Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
Cell Rep ; 43(4): 114047, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38607916

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.


Insulin Secretion , Insulin-Secreting Cells , L-Lactate Dehydrogenase , Lactic Acid , Humans , Insulin-Secreting Cells/metabolism , Animals , L-Lactate Dehydrogenase/metabolism , Mice , Lactic Acid/metabolism , Glucose/metabolism , Insulin/metabolism , Isoenzymes/metabolism , Citric Acid Cycle , Mice, Inbred C57BL , Male
2.
Diabet Med ; 40(12): e15220, 2023 12.
Article En | MEDLINE | ID: mdl-37669696

We previously developed, synthesized and tested light-activated sulfonylureas for optical control of KATP channels and pancreatic beta cell activity in vitro and in vivo. Such technology relies on installation of azobenzene photoswitches onto the sulfonylurea backbone, affording light-dependent isomerization, alteration in ligand affinity for SUR1 and hence KATP channel conductance. Inspired by molecular dynamics simulations and to further improve photoswitching characteristics, we set out to develop a novel push-pull closed ring azobenzene unit, before installing this on the sulfonylurea glimepiride as a small molecule recipient. Three fine-tuned, light-activated sulfonylureas were synthesized, encompassing azetidine, pyrrolidine and piperidine closed rings. Azetidine-, pyrrolidine- and piperidine-based sulfonylureas all increased beta cell Ca2+ -spiking activity upon continuous blue light illumination, similarly to first generation JB253. Notably, the pyrrolidine-based sulfonylurea showed superior switch OFF performance to JB253. As such, third generation sulfonylureas afford more precise optical control over primary pancreatic beta cells, and showcase the potential of pyrrolidine-azobenzenes as chemical photoswitches across drug classes.


Azetidines , Insulin-Secreting Cells , Humans , Sulfonylurea Compounds/therapeutic use , Adenosine Triphosphate , Piperidines , Pyrrolidines
3.
Cell Rep ; 42(5): 112529, 2023 05 30.
Article En | MEDLINE | ID: mdl-37200193

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.


Insulin-Secreting Cells , Islets of Langerhans , Male , Mice , Humans , Animals , Glucagon-Like Peptide 1/metabolism , Insulin-Secreting Cells/metabolism , Adenylyl Cyclases/metabolism , Receptors, Androgen/metabolism , Insulin/metabolism , Glucose/pharmacology , Glucose/metabolism , Testosterone , Islets of Langerhans/metabolism , Peptide Fragments/metabolism , Mammals/metabolism
4.
Clin Exp Immunol ; 212(1): 1-10, 2023 04 07.
Article En | MEDLINE | ID: mdl-36891817

Dysregulation of leukocyte trafficking, lipid metabolism, and other metabolic processes are the hallmarks that underpin and drive pathology in obesity. Current clinical management targets alternations in lifestyle choices (e.g. exercise, weight loss) to limit the impact of the disease. Crucially, re-gaining control over the pathogenic cellular and molecular processes may offer an alternative, complementary strategy for obese patients. Here we investigate the impact of the immunopeptide, PEPITEM, on pancreas homeostasis and leukocyte trafficking in mice on high-fed obesogenic diet (HFD). Both prophylactic and therapeutic treatment with PEPITEM alleviated the effects of HFD on the pancreas, reducing pancreatic beta cell size. Moreover, PEPITEM treatment also limited T-cell trafficking (CD4+ T-cells and KLRG1+ CD3+ T-cells) to obese visceral, but not subcutaneous, adipose tissue. Similarly, PEPITEM treatment reduced macrophage numbers within the peritoneal cavity of mice on HFD diet at both 6 and 12 weeks. By contrast, PEPITEM therapy elevated numbers of T and B cells were observed in the secondary lymphoid tissues (e.g. spleen and inguinal lymph node) when compared to the untreated HFD controls. Collectively our data highlights the potential for PEPITEM as a novel therapy to combat the systemic low-grade inflammation experienced in obesity and minimize the impact of obesity on pancreatic homeostasis. Thus, offering an alternative strategy to reduce the risk of developing obesity-related co-morbidities, such as type 2 diabetes mellitus, in individuals at high risk and struggling to control their weight through lifestyle modifications.


Diabetes Mellitus, Type 2 , Mice , Animals , Diabetes Mellitus, Type 2/metabolism , Obesity/complications , Obesity/metabolism , Obesity/pathology , Inflammation/pathology , Diet , CD4-Positive T-Lymphocytes/metabolism , Mice, Inbred C57BL , Adipose Tissue
5.
Nat Commun ; 14(1): 301, 2023 01 18.
Article En | MEDLINE | ID: mdl-36653347

The glucagon-like peptide-1 receptor (GLP1R) is a class B G protein-coupled receptor (GPCR) involved in glucose homeostasis and food intake. GLP1R agonists (GLP1RA) are widely used in the treatment of diabetes and obesity, yet visualizing the endogenous localization, organization and dynamics of a GPCR has so far remained out of reach. In the present study, we generate mice harboring an enzyme self-label genome-edited into the endogenous Glp1r locus. We also rationally design and test various fluorescent dyes, spanning cyan to far-red wavelengths, for labeling performance in tissue. By combining these technologies, we show that endogenous GLP1R can be specifically and sensitively detected in primary tissue using multiple colors. Longitudinal analysis of GLP1R dynamics reveals heterogeneous recruitment of neighboring cell subpopulations into signaling and trafficking, with differences observed between GLP1RA classes and dual agonists. At the nanoscopic level, GLP1Rs are found to possess higher organization, undergoing GLP1RA-dependent membrane diffusion. Together, these results show the utility of enzyme self-labels for visualization and interrogation of endogenous proteins, and provide insight into the biology of a class B GPCR in primary cells and tissue.


Glucagon-Like Peptide-1 Receptor , Obesity , Mice , Animals , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism
6.
Diabetes ; 72(2): 275-289, 2023 Feb 01.
Article En | MEDLINE | ID: mdl-36445949

GC-globulin (GC), or vitamin D-binding protein, is a multifunctional protein involved in the transport of circulating vitamin 25(OH)D and fatty acids, as well as actin scavenging. In the pancreatic islets, the gene encoding GC, GC/Gc, is highly localized to glucagon-secreting α-cells. Despite this, the role of GC in α-cell function is poorly understood. We previously showed that GC is essential for α-cell morphology, electrical activity, and glucagon secretion. We now show that loss of GC exacerbates α-cell failure during metabolic stress. High-fat diet-fed GC-/- mice have basal hyperglucagonemia, which is associated with decreased α-cell size, impaired glucagon secretion and Ca2+ fluxes, and changes in glucose-dependent F-actin remodelling. Impairments in glucagon secretion can be rescued using exogenous GC to replenish α-cell GC levels, increase glucagon granule area, and restore the F-actin cytoskeleton. Lastly, GC levels decrease in α-cells of donors with type 2 diabetes, which is associated with changes in α-cell mass, morphology, and glucagon expression. Together, these data demonstrate an important role for GC in α-cell adaptation to metabolic stress.


Diabetes Mellitus, Type 2 , Globulins , Animals , Mice , Diabetes Mellitus, Type 2/metabolism , Globulins/metabolism , Glucagon/metabolism , Stress, Physiological , Vitamin D-Binding Protein/genetics , Vitamin D-Binding Protein/metabolism
7.
J Endocrinol ; 253(1): 1-11, 2022 02 11.
Article En | MEDLINE | ID: mdl-35017316

Pancreatic ß-cells depend on the well-balanced regulation of cytosolic zinc concentrations, providing sufficient zinc ions for the processing and storage of insulin, but avoiding toxic effects. The zinc transporter ZnT8, encoded by SLC30A8,is a key player regarding islet cell zinc homeostasis, and polymorphisms in this gene are associated with altered type 2 diabetes susceptibility in man. The objective of this study was to investigate the role of ZnT8 and zinc in situations of cellular stress as hypoxia or inflammation. Isolated islets of WT and global ZnT8-/- mice were exposed to hypoxia or cytokines and cell death was measured. To explore the role of changing intracellular Zn2+ concentrations, WT islets were exposed to different zinc concentrations using zinc chloride or the zinc chelator N,N,N',N'-tetrakis(2-pyridinylmethyl)-1,2-ethanediamine (TPEN). Hypoxia or cytokine (TNF-α, IFN-γ, IL1-ß) treatment induced islet cell death, but to a lesser extent in islets from ZnT8-/- mice, which were shown to have a reduced zinc content. Similarly, chelation of zinc with TPEN reduced cell death in WT islets treated with hypoxia or cytokines, whereas increased zinc concentrations aggravated the effects of these stressors. This study demonstrates a reduced rate of cell death in islets from ZnT8-/- mice as compared to WT islets when exposed to two distinct cellular stressors, hypoxia or cytotoxic cytokines. This protection from cell death is, in part, mediated by a reduced zinc content in islet cells of ZnT8-/- mice. These findings may be relevant for altered diabetes burden in carriers of risk SLC30A8 alleles in man.


Diabetes Mellitus, Type 2/genetics , Genetic Predisposition to Disease/genetics , Islets of Langerhans/metabolism , Zinc Transporter 8/genetics , Animals , Apoptosis/genetics , Cell Death/drug effects , Cell Death/genetics , Cell Hypoxia , Cell Line , Cell Proliferation/genetics , Cells, Cultured , Cytokines/pharmacology , Diabetes Mellitus, Type 2/metabolism , Female , Insulin Secretion/drug effects , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Zinc/metabolism , Zinc/pharmacology , Zinc Transporter 8/metabolism
8.
Endocrinology ; 163(1)2022 01 01.
Article En | MEDLINE | ID: mdl-34718519

Pancreatic ß-cells can secrete insulin via 2 pathways characterized as KATP channel -dependent and -independent. The KATP channel-independent pathway is characterized by a rise in several potential metabolic signaling molecules, including the NADPH/NADP+ ratio and α-ketoglutarate (αKG). Prolyl hydroxylases (PHDs), which belong to the αKG-dependent dioxygenase superfamily, are known to regulate the stability of hypoxia-inducible factor α. In the current study, we assess the role of PHDs in vivo using the pharmacological inhibitor dimethyloxalylglycine (DMOG) and generated ß-cell-specific knockout (KO) mice for all 3 isoforms of PHD (ß-PHD1 KO, ß-PHD2 KO, and ß-PHD3 KO mice). DMOG inhibited in vivo insulin secretion in response to glucose challenge and inhibited the first phase of insulin secretion but enhanced the second phase of insulin secretion in isolated islets. None of the ß-PHD KO mice showed any significant in vivo defects associated with glucose tolerance and insulin resistance except for ß-PHD2 KO mice which had significantly increased plasma insulin during a glucose challenge. Islets from both ß-PHD1 KO and ß-PHD3 KO had elevated ß-cell apoptosis and reduced ß-cell mass. Isolated islets from ß-PHD1 KO and ß-PHD3 KO had impaired glucose-stimulated insulin secretion and glucose-stimulated increases in the ATP/ADP and NADPH/NADP+ ratio. All 3 PHD isoforms are expressed in ß-cells, with PHD3 showing the most distinct expression pattern. The lack of each PHD protein did not significantly impair in vivo glucose homeostasis. However, ß-PHD1 KO and ß-PHD3 KO mice had defective ß-cell mass and islet insulin secretion, suggesting that these mice may be predisposed to developing diabetes.


Insulin Secretion , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Prolyl Hydroxylases/metabolism , Protein Isoforms/chemistry , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis , Gene Expression Regulation , Glucose/metabolism , Glucose Tolerance Test , Homeostasis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ketoglutaric Acids/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NADP/metabolism , Oxidative Phosphorylation , Oxygen Consumption , Phenotype , Protein Domains
9.
J Endocrinol ; 251(1): 111-123, 2021 09 09.
Article En | MEDLINE | ID: mdl-34370682

Supplementation with precursors of NAD has been shown to prevent and reverse insulin resistance, mitochondrial dysfunction, and liver damage in mouse models of diet-induced obesity. We asked whether the beneficial effects of supplementation with the NAD precursor nicotinamide riboside (NR) are dependent on mouse strain. We compared the effects of NR supplementation on whole-body energy metabolism and mitochondrial function in mildly obese C57BL/6N and C57BL/6J mice, two commonly used strains to investigate metabolism. Male C57BL/6N and C57BL/6J mice were fed a high-fat diet (HFD) or standard chow with or without NR supplementation for 8 weeks. Body and organ weights, glucose tolerance, and metabolic parameters as well as mitochondrial O2 flux in liver and muscle fibers were assessed. We found that NR supplementation had no influence on body or organ weight, glucose metabolism or hepatic lipid accumulation, energy expenditure, or metabolic flexibility but increased mitochondrial respiration in soleus muscle in both mouse strains. Strain-dependent differences were detected for body and fat depot weight, fasting blood glucose, hepatic lipid accumulation, and energy expenditure. We conclude that, in mild obesity, NR supplementation does not alter metabolic phenotype in two commonly used laboratory mouse strains.


Energy Metabolism/drug effects , Niacinamide/analogs & derivatives , Obesity/drug therapy , Pyridinium Compounds/therapeutic use , Animals , Cell Respiration/drug effects , Diet, High-Fat , Disease Models, Animal , Drug Evaluation , Glucose Intolerance/prevention & control , Lipid Metabolism/drug effects , Liver/metabolism , Male , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Niacinamide/therapeutic use , Obesity/metabolism
10.
JCI Insight ; 6(16)2021 08 23.
Article En | MEDLINE | ID: mdl-34264866

The α-ketoglutarate-dependent dioxygenase, prolyl-4-hydroxylase 3 (PHD3), is an HIF target that uses molecular oxygen to hydroxylate peptidyl prolyl residues. Although PHD3 has been reported to influence cancer cell metabolism and liver insulin sensitivity, relatively little is known about the effects of this highly conserved enzyme in insulin-secreting ß cells in vivo. Here, we show that the deletion of PHD3 specifically in ß cells (ßPHD3KO) was associated with impaired glucose homeostasis in mice fed a high-fat diet. In the early stages of dietary fat excess, ßPHD3KO islets energetically rewired, leading to defects in the management of pyruvate fate and a shift from glycolysis to increased fatty acid oxidation (FAO). However, under more prolonged metabolic stress, this switch to preferential FAO in ßPHD3KO islets was associated with impaired glucose-stimulated ATP/ADP rises, Ca2+ fluxes, and insulin secretion. Thus, PHD3 might be a pivotal component of the ß cell glucose metabolism machinery in mice by suppressing the use of fatty acids as a primary fuel source during the early phases of metabolic stress.


Fatty Acids/adverse effects , Glucose/metabolism , Insulin Resistance , Insulin-Secreting Cells/enzymology , Procollagen-Proline Dioxygenase/metabolism , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Glycolysis , Humans , Insulin Secretion , Lipid Metabolism , Male , Mice , Mice, Knockout , Oxidation-Reduction , Procollagen-Proline Dioxygenase/genetics
12.
Nat Commun ; 12(1): 674, 2021 01 29.
Article En | MEDLINE | ID: mdl-33514698

Transcriptionally mature and immature ß-cells co-exist within the adult islet. How such diversity contributes to insulin release remains poorly understood. Here we show that subtle differences in ß-cell maturity, defined using PDX1 and MAFA expression, contribute to islet operation. Functional mapping of rodent and human islets containing proportionally more PDX1HIGH and MAFAHIGH ß-cells reveals defects in metabolism, ionic fluxes and insulin secretion. At the transcriptomic level, the presence of increased numbers of PDX1HIGH and MAFAHIGH ß-cells leads to dysregulation of gene pathways involved in metabolic processes. Using a chemogenetic disruption strategy, differences in PDX1 and MAFA expression are shown to depend on islet Ca2+ signaling patterns. During metabolic stress, islet function can be restored by redressing the balance between PDX1 and MAFA levels across the ß-cell population. Thus, preserving heterogeneity in PDX1 and MAFA expression, and more widely in ß-cell maturity, might be important for the maintenance of islet function.


Insulin Secretion/physiology , Insulin-Secreting Cells/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Female , Gene Knock-In Techniques , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Maf Transcription Factors, Large/genetics , Maf Transcription Factors, Large/metabolism , Male , Mice , Mice, Transgenic , Models, Animal , Primary Cell Culture , Trans-Activators/genetics , Trans-Activators/metabolism
13.
Cell Rep ; 33(9): 108466, 2020 12 01.
Article En | MEDLINE | ID: mdl-33264613

Pancreatic ß cell failure is key to type 2 diabetes (T2D) onset and progression. Here, we assess whether human ß cell dysfunction induced by metabolic stress is reversible, evaluate the molecular pathways underlying persistent or transient damage, and explore the relationships with T2D islet traits. Twenty-six islet preparations are exposed to several lipotoxic/glucotoxic conditions, some of which impair insulin release, depending on stressor type, concentration, and combination. The reversal of dysfunction occurs after washout for some, although not all, of the lipoglucotoxic insults. Islet transcriptomes assessed by RNA sequencing and expression quantitative trait loci (eQTL) analysis identify specific pathways underlying ß cell failure and recovery. Comparison of a large number of human T2D islet transcriptomes with those of persistent or reversible ß cell lipoglucotoxicity show shared gene expression signatures. The identification of mechanisms associated with human ß cell dysfunction and recovery and their overlap with T2D islet traits provide insights into T2D pathogenesis, fostering the development of improved ß cell-targeted therapeutic strategies.


Diabetes Mellitus, Type 2/genetics , Gene Expression/genetics , Insulin-Secreting Cells/metabolism , Stress, Physiological/genetics , Diabetes Mellitus, Type 2/pathology , Humans
15.
Cell Rep ; 31(11): 107761, 2020 06 16.
Article En | MEDLINE | ID: mdl-32553153

Vitamin-D-binding protein (DBP) or group-specific component of serum (GC-globulin) carries vitamin D metabolites from the circulation to target tissues. DBP is highly localized to the liver and pancreatic α cells. Although DBP serum levels, gene polymorphisms, and autoantigens have all been associated with diabetes risk, the underlying mechanisms remain unknown. Here, we show that DBP regulates α cell morphology, α cell function, and glucagon secretion. Deletion of DBP leads to smaller and hyperplastic α cells, altered Na+ channel conductance, impaired α cell activation by low glucose, and reduced rates of glucagon secretion both in vivo and in vitro. Mechanistically, this involves reversible changes in islet microfilament abundance and density, as well as changes in glucagon granule distribution. Defects are also seen in ß cell and δ cell function. Immunostaining of human pancreata reveals generalized loss of DBP expression as a feature of late-onset and long-standing, but not early-onset, type 1 diabetes. Thus, DBP regulates α cell phenotype, with implications for diabetes pathogenesis.


Cell Communication/physiology , Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Vitamin D-Binding Protein/metabolism , Vitamin D/metabolism , Animals , Biological Transport/physiology , Bodily Secretions/metabolism , Humans , Mice, Knockout , Phenotype
16.
Diabetologia ; 63(9): 1822-1835, 2020 09.
Article En | MEDLINE | ID: mdl-32472193

AIMS/HYPOTHESIS: During pregnancy, maternal metabolic disease and hormonal imbalance may alter fetal beta cell development and/or proliferation, thus leading to an increased risk for developing type 2 diabetes in adulthood. Although thyroid hormones play an important role in fetal endocrine pancreas development, the impact of maternal hypothyroidism on glucose homeostasis in adult offspring remains poorly understood. METHODS: We investigated this using a mouse model of hypothyroidism, induced by administration of an iodine-deficient diet supplemented with propylthiouracil during gestation. RESULTS: Here, we show that, when fed normal chow, adult mice born to hypothyroid mothers were more glucose-tolerant due to beta cell hyperproliferation (two- to threefold increase in Ki67-positive beta cells) and increased insulin sensitivity. However, following 8 weeks of high-fat feeding, these offspring gained 20% more body weight, became profoundly hyperinsulinaemic (with a 50% increase in fasting insulin concentration), insulin-resistant and glucose-intolerant compared with controls from euthyroid mothers. Furthermore, altered glucose metabolism was maintained in a second generation of animals. CONCLUSIONS/INTERPRETATION: Therefore, gestational hypothyroidism induces long-term alterations in endocrine pancreas function, which may have implications for type 2 diabetes prevention in affected individuals.


Blood Glucose/metabolism , Glucose Intolerance/metabolism , Hypothyroidism/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/embryology , Pregnancy Complications/metabolism , Prenatal Exposure Delayed Effects/metabolism , Animals , Antithyroid Agents/toxicity , Cell Proliferation , Diet, High-Fat , Disease Models, Animal , Female , Hyperinsulinism/metabolism , Insulin Resistance , Iodine/deficiency , Islets of Langerhans/metabolism , Mice , Pregnancy , Propylthiouracil/toxicity , Stress, Physiological
18.
Nat Commun ; 11(1): 467, 2020 01 24.
Article En | MEDLINE | ID: mdl-31980626

The glucagon-like peptide-1 receptor (GLP1R) is a class B G protein-coupled receptor (GPCR) involved in metabolism. Presently, its visualization is limited to genetic manipulation, antibody detection or the use of probes that stimulate receptor activation. Herein, we present LUXendin645, a far-red fluorescent GLP1R antagonistic peptide label. LUXendin645 produces intense and specific membrane labeling throughout live and fixed tissue. GLP1R signaling can additionally be evoked when the receptor is allosterically modulated in the presence of LUXendin645. Using LUXendin645 and LUXendin651, we describe islet, brain and hESC-derived ß-like cell GLP1R expression patterns, reveal higher-order GLP1R organization including membrane nanodomains, and track single receptor subpopulations. We furthermore show that the LUXendin backbone can be optimized for intravital two-photon imaging by installing a red fluorophore. Thus, our super-resolution compatible labeling probes allow visualization of endogenous GLP1R, and provide insight into class B GPCR distribution and dynamics both in vitro and in vivo.


Fluorescent Dyes , Glucagon-Like Peptide-1 Receptor/metabolism , Microscopy, Fluorescence, Multiphoton/methods , Amino Acid Sequence , Animals , Brain/metabolism , Cell Line , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Glucagon-Like Peptide-1 Receptor/deficiency , Glucagon-Like Peptide-1 Receptor/genetics , HEK293 Cells , Human Embryonic Stem Cells/metabolism , Humans , Islets of Langerhans/metabolism , Mice , Mice, Knockout , Models, Molecular , Molecular Structure , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Peptide Fragments/genetics , Signal Transduction , Tissue Distribution
19.
Mol Metab ; 27S: S49-S59, 2019 09.
Article En | MEDLINE | ID: mdl-31500831

BACKGROUND: Current therapeutic strategies for type 1 (T1DM) and type 2 diabetes mellitus (T2DM) rely on increasing or substituting endogenous insulin secretion in combination with lifestyle changes. ß-cell regeneration, a process whereby new ß-cells arise from progenitors, self-renewal or transdifferentiation, has the potential to become a viable route to insulin self-sufficiency. Current regeneration strategies capture many of the transcriptomic and protein features of native ß-cells, generating cells capable of glucose-dependent insulin secretion in vitro and alleviation of hyperglycemia in vivo. However, whether novel ß-cells display appreciable heterogeneity remains poorly understood, with potential consequences for long-term functional robustness. SCOPE OF REVIEW: The review brings together crucial discoveries in the ß-cell regeneration field with state-of-the-art knowledge regarding ß-cell heterogeneity. Aspects that might aid production of longer-lasting and more plastic regenerated ß-cells are highlighted and discussed. MAJOR CONCLUSIONS: Different ß-cell regeneration approaches result in a similar outcome: glucose-sensitive, insulin-positive cells that mimic the native ß-cell phenotype but which lack normal plasticity. The ß-cell subpopulations identified to date expand our understanding of ß-cell survival, proliferation and function, signposting the direction for future regeneration strategies. Therefore, regenerated ß-cells should exhibit stimulus-dependent differences in gene and protein expression, as well as establish a functional network with different ß-cells, all while coexisting with other cell types on a three-dimensional platform.


Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Animals , Cell Differentiation , Cell Survival , Humans
20.
J Mol Endocrinol ; 61(1): R43-R60, 2018 07.
Article En | MEDLINE | ID: mdl-29661799

It is becoming increasingly apparent that not all insulin-secreting beta cells are equal. Subtle differences exist at the transcriptomic and protein expression levels, with repercussions for beta cell survival/proliferation, calcium signalling and insulin release. Notably, beta cell heterogeneity displays plasticity during development, metabolic stress and type 2 diabetes mellitus (T2DM). Thus, heterogeneity or lack thereof may be an important contributor to beta cell failure during T2DM in both rodents and humans. The present review will discuss the molecular and cellular features of beta cell heterogeneity at both the single-cell and islet level, explore how this influences islet function and insulin release and look into the alterations that may occur during obesity and T2DM.


Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Humans , Insulin-Secreting Cells/physiology , Obesity/metabolism
...