80 results on '"Patrick Gilon"'
Search Results
2. Prolonged culture of human pancreatic islets under glucotoxic conditions changes their acute beta cell calcium and insulin secretion glucose response curves from sigmoid to bell-shaped
- Author
-
Mohammad Tariq, Arnaldo H. de Souza, Mohammed Bensellam, Heeyoung Chae, Manon Jaffredo, Anne-Françoise Close, Jean-Philippe Deglasse, Laila R. B. Santos, Antoine Buemi, Nizar I. Mourad, Anne Wojtusciszyn, Matthieu Raoux, Patrick Gilon, Christophe Broca, and Jean-Christophe Jonas
- Subjects
Endocrinology, Diabetes and Metabolism ,Internal Medicine - Abstract
The rapid remission of type 2 diabetes by a diet very low in energy correlates with a marked improvement in glucose-stimulated insulin secretion (GSIS), emphasising the role of beta cell dysfunction in the early stages of the disease. In search of novel mechanisms of beta cell dysfunction after long-term exposure to mild to severe glucotoxic conditions, we extensively characterised the alterations in insulin secretion and upstream coupling events in human islets cultured for 1-3 weeks at ~5, 8, 10 or 20 mmol/l glucose and subsequently stimulated by an acute stepwise increase in glucose concentration.Human islets from 49 non-diabetic donors (ND-islets) and six type 2 diabetic donors (T2D-islets) were obtained from five isolation centres. After shipment, the islets were precultured for 3-7 days in RPMI medium containing ~5 mmol/l glucose and 10% (vol/vol) heat-inactivated FBS with selective islet picking at each medium renewal. Islets were then cultured for 1-3 weeks in RPMI containing ~5, 8, 10 or 20 mmol/l glucose before measurement of insulin secretion during culture, islet insulin and DNA content, beta cell apoptosis and cytosolic and mitochondrial glutathione redox state, and assessment of dynamic insulin secretion and upstream coupling events during acute stepwise stimulation with glucose [NAD(P)H autofluorescence, ATP/(ATP+ADP) ratio, electrical activity, cytosolic CaCulture of ND-islets for 1-3 weeks at 8, 10 or 20 vs 5 mmol/l glucose did not significantly increase beta cell apoptosis or oxidative stress but decreased insulin content in a concentration-dependent manner and increased beta cell sensitivity to subsequent acute stimulation with glucose. Islet glucose responsiveness was higher after culture at 8 or 10 vs 5 mmol/l glucose and markedly reduced after culture at 20 vs 5 mmol/l glucose. In addition, the [CaProlonged culture of human islets under moderate to severe glucotoxic conditions markedly increased their glucose sensitivity and revealed a bell-shaped acute glucose response curve for changes in [Ca
- Published
- 2022
3. Chemerin as an Inducer of β Cell Proliferation Mediates Mitochondrial Homeostasis and Promotes β Cell Mass Expansion
- Author
-
Min Li, Ruifan Zhang, Qian Ge, Lingzhi Yue, Dan Ma, Firas Khattab, Wenhua Xie, Yewei Cui, Patrick Gilon, Xueya Zhao, Xi Li, Rui Cheng, and UCL - SSS/IREC/EDIN - Pôle d'endocrinologie, diabète et nutrition
- Subjects
chemerin ,β cell ,adipokine ,insulin secretion ,mitochondrial homeostasis ,type 2 diabetes ,Organic Chemistry ,General Medicine ,Catalysis ,Computer Science Applications ,Inorganic Chemistry ,Mice ,Diabetes Mellitus, Type 2 ,Adipokines ,Animals ,Intercellular Signaling Peptides and Proteins ,Homeostasis ,Receptors, Chemokine ,Chemokines ,Physical and Theoretical Chemistry ,Molecular Biology ,Spectroscopy ,Cell Proliferation - Abstract
Loss of the β cell population is a crucial feature of type 2 diabetes. Restoring the β cell mass by stimulating β cell proliferation and preventing its apoptosis was proposed as a therapeutic approach to treating diabetes. Therefore, researchers have been increasingly interested in identifying exogenous factors that can stimulate β cell proliferation in situ and in vitro. Adipokine chemerin, which is secreted from adipose tissue and the liver, has been identified as a chemokine that plays a critical role in the regulation of metabolism. In this study, we demonstrate that chemerin as a circulating adipokine promotes β cell proliferation in vivo and in vitro. Chemerin serum levels and the expression of the main receptors within islets are highly regulated under a variety of challenging conditions, including obesity and type 2 diabetes. As compared to their littermates, mice overexpressing chemerin had a larger islet area and increased β cell mass with both a normal and high-fat diet. Moreover, in chemerin-overexpressed mice, we observed improved mitochondrial homeostasis and increased insulin synthesis. In summary, our findings confirm the potential role of chemerin as an inducer of β cell proliferation, and they provide novel insights into the helpful strategy to expand β cell population.
- Published
- 2023
4. In depth functional characterization of human induced pluripotent stem cell-derived beta cells in vitro and in vivo
- Author
-
Federica Fantuzzi, Sanna Toivonen, Andrea Alex Schiavo, Heeyoung Chae, Mohammad Tariq, Toshiaki Sawatani, Nathalie Pachera, Ying Cai, Chiara Vinci, Enrico Virgilio, Laurence Ladriere, Mara Suleiman, Piero Marchetti, Jean-Christophe Jonas, Patrick Gilon, Décio L. Eizirik, Mariana Igoillo-Esteve, Miriam Cnop, and UCL - SSS/IREC/EDIN - Pôle d'endocrinologie, diabète et nutrition
- Subjects
Cell Biology ,Developmental Biology - Abstract
In vitro differentiation of human induced pluripotent stem cells (iPSCs) into beta cells represents an important cell source for diabetes research. Here, we fully characterized iPSC-derived beta cell function in vitro and in vivo in humanized mice. Using a 7-stage protocol, human iPSCs were differentiated into islet-like aggregates with a yield of insulin-positive beta cells comparable to that of human islets. The last three stages of differentiation were conducted with two different 3D culture systems, rotating suspension or static microwells. In the latter, homogeneously small-sized islet-like aggregates were obtained, while in rotating suspension size was heterogeneous and aggregates often clumped. In vitro function was assessed by glucose-stimulated insulin secretion, NAD(P)H and calcium fluctuations. Stage 7 aggregates slightly increased insulin release in response to glucose in vitro. Aggregates were transplanted under the kidney capsule of NOD-SCID mice to allow for further in vivo beta cell maturation. In transplanted mice, grafts showed glucose-responsiveness and maintained normoglycemia after streptozotocin injection. In situ kidney perfusion assays showed modulation of human insulin secretion in response to different secretagogues. In conclusion, iPSCs differentiated with equal efficiency into beta cells in microwells compared to rotating suspension, but the former had a higher experimental success rate. In vitro differentiation generated aggregates lacking fully mature beta cell function. In vivo, beta cells acquired the functional characteristics typical of human islets. With this technology an unlimited supply of islet-like organoids can be generated from human iPSCs that will be instrumental to study beta cell biology and dysfunction in diabetes.
- Published
- 2022
- Full Text
- View/download PDF
5. Glucose inhibits glucagon secretion by decreasing [Ca
- Author
-
Bilal, Singh, Firas, Khattab, and Patrick, Gilon
- Subjects
Mice ,Adenosine Triphosphate ,Glucose ,KATP Channels ,Cations, Divalent ,Glucagon-Secreting Cells ,Animals ,Calcium ,Glucagon ,Somatostatin ,Exocytosis - Abstract
The mechanisms by which glucose stimulates insulin secretion from β-cells are well established and involve inhibition of ATP-sensitive KWe tested the effect of a decrease or an increase of glucose concentration (Gx, with x = concentration in mM) on α-cell [CaBlockade of KWe propose a model according to which glucose controls α-cell [Ca
- Published
- 2021
6. The endoplasmic reticulum-plasma membrane tethering protein TMEM24 is a regulator of cellular Ca2+ homeostasis
- Author
-
Styliani Panagiotou, Beichen Xie, Olof Idevall-Hagren, Patrick Gilon, Peter Bergsten, and Jing Cen
- Subjects
Cytosol ,chemistry.chemical_compound ,Membrane ,chemistry ,Endoplasmic reticulum ,Phosphatidylinositol ,Mitochondrion ,Plant lipid transfer proteins ,Lipid Transport ,Diacylglycerol kinase ,Cell biology - Abstract
Endoplasmic reticulum (ER) - plasma membrane (PM) contacts are sites of lipid exchange and Ca2+ transport, and both lipid transport proteins and Ca2+ channels specifically accumulate at these locations. In pancreatic β-cells, both lipid- and Ca2+ signaling are essential for insulin secretion. The recently characterized lipid transfer protein TMEM24 dynamically localize to ER-PM contact sites and provide phosphatidylinositol, a precursor of PI(4)P and PI(4,5)P2, to the plasma membrane. β-cells lacking TMEM24 exhibit markedly suppressed glucose-induced Ca2+ oscillations and insulin secretion but the underlying mechanism is not known. We now show that TMEM24 only weakly interact with the PM, and dissociates in response to both diacylglycerol and nanomolar elevations of cytosolic Ca2+. Release of TMEM24 into the bulk ER membrane also enables direct interactions with mitochondria, and we report that loss of TMEM24 results in excessive accumulation of Ca2+ in both the ER and mitochondria and in impaired mitochondria function.
- Published
- 2021
7. Glucose Acutely Reduces Cytosolic and Mitochondrial H2O2in Rat Pancreatic Beta Cells
- Author
-
Jean Philippe Deglasse, Daniel Pastor-Flores, Patrick Gilon, Tobias P. Dick, Leticia Prates Roma, Jean-Christophe Jonas, UCL - SSS/IREC - Institut de recherche expérimentale et clinique, and UCL - SSS/IREC/EDIN - Pôle d'endocrinologie, diabète et nutrition
- Subjects
inorganic chemicals ,0301 basic medicine ,medicine.medical_specialty ,030102 biochemistry & molecular biology ,Physiology ,Clinical Biochemistry ,Stimulation ,Cell Biology ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Cytosol ,030104 developmental biology ,Endocrinology ,Menadione ,chemistry ,Internal medicine ,Pancreatic beta Cells ,medicine ,General Earth and Planetary Sciences ,Hydrogen peroxide ,Insulin secretion ,Molecular Biology ,General Environmental Science - Abstract
Aims: Whether H2O2 contributes to the glucose-dependent stimulation of insulin secretion by pancreatic β-cells is highly controversial. We used two H2O2-sensitive probes, roGFP2-Orp1 and HyPer with its pH-control SypHer, to test the acute effects of glucose, monomethylsuccinate, leucine with glutamine, and α-ketoisocaproate, on β-cell cytosolic and mitochondrial H2O2 concentrations. We then tested the effects of low H2O2 and menadione concentrations on insulin secretion. Results: RoGFP2-Orp1 was more sensitive than HyPer to H2O2 (response at 2-5 vs. 10µM) and less pH-sensitive. Under control conditions, stimulation with glucose reduced mitochondrial roGFP2-Orp1 oxidation without affecting cytosolic roGFP2-Orp1 and HyPer fluorescence ratios, except for the pH-dependent effects on HyPer. However, stimulation with glucose decreased the oxidation of both cytosolic probes by 15µM exogenous H2O2. The glucose effects were not affected by overexpression of catalase, mitochondrial catalase or superoxide dismutase 1 and 2. They followed the increase in NAD(P)H autofluorescence, were maximal at 5mM glucose in the cytosol and 10mM glucose in the mitochondria, and were partly mimicked by the other nutrients. Exogenous H2O2 (1-15µM) did not affect insulin secretion. By contrast, menadione (1-5µM) did not increase basal insulin secretion but reduced the stimulation of insulin secretion by 20mM glucose. Innovation: Subcellular changes in β-cell H2O2 levels are better monitored with roGFP2-Orp1 than HyPer/SypHer. Nutrients acutely lower mitochondrial H2O2 levels in β-cells and promote degradation of exogenously supplied H2O2 in both cytosolic and mitochondrial compartments. Conclusion: The glucose-dependent stimulation of insulin secretion occurs independently of a detectable increase in β-cell cytosolic or mitochondrial H2O2 levels.
- Published
- 2019
8. Glucose inhibits glucagon secretion by decreasing [Ca2+]c and by reducing the efficacy of Ca2+ on exocytosis via somatostatin-dependent and independent mechanisms
- Author
-
Bilal Singh, Firas Khattab, and Patrick Gilon
- Subjects
Cell Biology ,Molecular Biology - Published
- 2022
9. SGLT2 is not expressed in pancreatic α- and β-cells, and its inhibition does not directly affect glucagon and insulin secretion in rodents and humans
- Author
-
Nancy Antoine, Lucie Ruiz, Holger Klein, Patrick Gilon, Anne Wojtusciszyn, Fiona M. Gribble, Frank Reimann, Robert Augustin, Bao-Khanh Lai, Bilal Singh, Michael P. Pieper, Eva Gatineau, Nano Rita, Mohammed Bensellam, Pedro Luis Herrera, Birgit Stierstorfer, Firas Khattab, Heeyoung Chae, Lorenzo Piemonti, Davide Brusa, Michael Mark, Eric Mayoux, Christophe Broca, Université Catholique de Louvain = Catholic University of Louvain (UCL), Boehringer Ingelheim Pharma GmbH & Co. KG, University of Geneva [Switzerland], Addenbrooke's Hospital, Cambridge University NHS Trust, Centre Hospitalier Régional Universitaire [Montpellier] (CHRU Montpellier), Lausanne University Hospital, IRCCS Ospedale San Raffaele [Milan, Italy], Chae, Heeyoung, Augustin, Robert, Gatineau, Eva, Mayoux, Eric, Bensellam, Mohammed, Antoine, Nancy, Khattab, Fira, Lai, Bao-Khanh, Brusa, Davide, Stierstorfer, Birgit, Klein, Holger, Singh, Bilal, Ruiz, Lucie, Pieper, Michael, Mark, Michael, Herrera, Pedro L, Gribble, Fiona M, Reimann, Frank, Wojtusciszyn, Anne, Broca, Christophe, Rita, Nano, Piemonti, Lorenzo, Gilon, Patrick, Gribble, Fiona [0000-0002-4232-2898], Reimann, Frank [0000-0001-9399-6377], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Blood Glucose ,medicine.medical_treatment ,IC50, half maximal inhibitory concentration ,SGLT2i, sodium-glucose cotransporter 2 inhibitors ,MESH: Insulin Secretion ,MESH: Glucosides ,MESH: Insulin-Secreting Cells ,chemistry.chemical_compound ,Mice ,αMG, α-methyl-D-glucopyranoside ,EGP, endogenous glucose production ,0302 clinical medicine ,Glucosides ,Glucagon-Like Peptide 1 ,Insulin-Secreting Cells ,FACS, fluorescence-activated cell sorting ,Insulin Secretion ,Insulin ,ddc:576.5 ,MESH: Animals ,Dapagliflozin ,geography.geographical_feature_category ,Chemistry ,MESH: Glucagon ,BW, body weight ,Diabetes ,SGLT2 inhibitor ,MESH: Pancreas ,[SDV.MHEP.EM]Life Sciences [q-bio]/Human health and pathology/Endocrinology and metabolism ,Islet ,3. Good health ,SGLT, sodium-glucose cotransporter ,MESH: Glucose ,MESH: Sodium-Glucose Transporter 2 ,No direct effect of SGLT2i on α- and β-cells ,MESH: Sodium-Glucose Transporter 2 Inhibitors ,Ketone bodies ,Original Article ,medicine.medical_specialty ,lcsh:Internal medicine ,endocrine system ,MESH: Rats ,030209 endocrinology & metabolism ,MESH: Insulin ,Glucagon ,03 medical and health sciences ,Islets of Langerhans ,Gliflozins ,TPM, transcripts per million ,Sodium-Glucose Transporter 2 ,G, glucose ,In vivo ,Internal medicine ,Empagliflozin ,medicine ,MESH: Benzhydryl Compounds ,Animals ,Humans ,Benzhydryl Compounds ,lcsh:RC31-1245 ,Molecular Biology ,MESH: Mice ,Pancreas ,Sodium-Glucose Transporter 2 Inhibitors ,geography ,MESH: Humans ,MESH: Islets of Langerhans ,Glucose transporter ,MESH: Glucagon-Like Peptide 1 ,Cell Biology ,Rats ,030104 developmental biology ,Endocrinology ,Glucose ,Glucagon-Secreting Cells ,MESH: Blood Glucose ,MESH: Glucagon-Secreting Cells ,Cmax, maximum serum concentration ,diabetes ,glucagon ,insulin - Abstract
Objective Sodium-glucose cotransporter 2 (SGLT2) inhibitors (SGLT2i), or gliflozins, are anti-diabetic drugs that lower glycemia by promoting glucosuria, but they also stimulate endogenous glucose and ketone body production. The likely causes of these metabolic responses are increased blood glucagon levels, and decreased blood insulin levels, but the mechanisms involved are hotly debated. This study verified whether or not SGLT2i affect glucagon and insulin secretion by a direct action on islet cells in three species, using multiple approaches. Methods We tested the in vivo effects of two selective SGLT2i (dapagliflozin, empagliflozin) and a SGLT1/2i (sotagliflozin) on various biological parameters (glucosuria, glycemia, glucagonemia, insulinemia) in mice. mRNA expression of SGLT2 and other glucose transporters was assessed in rat, mouse, and human FACS-purified α- and β-cells, and by analysis of two human islet cell transcriptomic datasets. Immunodetection of SGLT2 in pancreatic tissues was performed with a validated antibody. The effects of dapagliflozin, empagliflozin, and sotagliflozin on glucagon and insulin secretion were assessed using isolated rat, mouse and human islets and the in situ perfused mouse pancreas. Finally, we tested the long-term effect of SGLT2i on glucagon gene expression. Results SGLT2 inhibition in mice increased the plasma glucagon/insulin ratio in the fasted state, an effect correlated with a decline in glycemia. Gene expression analyses and immunodetections showed no SGLT2 mRNA or protein expression in rodent and human islet cells, but moderate SGLT1 mRNA expression in human α-cells. However, functional experiments on rat, mouse, and human (29 donors) islets and the in situ perfused mouse pancreas did not identify any direct effect of dapagliflozin, empagliflozin or sotagliflozin on glucagon and insulin secretion. SGLT2i did not affect glucagon gene expression in rat and human islets. Conclusions The data indicate that the SGLT2i-induced increase of the plasma glucagon/insulin ratio in vivo does not result from a direct action of the gliflozins on islet cells., Highlights • Gliflozins (SGLT2 and SGLT1/2 inhibitors) increase plasma glucagon/insulin ratio. • SGLT2 is not expressed in rodent and human pancreatic α- and β-cells. • SGLT1 is however expressed in human α-cells. • SGLT2 and SGLT1/2 inhibitors do not directly affect glucagon and insulin secretion.
- Published
- 2020
10. Impaired Store-Operated Calcium Entry and STIM1 Loss Lead to Reduced Insulin Secretion and Increased Endoplasmic Reticulum Stress in the Diabetic β-Cell
- Author
-
Chih-Chun Lee, Michael W. Roe, Tatsuyoshi Kono, Paul Sohn, Xin Tong, Hitoshi Iida, Patrick Gilon, Solaema Taleb, Carmella Evans-Molina, and Robert N. Bone
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Type 2 diabetes ,Cell Line ,Diabetes Mellitus, Experimental ,Proinflammatory cytokine ,Mice ,03 medical and health sciences ,Insulin-Secreting Cells ,Internal medicine ,Internal Medicine ,medicine ,Animals ,Humans ,Insulin ,Stromal Interaction Molecule 1 ,Calcium metabolism ,geography ,geography.geographical_feature_category ,Chemistry ,Endoplasmic reticulum ,STIM1 ,Endoplasmic Reticulum Stress ,Islet ,medicine.disease ,Store-operated calcium entry ,Rats ,Glucose ,030104 developmental biology ,Endocrinology ,Islet Studies ,Unfolded protein response ,Calcium - Abstract
Store-operated Ca2+ entry (SOCE) is a dynamic process that leads to refilling of endoplasmic reticulum (ER) Ca2+ stores through reversible gating of plasma membrane Ca2+ channels by the ER Ca2+ sensor Stromal Interaction Molecule 1 (STIM1). Pathogenic reductions in β-cell ER Ca2+ have been observed in diabetes. However, a role for impaired SOCE in this phenotype has not been tested. We measured the expression of SOCE molecular components in human and rodent models of diabetes and found a specific reduction in STIM1 mRNA and protein levels in human islets from donors with type 2 diabetes (T2D), islets from hyperglycemic streptozotocin-treated mice, and INS-1 cells (rat insulinoma cells) treated with proinflammatory cytokines and palmitate. Pharmacologic SOCE inhibitors led to impaired islet Ca2+ oscillations and insulin secretion, and these effects were phenocopied by β-cell STIM1 deletion. STIM1 deletion also led to reduced ER Ca2+ storage and increased ER stress, whereas STIM1 gain of function rescued β-cell survival under proinflammatory conditions and improved insulin secretion in human islets from donors with T2D. Taken together, these data suggest that the loss of STIM1 and impaired SOCE contribute to ER Ca2+ dyshomeostasis under diabetic conditions, whereas efforts to restore SOCE-mediated Ca2+ transients may have the potential to improve β-cell health and function.
- Published
- 2018
11. KATP channel blockers control glucagon secretion by distinct mechanisms: A direct stimulation of α-cells involving a [Ca2+]c rise and an indirect inhibition mediated by somatostatin
- Author
-
Patrick Gilon, Bilal Singh, Pedro Luis Herrera, Firas Khattab, Heeyoung Chae, Lieven Desmet, UCL - SSS/IREC/EDIN - Pôle d'endocrinologie, diabète et nutrition, and UCL - SSH/LIDAM/SMCS - Support en méthodologie et calcul statistique (plate-forme technologique)
- Subjects
0301 basic medicine ,[Ca2+]c, free cytosolic Ca2+ concentration ,medicine.medical_treatment ,Mice ,0302 clinical medicine ,KATP Channels ,Sulfonylureas ,TPNQ, tertiapin-Q ,Internal medicine ,GIRK channels, G protein-gated inwardly rectifying K+ channels ,Mice, Knockout ,Chemistry ,Glucagon secretion ,Ca2+ ,Somatostatin ,medicine.anatomical_structure ,Gliclazide ,Original Article ,Ca(2+) ,hormones, hormone substitutes, and hormone antagonists ,medicine.drug ,endocrine system ,medicine.medical_specialty ,medicine.drug_class ,Tolbutamide ,Pancreatic islets ,KATP channels, ATP-sensitive K+ channels ,Mice, Transgenic ,030209 endocrinology & metabolism ,Hypoglycemia ,Glucagon ,SSTR, somatostatin receptors ,03 medical and health sciences ,Potassium Channel Blockers ,medicine ,Animals ,VGCC, voltage-gated Ca2+ channels ,Molecular Biology ,Insulin ,Cell Biology ,medicine.disease ,RC31-1245 ,Sulfonylurea ,030104 developmental biology ,Endocrinology ,Glucagon-Secreting Cells ,Calcium ,SST, somatostatin - Abstract
Objective Glucagon is secreted by pancreatic α-cells in response to hypoglycemia and its hyperglycemic effect helps to restore normal blood glucose. Insulin and somatostatin (SST) secretions from β- and δ-cells, respectively, are stimulated by glucose by mechanisms involving an inhibition of their ATP-sensitive K+ (KATP) channels, leading to an increase in [Ca2+]c that triggers exocytosis. Drugs that close KATP channels, such as sulfonylureas, are used to stimulate insulin release in type 2 diabetic patients. α-cells also express KATP channels. However, the mechanisms by which sulfonylureas control glucagon secretion are still largely debated and were addressed in the present study. In particular, we studied the effects of KATP channel blockers on α-cell [Ca2+]c and glucagon secretion in the presence of a low (1 mM) or a high (15 mM) glucose concentration and evaluated the role of SST in these effects. Methods Using a transgenic mouse model expressing the Ca2+-sensitive fluorescent protein, GCaMP6f, specifically in α-cells, we measured [Ca2+]c in α-cells either dispersed or within whole islets (by confocal microscopy). By measuring [Ca2+]c in α-cells within islets and glucagon secretion using the same perifusion protocols, we tested whether glucagon secretion correlated with changes in [Ca2+]c in response to sulfonylureas. We studied the role of SST in the effects of sulfonylureas using multiple approaches including genetic ablation of SST, or application of SST-14 and SST receptor antagonists. Results Application of the sulfonylureas, tolbutamide, or gliclazide, to a medium containing 1 mM or 15 mM glucose increased [Ca2+]c in α-cells by a direct effect as in β-cells. At low glucose, sulfonylureas inhibited glucagon secretion of islets despite the rise in α-cell [Ca2+]c that they triggered. This glucagonostatic effect was indirect and attributed to SST because, in the islets of SST-knockout mice, sulfonylureas induced a stimulation of glucagon secretion which correlated with an increase in α-cell [Ca2+]c. Experiments with exogenous SST-14 and SST receptor antagonists indicated that the glucagonostatic effect of sulfonylureas mainly resulted from an inhibition of the efficacy of cytosolic Ca2+ on exocytosis. Although SST-14 was also able to inhibit glucagon secretion by decreasing α-cell [Ca2+]c, no decrease in [Ca2+]c occurred during sulfonylurea application because it was largely counterbalanced by the direct stimulatory effect of these drugs on α-cell [Ca2+]c. At high glucose, i.e., in conditions where glucagon release was already low, sulfonylureas stimulated glucagon secretion because their direct stimulatory effect on α-cells exceeded the indirect effect by SST. Our results also indicated that, unexpectedly, SST-14 poorly decreased the efficacy of Ca2+ on exocytosis in β-cells. Conclusions Sulfonylureas exert two opposite actions on α-cells: a direct stimulation as in β-cells and an indirect inhibition by SST. This suggests that any alteration of SST paracrine influence, as described in diabetes, will modify the effect of sulfonylureas on glucagon release. In addition, we suggest that δ-cells inhibit α-cells more efficiently than β-cells., Highlights • KATP channel blockers control glucagon secretion by two mechanisms. • The first one is the direct stimulation of α-cell by a [Ca2+]c rise, as in β-cells. • The second one is an indirect inhibition mediated by δ-cells releasing somatostatin. • Somatostatin mainly reduces the efficacy of Ca2+ on exocytosis in α-cells. • Somatostatin more potently inhibits glucagon than insulin secretion.
- Published
- 2021
12. Identification of islet-enriched long non-coding RNAs contributing to β-cell failure in type 2 diabetes
- Author
-
Romano Regazzi, Ana Gómez-Ruiz, Frédéric Burdet, Marc Prentki, Patrick Gilon, Mark Ibberson, Sonia Gattesco, Jonathan L.S. Esguerra, Lena Eliasson, D. Ross Laybutt, Anna Motterle, Marie-Line Peyot, and UCL - SSS/IREC/EDIN - Pôle d'endocrinologie, diabète et nutrition
- Subjects
Male ,0301 basic medicine ,lcsh:Internal medicine ,medicine.medical_specialty ,medicine.medical_treatment ,Gene Expression ,Mice, Obese ,030209 endocrinology & metabolism ,Type 2 diabetes ,Biology ,Diet, High-Fat ,Pancreatic islet ,Transcriptome ,Islets of Langerhans ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Insulin-Secreting Cells ,Diabetes mellitus ,Internal medicine ,microRNA ,Gene expression ,medicine ,Animals ,Insulin ,Obesity ,lcsh:RC31-1245 ,Molecular Biology ,geography ,geography.geographical_feature_category ,Sequence Analysis, RNA ,Diabetes ,Cell Biology ,medicine.disease ,Islet ,Cell biology ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Diabetes Mellitus, Type 2 ,RNA, Long Noncoding ,Original Article ,Diabetes Mellitus, Type 2/genetics ,Diabetes Mellitus, Type 2/metabolism ,Gene Expression/genetics ,Insulin/metabolism ,Insulin-Secreting Cells/metabolism ,Islets of Langerhans/metabolism ,RNA, Long Noncoding/genetics ,RNA, Long Noncoding/metabolism - Abstract
Objective Non-coding RNAs constitute a major fraction of the β-cell transcriptome. While the involvement of microRNAs is well established, the contribution of long non-coding RNAs (lncRNAs) in the regulation of β-cell functions and in diabetes development remains poorly understood. The aim of this study was to identify novel islet lncRNAs differently expressed in type 2 diabetes models and to investigate their role in β-cell failure and in the development of the disease. Methods Novel transcripts dysregulated in the islets of diet-induced obese mice were identified by high throughput RNA-sequencing coupled with de novo annotation. Changes in the level of the lncRNAs were assessed by real-time PCR. The functional role of the selected lncRNAs was determined by modifying their expression in MIN6 cells and primary islet cells. Results We identified about 1500 novel lncRNAs, a number of which were differentially expressed in obese mice. The expression of two lncRNAs highly enriched in β-cells, βlinc2, and βlinc3, correlated to body weight gain and glycemia levels in obese mice and was also modified in diabetic db/db mice. The expression of both lncRNAs was also modulated in vitro in isolated islet cells by glucolipotoxic conditions. Moreover, the expression of the human orthologue of βlinc3 was altered in the islets of type 2 diabetic patients and was associated to the BMI of the donors. Modulation of the level of βlinc2 and βlinc3 by overexpression or downregulation in MIN6 and mouse islet cells did not affect insulin secretion but increased β-cell apoptosis. Conclusions Taken together, the data show that lncRNAs are modulated in a model of obesity-associated type 2 diabetes and that variations in the expression of some of them may contribute to β-cell failure during the development of the disease., Highlights • Mouse pancreatic islets express a large number of novel long non-coding RNAs. • Many long non-coding RNAs are differentially expressed in the islets of obese mice. • The level of two islet long non-coding RNAs correlates to body weight and glycemia. • The expression of these islet long non-coding RNAs is altered in Type 2 diabetes. • Altered expression of these long non-coding RNAs sensitise β-cells to apoptosis.
- Published
- 2017
13. Metallothionein 1 negatively regulates glucose-stimulated insulin secretion and is differentially expressed in conditions of beta cell compensation and failure in mice and humans
- Author
-
D. Ross Laybutt, Mohammed Bensellam, Helen E. Thomas, Evan G Pappas, Michel Abou-Samra, Heeyoung Chae, Jeng Yie Chan, Patrick Gilon, Yan-Chuan Shi, Jean-Christophe Jonas, UCL - SSS/IREC/EDIN - Pôle d'endocrinologie, diabète et nutrition, and UCL - SSS/IREC - Institut de recherche expérimentale et clinique
- Subjects
0301 basic medicine ,Blood Glucose ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Gene Expression ,030209 endocrinology & metabolism ,Type 2 diabetes ,Diet, High-Fat ,Cell Line ,Prediabetic State ,03 medical and health sciences ,Islets of Langerhans ,Mice ,0302 clinical medicine ,Endocrinology ,Downregulation and upregulation ,Diabetes mellitus ,Internal medicine ,Insulin-Secreting Cells ,Insulin Secretion ,medicine ,Internal Medicine ,Glucose homeostasis ,Animals ,Humans ,Insulin ,Prediabetes ,Obesity ,Glucose tolerance test ,medicine.diagnostic_test ,Chemistry ,Phenyl Ethers ,Glucose Tolerance Test ,medicine.disease ,Diabetes and Metabolism ,030104 developmental biology ,Glucose ,Acrylates ,Diabetes Mellitus, Type 2 ,Female ,Metallothionein ,Beta cell - Abstract
Aims/hypothesis: The mechanisms responsible for beta cell compensation in obesity and for beta cell failure in type 2 diabetes are poorly defined. The mRNA levels of several metallothionein (MT) genes are upregulated in islets from individuals with type 2 diabetes, but their role in beta cells is not clear. Here we examined: 1) the temporal changes of islet Mt1 and Mt2 gene expression in mouse models of beta cell compensation and failure; and 2) the role of Mt1 and Mt2 in beta cell function and glucose homeostasis in mice. Methods: Mt1 and Mt2 expression was assessed in islets from: (1) control lean (chow diet-fed) and diet-induced obese (high-fat diet-fed for 6 weeks) mice; (2) mouse models of prediabetes (6-week-old db/db mice) and diabetes (16-week-old db/db mice) and age-matched db/+ (control) mice; and (3) obese non-diabetic ob/ob mice (16-week-old) and age-matched ob/+ (control) mice. MT1E, MT1X and MT2A expression was assessed in islets from humans with and without type 2 diabetes. Mt1-Mt2 double-knockout (KO) mice, transgenic mice overexpressing Mt1 under the control of its natural promoter (Tg-Mt1) and corresponding control mice were also studied. In MIN6 cells, MT1 and MT2 were inhibited by small interfering RNAs. mRNA levels were assessed by real-time RT-PCR, plasma insulin and islet MT levels by ELISA, glucose tolerance by i.p. glucose tolerance tests and fasting 1 h refeeding tests, insulin tolerance by i.p. insulin tolerance tests, insulin secretion by RIA, cytosolic free Ca2+ concentration with Fura-2 leakage resistant (Fura-2 LR), cytosolic free Zn2+ concentration with Fluozin-3, and NAD(P)H by autofluorescence. Results: Mt1 and Mt2 mRNA levels were reduced in islets of murine models of beta cell compensation, whereas they were increased in diabetic db/db mice. In humans, MT1X mRNA levels were significantly upregulated in islets from individuals with type 2 diabetes in comparison with non-diabetic donors, while MT1E and MT2A mRNA levels were unchanged. Ex vivo, islet Mt1 and Mt2 mRNA and MT1 and MT2 protein levels were downregulated after culture with glucose at 10–30 mmol/l vs 2–5 mmol/l, in association with increased insulin secretion. In human islets, mRNA levels of MT1E, MT1X and MT2A were downregulated by stimulation with physiological and supraphysiological levels of glucose. In comparison with wild-type (WT) mice, Mt1-Mt2 double-KO mice displayed improved glucose tolerance in association with increased insulin levels and enhanced insulin release from isolated islets. In contrast, isolated islets from Tg-Mt1 mice displayed impaired glucose-stimulated insulin secretion (GSIS). In both Mt1-Mt2 double-KO and Tg-Mt1 models, the changes in GSIS occurred despite similar islet insulin content, rises in cytosolic free Ca2+ concentration and NAD(P)H levels, or intracellular Zn2+ concentration vs WT mice. In MIN6 cells, knockdown of MT1 but not MT2 potentiated GSIS, suggesting that Mt1 rather than Mt2 affects beta cell function. Conclusions/interpretation: These findings implicate Mt1 as a negative regulator of insulin secretion. The downregulation of Mt1 is associated with beta cell compensation in obesity, whereas increased Mt1 accompanies beta cell failure and type 2 diabetes.
- Published
- 2019
14. Paracrine nitric oxide induces expression of cardiac sarcomeric proteins in adult progenitor cells through soluble guanylyl cyclase/cyclic-guanosine monophosphate and Wnt/β-catenin inhibition
- Author
-
Hrag Esfahani, Peter Brouckaert, Ann Friart, Adrien Strapart, Stefan Janssens, Pierre Sonveaux, Patrick Gilon, Aurelia De Pauw, Ruben Martherus, Belaid Sekkali, Paul Massion, Caroline Dubroca, Valéry Payen, Emilie Andre, Jean-Luc Balligand, Jana Kmecova, Catherine Sibille, Delphine De Mulder, and Caroline Bouzin
- Subjects
Male ,0301 basic medicine ,Time Factors ,Physiology ,chemistry.chemical_compound ,Soluble Guanylyl Cyclase ,Antigens, Ly ,Myocytes, Cardiac ,Cyclic GMP ,Wnt Signaling Pathway ,Cells, Cultured ,beta Catenin ,Mice, Knockout ,biology ,GUCY1A3 ,Wnt signaling pathway ,Cell Differentiation ,Cell biology ,Nitric oxide synthase ,Adult Stem Cells ,Biochemistry ,cardiovascular system ,Female ,Cardiology and Cardiovascular Medicine ,Signal Transduction ,Sarcomeres ,Nitric Oxide Synthase Type III ,macromolecular substances ,Nitric Oxide ,Transfection ,Nitric oxide ,03 medical and health sciences ,Paracrine signalling ,Physiology (medical) ,Paracrine Communication ,Animals ,Cell Lineage ,Nitric Oxide Donors ,Cyclic guanosine monophosphate ,Dose-Response Relationship, Drug ,Immunomagnetic Separation ,technology, industry, and agriculture ,Membrane Proteins ,Coculture Techniques ,Mice, Inbred C57BL ,030104 developmental biology ,chemistry ,Catenin ,biology.protein ,Soluble guanylyl cyclase - Abstract
Aim Cardiac progenitor cells (CPC) from adult hearts can differentiate to several cell types composing the myocardium but the underlying molecular pathways are poorly characterized. We examined the role of paracrine nitric oxide (NO) in the specification of CPC to the cardiac lineage, particularly through its inhibition of the canonical Wnt/ β -catenin pathway, a critical step preceding cardiac differentiation. Methods and results Sca1 + CPC from adult mouse hearts were isolated by magnetic-activated cell sorting and clonally expanded. Pharmacologic NO donors increased their expression of cardiac myocyte-specific sarcomeric proteins in a concentration and time-dependent manner. The optimal time window for NO efficacy coincided with up-regulation of CPC expression of Gucy1a3 (coding the alpha1 subunit of guanylyl cyclase). The effect of paracrine NO was reproduced in vitro upon co-culture of CPC with cardiac myocytes expressing a transgenic NOS3 (endothelial nitric oxide synthase) and in vivo upon injection of CPC in infarcted hearts from cardiac-specific NOS3 transgenic mice. In mono- and co-cultures, this effect was abrogated upon inhibition of soluble guanylyl cyclase or nitric oxide synthase, and was lost in CPC genetically deficient in Gucy1a3 . Mechanistically, NO inhibits the constitutive activity of the canonical Wnt/ β -catenin in CPC and in cell reporter assays in a guanylyl cyclase-dependent fashion. This was paralleled with decreased expression of β -catenin and down-regulation of Wnt target genes in CPC and abrogated in CPC with a stabilized, non-inhibitable β -catenin. Conclusions Exogenous or paracrine sources of NO promote the specification towards the myocyte lineage and expression of cardiac sarcomeric proteins of adult CPC. This is contingent upon the expression and activity of the alpha1 subunit of guanylyl cyclase in CPC that is necessary for NO-mediated inhibition of the canonical Wnt/ β -catenin pathway.
- Published
- 2016
15. SERCA2 Deficiency Impairs Pancreatic β-Cell Function in Response to Diet-Induced Obesity
- Author
-
Emily Anderson-Baucum, Richard O. Day, Tatsuyoshi Kono, Wataru Yamamoto, Xin Tong, Djamel Lebeche, Patrick Gilon, Gary E. Shull, and Carmella Evans-Molina
- Subjects
Blood Glucose ,Male ,0301 basic medicine ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,ATPase ,Biology ,Diet, High-Fat ,Endoplasmic Reticulum ,Sarcoplasmic Reticulum Calcium-Transporting ATPases ,Islets of Langerhans ,Mice ,03 medical and health sciences ,Cytosol ,Insulin-Secreting Cells ,Internal medicine ,Diabetes mellitus ,Internal Medicine ,medicine ,Animals ,Homeostasis ,Insulin ,Glucose homeostasis ,Obesity ,Cell Proliferation ,Proinsulin ,geography ,geography.geographical_feature_category ,Islet ,medicine.disease ,030104 developmental biology ,Endocrinology ,Islet Studies ,Unfolded protein response ,biology.protein ,Calcium ,Insulin Resistance - Abstract
The sarcoendoplasmic reticulum (ER) Ca2+ ATPase 2 (SERCA2) pump is a P-type ATPase tasked with the maintenance of ER Ca2+ stores. Whereas β-cell SERCA2 expression is reduced in diabetes, the role of SERCA2 in the regulation of whole-body glucose homeostasis has remained uncharacterized. To this end, SERCA2 heterozygous mice (S2HET) were challenged with a high-fat diet (HFD) containing 45% of kilocalories from fat. After 16 weeks of the HFD, S2HET mice were hyperglycemic and glucose intolerant, but adiposity and insulin sensitivity were not different between HFD-fed S2HET mice and HFD-fed wild-type controls. Consistent with a defect in β-cell function, insulin secretion, glucose-induced cytosolic Ca2+ mobilization, and the onset of steady-state glucose-induced Ca2+ oscillations were impaired in HFD-fed S2HET islets. Moreover, HFD-fed S2HET mice exhibited reduced β-cell mass and proliferation, altered insulin production and proinsulin processing, and increased islet ER stress and death. In contrast, SERCA2 activation with a small molecule allosteric activator increased ER Ca2+ storage and rescued tunicamycin-induced β-cell death. In aggregate, these data suggest a critical role for SERCA2 and the regulation of ER Ca2+ homeostasis in the β-cell compensatory response to diet-induced obesity.
- Published
- 2016
16. Cocaine- and amphetamine-regulated transcript: a novel regulator of energy homeostasis expressed in a subpopulation of pancreatic islet cells
- Author
-
Patrick Gilon
- Subjects
0301 basic medicine ,Cart ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Nerve Tissue Proteins ,030209 endocrinology & metabolism ,Biology ,Glucagon ,Cocaine and amphetamine regulated transcript ,Energy homeostasis ,Islets of Langerhans ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Internal medicine ,Internal Medicine ,medicine ,Animals ,Homeostasis ,Humans ,Insulin ,Glucose homeostasis ,geography ,geography.geographical_feature_category ,Brain ,Islet ,030104 developmental biology ,Endocrinology ,Diabetes Mellitus, Type 2 ,Hyperglycemia - Abstract
Type 2 diabetes is characterised by chronic hyperglycaemia and its incidence is highly increased by exaggerated food consumption. It results from a lack of insulin action/production, but growing evidence suggests that it might also involve hyperglucagonaemia and impaired control of glucose homeostasis by the brain. In recent years, the cocaine and amphetamine-regulated transcript (CART) peptides have generated a lot of interest in the battle against obesity because, via the brain, they exert anorexic effects and they increase energy expenditure. They are also localised, outside the brain, in discrete regions of the body and play a hormonal role in controlling various functions. In this issue of Diabetologia, the Wierup group (doi: 10.1007/s00125-016-4020-6 ) shows that CART peptides are expressed heterogeneously in islet cells of various species, including humans, and that their expression is upregulated in diabetes. The authors also shine a spotlight on some interesting effects of CART peptides on islet function, including stimulation of insulin secretion and inhibition of glucagon release. CART peptides would thus be at the centre of a cooperation between the brain and the endocrine pancreas to control glucose homeostasis. Although the mechanisms of action of CART peptides remain enigmatic because no specific receptor for these peptides has so far been discovered, their potential therapeutic use is evident and represents a new challenge for future research.
- Published
- 2016
17. Inter-domain tagging implicates caveolin-1 in insulin receptor trafficking and Erk signaling bias in pancreatic beta-cells
- Author
-
Patrick Gilon, Ivan R. Nabi, James D. Johnson, Søs Skovsø, Gareth E. Lim, Shu Nan Li, Tobias Boothe, Haoning Cen, and Micah Piske
- Subjects
0301 basic medicine ,lcsh:Internal medicine ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Insulin resistance ,Pancreatic islet beta-cells ,Insulin receptor substrate ,medicine ,lcsh:RC31-1245 ,Receptor ,Molecular Biology ,Protein kinase B ,Insulin receptor internalization ,GRB10 ,Cell Biology ,medicine.disease ,IRS2 ,Cell biology ,Insulin receptor ,030104 developmental biology ,Biochemistry ,030220 oncology & carcinogenesis ,biology.protein ,Original Article ,Autocrine insulin signaling - Abstract
Objective The role and mechanisms of insulin receptor internalization remain incompletely understood. Previous trafficking studies of insulin receptors involved fluorescent protein tagging at their termini, manipulations that may be expected to result in dysfunctional receptors. Our objective was to determine the trafficking route and molecular mechanisms of functional tagged insulin receptors and endogenous insulin receptors in pancreatic beta-cells. Methods We generated functional insulin receptors tagged with pH-resistant fluorescent proteins between domains. Confocal, TIRF and STED imaging revealed a trafficking pattern of inter-domain tagged insulin receptors and endogenous insulin receptors detected with antibodies. Results Surprisingly, interdomain-tagged and endogenous insulin receptors in beta-cells bypassed classical Rab5a- or Rab7-mediated endocytic routes. Instead, we found that removal of insulin receptors from the plasma membrane involved tyrosine-phosphorylated caveolin-1, prior to trafficking within flotillin-1-positive structures to lysosomes. Multiple methods of inhibiting caveolin-1 significantly reduced Erk activation in vitro or in vivo, while leaving Akt signaling mostly intact. Conclusions We conclude that phosphorylated caveolin-1 plays a role in insulin receptor internalization towards lysosomes through flotillin-1-positive structures and that caveolin-1 helps bias physiological beta-cell insulin signaling towards Erk activation., Highlights • Insulin receptors are tagged between domains to maintain the functionality and endogenous intracellular trafficking patterns. • Insulin receptors have virtually no co-localization with classical endocytosis markers, but co-localize with Cav11. • Insulin receptor internalization and anti-apoptotic Erk signaling are modulated by phosphorylated Cav1.
- Published
- 2016
- Full Text
- View/download PDF
18. Glucose Acutely Reduces Cytosolic and Mitochondrial H
- Author
-
Jean-Philippe, Deglasse, Leticia Prates, Roma, Daniel, Pastor-Flores, Patrick, Gilon, Tobias P, Dick, and Jean-Christophe, Jonas
- Subjects
Male ,Cytosol ,Glucose ,Insulin-Secreting Cells ,Animals ,Hydrogen Peroxide ,Rats, Wistar ,Oxidation-Reduction ,Mitochondria ,Rats - Published
- 2018
19. Somatostatin Is Only Partly Required for the Glucagonostatic Effect of Glucose but Is Necessary for the Glucagonostatic Effect of K
- Author
-
Bao-Khanh, Lai, Heeyoung, Chae, Ana, Gómez-Ruiz, Panpan, Cheng, Paola, Gallo, Nancy, Antoine, Christophe, Beauloye, Jean-Christophe, Jonas, Victor, Seghers, Susumu, Seino, and Patrick, Gilon
- Subjects
Mice, Knockout ,Islets of Langerhans ,Mice ,Glucose ,KATP Channels ,Potassium Channel Blockers ,Animals ,Glucagon ,Somatostatin ,Pancreas - Abstract
The mechanisms of control of glucagon secretion are largely debated. In particular, the paracrine role of somatostatin (SST) is unclear. We studied its role in the control of glucagon secretion by glucose and K
- Published
- 2017
20. Steviol glycosides enhance pancreatic beta-cell function and taste sensation by potentiation of TRPM5 channel activity
- Author
-
Silvia Pinto, William Sones, Nancy Antoine, Katleen Lemaire, Koenraad Philippaert, Chantal Mathieu, Andy Pironet, Patrik Rorsman, Patrick Gilon, Margot Mesuere, Conny Gysemans, Eva Cuypers, Rudi Vennekens, Frans Schuit, Sara Kerselaers, Andrei Segal, Laura Vermeiren, Jos Laureys, Jan Tytgat, Karel Talavera, Thomas Voets, and UCL - SSS/IREC/EDIN - Pôle d'endocrinologie, diabète et nutrition
- Subjects
0301 basic medicine ,Blood Glucose ,Male ,Taste ,Patch-Clamp Techniques ,Science ,General Physics and Astronomy ,TRPM Cation Channels ,Steviol ,Umami ,Diet, High-Fat ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Glucosides ,stomatognathic system ,Taste receptor ,Insulin-Secreting Cells ,Insulin Secretion ,Animals ,Humans ,Insulin ,TRPM5 ,Stevioside ,Mice, Knockout ,Multidisciplinary ,Chemistry ,digestive, oral, and skin physiology ,food and beverages ,General Chemistry ,Stevia rebaudiana ,030104 developmental biology ,HEK293 Cells ,Biochemistry ,Diabetes Mellitus, Type 2 ,Sweetening Agents ,Female ,Diterpenes, Kaurane ,Rebaudioside A ,030217 neurology & neurosurgery - Abstract
Steviol glycosides (SGs), such as stevioside and rebaudioside A, are natural, non-caloric sweet-tasting organic molecules, present in extracts of the scrub plant Stevia rebaudiana, which are widely used as sweeteners in consumer foods and beverages. TRPM5 is a Ca2+-activated cation channel expressed in type II taste receptor cells and pancreatic β-cells. Here we show that stevioside, rebaudioside A and their aglycon steviol potentiate the activity of TRPM5. We find that SGs potentiate perception of bitter, sweet and umami taste, and enhance glucose-induced insulin secretion in a Trpm5-dependent manner. Daily consumption of stevioside prevents development of high-fat-diet-induced diabetic hyperglycaemia in wild-type mice, but not in Trpm5−/− mice. These results elucidate a molecular mechanism of action of SGs and identify TRPM5 as a potential target to prevent and treat type 2 diabetes., Steviol glycosides are sweet-tasting compounds isolated from a South American shrub and are increasingly used as sweeteners in foods and beverages. Philippaert et al. demonstrate that steviol glycosides potentiate Ca2+-dependent TRPM5 activity and promote glucose-induced insulin secretion and glucose tolerance.
- Published
- 2017
21. AMPK activation by glucagon-like peptide-1 prevents NADPH oxidase activation induced by hyperglycemia in adult cardiomyocytes
- Author
-
Chantal Dessy, Gaetane Behets-Wydemans, Anne Van Steenbergen, Jean-Louis Vanoverschelde, Aurélie Timmermans, Magali Balteau, Sandrine Horman, Luc Bertrand, Christophe Beauloye, Patrick Gilon, Diego Castanares-Zapatero, Nicolas Tajeddine, and Louis Hue
- Subjects
Male ,medicine.medical_specialty ,Physiology ,Glucose uptake ,Thiophenes ,AMP-Activated Protein Kinases ,Phenformin ,chemistry.chemical_compound ,AMP-activated protein kinase ,Glucagon-Like Peptide 1 ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Hypoglycemic Agents ,Myocytes, Cardiac ,Rats, Wistar ,Cells, Cultured ,Protein Kinase C ,Membrane Glycoproteins ,NADPH oxidase ,biology ,Biphenyl Compounds ,Glucose transporter ,Methylglucosides ,NADPH Oxidases ,AMPK ,Glucose analog ,Angiotensin II ,Rats ,Cell biology ,Protein Transport ,Glucose ,Endocrinology ,chemistry ,Pyrones ,NADPH Oxidase 2 ,biology.protein ,Reactive Oxygen Species ,Cardiology and Cardiovascular Medicine - Abstract
Exposure of cardiomyocytes to high glucose concentrations (HG) stimulates reactive oxygen species (ROS) production by NADPH oxidase (NOX2). NOX2 activation is triggered by enhanced glucose transport through a sodium-glucose cotransporter (SGLT) but not by a stimulation of glucose metabolism. The aim of this work was to identify potential therapeutic approaches to counteract this glucotoxicity. In cultured adult rat cardiomyocytes incubated with 21 mM glucose (HG), AMP-activated protein kinase (AMPK) activation by A769662 or phenformin nearly suppressed ROS production. Interestingly, glucagon-like peptide 1 (GLP-1), a new antidiabetic drug, concomitantly induced AMPK activation and prevented the HG-mediated ROS production (maximal effect at 100 nM). α2-AMPK, the major isoform expressed in cardiomyocytes (but not α1-AMPK), was activated in response to GLP-1. Anti-ROS properties of AMPK activators were not related to changes in glucose uptake or glycolysis. Using in situ proximity ligation assay, we demonstrated that AMPK activation prevented the HG-induced p47phox translocation to caveolae, whatever the AMPK activators used. NOX2 activation by either α-methyl-d-glucopyranoside, a glucose analog transported through SGLT, or angiotensin II was also counteracted by GLP-1. The crucial role of AMPK in limiting HG-mediated NOX2 activation was demonstrated by overexpressing a constitutively active form of α2-AMPK using adenoviral infection. This overexpression prevented NOX2 activation in response to HG, whereas GLP-1 lost its protective action in α2-AMPK-deficient mouse cardiomyocytes. Under HG, the GLP-1/AMPK pathway inhibited PKC-β2 phosphorylation, a key element mediating p47phox translocation. In conclusion, GLP-1 induces α2-AMPK activation and blocks HG-induced p47phox translocation to the plasma membrane, thereby preventing glucotoxicity.
- Published
- 2014
22. Glucose regulation of glucagon secretion
- Author
-
Patrick Gilon, Erik Gylfe, and UCL - SSS/IREC/EDIN - Pôle d'endocrinologie, diabète et nutrition
- Subjects
endocrine system ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Glucagon ,Islets of Langerhans ,Endocrinology ,Diabetes mellitus ,Internal medicine ,Internal Medicine ,Animals ,Humans ,Medicine ,business.industry ,Pancreatic islets ,Insulin ,digestive, oral, and skin physiology ,Glucagon secretion ,General Medicine ,medicine.disease ,Glucose ,medicine.anatomical_structure ,Somatostatin ,Sweetening Agents ,Blood sugar regulation ,business ,hormones, hormone substitutes, and hormone antagonists ,Hyperglucagonemia - Abstract
Glucagon secreted by pancreatic α-cells is the major hyperglycemic hormone correcting acute hypoglycaemia (glucose counterregulation). In diabetes the glucagon response to hypoglycaemia becomes compromised and chronic hyperglucagonemia appears. There is increasing awareness that glucagon excess may underlie important manifestations of diabetes. However opinions differ widely how glucose controls glucagon secretion. The autonomous nervous system plays an important role in the glucagon response to hypoglycaemia. But it is clear that glucose controls glucagon secretion also by mechanisms involving direct effects on α-cells or indirect effects via paracrine factors released from non-α-cells within the pancreatic islets. The present review discusses these mechanisms and argues that different regulatory processes are involved in a glucose concentration-dependent manner. Direct glucose effects on the α-cell and autocrine mechanisms are probably most significant for the glucagon response to hypoglycaemia. During hyperglycaemia, when secretion from β- and δ-cells is stimulated, paracrine inhibitory factors generate pulsatile glucagon release in opposite phase to pulsatile release of insulin and somatostatin. High concentrations of glucose have also stimulatory effects on glucagon secretion that tend to balance and even exceed the inhibitory influence. The latter actions might underlie the paradoxical hyperglucagonemia that aggravates hyperglycaemia in persons with diabetes.
- Published
- 2014
23. Tolbutamide Controls Glucagon Release From Mouse Islets Differently Than Glucose
- Author
-
Heeyoung Chae, Rui Cheng-Xue, Laura A. Noël, Patrick Gilon, Magalie A. Ravier, Ana Gómez-Ruiz, Frans Schuit, Nancy Antoine, and Fabrice Chimienti
- Subjects
Somatostatin-Secreting Cells ,endocrine system ,medicine.medical_specialty ,Receptors, Drug ,Tolbutamide ,Endocrinology, Diabetes and Metabolism ,Zinc Transporter 8 ,Carbohydrate metabolism ,Biology ,Sulfonylurea Receptors ,Glucagon ,Tissue Culture Techniques ,Islets of Langerhans ,Mice ,KATP Channels ,Insulin-Secreting Cells ,Membrane Transport Modulators ,Internal medicine ,Potassium Channel Blockers ,Internal Medicine ,medicine ,Diazoxide ,Animals ,Hypoglycemic Agents ,Potassium Channels, Inwardly Rectifying ,Cation Transport Proteins ,Crosses, Genetic ,Original Research ,Mice, Knockout ,Delta cell ,Osmolar Concentration ,Glucagon secretion ,Glucose ,Somatostatin ,Endocrinology ,Islet Studies ,Commentary ,Sulfonylurea receptor ,ATP-Binding Cassette Transporters ,hormones, hormone substitutes, and hormone antagonists ,medicine.drug - Abstract
We evaluated the role of ATP-sensitive K+ (KATP) channels, somatostatin, and Zn2+ in the control of glucagon secretion from mouse islets. Switching from 1 to 7 mmol/L glucose inhibited glucagon release. Diazoxide did not reverse the glucagonostatic effect of glucose. Tolbutamide decreased glucagon secretion at 1 mmol/L glucose (G1) but stimulated it at 7 mmol/L glucose (G7). The reduced glucagon secretion produced by high concentrations of tolbutamide or diazoxide, or disruption of KATP channels (Sur1−/− mice) at G1 could be inhibited further by G7. Removal of the somatostatin paracrine influence (Sst−/− mice or pretreatement with pertussis toxin) strongly increased glucagon release, did not prevent the glucagonostatic effect of G7, and unmasked a marked glucagonotropic effect of tolbutamide. Glucose inhibited glucagon release in the absence of functional KATP channels and somatostatin signaling. Knockout of the Zn2+ transporter ZnT8 (ZnT8−/− mice) did not prevent the glucagonostatic effect of glucose. In conclusion, glucose can inhibit glucagon release independently of Zn2+, KATP channels, and somatostatin. Closure of KATP channels controls glucagon secretion by two mechanisms, a direct stimulation of α-cells and an indirect inhibition via somatostatin released from δ-cells. The net effect on glucagon release results from a balance between both effects.
- Published
- 2013
24. In Situ Electrophysiological Examination of Pancreatic α Cells in the Streptozotocin-Induced Diabetes Model, Revealing the Cellular Basis of Glucagon Hypersecretion
- Author
-
Pedro Luis Herrera, Negar Karimian, Patrick Gilon, Ya-Chi Huang, Marjan Slak Rupnik, Herbert Y. Gaisano, and Zhong-Ping Feng
- Subjects
Action Potentials/physiology ,endocrine system diseases ,Endocrinology, Diabetes and Metabolism ,Hyperglycemia/blood/physiopathology ,Action Potentials ,Stimulation ,Voltage-Gated Sodium Channels ,Glucagon-Secreting Cells/pathology/physiology ,Mice ,0302 clinical medicine ,Exocytosis/physiology ,ddc:576.5 ,Cells, Cultured ,0303 health sciences ,Chemistry ,Potassium channel ,medicine.anatomical_structure ,Diabetes Mellitus, Experimental/physiopathology ,Potassium Channels, Voltage-Gated ,Pancreas ,medicine.drug ,medicine.medical_specialty ,Potassium Channels, Voltage-Gated/physiology ,Glucagon/analysis/blood/secretion ,030209 endocrinology & metabolism ,Glucagon ,Exocytosis ,Diabetes Mellitus, Experimental ,Glucose Intolerance/physiopathology ,03 medical and health sciences ,Diabetes Mellitus, Type 1/chemically induced/physiopathology ,Diabetes mellitus ,Internal medicine ,Glucose Intolerance ,Internal Medicine ,medicine ,Animals ,030304 developmental biology ,Secretory Vesicles ,Sodium channel ,nutritional and metabolic diseases ,Secretory Vesicles/physiology ,medicine.disease ,Streptozotocin ,Electrophysiological Phenomena ,Diabetes Mellitus, Type 1 ,Endocrinology ,Islet Studies ,Glucagon-Secreting Cells ,Hyperglycemia ,Voltage-Gated Sodium Channels/physiology ,Hyperglucagonemia - Abstract
Early-stage type 1 diabetes (T1D) exhibits hyperglucagonemia by undefined cellular mechanisms. Here we characterized α-cell voltage-gated ion channels in a streptozotocin (STZ)-induced diabetes model that lead to increased glucagon secretion mimicking T1D. GYY mice expressing enhanced yellow fluorescence protein in α cells were used to identify α cells within pancreas slices. Mice treated with low-dose STZ exhibited hyperglucagonemia, hyperglycemia, and glucose intolerance, with 71% reduction of β-cell mass. Although α-cell mass of STZ-treated mice remained unchanged, total pancreatic glucagon content was elevated, coinciding with increase in size of glucagon granules. Pancreas tissue slices enabled in situ examination of α-cell electrophysiology. α cells of STZ-treated mice exhibited the following: 1) increased exocytosis (serial depolarization-induced capacitance), 2) enhanced voltage-gated Na+ current density, 3) reduced voltage-gated K+ current density, and 4) increased action potential (AP) amplitude and firing frequency. Hyperglucagonemia in STZ-induced diabetes is thus likely due to increased glucagon content arising from enlarged glucagon granules and increased AP firing frequency and amplitude coinciding with enhanced Na+ and reduced K+ currents. These alterations may prime α cells in STZ-treated mice for more glucagon release per cell in response to low glucose stimulation. Thus, our study provides the first insight that STZ treatment sensitizes release mechanisms of α cells.
- Published
- 2013
25. Subplasmalemmal Ca2+ measurements in mouse pancreatic beta cells support the existence of an amplifying effect of glucose on insulin secretion
- Author
-
Amy E. Palmer, R. Cheng-Xue, Jean-Claude Henquin, Patrick Gilon, and Magalie A. Ravier
- Subjects
medicine.medical_specialty ,Ratón ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,chemistry.chemical_element ,In Vitro Techniques ,Calcium ,Biology ,Article ,Mice ,Insulin-Secreting Cells ,Internal medicine ,Insulin Secretion ,Internal Medicine ,medicine ,Animals ,Insulin ,Secretion ,Cells, Cultured ,Pancreatic hormone ,geography ,Microscopy, Confocal ,geography.geographical_feature_category ,Islet ,Immunohistochemistry ,Cytosol ,Glucose ,Endocrinology ,Microscopy, Fluorescence ,chemistry ,Female ,Beta cell - Abstract
Glucose-induced insulin secretion is attributed to a rise of beta cell cytosolic free [Ca(2+)] ([Ca(2+)](c)) (triggering pathway) and amplification of the action of Ca(2+). This concept of amplification rests on observations that glucose can increase Ca(2+)-induced insulin secretion without further elevating an imposed already high [Ca(2+)](c). However, it remains possible that this amplification results from an increase in [Ca(2+)] just under the plasma membrane ([Ca(2+)](SM)), which escaped detection by previous measurements of global [Ca(2+)](c). This was the hypothesis that we tested here by measuring [Ca(2+)](SM).The genetically encoded Ca(2+) indicators D3-cpv (untargeted) and LynD3-cpv (targeted to plasma membrane) were expressed in clusters of mouse beta cells. LynD3-cpv was also expressed in beta cells within intact islets. [Ca(2+)](SM) changes were monitored using total internal reflection fluorescence microscopy. Insulin secretion was measured in parallel.Beta cells expressing D3cpv or LynD3cpv displayed normal [Ca(2+)] changes and insulin secretion in response to glucose. Distinct [Ca(2+)](SM) fluctuations were detected during repetitive variations of KCl between 30 and 32-35 mmol/l, attesting to the adequate sensitivity of our system. When the amplifying pathway was evaluated (high KCl + diazoxide), increasing glucose from 3 to 15 mmol/l consistently lowered [Ca(2+)](SM) while stimulating insulin secretion approximately two fold. Blocking Ca(2+) uptake by the endoplasmic reticulum largely attenuated the [Ca(2+)](SM) decrease produced by high glucose but did not unmask localised [Ca(2+)](SM) increases.Glucose can increase Ca(2+)-induced insulin secretion without causing further elevation of beta cell [Ca(2+)](SM). The phenomenon is therefore a true amplification of the triggering action of Ca(2+).
- Published
- 2010
26. Insulin crystallization depends on zinc transporter ZnT8 expression, but is not required for normal glucose homeostasis in mice
- Author
-
Katleen Lemaire, Fabrice Chimienti, Patrick Gilon, John W.M. Creemers, Magalie A. Ravier, Guy A. Rutter, Etienne Waelkens, P. In't Veld, L. Van Lommel, Mikaela Granvik, R Van de Plas, Anica Schraenen, Frans Schuit, and UCL - MD/FSIO - Département de physiologie et pharmacologie
- Subjects
medicine.medical_specialty ,medicine.medical_treatment ,Zinc Transporter 8 ,Carbohydrate metabolism ,Islets of Langerhans ,Mice ,Microscopy, Electron, Transmission ,Diabetes mellitus ,Internal medicine ,Glucose Intolerance ,medicine ,Animals ,Insulin ,Glucose homeostasis ,Cation Transport Proteins ,Mice, Knockout ,Multidisciplinary ,diabetes ,biology ,SLC30A8 ,zinc ,Biological Sciences ,medicine.disease ,Insulin oscillation ,Mice, Inbred C57BL ,Zinc ,Insulin receptor ,Glucose ,Endocrinology ,biology.protein ,Calcium ,dense core granule ,Crystallization - Abstract
Zinc co-crystallizes with insulin in dense core secretory granules, but its role in insulin biosynthesis, storage and secretion is unknown. In this study we assessed the role of the zinc transporter ZnT8 using ZnT8-knockout ( ZnT8 −/− ) mice. Absence of ZnT8 expression caused loss of zinc release upon stimulation of exocytosis, but normal rates of insulin biosynthesis, normal insulin content and preserved glucose-induced insulin release. Ultrastructurally, mature dense core insulin granules were rare in ZnT8 −/− beta cells and were replaced by immature, pale insulin “progranules,” which were larger than in ZnT8 +/+ islets. When mice were fed a control diet, glucose tolerance and insulin sensitivity were normal. However, after high-fat diet feeding, the ZnT8 −/− mice became glucose intolerant or diabetic, and islets became less responsive to glucose. Our data show that the ZnT8 transporter is essential for the formation of insulin crystals in beta cells, contributing to the packaging efficiency of stored insulin. Interaction between the ZnT8 −/− genotype and diet to induce diabetes is a model for further studies of the mechanism of disease of human ZNT8 gene mutations.
- Published
- 2009
27. Glucose and Pharmacological Modulators of ATP-Sensitive K+ Channels Control [Ca2+]c by Different Mechanisms in Isolated Mouse α-Cells
- Author
-
Patrick Gilon, Rui Cheng-Xue, Mélanie C. Beauvois, Jean-Claude Henquin, Nicolas Quoix, Ziad Zeinoun, Laurine Mattart, Yves Guiot, UCL - MD/MNOP - Département de morphologie normale et pathologique, UCL - (SLuc) Service d'anatomie pathologique, UCL - MD/FSIO - Département de physiologie et pharmacologie, and UCL - (SLuc) Service d'endocrinologie et de nutrition
- Subjects
Azides ,medicine.medical_specialty ,Tolbutamide ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,chemistry.chemical_element ,Mice, Inbred Strains ,Calcium ,Biology ,gamma-Aminobutyric acid ,Mice ,chemistry.chemical_compound ,KATP Channels ,Insulin-Secreting Cells ,Internal medicine ,Internal Medicine ,medicine ,Diazoxide ,Animals ,gamma-Aminobutyric Acid ,Insulin ,Glucagon secretion ,Glucose ,Endocrinology ,Islet Studies ,chemistry ,L-Glucose ,Glucagon-Secreting Cells ,Nimodipine ,NAD+ kinase ,NADP ,medicine.drug - Abstract
OBJECTIVE: We studied how glucose and ATP-sensitive K(+) (K(ATP)) channel modulators affect alpha-cell [Ca(2+)](c). RESEARCH DESIGN AND METHODS: GYY mice (expressing enhanced yellow fluorescent protein in alpha-cells) and NMRI mice were used. [Ca(2+)](c), the K(ATP) current (I(KATP), perforated mode) and cell metabolism [NAD(P)H fluorescence] were monitored in single alpha-cells and, for comparison, in single beta-cells. RESULTS: In 0.5 mmol/l glucose, [Ca(2+)](c) oscillated in some alpha-cells and was basal in the others. Increasing glucose to 15 mmol/l decreased [Ca(2+)](c) by approximately 30% in oscillating cells and was ineffective in the others. alpha-Cell I(KATP) was inhibited by tolbutamide and activated by diazoxide or the mitochondrial poison azide, as in beta-cells. Tolbutamide increased alpha-cell [Ca(2+)](c), whereas diazoxide and azide abolished [Ca(2+)](c) oscillations. Increasing glucose from 0.5 to 15 mmol/l did not change I(KATP) and NAD(P)H fluorescence in alpha-cells in contrast to beta-cells. The use of nimodipine showed that L-type Ca(2+) channels are the main conduits for Ca(2+) influx in alpha-cells. gamma-Aminobutyric acid and zinc did not decrease alpha-cell [Ca(2+)](c), and insulin, although lowering [Ca(2+)](c) very modestly, did not affect glucagon secretion. CONCLUSIONS: alpha-Cells display similarities with beta-cells: K(ATP) channels control Ca(2+) influx mainly through L-type Ca(2+) channels. However, alpha-cells have distinct features from beta-cells: Most K(ATP) channels are already closed at low glucose, glucose does not affect cell metabolism and I(KATP), and it slightly decreases [Ca(2+)](c). Hence, glucose and K(ATP) channel modulators exert distinct effects on alpha-cell [Ca(2+)](c). The direct small glucose-induced drop in alpha-cell [Ca(2+)](c) contributes likely only partly to the strong glucose-induced inhibition of glucagon secretion in islets.
- Published
- 2009
28. Glucose Acutely Decreases pH of Secretory Granules in Mouse Pancreatic Islets
- Author
-
Jean-Claude Henquin, Patrick Stiernet, Yves Guiot, and Patrick Gilon
- Subjects
medicine.medical_specialty ,Methylamine ,Insulin ,medicine.medical_treatment ,Pancreatic islets ,Granule (cell biology) ,Cell Biology ,Biology ,Biochemistry ,Exocytosis ,Insulin oscillation ,Cytosol ,chemistry.chemical_compound ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Internal medicine ,medicine ,Secretion ,Molecular Biology - Abstract
Glucose-induced insulin secretion requires a rise in beta-cell cytosolic Ca2+ ([Ca2+]c) that triggers exocytosis and a mechanistically unexplained amplification of the action of [Ca2+]c. Insulin granules are kept acidic by luminal pumping of protons with simultaneous Cl- uptake to maintain electroneutrality. Experiments using patched, dialyzed beta-cells prompted the suggestion that acute granule acidification by glucose underlies amplification of insulin secretion. However, others found glucose to increase granular pH in intact islets. In this study, we measured islet granular pH with Lysosensor DND-160, a fluorescent dye that permits ratiometric determination of pH methylamine (weak base) > Cl- omission. Concanamycin and methylamine did not alter glucose-induced [Ca2+]c increase in islets but strongly inhibited the two phases of insulin secretion. Omission of Cl- did not affect the first phase but decreased the second phase of both [Ca2+]c and insulin responses. Neither experimental condition affected the [Ca2+]c rise induced by 30 mM KCl, but the insulin responses were inhibited by concanamycin > methylamine and not affected by Cl- omission. The amplification of insulin secretion by glucose was not suppressed. We conclude that an acidic granular pH is important for insulin secretion but that the acute further acidification produced by glucose is not essential for the augmentation of secretion via the amplifying pathway.
- Published
- 2006
29. Atypical Ca2+-induced Ca2+release from a sarco-endoplasmic reticulum Ca2+-ATPase 3-dependent Ca2+pool in mouse pancreatic β-cells
- Author
-
Abdelilah Arredouani, Jean-Christophe Jonas, Jean-Claude Henquin, Jean François Rolland, Patrick Gilon, F. Schuit, and Mélanie C. Beauvois
- Subjects
medicine.medical_specialty ,Thapsigargin ,Physiology ,Ryanodine receptor ,Endoplasmic reticulum ,Depolarization ,Stimulation ,Biology ,Ryanodine receptor 2 ,chemistry.chemical_compound ,Endocrinology ,chemistry ,Internal medicine ,cardiovascular system ,medicine ,Diazoxide ,Inositol ,medicine.drug - Abstract
The contribution of Ca(2+) release from intracellular stores to the rise in the free cytosolic Ca(2+) concentration ([Ca(2+)](c)) triggered by Ca(2+) influx was investigated in mouse pancreatic beta-cells. Depolarization of beta-cells by 45 mm K(+) (in the presence of 15 mm glucose and 0.1 mm diazoxide) evoked two types of [Ca(2+)](c) responses: a monotonic and sustained elevation; or a sustained elevation superimposed by a transient [Ca(2+)](c) peak (TCP) (40-120 s after the onset of depolarization). Simultaneous measurements of [Ca(2+)](c) and voltage-dependent Ca(2+) current established that the TCP did not result from a larger Ca(2+) current. Abolition of the TCP by thapsigargin and its absence in sarco-endoplasmic reticulum Ca(2+)-ATPase 3 (SERCA3) knockout mice show that it is caused by Ca(2+) mobilization from the endoplasmic reticulum. A TCP could not be evoked by the sole depolarization of beta-cells but required a rise in [Ca(2+)](c) pointing to a Ca(2+)-induced Ca(2+) release (CICR). This CICR did not involve inositol 1,4,5-trisphosphate (IP(3)) receptors (IP(3)Rs) because it was resistant to heparin. Nor did it involve ryanodine receptors (RyRs) because it persisted after blockade of RyRs with ryanodine, and was not mimicked by caffeine, a RyR agonist. Moreover, RyR1 and RyR2 mRNA were not found and RyR3 mRNA was only slightly expressed in purified beta-cells. A CICR could also be detected in a limited number of cells in response to glucose. Our data demonstrate, for the first time in living cells, the existence of an atypical CICR that is independent from the IP(3)R and the RyR. This CICR is prominent in response to a supraphysiological stimulation with high K(+), but plays little role in response to glucose in non-obese mouse pancreatic beta-cells.
- Published
- 2004
30. Hierarchy of the β-cell signals controlling insulin secretion
- Author
-
Jean-Claude Henquin, Patrick Gilon, Myriam Nenquin, Jean-Christophe Jonas, and Magalie A. Ravier
- Subjects
medicine.medical_specialty ,Chemistry ,Insulin ,medicine.medical_treatment ,Clinical Biochemistry ,General Medicine ,Carbohydrate metabolism ,Biochemistry ,Endocrinology ,Internal medicine ,Extracellular ,medicine ,Glucose homeostasis ,Hormone metabolism ,Secretion ,Beta cell ,Intracellular - Abstract
The main function of pancreatic β cells is to synthesize and secrete insulin at appropriate rates to limit blood glucose fluctuations within a narrow range. Any alteration in β -cell functioning has a profound impact on glucose homeostasis: excessive secretion of insulin causes hypoglycaemia, and insufficient secretion leads to diabetes. It is therefore not surprising that insulin secretion is subject to very tight control. This control is primarily ensured by glucose itself but also involves an array of metabolic, neural, hormonal and sometimes pharmacological factors (Fig. 1). To integrate all these stimulatory and inhibitory influences, β cells rely on an astonishingly complex stimulus-secretion coupling. This review discusses how the hierarchy between two intracellular pathways, producing triggering and amplifying signals [1], optimizes adequate insulin secretion to changes in blood glucose concentration and enables the β cell to grade the numerous extracellular messages that it receives.
- Published
- 2003
31. SERCA3 Ablation Does Not Impair Insulin Secretion but Suggests Distinct Roles of Different Sarcoendoplasmic Reticulum Ca2+ Pumps for Ca2+ Homeostasis in Pancreatic β-cells
- Author
-
Lynne H. Liu, Yves Guiot, Frank Wuytack, Jacques Rahier, Abdelilah Arredouani, José A. Pertusa, Jean François Rolland, Gary E. Shull, Jean-Claude Henquin, Myriam Nenquin, Patrick Gilon, Martine Stevens, Jean-Christophe Jonas, UCL - MD/FSIO - Département de physiologie et pharmacologie, and UCL - MD/MNOP - Département de morphologie normale et pathologique
- Subjects
medicine.medical_specialty ,Time Factors ,Thapsigargin ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Mice, Inbred Strains ,Calcium-Transporting ATPases ,Biology ,Sarcoplasmic Reticulum Calcium-Transporting ATPases ,Islets of Langerhans ,Mice ,chemistry.chemical_compound ,Internal medicine ,Insulin Secretion ,Internal Medicine ,medicine ,Animals ,Homeostasis ,Insulin ,Glucose homeostasis ,Calcium Signaling ,Mice, Knockout ,Calcium metabolism ,Reverse Transcriptase Polymerase Chain Reaction ,Endoplasmic reticulum ,Pancreatic islets ,Exons ,Immunohistochemistry ,Isoenzymes ,Kinetics ,Glucose ,Endocrinology ,medicine.anatomical_structure ,chemistry ,L-Glucose ,Calcium - Abstract
Two sarcoendoplasmic reticulum Ca(2+)-ATPases, SERCA3 and SERCA2b, are expressed in pancreatic islets. Immunocytochemistry showed that SERCA3 is restricted to beta-cells in the mouse pancreas. Control and SERCA3-deficient mice were used to evaluate the role of SERCA3 in beta-cell cytosolic-free Ca(2+) concentration ([Ca(2+)](c)) regulation, insulin secretion, and glucose homeostasis. Basal [Ca(2+)](c) was not increased by SERCA3 ablation. Stimulation with glucose induced a transient drop in basal [Ca(2+)](c) that was suppressed by inhibition of all SERCAs with thapsigargin (TG) but unaffected by selective SERCA3 ablation. Ca(2+) mobilization by acetylcholine was normal in SERCA3-deficient beta-cells. In contrast, [Ca(2+)](c) oscillations resulting from intermittent glucose-stimulated Ca(2+) influx and [Ca(2+)](c) transients induced by pulses of high K(+) were similarly affected by SERCA3 ablation or TG pretreatment of control islets; their amplitude was increased and their slow descending phase suppressed. This suggests that, during the decay of each oscillation, the endoplasmic reticulum releases Ca(2+) that was pumped by SERCA3 during the upstroke phase. SERCA3 ablation increased the insulin response of islets to 15 mmol/l glucose. However, basal and postprandial plasma glucose and insulin concentrations in SERCA3-deficient mice were normal. In conclusion, SERCA2b, but not SERCA3, is involved in basal [Ca(2+)](c) regulation in beta-cells. SERCA3 becomes operative when [Ca(2+)](c) rises and is required for normal [Ca(2+)](c) oscillations in response to glucose. However, a lack of SERCA3 is insufficient in itself to alter glucose homeostasis or impair insulin secretion in mice.
- Published
- 2002
32. G Protein-independent Activation of an Inward Na+Current by Muscarinic Receptors in Mouse Pancreatic β-Cells
- Author
-
Patrick Gilon, Jean-Claude Henquin, and Jean-François Rolland
- Subjects
medicine.medical_specialty ,Patch-Clamp Techniques ,Biochemistry ,Sodium Channels ,Membrane Potentials ,Islets of Langerhans ,Mice ,GTP-Binding Proteins ,Internal medicine ,Muscarinic acetylcholine receptor M5 ,Muscarinic acetylcholine receptor ,medicine ,Animals ,Reversal potential ,Molecular Biology ,Cells, Cultured ,Chemistry ,Sodium ,Cell Biology ,Muscarinic acetylcholine receptor M1 ,Hyperpolarization (biology) ,Receptors, Muscarinic ,Acetylcholine ,Endocrinology ,Biophysics ,Ligand-gated ion channel ,Calcium ,Alpha-4 beta-2 nicotinic receptor ,medicine.drug - Abstract
Depolarization of pancreatic beta-cells is critical for stimulation of insulin secretion by acetylcholine but remains unexplained. Using voltage-clamped beta-cells, we identified a small inward current produced by acetylcholine, which was suppressed by atropine or external Na(+) omission, but was not mimicked by nicotine, and was insensitive to nicotinic antagonists, tetrodotoxin, 4,4'-diisothiocyanostilbene-2,2'-disulfonic acid (DiDS), thapsigargin pretreatment, and external Ca(2+) and K(+) removal. This suggests that muscarinic receptor stimulation activates voltage-insensitive Na(+) channels distinct from store-operated channels. No outward Na(+) current was produced by acetylcholine when the electrochemical Na(+) gradient was reversed, indicating that the channels are inward rectifiers. No outward K(+) current occurred either, and the reversal potential of the current activated by acetylcholine in the presence of Na(+) and K(+) was close to that expected for a Na(+)-selective membrane, suggesting that the channels opened by acetylcholine are specific for Na(+). Overnight pretreatment with pertussis toxin or the addition of guanosine 5'-O-(3-thiotriphosphate) (GTP-gamma-S) or guanosine-5'-O-(2-thiodiphosphate) (GDP-beta-S) instead of GTP to the pipette solution did not alter this current, excluding involvement of G proteins. Injection of a current of a similar amplitude to that induced by acetylcholine elicited electrical activity in beta-cells perifused with a subthreshold glucose concentration. These results demonstrate that muscarinic receptor activation in pancreatic beta-cells triggers, by a G protein-independent mechanism, a selective Na(+) current that explains the plasma membrane depolarization.
- Published
- 2002
33. Contribution of the endoplasmic reticulum to the glucose-induced [Ca2+]cresponse in mouse pancreatic islets
- Author
-
Patrick Gilon, Abdelilah Arredouani, and Jean-Claude Henquin
- Subjects
medicine.medical_specialty ,Thapsigargin ,Physiology ,Vasodilator Agents ,Endocrinology, Diabetes and Metabolism ,chemistry.chemical_element ,Mice, Inbred Strains ,In Vitro Techniques ,Biology ,Calcium ,Endoplasmic Reticulum ,Membrane Potentials ,Islets of Langerhans ,Mice ,chemistry.chemical_compound ,Physiology (medical) ,Internal medicine ,medicine ,Diazoxide ,Extracellular ,Animals ,Calcium Signaling ,Enzyme Inhibitors ,Cation Transport Proteins ,Adenosine Triphosphatases ,Membrane potential ,Endoplasmic reticulum ,Pancreatic islets ,Depolarization ,Electrophysiology ,Glucose ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Potassium ,Female ,medicine.drug - Abstract
Thapsigargin (TG), a blocker of Ca2+uptake by the endoplasmic reticulum (ER), was used to evaluate the contribution of the organelle to the oscillations of cytosolic Ca2+concentration ([Ca2+]c) induced by repetitive Ca2+influx in mouse pancreatic β-cells. Because TG depolarized the plasma membrane in the presence of glucose alone, extracellular K+was alternated between 10 and 30 mM in the presence of diazoxide to impose membrane potential (MP) oscillations. In control islets, pulses of K+, mimicking regular MP oscillations elicited by 10 mM glucose, induced [Ca2+]coscillations whose nadir remained higher than basal [Ca2+]c. Increasing the depolarization phase of the pulses while keeping their frequency constant (to mimic the effects of a further rise of the glucose concentration on MP) caused an upward shift of the nadir of [Ca2+]coscillations that was reproduced by raising extracellular Ca2+(to increase Ca2+influx) without changing the pulse protocol. In TG-pretreated islets, the imposed [Ca2+]coscillations were of much larger amplitude than in control islets and occurred on basal levels. During intermittent trains of depolarizations, control islets displayed mixed [Ca2+]coscillations characterized by a summation of fast oscillations on top of slow ones, whereas no progressive summation of the fast oscillations was observed in TG-pretreated islets. In conclusion, the buffering capacity of the ER in pancreatic β-cells limits the amplitude of [Ca2+]coscillations and may explain how the nadir between oscillations remains above baseline during regular oscillations or gradually increases during mixed [Ca2+]coscillations, two types of response observed during glucose stimulation.
- Published
- 2002
34. How stable is repression of disallowed genes in pancreatic islets in response to metabolic stress?
- Author
-
Patrick Gilon, Peter In 'T Veld, Anica Schraenen, Leentje Van Lommel, Frans Schuit, Lotte Goyvaerts, Katleen Lemaire, Mikaela Granvik, Ana Gómez-Ruiz, Medical Biochemistry, Pathology/molecular and cellular medicine, Diabetes Pathology & Therapy, Experimental Pathology, and UCL - SSS/IREC/EDIN - Pôle d'endocrinologie, diabète et nutrition
- Subjects
0301 basic medicine ,Aging ,Maternal Health ,Gene Expression ,lcsh:Medicine ,Mice, Obese ,Biochemistry ,DNA Methyltransferase 3A ,Fats ,Endocrinology ,Insulin-Secreting Cells ,Medicine and Health Sciences ,Insulin ,Histone code ,DNA (Cytosine-5-)-Methyltransferases ,lcsh:Science ,DNA methylation ,Multidisciplinary ,Obstetrics and Gynecology ,Animal Models ,Lipids ,Phenotype ,Type 2 Diabetes ,Housekeeping gene ,Histone Code ,Histone ,medicine.anatomical_structure ,Experimental Organism Systems ,diabetes mellitus ,Epigenetics ,Female ,pregnancy ,Research Article ,medicine.medical_specialty ,mice ,Endocrine Disorders ,Mouse Models ,Biology ,Research and Analysis Methods ,Diet, High-Fat ,03 medical and health sciences ,Model Organisms ,Stress, Physiological ,Internal medicine ,Genetics ,Journal Article ,medicine ,Animals ,Psychological repression ,Nutrition ,Diabetic Endocrinology ,Delta cell ,Pancreatic islets ,lcsh:R ,Biology and Life Sciences ,Glucose Tolerance Test ,Hormones ,Diet ,Mice, Inbred C57BL ,030104 developmental biology ,Diet and Type 2 Diabetes ,Metabolic Disorders ,biology.protein ,Women's Health ,lcsh:Q - Abstract
The specific phenotype of mature differentiated beta cells not only depends on the specific presence of genes that allow beta cell function but also on the selective absence of housekeeping genes (“disallowed genes”) that would interfere with this function. Recent studies have shown that both histone modifications and DNA methylation via the de novo methyltransferase DNMT3A are involved in repression of disallowed genes in neonatal beta cells when these cells acquire their mature phenotype. It is unknown, however, if the environmental influence of advanced age, pregnancy and the metabolic stress of high fat diet or diabetes could alter the repression of disallowed genes in beta cells. In the present study, we show that islet disallowed genes—which are also deeply repressed in FACS-purified beta cells—remain deeply repressed in animals of advanced age and in pregnant females. Moreover, the stability of this repression was correlated with strong and stable histone repression marks that persisted in islets isolated from 2 year old mice and with overall high expression of Dnmt3a in islets. Furthermore, repression of disallowed genes was unaffected by the metabolic stress of high fat diet. However, repression of about half of the disallowed genes was weakened in 16 week-old diabetic db/db mice. In conclusion, we show that the disallowed status of islet genes is stable under physiological challenging conditions (advanced age, pregnancy, high fat diet) but partially lost in islets from diabetic animals.
- Published
- 2017
35. Involvement of nitric oxide in iodine deficiency-induced microvascular remodeling in the thyroid gland: role of nitric oxide synthase 3 and ryanodine receptors
- Author
-
Jessica Vanderstraeten, Patrick Gilon, Anne Catherine Gérard, Marie-Christine Many, Julie Craps, B. De Jongh, Pierre Sonveaux, Ides M. Colin, Jean-Luc Balligand, Cindy Wilvers, Virginie Joris, and Irina Lobysheva
- Subjects
Male ,Vascular Endothelial Growth Factor A ,medicine.medical_specialty ,Thapsigargin ,Nitric Oxide Synthase Type III ,medicine.medical_treatment ,Thyroid Gland ,Nitric Oxide ,Ryanodine receptor 2 ,Nitric oxide ,Cell Line ,chemistry.chemical_compound ,Mice ,Endocrinology ,In vivo ,Internal medicine ,medicine ,Animals ,Humans ,RYR1 ,biology ,Ryanodine receptor ,Growth factor ,Ryanodine Receptor Calcium Release Channel ,Hypoxia-Inducible Factor 1, alpha Subunit ,Rats ,Nitric oxide synthase ,chemistry ,cardiovascular system ,biology.protein ,Calcium ,Female ,Reactive Oxygen Species ,Iodine - Abstract
Iodine deficiency (ID) induces microvascular changes in the thyroid gland via a TSH-independent reactive oxygen species-hypoxia inducible factor (HIF)-1α-vascular endothelial growth factor (VEGF) pathway. The involvement of nitric oxide (NO) in this pathway and the role of calcium (Ca(2+)) and of ryanodine receptors (RYRs) in NO synthase 3 (NOS3) activation were investigated in a murine model of goitrogenesis and in three in vitro models of ID including primary cultures of human thyrocytes. ID activated NOS3 and the production of NO in thyrocytes in vitro and increased the thyroid blood flow in vivo. Using bevacizumab (a blocking antibody against VEGF-A) in mice, it appeared that NOS3 is activated upstream of VEGF-A. L-nitro arginine methyl ester (L-NAME, a NOS inhibitor) blocked the ID-induced increase in thyroid blood flow in vivo and NO production in vitro, as well as ID-induced VEGF-A mRNA and HIF-1α expression in vitro, while S-nitroso-acetyl-penicillamine (SNAP, a NO donor) did the opposite. Ca(2+) is involved in this pathway as intracellular Ca(2+) flux increased after ID, and thapsigargin activated NOS3 and increased VEGF-A mRNA expression. Two of the three known mammalian RYR isoforms (RYR1 and RYR2) were shown to be expressed in thyrocytes. RYR inhibition using ryanodine at 10 μ M decreased ID-induced NOS3 activation, HIF-1α and VEGF-A expression, while RYR activation with ryanodine at 1nM increased NOS3 activation and VEGF-A mRNA expression. In conclusion, during the early phase of TSH-independent ID-induced microvascular activation, ID sequentially activates RYRs and NOS3, thereby supporting ID-induced activation of the NO/HIF-1α/VEGF-A pathway in thyrocytes.
- Published
- 2014
36. Pancreatic and duodenal homeobox protein 1 (Pdx-1) maintains endoplasmic reticulum calcium levels through transcriptional regulation of sarco-endoplasmic reticulum calcium ATPase 2b (SERCA2b) in the islet β cell
- Author
-
Leslie S. Satin, Tatsuyoshi Kono, Xin Tong, Matthew J. Merrins, Patrick Gilon, Angel Zarain-Herzberg, Wataru Yamamoto, Carmella Evans-Molina, and Justin S. Johnson
- Subjects
endocrine system ,medicine.medical_specialty ,XBP1 ,animal structures ,ATPase ,Immunoblotting ,Gene Expression ,Endoplasmic Reticulum ,digestive system ,Biochemistry ,Sarcoplasmic Reticulum Calcium-Transporting ATPases ,Mice ,Internal medicine ,Cell Line, Tumor ,Insulin-Secreting Cells ,medicine ,Diabetes Mellitus ,Animals ,Humans ,Molecular Biology ,Transcription factor ,Homeodomain Proteins ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Endoplasmic reticulum ,Cell Biology ,Mice, Mutant Strains ,Cell biology ,Calcium ATPase ,Mice, Inbred C57BL ,Endocrinology ,HEK293 Cells ,Metabolism ,Gene Expression Regulation ,Microscopy, Fluorescence ,Unfolded protein response ,biology.protein ,cardiovascular system ,NIH 3T3 Cells ,Trans-Activators ,PDX1 ,Calcium ,RNA Interference ,Chromatin immunoprecipitation - Abstract
Although the pancreatic duodenal homeobox 1 (Pdx-1) transcription factor is known to play an indispensable role in β cell development and secretory function, recent data also implicate Pdx-1 in the maintenance of endoplasmic reticulum (ER) health. The sarco-endoplasmic reticulum Ca(2+) ATPase 2b (SERCA2b) pump maintains a steep Ca(2+) gradient between the cytosol and ER lumen. In models of diabetes, our data demonstrated loss of β cell Pdx-1 that occurs in parallel with altered SERCA2b expression, whereas in silico analysis of the SERCA2b promoter revealed multiple putative Pdx-1 binding sites. We hypothesized that Pdx-1 loss under inflammatory and diabetic conditions leads to decreased SERCA2b levels and activity with concomitant alterations in ER health. To test this, siRNA-mediated knockdown of Pdx-1 was performed in INS-1 cells. The results revealed reduced SERCA2b expression and decreased ER Ca(2+), which was measured using fluorescence lifetime imaging microscopy. Cotransfection of human Pdx-1 with a reporter fused to the human SERCA2 promoter increased luciferase activity 3- to 4-fold relative to an empty vector control, and direct binding of Pdx-1 to the proximal SERCA2 promoter was confirmed by chromatin immunoprecipitation. To determine whether restoration of SERCA2b could rescue ER stress induced by Pdx-1 loss, Pdx1(+/-) mice were fed a high-fat diet. Isolated islets demonstrated an increased spliced-to-total Xbp1 ratio, whereas SERCA2b overexpression reduced the Xbp1 ratio to that of wild-type controls. Together, these results identify SERCA2b as a novel transcriptional target of Pdx-1 and define a role for altered ER Ca(2+) regulation in Pdx-1-deficient states.
- Published
- 2014
37. Inhibition of Protein Synthesis Sequentially Impairs Distinct Steps of Stimulus-secretion Coupling in Pancreatic β Cells1
- Author
-
Patrick Gilon, Jean-Claude Henquin, María José García-Barrado, Jean-François Rolland, Myriam Nenquin, and Magalie A. Ravier
- Subjects
medicine.medical_specialty ,Insulin ,medicine.medical_treatment ,Carbohydrate metabolism ,Cycloheximide ,Biology ,Exocytosis ,chemistry.chemical_compound ,Endocrinology ,Tolbutamide ,chemistry ,Internal medicine ,medicine ,Secretion ,Glycolysis ,NAD+ kinase ,medicine.drug - Abstract
Proteins with a short half-life are potential sites of pancreatic ss cell dysfunction under pathophysiological conditions. In this study, mouse islets were used to establish which step in the regulation of insulin secretion is most sensitive to inhibition of protein synthesis by 10 microM cycloheximide (CHX). Although islet protein synthesis was inhibited approximately 95% after 1 h, the inhibition of insulin secretion was delayed and progressive. After long (18-20 h) CHX-treatment, the strong (80%) inhibition of glucose-, tolbutamide-, and K(+)-induced insulin secretion was not due to lower insulin stores, to any marked impairment of glucose metabolism or to altered function of K(+)-ATP channels (total K(+)-ATP currents were however decreased). It was partly caused by a decreased Ca(2+) influx (whole-cell Ca(2+) current) resulting in a smaller rise in cytosolic Ca(2+) ([Ca(2+)](i)). The situation was very different after short (2-5 h) CHX-treatment. Insulin secretion was 50-60% inhibited although islet glucose metabolism was unaffected and stimulus-induced [Ca(2+)](i) rise was not (2 h) or only marginally (5 h) decreased. The efficiency of Ca(2+) on secretion was thus impaired. The inhibition of insulin secretion by 15 h of CHX treatment was more slowly reversible (>4 h) than that of protein synthesis. This reversibility of secretion was largely attributable to recovery of a normal Ca(2+) efficiency. In conclusion, inhibition of protein synthesis in islets inhibits insulin secretion in two stages: a rapid decrease in the efficiency of Ca(2+) on exocytosis, followed by a decrease in the Ca(2+) signal mediated by a slower loss of functional Ca(2+) channels. Glucose metabolism and the regulation of K(+)-ATP channels are more resistant. Proteins with a short half-life appear to be important to ensure optimal Ca(2+) effects on exocytosis, and are the potential Achille's heel of stimulus-secretion coupling.
- Published
- 2001
38. Alterations of insulin secretion from mouse islets treated with sulphonylureas: perturbations of Ca2+regulation prevail over changes in insulin content
- Author
-
Patrick Gilon, Jean-Claude Henquin, and Marcello Anello
- Subjects
Pharmacology ,medicine.medical_specialty ,Insulin ,medicine.medical_treatment ,Pancreatic islets ,Carbohydrate metabolism ,Biology ,Insulin oscillation ,Glibenclamide ,Endocrinology ,medicine.anatomical_structure ,Tolbutamide ,Internal medicine ,medicine ,Diazoxide ,Blood sugar regulation ,medicine.drug - Abstract
1. To determine how pretreatment with sulphonylureas alters the beta cell function, mouse islets were cultured (18 - 20 h) without (controls) or with (test) 0.01 microM glibenclamide. Acute responses to glucose were then determined in the absence of glibenclamide. 2. Test islets were insensitive to drugs (sulphonylureas and diazoxide) acting on K+-ATP channels, and their [Ca2+]i was already elevated in the absence of stimulation. 3. Insulin secretion was increased in the absence of glucose, and mainly stimulated between 0 - 10 instead of 7 - 20 mM glucose in controls. The maximum response was halved, but this difference disappeared after correction for the 45% decrease in the islet insulin content. 4. The first phase of glucose-induced insulin secretion was abrogated because of a paradoxical decrease of the high basal [Ca2+]i in beta cells. The second phase was preserved but occurred with little rise of [Ca2+]i. These abnormalities did not result from alterations of glucose metabolism (NADPH fluorescence). 5. In islets cultured with 50 microM tolbutamide, glucose induced biphasic increases in [Ca2+]i and insulin secretion. The decrease in the secretory response was matched by the decrease in insulin content (45%) except at maximal glucose concentrations. Islets pretreated with tolbutamide, however, behaved like those cultured with glibenclamide if tolbutamide was also present during the acute functional tests. 6. In conclusion, treatment with a low glibenclamide concentration causes long-lasting blockade of K+-ATP channels and rise of [Ca2+]i in beta cells. Glucose-induced insulin secretion occurs at lower concentrations, is delayed and is largely mediated by a modulation of Ca2+ action on exocytosis. It is suggested that glucose regulation of insulin secretion mainly depends on a K+-ATP channel-independent pathway during in vivo sulphonylurea treatment.
- Published
- 1999
39. G protein-dependent inhibition of L-type Ca2+ currents by acetylcholine in mouse pancreatic B-cells
- Author
-
Jean-Claude Henquin, P Rorsman, Patrick Gilon, Jesper Gromada, Yu Zhu, Jerrel Yakel, UCL - MD/FSIO - Département de physiologie et pharmacologie, and UCL - (SLuc) Service d'endocrinologie et de nutrition
- Subjects
Agonist ,Dose-Response Relationship, Drug ,GTP' ,Physiology ,medicine.drug_class ,G protein ,Stereochemistry ,Cholera toxin ,Inhibitory postsynaptic potential ,medicine.disease_cause ,Acetylcholine ,Mice ,EGTA ,chemistry.chemical_compound ,chemistry ,GTP-Binding Proteins ,Muscarinic acetylcholine receptor ,medicine ,Biophysics ,Animals ,Calcium Channels ,Pancreas ,Research Article ,medicine.drug - Abstract
1. The effect of acetylcholine (ACh) on voltage-dependent Ca2+ currents in mouse pancreatic B-cells was studied using the whole-cell configuration of the patch-clamp technique. 2. ACh (0.25-250 microM) reversibly and dose-dependently inhibited the Ca2+ current elicited by depolarizations from -80 mV to +10 mV. Maximal inhibition was observed at concentrations > 25 microM where it amounted to approximately 35%. The effect was voltage independent and prevented by atropine (10 microM) suggesting that it was mediated by muscarinic receptors. 3. The inhibitory action of ACh on the Ca2+ current was abolished when the cytoplasmic solution contained GDP beta S (2 mM) and became irreversible when the non-hydrolysable GTP analogue GTP gamma S (10 microM) was included in the pipette. This indicates the participation of G proteins in the inhibitory effect of ACh but pretreatment of the cells with either pertussis or cholera toxin failed to prevent the effect of ACh on the Ca2+ current. 4. ACh remained equally effective as an inhibitor of the whole-cell Ca2+ current in the presence of the L-type Ca2+ channel agonist (-)-Bay K 8644 and after partial inhibition of the current by nifedipine. Addition of omega-agatoxin IVA, omega-conotoxin GVIA or omega-conotoxin MVIIC neither affected the peak Ca2+ current amplitude nor the extent of inhibition produced by ACh. These pharmacological properties indicate that ACh acts by inhibiting L-type Ca2+ channels. 5. The inhibitory action of ACh on the B-cell Ca2+ current was not secondary to elevation of [Ca2+]i and ACh remained equally effective as an inhibitor when Ba2+ was used as the charge carrier, when [Ca2+]i was buffered to low concentrations using EGTA and under experimental conditions preventing the mobilization of Ca2+ from intracellular stores. 6. These results suggest that ACh reduces the whole-cell Ca2+ current in the B-cell through a G protein-regulated, voltage- and Ca(2+)-independent inhibition of L-type Ca2+ channels.
- Published
- 1997
40. No evidence for a role of reverse Na(+)-Ca2+ exchange in insulin release from mouse pancreatic islets
- Author
-
Myriam Nenquin, Patrick Gilon, María José García-Barrado, Yoshihiko Sato, and Jean-Claude Henquin
- Subjects
medicine.medical_specialty ,Physiology ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Membrane Potentials ,Islets of Langerhans ,Mice ,Culture Techniques ,Physiology (medical) ,Internal medicine ,Insulin Secretion ,medicine ,Diazoxide ,Extracellular ,Animals ,Insulin ,Na+/K+-ATPase ,Membrane potential ,Ion Transport ,Chemistry ,Pancreatic islets ,Sodium ,Hyperpolarization (biology) ,Endocrinology ,medicine.anatomical_structure ,Calcium ,Intracellular ,medicine.drug - Abstract
We studied whether reverse Na(+)-Ca2+ exchange can increase cytoplasmic Ca2+ ([Ca2+]i) in mouse islets and contribute to insulin release. The exchange was stimulated by replacing Na+ with choline, sucrose, or lithium in a medium containing 15 mM glucose. Na+ omission increased electrical activity in B cells, [Ca2+]i, and insulin release. When voltage-dependent Ca2+ channels were blocked by nimodipine or closed by holding the membrane polarized with diazoxide, Na+ omission caused a slight hyperpolarization, a small rise in [Ca2+]i, and a marginal increase in insulin release (the latter only with choline). This small rise in [Ca2+]i was dependent on extracellular Ca2+ but was hardly augmented when intracellular Na+ was raised with alanine. When B cells were depolarized by 30 mM K+, Na+ omission did not affect the membrane potential but increased [Ca2+]i and insulin release. If Ca2+ channels were blocked by nimodipine, only marginal increases in Ca2+ and insulin release persisted, which were not different from those observed when the cells were not depolarized. This indicates that Ca2+ influx through voltage-dependent Ca2+ channels rather than via reverse Na(+)-Ca2+ exchange underlies the rise in [Ca2+]i and in insulin release produced by Na+ removal. No decisive support for Ca2+ influx by reverse Na(+)-Ca2+ exchange could be found.
- Published
- 1996
41. Abstracts of the Summer Meeting 6 June 1996, Louvain-en-Woluwé, Belgium
- Author
-
Sonia Brichard, Myriam Nenquin, F. Kolar, A. Houvenaghel, I. Mountian, G. Maréchal, Lumbe Ongemba, Pierre Lekeux, W. Van Driessche, Luc Leybaert, J. Weyne, D. Marechal, D. Votion, Jean-Claude Henquin, B. S. Kelemen, H. Duvivier, Dominique J. Becker, Patrick Gilon, Lei Zheng, André Boland, E. Van Erck, Giovanna Mancuso, Willy Eechaute, Mehdi Maleki, Yoshikazu Miura, Bénédicte Reul, Jan Tytgat, N. C. Gao, P Calders, Theophile Godfraind, Sandrine Vandenput, S. Tomasovic, Maurice Wibo, K. Paemeleire, V. Rousseau, Tatiana Art, Ezio Tirelli, F. Frankenne, Jean-Marc Kaufman, Albert Dresse, E. Wechsung, C. Lambillotte, Olivier Feron, V. Decostre, W. Dhooghe, D. Delapierre, E. Lacroix, Ludwig Missiaen, P. Daenens, and D. H. Duvivier
- Subjects
Physiology ,Physiology (medical) ,Clinical Biochemistry ,Library science ,Human physiology ,Biology - Published
- 1996
42. Ketoisocaproic acid and leucine increase cytoplasmic pH in mouse pancreatic B cells: role of cytoplasmic Ca2+ and pH-regulating exchangers
- Author
-
R. M. Shepherd, Jean-Claude Henquin, and Patrick Gilon
- Subjects
Cytoplasm ,medicine.medical_specialty ,Sodium-Hydrogen Exchangers ,Fura-2 ,Mice, Inbred Strains ,Antiporters ,Islets of Langerhans ,Mice ,chemistry.chemical_compound ,Endocrinology ,Leucine ,Internal medicine ,medicine ,Diazoxide ,Animals ,Insulin ,Chloride-Bicarbonate Antiporters ,HEPES ,Osmolar Concentration ,Metabolism ,Hydrogen-Ion Concentration ,Keto Acids ,Sodium–hydrogen antiporter ,chemistry ,DIDS ,Calcium ,Female ,NAD+ kinase ,Hydrogen ,medicine.drug - Abstract
The effects of nonglucose nutrient insulin secretagogues on cytoplasmic pH (pHi) in pancreatic B cells are unclear. These were studied with intact mouse islets loaded with BCECF and stimulated with ketoisocaproic acid (KIC) or leucine, which, unlike glucose, are exclusively metabolized in mitochondria. The changes in pHi were compared to those in cytoplasmic Ca2+ ([Ca2+]i; islets loaded with fura-2), metabolism [NAD(P)H fluorescence], and insulin release. In HCO3- buffer containing 3 mM glucose, 10 mM KIC produced a rapid and sustained increase in metabolism, [Ca2+]i, pHi, and insulin release. In HEPES buffer, the increase in metabolism, [Ca2+]i, and release were also rapid but not as sustained, whereas the alkalinization was delayed. The changes in release thus follow a time course more similar to that of [Ca2+]i and metabolism than to that of pHi. The role of [Ca2+]i in pHi changes was next examined. A similar rapid rise in pHi was produced by KIC in both HCO3- and HEPES buffers when its effects on [Ca2+]i were prevented, whether [Ca2+]i was kept low (4.8 mM KCl plus diazoxide) or high (30 mM KCl plus diazoxide). When the Na(+):H+ exchanger was blocked by dimethylamiloride, the alkalinizing effect of KIC was unaffected in HCO3- buffer, indicating that it does not result from an activation of this exchanger. In HEPES buffer, however, KIC strongly decreased pHi unless the rise in [Ca2+]i was prevented, in which case KIC increased pHi. When the HCO3-/Cl-exchanger was blocked by 4,4'-diisothiocyanostilbene-2,2'-disulfonic acid (DIDS), the effect of KIC in HCO3- buffer became similar to that in HEPES buffer without or with DIDS. The effects of leucine on pHi were similar to those of KIC. In conclusion, the effect of KIC and leucine on islet cell pHi, like that of glucose, is the complex result of an alkalinizing action of their metabolism and an acidifying action of the [Ca2+]i rise that they also produce. Compensation of this acidification is achieved by operation of the pHi-regulating exchangers, of which the HCO3-/Cl- exchanger plays a predominant role.
- Published
- 1996
43. Distinct effects of glucose on the synchronous oscillations of insulin release and cytoplasmic Ca2+ concentration measured simultaneously in single mouse islets
- Author
-
Patrick Gilon and Jean-Claude Henquin
- Subjects
Cytoplasm ,medicine.medical_specialty ,Fura-2 ,medicine.medical_treatment ,Stimulation ,Biology ,Islets of Langerhans ,Mice ,chemistry.chemical_compound ,Endocrinology ,Internal medicine ,Insulin Secretion ,medicine ,Animals ,Insulin ,Secretion ,B cell ,Calcium metabolism ,geography ,geography.geographical_feature_category ,Islet ,Insulin oscillation ,Glucose ,medicine.anatomical_structure ,chemistry ,Calcium - Abstract
There exists a good temporal correlation between the oscillations of cytoplasmic Ca2+ ([Ca2+]i) in pancreatic B cells and insulin release. Here, single mouse islets loaded with fura-2 were used to investigate whether there also exists a quantitative correlation between both events during stimulation by different glucose concentrations. The frequency of [Ca2+]i oscillations was decreased by raising the CaCl2 concentration to 10 mM in the perifusion medium (to ensure adequate resolution of insulin oscillations by the immunoassay), and the glucose concentration was increased from 10 to 20 or from 15 to 30 mM. Raising the glucose level was followed by changes in [Ca2+]i oscillations. Their duration increased slightly, and that of the intervals decreased, whereas the nadir between the oscillations became less deep. On the other hand, the peak of [Ca2+]i oscillations did not change. As a result, the rhythm was accelerated, and the average [Ca2+]i was increased. The concomitant increase in insulin release resulted from similar changes, with, in addition, a marked increase in the peak of insulin oscillations. In two thirds of the islets, variations in the amplitude of successive [Ca2+]i oscillations occurred during stimulation with 10 mM glucose and disappeared at higher glucose levels. This was due to temporal variations in the responsiveness of all regions of the islet, rather than to the existence of nonresponsive regions that would be recruited into an active state by high glucose. In conclusion, there exists a good temporal correlation between insulin and [Ca2+]i oscillations in islets stimulated by various glucose concentrations. The quantitative correlation is not as close, indicating that the relationship between the two phenomena is nonlinear and supporting previous evidence that glucose also increases the efficacy of Ca2+ on secretion. This mechanism, rather than the development of a Ca2+ rise in nonresponsive cells, might underlie B cell recruitment in intact islets.
- Published
- 1995
44. Muscarinic stimulation exerts both stimulatory and inhibitory effects on the concentration of cytoplasmic Ca2+ in the electrically excitable pancreatic B-cell
- Author
-
Patrick Gilon, Myriam Nenquin, Jean-Claude Henquin, and UCL - MD/FSIO - Département de physiologie et pharmacologie
- Subjects
Atropine ,Cytoplasm ,medicine.medical_specialty ,Thapsigargin ,Action Potentials ,Stimulation ,Inhibitory postsynaptic potential ,Biochemistry ,Islets of Langerhans ,Mice ,chemistry.chemical_compound ,Internal medicine ,Muscarinic acetylcholine receptor ,Diazoxide ,medicine ,Extracellular ,Animals ,Gallopamil ,Molecular Biology ,Terpenes ,Sodium ,Depolarization ,Cell Biology ,Calcium Channel Blockers ,Receptors, Muscarinic ,Acetylcholine ,Electric Stimulation ,Endocrinology ,chemistry ,Potassium ,Calcium ,Female ,Research Article ,medicine.drug - Abstract
Mouse pancreatic islets were used to investigate how muscarinic stimulation influences the cytoplasmic Ca2+ concentration ([Ca2+]i) in insulin-secreting B-cells. In the absence of extracellular Ca2+, acetylcholine (ACh) triggered a transient, concentration-dependent and thapsigargin-inhibited increase in [Ca2+]i. In the presence of extracellular Ca2+ and 15 mM glucose, ACh induced a biphasic rise in [Ca2+]i. The initial, transient phase increased with the concentration of ACh, whereas the second, sustained, phase was higher at low (0.1-1 microM) than at high (> or = 10 microM) concentrations of ACh. Thapsigargin attenuated (did not suppress) the first phase of the [Ca2+]i rise and did not affect the sustained response. This sustained rise was inhibited by omission of extracellular Na+ (which prevents the depolarizing action of ACh) and by D600 or diazoxide (which prevent activation of voltage-dependent Ca2+ channels). During steady-state stimulation, the Ca2+ action potentials in B-cells were stimulated by 1 microM ACh but inhibited by 100 microM ACh. When B-cells were depolarized by 45 mM K+, ACh induced a concentration-dependent, biphasic change in [Ca2+]i, consisting of a first peak rapidly followed by a decrease. Thapsigargin suppressed the peak without affecting the drop in [Ca2+]i. Measurements of 45Ca2+ efflux under similar conditions indicated that ACh decreases Ca2+ influx and slightly increases the efflux. All effects of ACh were blocked by atropine. In conclusion, three mechanisms at least are involved in the biphasic change in [Ca2+]i that muscarinic stimulation exerts in excitable pancreatic B-cells. A mobilization of Ca2+ from the endoplasmic reticulum contributes significantly to the first peak, but little to the steady-state rise in [Ca2+]i. This second phase results from an influx of Ca2+ through voltage-dependent Ca2+ channels activated by a Na(+)-dependent depolarization. However, when high concentrations of ACh are used, Ca2+ influx is attenuated.
- Published
- 1995
45. The Ca2+-mobilizing Actions of a Jurkat Cell Extract on Mammalian Cells and Xenopus laevis Oocytes
- Author
-
James W. Putney, Patrick Gilon, Xiaopeng Bian, Gary S. Bird, and Jerry L. Yakel
- Subjects
Cell Extracts ,Cell type ,medicine.medical_specialty ,T-Lymphocytes ,Xenopus ,chemistry.chemical_element ,Biology ,Calcium ,Lacrimal apparatus ,Biochemistry ,Jurkat cells ,Cell Line ,Xenopus laevis ,Internal medicine ,Tumor Cells, Cultured ,medicine ,Extracellular ,Animals ,Humans ,Molecular Biology ,Cells, Cultured ,Lacrimal Apparatus ,Cell Biology ,biology.organism_classification ,Cell biology ,medicine.anatomical_structure ,Endocrinology ,Liver ,chemistry ,Cell culture ,Oocytes ,Intracellular - Abstract
Randriamampita and Tsien (Randriamampita, C., and Tsien, R. Y. (1993) Nature 364, 809-814) suggested that an acid-extracted fraction from a Jurkat cell line contains a messenger responsible for the coupling of calcium entry to the depletion of intracellular stores, i.e. capacitative calcium entry. We found that the extract, prepared as described by Randriamampita and Tsien, caused Ca2+ entry in 1321N1 astrocytoma cells which was not blocked by the D-myo-1,4,5-trisphosphate-receptor antagonist, heparin. In contrast to astrocytoma cells, when applied to mouse lacrimal acinar cells and rat hepatocytes the Jurkat extract always caused the release of intracellular Ca2+, followed by Ca2+ entry across the plasma membrane. This activity of the extract on lacrimal cells was blocked by either intracellular injection of heparin or extracellular atropine. Similarly prepared lacrimal cell extracts gave Ca2+ responses when applied to astrocytoma cells or lacrimal cells which were similar to those for Jurkat-derived extract. However, extracts from hepatocytes had no effect. In most Xenopus oocytes, the Jurkat extract had no effect, while in a few oocytes, the extract gave a [Ca2+]i response similar to that seen in lacrimal cells, that is, release of Ca2+ followed by Ca2+ entry. We conclude that the actions of the Jurkat cell extract are not consistent with its containing the long sought messenger for capacitative calcium entry. It is likely that this fraction contains a number of factors that mediate Ca2+ response in different cell types, possibly through receptor-mediated mechanisms.
- Published
- 1995
46. Frequency-dependent mitochondrial Ca(2+) accumulation regulates ATP synthesis in pancreatic β cells
- Author
-
Guy A. Rutter, Francesca Semplici, Rosario Rizzuto, Daliang Li, Patrick Gilon, Magalie A. Ravier, and Andrei I. Tarasov
- Subjects
Physiology ,Signaling and Cell Physiology ,Clinical Biochemistry ,Action Potentials ,Biology ,Mitochondrion ,Endoplasmic Reticulum ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Adenosine Triphosphate ,Cytosol ,Physiology (medical) ,Insulin-Secreting Cells ,Animals ,Insulin ,Calcium Signaling ,Uniporter ,Cells, Cultured ,030304 developmental biology ,Calcium signaling ,0303 health sciences ,Voltage-dependent calcium channel ,ATP synthase ,Endoplasmic reticulum ,Insulin secretion ,Mitochondria ,Adenosine Diphosphate ,ATP ,Adenosine diphosphate ,Oscillation ,Glucose ,MCU ,chemistry ,Biochemistry ,Biophysics ,biology.protein ,Calcium ,Female ,Calcium Channels ,Adenosine triphosphate ,030217 neurology & neurosurgery - Abstract
Pancreatic β cells respond to increases in glucose concentration with enhanced metabolism, the closure of ATP-sensitive K+ channels and electrical spiking. The latter results in oscillatory Ca2+ influx through voltage-gated Ca2+ channels and the activation of insulin release. The relationship between changes in cytosolic and mitochondrial free calcium concentration ([Ca2+]cyt and [Ca2+]mit, respectively) during these cycles is poorly understood. Importantly, the activation of Ca2+-sensitive intramitochondrial dehydrogenases, occurring alongside the stimulation of ATP consumption required for Ca2+ pumping and other processes, may exert complex effects on cytosolic ATP/ADP ratios and hence insulin secretion. To explore the relationship between these parameters in single primary β cells, we have deployed cytosolic (Fura red, Indo1) or green fluorescent protein-based recombinant-targeted (Pericam, 2mt8RP for mitochondria; D4ER for the ER) probes for Ca2+ and cytosolic ATP/ADP (Perceval) alongside patch-clamp electrophysiology. We demonstrate that: (1) blockade of mitochondrial Ca2+ uptake by shRNA-mediated silencing of the uniporter MCU attenuates glucose- and essentially blocks tolbutamide-stimulated, insulin secretion; (2) during electrical stimulation, mitochondria decode cytosolic Ca2+ oscillation frequency as stable increases in [Ca2+]mit and cytosolic ATP/ADP; (3) mitochondrial Ca2+ uptake rates remained constant between individual spikes, arguing against activity-dependent regulation (“plasticity”) and (4) the relationship between [Ca2+]cyt and [Ca2+]mit is essentially unaffected by changes in endoplasmic reticulum Ca2+ ([Ca2+]ER). Our findings thus highlight new aspects of Ca2+ signalling in β cells of relevance to the actions of both glucose and sulphonylureas. Electronic supplementary material The online version of this article (doi:10.1007/s00424-012-1177-9) contains supplementary material, which is available to authorized users.
- Published
- 2012
47. Culture duration and conditions affect the oscillations of cytoplasmic calcium concentration induced by glucose in mouse pancreatic islets
- Author
-
Jean-Claude Henquin, Jean-Christophe Jonas, and Patrick Gilon
- Subjects
Cytoplasm ,medicine.medical_specialty ,Time Factors ,Endocrinology, Diabetes and Metabolism ,chemistry.chemical_element ,Calcium ,Biology ,Islets of Langerhans ,Mice ,chemistry.chemical_compound ,Internal medicine ,Mole ,Internal Medicine ,medicine ,Animals ,Beta (finance) ,Cells, Cultured ,Calcium metabolism ,geography ,geography.geographical_feature_category ,Dose-Response Relationship, Drug ,Pancreatic islets ,Islet ,Dose–response relationship ,Glucose ,Endocrinology ,medicine.anatomical_structure ,chemistry ,L-Glucose ,Female - Abstract
The pattern of the increase in cytoplasmic Cai2+ that glucose produces in beta cells has been reported to be highly variable. Here, we evaluated the influence of the culture duration (1-4 days) and conditions (5-10 mmol/l glucose) on Cai2+ in normal mouse islets stimulated by glucose. After 1 day of culture in 10 mmol/l glucose, a rise of the glucose concentration from 3 to 15 mmol/l induced a triphasic change of Cai2+ in the islets. A small initial decrease was followed by a large peak increase and then by regular fast oscillations (approximately 2.5/min). When the culture was prolonged to 2, 3 and 4 days, the initial decrease became inconsistent and the peak occurred earlier, whereas the oscillations decreased in frequency, increased in duration and eventually disappeared; on day 4 the Cai2+ rise was sustained. After culture in 5 mmol/l glucose, the pattern of Cai2+ changes induced by 15 mmol/l glucose was different. The initial decrease was very pronounced, the first peak was delayed and clearly separated from the subsequent oscillations. These were of a mixed type (fast Ca2+ transients on top of slow ones) after 1 day, and of a slow type only after 4 days. These alterations in the Cai2+ oscillations triggered by glucose could not be ascribed to desynchronization of the signal between different regions of the islets. In conclusion, culturing normal mouse islets in 5 or 10 mmol/l glucose for 1-4 days, markedly alters the characteristics of the changes in Cai2+ produced by glucose.(ABSTRACT TRUNCATED AT 250 WORDS)
- Published
- 1994
48. Dynamic measurements of mitochondrial hydrogen peroxide concentration and glutathione redox state in rat pancreatic β-cells using ratiometric fluorescent proteins: confounding effects of pH with HyPer but not roGFP1
- Author
-
Leticia Prates Roma, Jessica Duprez, Patrick Gilon, Andreas Wiederkehr, Hilton Kenji Takahashi, and Jean-Christophe Jonas
- Subjects
Male ,Antioxidant ,medicine.medical_treatment ,Green Fluorescent Proteins ,Mitochondrion ,medicine.disease_cause ,Biochemistry ,Redox ,Sensitivity and Specificity ,Green fluorescent protein ,chemistry.chemical_compound ,Insulin-Secreting Cells ,medicine ,Animals ,Humans ,Rats, Wistar ,Hydrogen peroxide ,Molecular Biology ,chemistry.chemical_classification ,Reactive oxygen species ,Osmolar Concentration ,Cell Biology ,Glutathione ,Hydrogen Peroxide ,Hydrogen-Ion Concentration ,Mitochondria ,Rats ,Kinetics ,HEK293 Cells ,chemistry ,Luminescent Measurements ,Reactive Oxygen Species ,Oxidation-Reduction ,Oxidative stress - Abstract
Using the ROS (reactive oxygen species)-sensitive fluorescent dyes dichlorodihydrofluorescein and dihydroethidine, previous studies yielded opposite results about the glucose regulation of oxidative stress in insulin-secreting pancreatic β-cells. In the present paper, we used the ratiometric fluorescent proteins HyPer and roGFP1 (redox-sensitive green fluorescent protein 1) targeted to mitochondria [mt-HyPer (mitochondrial HyPer)/mt-roGFP1 (mitochondrial roGFP1)] to monitor glucose-induced changes in mitochondrial hydrogen peroxide concentration and glutathione redox state in adenovirus-infected rat islet cell clusters. Because of the reported pH sensitivity of HyPer, the results were compared with those obtained with the mitochondrial pH sensors mt-AlpHi and mt-SypHer. The fluorescence ratio of the mitochondrial probes slowly decreased (mt-HyPer) or increased (mt-roGFP1) in the presence of 10 mmol/l glucose. Besides its expected sensitivity to H2O2, mt-HyPer was also highly pH sensitive. In agreement, changes in mitochondrial metabolism similarly affected mt-HyPer, mt-AlpHi and mt-SypHer fluorescence signals. In contrast, the mt-roGFP1 fluorescence ratio was only slightly affected by pH and reversibly increased when glucose was lowered from 10 to 2 mmol/l. This increase was abrogated by the catalytic antioxidant Mn(III) tetrakis (4-benzoic acid) porphyrin but not by N-acetyl-L-cysteine. In conclusion, due to its pH sensitivity, mt-HyPer is not a reliable indicator of mitochondrial H2O2 in β-cells. In contrast, the mt-roGFP1 fluorescence ratio monitors changes in β-cell mitochondrial glutathione redox state with little interference from pH changes. Our results also show that glucose acutely decreases rather than increases mitochondrial thiol oxidation in rat β-cells.
- Published
- 2011
49. Oscillations of secretion driven by oscillations of cytoplasmic Ca2+ as evidences in single pancreatic islets
- Author
-
Rm. Shepherd, Patrick Gilon, and Jean-Claude Henquin
- Subjects
medicine.medical_specialty ,Pulsatile insulin ,Pancreatic islets ,Insulin ,medicine.medical_treatment ,Stimulation ,Cell Biology ,Biology ,Biochemistry ,Insulin oscillation ,Tolbutamide ,Endocrinology ,medicine.anatomical_structure ,Internal medicine ,medicine ,Diazoxide ,Secretion ,Molecular Biology ,medicine.drug - Abstract
It is often assumed, but has not been demonstrated, that the oscillations of cytoplasmic calcium (Ca2+i) that occur in various secretory cells induce oscillations in secretion. Here, we have used single pancreatic islets from normal mice to monitor simultaneously insulin release and Ca2+i in beta-cells for periods up to 25 min. Ca2+i and insulin secretion were found to oscillate in synchrony during stimulation by glucose. This synchrony persisted when the frequency of both events changed spontaneously or upon addition of the hypoglycaemic sulfonylurea tolbutamide, or when their relative amplitudes varied. Repolarizing the beta-cell membrane with diazoxide abolished both Ca2+i and insulin oscillations. In contrast, epinephrine suppressed insulin oscillations in spite of the persistence of Ca2+i oscillations. This study is the first direct demonstration that, in an intact organ, sustained oscillations of Ca2+i induced by a physiological stimulus entrain synchronous oscillations of the functional response, and that both events can vary simultaneously or be dissociated depending on the experimental conditions.
- Published
- 1993
50. Activation of muscarinic receptors increases the concentration of free Na+in mouse pancreatic B-cells
- Author
-
Jean-Claude Henquin and Patrick Gilon
- Subjects
medicine.medical_specialty ,Membrane permeability ,Sodium ,Biophysics ,chemistry.chemical_element ,Tetrodotoxin ,Insulin release ,Biochemistry ,Sodium Channels ,Ouabain ,Islets of Langerhans ,Mice ,Pancreatic B-cell ,Structural Biology ,Internal medicine ,Muscarinic acetylcholine receptor ,Genetics ,medicine ,Extracellular ,Animals ,Na+/K+-ATPase ,Molecular Biology ,Muscarinic receptor ,Depolarization ,Cell Biology ,Receptors, Muscarinic ,Acetylcholine ,Endocrinology ,chemistry ,Female ,Cytosolic sodium ,medicine.drug - Abstract
The fluorescent probe SBFI was used to monitor the influence of acetylcholine (ACh) on the cytosolic concentration of free Na+ (Na+i) in single mouse pancreatic B-cells. In the presence of 3 mM glucose and 135 mM extracellular Na+, Na+i averaged 16.6 mM. ACh (100 microM) increased Na+i by approximately 80%. This rise was prevented by atropine, a blocker of muscarinic receptors, and by omission of extracellular Na+, but still occurred if the sodium pump was blocked by ouabain. It was unaffected by tetrodotoxin, a blocker of voltage-sensitive Na+ channels, and was not mimicked by depolarization of the cells with high K+. It is concluded that activation of muscarinic receptors increases the membrane permeability to Na+ in pancreatic B-cells.
- Published
- 1993
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.