Anglais | MEDLINE | ID: mdl-35997256

RÉSUMÉ

Pyruvate kinase (PK) and the phosphoenolpyruvate (PEP) cycle play key roles in nutrient-stimulated KATP channel closure and insulin secretion. To identify the PK isoforms involved, we generated mice lacking ß-cell PKm1, PKm2, and mitochondrial PEP carboxykinase (PCK2) that generates mitochondrial PEP. Glucose metabolism was found to generate both glycolytic and mitochondrially derived PEP, which triggers KATP closure through local PKm1 and PKm2 signaling at the plasma membrane. Amino acids, which generate mitochondrial PEP without producing glycolytic fructose 1,6-bisphosphate to allosterically activate PKm2, signal through PKm1 to raise ATP/ADP, close KATP channels, and stimulate insulin secretion. Raising cytosolic ATP/ADP with amino acids is insufficient to close KATP channels in the absence of PK activity or PCK2, indicating that KATP channels are primarily regulated by PEP that provides ATP via plasma membrane-associated PK, rather than mitochondrially derived ATP. Following membrane depolarization, the PEP cycle is involved in an 'off-switch' that facilitates KATP channel reopening and Ca2+ extrusion, as shown by PK activation experiments and ß-cell PCK2 deletion, which prolongs Ca2+ oscillations and increases insulin secretion. In conclusion, the differential response of PKm1 and PKm2 to the glycolytic and mitochondrial sources of PEP influences the ß-cell nutrient response, and controls the oscillatory cycle regulating insulin secretion.


Sujet(s)
Adénosine triphosphate , Pyruvate kinase , ADP , Adénosine triphosphate/métabolisme , Acides aminés , Animaux , Souris , Nutriments , Isoformes de protéines , Pyruvate kinase/génétique , Pyruvate kinase/métabolisme
12.
Cell Metab ; 34(7): 947-968, 2022 07 05.
Article de Anglais | MEDLINE | ID: mdl-35728586

RÉSUMÉ

In this review, we focus on recent developments in our understanding of nutrient-induced insulin secretion that challenge a key aspect of the "canonical" model, in which an oxidative phosphorylation-driven rise in ATP production closes KATP channels. We discuss the importance of intrinsic ß cell metabolic oscillations; the phasic alignment of relevant metabolic cycles, shuttles, and shunts; and how their temporal and compartmental relationships align with the triggering phase or the secretory phase of pulsatile insulin secretion. Metabolic signaling components are assigned regulatory, effectory, and/or homeostatic roles vis-à-vis their contribution to glucose sensing, signal transmission, and resetting the system. Taken together, these functions provide a framework for understanding how allostery, anaplerosis, and oxidative metabolism are integrated into the oscillatory behavior of the secretory pathway. By incorporating these temporal as well as newly discovered spatial aspects of ß cell metabolism, we propose a much-refined MitoCat-MitoOx model of the signaling process for the field to evaluate.


Sujet(s)
Ilots pancréatiques , Adénosine triphosphate/métabolisme , Glucose/métabolisme , Insuline/métabolisme , Sécrétion d'insuline , Ilots pancréatiques/métabolisme
13.
JCI Insight ; 7(10)2022 05 23.
Article de Anglais | MEDLINE | ID: mdl-35603790

RÉSUMÉ

Insulin secretion from pancreatic ß cells is essential for glucose homeostasis. An insufficient response to the demand for insulin results in diabetes. We previously showed that ß cell-specific deletion of Zfp148 (ß-Zfp148KO) improves glucose tolerance and insulin secretion in mice. Here, we performed Ca2+ imaging of islets from ß­Zfp148KO and control mice fed both a chow and a Western-style diet. ß-Zfp148KO islets demonstrated improved sensitivity and sustained Ca2+ oscillations in response to elevated glucose levels. ß-Zfp148KO islets also exhibited elevated sensitivity to amino acid-induced Ca2+ influx under low glucose conditions, suggesting enhanced mitochondrial phosphoenolpyruvate-dependent (PEP-dependent), ATP-sensitive K+ channel closure, independent of glycolysis. RNA-Seq and proteomics of ß-Zfp148KO islets revealed altered levels of enzymes involved in amino acid metabolism (specifically, SLC3A2, SLC7A8, GLS, GLS2, PSPH, PHGDH, and PSAT1) and intermediary metabolism (namely, GOT1 and PCK2), consistent with altered PEP cycling. In agreement with this, ß-Zfp148KO islets displayed enhanced insulin secretion in response to l-glutamine and activation of glutamate dehydrogenase. Understanding pathways controlled by ZFP148 may provide promising strategies for improving ß cell function that are robust to the metabolic challenge imposed by a Western diet.


Sujet(s)
Cellules à insuline , Ilots pancréatiques , Animaux , Calcium/métabolisme , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Glucose/métabolisme , Glutamine/métabolisme , Cellules à insuline/métabolisme , Ilots pancréatiques/métabolisme , Souris , Nutriments , Facteurs de transcription/métabolisme
14.
Diabetes ; 70(10): 2174-2182, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34593535

RÉSUMÉ

We hypothesize that basal hyperinsulinemia is synergistically mediated by an interplay between increased oxidative stress and excess lipid in the form of reactive oxygen species (ROS) and long-chain acyl-CoA esters (LC-CoA). In addition, ROS production may increase in response to inflammatory cytokines and certain exogenous environmental toxins that mislead ß-cells into perceiving nutrient excess when none exists. Thus, basal hyperinsulinemia is envisioned as an adaptation to sustained real or perceived nutrient excess that only manifests as a disease when the excess demand can no longer be met by an overworked ß-cell. In this article we will present a testable hypothetical mechanism to explain the role of lipids and ROS in basal hyperinsulinemia and how they differ from glucose-stimulated insulin secretion (GSIS). The model centers on redox regulation, via ROS, and S-acylation-mediated trafficking via LC-CoA. These pathways are well established in neural systems but not ß-cells. During GSIS, these signals rise and fall in an oscillatory pattern, together with the other well-established signals derived from glucose metabolism; however, their precise roles have not been defined. We propose that failure to either increase or decrease ROS or LC-CoA appropriately will disturb ß-cell function.


Sujet(s)
Hyperinsulinisme/étiologie , Sécrétion d'insuline/physiologie , Animaux , Glucose/métabolisme , Glucose/pharmacologie , Humains , Hyperinsulinisme/métabolisme , Insuline/métabolisme , Sécrétion d'insuline/effets des médicaments et des substances chimiques , Cellules à insuline/effets des médicaments et des substances chimiques , Cellules à insuline/métabolisme , Lipides/physiologie , Oxydoréduction , Stress oxydatif/physiologie , Espèces réactives de l'oxygène/métabolisme
15.
Diabetes ; 70(11): 2626-2638, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34462260

RÉSUMÉ

SUMOylation reduces oxidative stress and preserves islet mass at the expense of robust insulin secretion. To investigate a role for the deSUMOylating enzyme sentrin-specific protease 1 (SENP1) following metabolic stress, we put pancreas/gut-specific SENP1 knockout (pSENP1-KO) mice on a high-fat diet (HFD). Male pSENP1-KO mice were more glucose intolerant following HFD than littermate controls but only in response to oral glucose. A similar phenotype was observed in females. Plasma glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide 1 (GLP-1) responses were identical in pSENP1-KO and wild-type littermates, including the HFD-induced upregulation of GIP responses. Islet mass was not different, but insulin secretion and ß-cell exocytotic responses to the GLP-1 receptor agonist exendin-4 (Ex4) and GIP were impaired in islets lacking SENP1. Glucagon secretion from pSENP1-KO islets was also reduced, so we generated ß-cell-specific SENP1 KO mice. These phenocopied the pSENP1-KO mice with selective impairment in oral glucose tolerance following HFD, preserved islet mass expansion, and impaired ß-cell exocytosis and insulin secretion to Ex4 and GIP without changes in cAMP or Ca2+ levels. Thus, ß-cell SENP1 limits oral glucose intolerance following HFD by ensuring robust insulin secretion at a point downstream of incretin signaling.


Sujet(s)
Cysteine endopeptidases/métabolisme , Alimentation riche en graisse/effets indésirables , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Cellules à insuline/métabolisme , Animaux , Cysteine endopeptidases/génétique , Glucose/pharmacologie , Intolérance au glucose , Hyperglycémie provoquée , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Incrétines , Insuline ordinaire humaine/pharmacologie , Souris , Souris knockout , Transactivateurs/génétique , Transactivateurs/métabolisme
16.
Elife ; 102021 07 07.
Article de Anglais | MEDLINE | ID: mdl-34231467

RÉSUMÉ

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.


Sujet(s)
Sécrétion d'insuline/physiologie , Cellules à insuline/physiologie , Protéines de tissu nerveux/effets des médicaments et des substances chimiques , Protéines de tissu nerveux/métabolisme , Récepteurs immunologiques/déficit , Récepteurs immunologiques/métabolisme , Animaux , Connexines/génétique , Connexines/métabolisme , Jonctions communicantes/métabolisme , Souris , Souris knockout , Protéines de tissu nerveux/génétique , Récepteurs immunologiques/génétique , ,
17.
Cell Rep ; 34(4): 108690, 2021 01 26.
Article de Anglais | MEDLINE | ID: mdl-33503433

RÉSUMÉ

Hallmarks of mature ß cells are restricted proliferation and a highly energetic secretory state. Paradoxically, cyclin-dependent kinase 2 (CDK2) is synthesized throughout adulthood, its cytosolic localization raising the likelihood of cell cycle-independent functions. In the absence of any changes in ß cell mass, maturity, or proliferation, genetic deletion of Cdk2 in adult ß cells enhanced insulin secretion from isolated islets and improved glucose tolerance in vivo. At the single ß cell level, CDK2 restricts insulin secretion by increasing KATP conductance, raising the set point for membrane depolarization in response to activation of the phosphoenolpyruvate (PEP) cycle with mitochondrial fuels. In parallel with reduced ß cell recruitment, CDK2 restricts oxidative glucose metabolism while promoting glucose-dependent amplification of insulin secretion. This study provides evidence of essential, non-canonical functions of CDK2 in the secretory pathways of quiescent ß cells.


Sujet(s)
Lymphocytes B/métabolisme , Kinase-2 cycline-dépendante/usage thérapeutique , Canaux KATP/effets des médicaments et des substances chimiques , Animaux , Kinase-2 cycline-dépendante/pharmacologie , Humains , Souris
18.
Cell Metab ; 32(5): 736-750.e5, 2020 11 03.
Article de Anglais | MEDLINE | ID: mdl-33147484

RÉSUMÉ

Pancreatic ß cells couple nutrient metabolism with appropriate insulin secretion. Here, we show that pyruvate kinase (PK), which converts ADP and phosphoenolpyruvate (PEP) into ATP and pyruvate, underlies ß cell sensing of both glycolytic and mitochondrial fuels. Plasma membrane-localized PK is sufficient to close KATP channels and initiate calcium influx. Small-molecule PK activators increase the frequency of ATP/ADP and calcium oscillations and potently amplify insulin secretion. PK restricts respiration by cyclically depriving mitochondria of ADP, which accelerates PEP cycling until membrane depolarization restores ADP and oxidative phosphorylation. Our findings support a compartmentalized model of ß cell metabolism in which PK locally generates the ATP/ADP required for insulin secretion. Oscillatory PK activity allows mitochondria to perform synthetic and oxidative functions without any net impact on glucose oxidation. These findings suggest a potential therapeutic route for diabetes based on PK activation that would not be predicted by the current consensus single-state model of ß cell function.


Sujet(s)
Insuline/métabolisme , Pyruvate kinase/métabolisme , Animaux , Lignée cellulaire , Humains , Sécrétion d'insuline , Mâle , Souris , Souris de lignée C57BL
19.
Cell Metab ; 32(5): 751-766.e11, 2020 11 03.
Article de Anglais | MEDLINE | ID: mdl-33147485

RÉSUMÉ

The mitochondrial GTP (mtGTP)-dependent phosphoenolpyruvate (PEP) cycle couples mitochondrial PEPCK (PCK2) to pyruvate kinase (PK) in the liver and pancreatic islets to regulate glucose homeostasis. Here, small molecule PK activators accelerated the PEP cycle to improve islet function, as well as metabolic homeostasis, in preclinical rodent models of diabetes. In contrast, treatment with a PK activator did not improve insulin secretion in pck2-/- mice. Unlike other clinical secretagogues, PK activation enhanced insulin secretion but also had higher insulin content and markers of differentiation. In addition to improving insulin secretion, acute PK activation short-circuited gluconeogenesis to reduce endogenous glucose production while accelerating red blood cell glucose turnover. Four-week delivery of a PK activator in vivo remodeled PK phosphorylation, reduced liver fat, and improved hepatic and peripheral insulin sensitivity in HFD-fed rats. These data provide a preclinical rationale for PK activation to accelerate the PEP cycle to improve metabolic homeostasis and insulin sensitivity.


Sujet(s)
Mitochondries/métabolisme , Phosphoénolpyruvate/métabolisme , Animaux , Homéostasie , Insuline/métabolisme , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Pyruvate kinase/métabolisme , Rats , Rat Sprague-Dawley
20.
Sci Rep ; 10(1): 2823, 2020 02 18.
Article de Anglais | MEDLINE | ID: mdl-32071395

RÉSUMÉ

Glucagon-like peptide 1 (GLP-1) and cholecystokinin (CCK) are gut-derived peptide hormones known to play important roles in the regulation of gastrointestinal motility and secretion, appetite, and food intake. We have previously demonstrated that both GLP-1 and CCK are produced in the endocrine pancreas of obese mice. Interestingly, while GLP-1 is well known to stimulate insulin secretion by the pancreatic ß-cells, direct evidence of CCK promoting insulin release in human islets remains to be determined. Here, we tested whether islet-derived GLP-1 or CCK is necessary for the full stimulation of insulin secretion. We confirm that mouse pancreatic islets secrete GLP-1 and CCK, but only GLP-1 acts locally within the islet to promote insulin release ex vivo. GLP-1 is exclusively produced in approximately 50% of α-cells in lean mouse islets and 70% of α-cells in human islets, suggesting a paracrine α to ß-cell signaling through the ß-cell GLP-1 receptor. Additionally, we provide evidence that islet CCK expression is regulated by glucose, but its receptor signaling is not required during glucose-stimulated insulin secretion (GSIS). We also see no increase in GSIS in response to CCK peptides. Importantly, all these findings were confirmed in islets from non-diabetic human donors. In summary, our data suggest no direct role for CCK in stimulating insulin secretion and highlight the critical role of intra-islet GLP-1 signaling in the regulation of human ß-cell function.


Sujet(s)
Cholécystokinine/physiologie , Glucagon-like peptide 1/physiologie , Sécrétion d'insuline , Cellules à insuline/cytologie , Animaux , Humains , Mâle , Souris , Souris de lignée C57BL
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...