Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Peptides ; 147: 170704, 2022 01.
Article in English | MEDLINE | ID: mdl-34826505

ABSTRACT

The pancreatic islets contain beta-cells and alpha-cells, which are responsible for secreting two principal gluco-regulatory hormones; insulin and glucagon, respectively. However, they also contain delta-cells, a relatively sparse cell type that secretes somatostatin (SST). These cells have a complex morphology allowing them to establish an extensive communication network throughout the islet, despite their scarcity. Delta-cells are electrically excitable cells, and SST secretion is released in a glucose- and KATP-dependent manner. SST hyperpolarises the alpha-cell membrane and suppresses exocytosis. In this way, islet SST potently inhibits glucagon release. Recent studies investigating the activity of delta-cells have revealed they are electrically coupled to beta-cells via gap junctions, suggesting the delta-cell is more than just a paracrine inhibitor. In this Review, we summarize delta-cell morphology, function, and the role of SST signalling for regulating islet hormonal output. A distinguishing feature of this Review is that we attempt to use the discovery of this gap junction pathway, together with what is already known about delta-cells, to reframe the role of these cells in both health and disease. In particular, we argue that the discovery of gap junction communication between delta-cells and beta-cells provides new insights into the contribution of delta-cells to the islet hormonal defects observed in both type 1 and type 2 diabetes. This reappraisal of the delta-cell is important as it may offer novel insights into how the physiology of this cell can be utilised to restore islet function in diabetes.


Subject(s)
Diabetes Mellitus/pathology , Gap Junctions/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/physiology , Animals , Glucagon/metabolism , Humans , Insulin/metabolism , Islets of Langerhans/ultrastructure , Somatostatin/metabolism
2.
Nat Metab ; 2(1): 32-40, 2020 01.
Article in English | MEDLINE | ID: mdl-31993555

ABSTRACT

Pancreatic islets are complex micro-organs consisting of at least three different cell types: glucagon-secreting α-, insulin-producing ß- and somatostatin-releasing δ-cells1. Somatostatin is a powerful paracrine inhibitor of insulin and glucagon secretion2. In diabetes, increased somatostatinergic signalling leads to defective counter-regulatory glucagon secretion3. This increases the risk of severe hypoglycaemia, a dangerous complication of insulin therapy4. The regulation of somatostatin secretion involves both intrinsic and paracrine mechanisms5 but their relative contributions and whether they interact remains unclear. Here we show that dapagliflozin-sensitive glucose- and insulin-dependent sodium uptake stimulates somatostatin secretion by elevating the cytoplasmic Na+ concentration ([Na+]i) and promoting intracellular Ca2+-induced Ca2+ release (CICR). This mechanism also becomes activated when [Na+]i is elevated following the inhibition of the plasmalemmal Na+-K+ pump by reductions of the extracellular K+ concentration emulating those produced by exogenous insulin in vivo 6. Islets from some donors with type-2 diabetes hypersecrete somatostatin, leading to suppression of glucagon secretion that can be alleviated by a somatostatin receptor antagonist. Our data highlight the role of Na+ as an intracellular second messenger, illustrate the significance of the intraislet paracrine network and provide a mechanistic framework for pharmacological correction of the hormone secretion defects associated with diabetes that selectively target the δ-cells.


Subject(s)
Calcium/metabolism , Sodium/metabolism , Somatostatin-Secreting Cells/metabolism , Somatostatin/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Glucagon/metabolism , Glucose/metabolism , Humans , Hypoglycemia/metabolism , Insulin/metabolism , Mice
3.
J Vis Exp ; (153)2019 11 26.
Article in English | MEDLINE | ID: mdl-31840667

ABSTRACT

Pancreatic islet hormones regulate blood glucose homeostasis. Changes in blood glucose induce oscillations of cytosolic calcium in pancreatic islet cells that trigger secretion of three main hormones: insulin (from ß-cells), glucagon (α-cells) and somatostatin (δ-cells). ß-Cells, which make up the majority of islet cells and are electrically coupled to each other, respond to the glucose stimulus as one single entity. The excitability of the minor subpopulations, α-cells and δ-cells (making up around 20% (30%) and 4% (10%) of the total rodent1 (human2) islet cell numbers, respectively) is less predictable and is therefore of special interest. Calcium sensors are delivered into the peripheral layer of cells within the isolated islet. The islet or a group of islets is then immobilized and imaged using a fluorescence microscope. The choice of the imaging mode is between higher throughput (wide-field) and better spatial resolution (confocal). Conventionally, laser scanning confocal microscopy is used for imaging tissue, as it provides the best separation of the signal between the neighboring cells. A wide-field system can be utilized too, if the contaminating signal from the dominating population of ß-cells is minimized. Once calcium dynamics in response to specific stimuli have been recorded, data are expressed in numerical form as fluorescence intensity vs. time, normalized to the initial fluorescence and baseline-corrected, to remove the effects linked to bleaching of the fluorophore. Changes in the spike frequency or partial area under the curve (pAUC) are computed vs. time, to quantify the observed effects. pAUC is more sensitive and quite robust whereas spiking frequency provides more information on the mechanism of calcium increase. Minor cell subpopulations can be identified using functional responses to marker compounds, such as adrenaline and ghrelin, that induce changes in cytosolic calcium in a specific populations of islet cells.


Subject(s)
Calcium/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Molecular Imaging/methods , Animals , Calcium Signaling , Mice , Microscopy, Fluorescence/methods
4.
J Gen Physiol ; 151(9): 1094-1115, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31358556

ABSTRACT

Somatostatin secretion from pancreatic islet δ-cells is stimulated by elevated glucose levels, but the underlying mechanisms have only partially been elucidated. Here we show that glucose-induced somatostatin secretion (GISS) involves both membrane potential-dependent and -independent pathways. Although glucose-induced electrical activity triggers somatostatin release, the sugar also stimulates GISS via a cAMP-dependent stimulation of CICR and exocytosis of somatostatin. The latter effect is more quantitatively important and in mouse islets depolarized by 70 mM extracellular K+ , increasing glucose from 1 mM to 20 mM produced an ∼3.5-fold stimulation of somatostatin secretion, an effect that was mimicked by the application of the adenylyl cyclase activator forskolin. Inhibiting cAMP-dependent pathways with PKI or ESI-05, which inhibit PKA and exchange protein directly activated by cAMP 2 (Epac2), respectively, reduced glucose/forskolin-induced somatostatin secretion. Ryanodine produced a similar effect that was not additive to that of the PKA or Epac2 inhibitors. Intracellular application of cAMP produced a concentration-dependent stimulation of somatostatin exocytosis and elevation of cytoplasmic Ca2+ ([Ca2+]i). Both effects were inhibited by ESI-05 and thapsigargin (an inhibitor of SERCA). By contrast, inhibition of PKA suppressed δ-cell exocytosis without affecting [Ca2+]i Simultaneous recordings of electrical activity and [Ca2+]i in δ-cells expressing the genetically encoded Ca2+ indicator GCaMP3 revealed that the majority of glucose-induced [Ca2+]i spikes did not correlate with δ-cell electrical activity but instead reflected Ca2+ release from the ER. These spontaneous [Ca2+]i spikes are resistant to PKI but sensitive to ESI-05 or thapsigargin. We propose that cAMP links an increase in plasma glucose to stimulation of somatostatin secretion by promoting CICR, thus evoking exocytosis of somatostatin-containing secretory vesicles in the δ-cell.


Subject(s)
Calcium/metabolism , Cyclic AMP/metabolism , Glucose/pharmacology , Pancreas/cytology , Somatostatin-Secreting Cells/drug effects , Somatostatin/metabolism , Adjuvants, Immunologic/pharmacology , Animals , Cell Membrane/physiology , Colforsin/pharmacology , Gene Expression Regulation/drug effects , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Somatostatin-Secreting Cells/metabolism , Thapsigargin/pharmacology
5.
Nat Commun ; 10(1): 139, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30635569

ABSTRACT

Hypoglycaemia (low plasma glucose) is a serious and potentially fatal complication of insulin-treated diabetes. In healthy individuals, hypoglycaemia triggers glucagon secretion, which restores normal plasma glucose levels by stimulation of hepatic glucose production. This counterregulatory mechanism is impaired in diabetes. Here we show in mice that therapeutic concentrations of insulin inhibit glucagon secretion by an indirect (paracrine) mechanism mediated by stimulation of intra-islet somatostatin release. Insulin's capacity to inhibit glucagon secretion is lost following genetic ablation of insulin receptors in the somatostatin-secreting δ-cells, when insulin-induced somatostatin secretion is suppressed by dapagliflozin (an inhibitor of sodium-glucose co-tranporter-2; SGLT2) or when the action of secreted somatostatin is prevented by somatostatin receptor (SSTR) antagonists. Administration of these compounds in vivo antagonises insulin's hypoglycaemic effect. We extend these data to isolated human islets. We propose that SSTR or SGLT2 antagonists should be considered as adjuncts to insulin in diabetes therapy.


Subject(s)
Diabetes Mellitus/pathology , Glucagon/metabolism , Hypoglycemia/pathology , Insulin/metabolism , Sodium-Glucose Transporter 2/metabolism , Somatostatin/metabolism , Animals , Benzhydryl Compounds/pharmacology , Blood Glucose/analysis , Diabetes Mellitus/drug therapy , Female , Glucagon-Secreting Cells/drug effects , Glucosides/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Insulin/genetics , Receptors, Somatostatin/antagonists & inhibitors , Sodium-Glucose Transporter 2 Inhibitors/pharmacology
6.
Lab Chip ; 18(18): 2838-2848, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30083680

ABSTRACT

High-content real-time imaging of hormone secretion in tissues or cell populations is a challenging task, which is unlikely to be resolved directly, despite immense translational value. We approach this problem indirectly, using compensatory endocytosis, a process that closely follows exocytosis in the cell, as a surrogate read-out for secretion. The tissue is immobilized in an open-air perifusion chamber and imaged using a two-photon microscope. A fluorescent polar tracer, perifused through the experimental circuit, gets trapped into the cells via endocytosis, and is quantified using a feature-detection algorithm. The signal of the tracer that accumulates into the endocytotic system reliably reflects stimulated exocytosis, which is demonstrated via co-imaging of the latter using existing reporters. A high signal-to-noise ratio and compatibility with multisensor imaging affords the real-time quantification of the secretion at the tissue/population level, whereas the cumulative nature of the signal allows imprinting of the "secretory history" within each cell. The technology works for several cell types, reflects disease progression and can be used for human tissue.


Subject(s)
Endocytosis , Hormones/metabolism , Imaging, Three-Dimensional , Animals , Humans , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Kinetics , Mice , Neurons/cytology , Neurons/metabolism , Perfusion
7.
J R Soc Interface ; 14(128)2017 03.
Article in English | MEDLINE | ID: mdl-28275121

ABSTRACT

The α-, ß- and δ-cells of the pancreatic islet exhibit different electrophysiological features. We used a large dataset of whole-cell patch-clamp recordings from cells in intact mouse islets (N = 288 recordings) to investigate whether it is possible to reliably identify cell type (α, ß or δ) based on their electrophysiological characteristics. We quantified 15 electrophysiological variables in each recorded cell. Individually, none of the variables could reliably distinguish the cell types. We therefore constructed a logistic regression model that included all quantified variables, to determine whether they could together identify cell type. The model identified cell type with 94% accuracy. This model was applied to a dataset of cells recorded from hyperglycaemic ßV59M mice; it correctly identified cell type in all cells and was able to distinguish cells that co-expressed insulin and glucagon. Based on this revised functional identification, we were able to improve conductance-based models of the electrical activity in α-cells and generate a model of δ-cell electrical activity. These new models could faithfully emulate α- and δ-cell electrical activity recorded experimentally.


Subject(s)
Electrophysiological Phenomena , Hyperglycemia/physiopathology , Islets of Langerhans/physiopathology , Models, Biological , Animals , Hyperglycemia/genetics , Mice , Mice, Knockout
8.
Ups J Med Sci ; 121(2): 113-9, 2016 May.
Article in English | MEDLINE | ID: mdl-27044683

ABSTRACT

Type 2 diabetes involves a ménage à trois of impaired glucose regulation of pancreatic hormone release: in addition to impaired glucose-induced insulin secretion, the release of the hyperglycaemic hormone glucagon becomes dysregulated; these last-mentioned defects exacerbate the metabolic consequences of hypoinsulinaemia and are compounded further by hypersecretion of somatostatin (which inhibits both insulin and glucagon secretion). Glucagon secretion has been proposed to be regulated by either intrinsic or paracrine mechanisms, but their relative significance and the conditions under which they operate are debated. Importantly, the paracrine and intrinsic modes of regulation are not mutually exclusive; they could operate in parallel to control glucagon secretion. Here we have applied mathematical modelling of α-cell electrical activity as a novel means of dissecting the processes that underlie metabolic regulation of glucagon secretion. Our analyses indicate that basal hypersecretion of somatostatin and/or increased activity of somatostatin receptors may explain the loss of adequate counter-regulation under hypoglycaemic conditions, as well as the physiologically inappropriate stimulation of glucagon secretion during hyperglycaemia seen in diabetic patients. We therefore advocate studying the interaction of the paracrine and intrinsic mechanisms; unifying these processes may give a more complete picture of the regulation of glucagon secretion from α-cells than studying the individual parts.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Hyperglycemia/metabolism , Animals , Diabetes Mellitus, Type 2/blood , Electrophysiology , Glucose/metabolism , Humans , Insulin/metabolism , Islets of Langerhans/metabolism , Mice , Models, Theoretical , Rats , Receptors, Somatostatin/metabolism , Signal Transduction , Somatostatin/metabolism
9.
J Clin Invest ; 125(12): 4714-28, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26571400

ABSTRACT

Strategies aimed at mimicking or enhancing the action of the incretin hormone glucagon-like peptide 1 (GLP-1) therapeutically improve glucose-stimulated insulin secretion (GSIS); however, it is not clear whether GLP-1 directly drives insulin secretion in pancreatic islets. Here, we examined the mechanisms by which GLP-1 stimulates insulin secretion in mouse and human islets. We found that GLP-1 enhances GSIS at a half-maximal effective concentration of 0.4 pM. Moreover, we determined that GLP-1 activates PLC, which increases submembrane diacylglycerol and thereby activates PKC, resulting in membrane depolarization and increased action potential firing and subsequent stimulation of insulin secretion. The depolarizing effect of GLP-1 on electrical activity was mimicked by the PKC activator PMA, occurred without activation of PKA, and persisted in the presence of PKA inhibitors, the KATP channel blocker tolbutamide, and the L-type Ca(2+) channel blocker isradipine; however, depolarization was abolished by lowering extracellular Na(+). The PKC-dependent effect of GLP-1 on membrane potential and electrical activity was mediated by activation of Na(+)-permeable TRPM4 and TRPM5 channels by mobilization of intracellular Ca(2+) from thapsigargin-sensitive Ca(2+) stores. Concordantly, GLP-1 effects were negligible in Trpm4 or Trpm5 KO islets. These data provide important insight into the therapeutic action of GLP-1 and suggest that circulating levels of this hormone directly stimulate insulin secretion by ß cells.


Subject(s)
Glucagon-Like Peptide 1/pharmacology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Protein Kinase C/metabolism , TRPM Cation Channels/metabolism , Animals , Humans , Insulin/genetics , Insulin Secretion , Insulin-Secreting Cells/cytology , Ion Transport/drug effects , Ion Transport/genetics , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Knockout , Protein Kinase C/genetics , TRPM Cation Channels/genetics , Tetradecanoylphorbol Acetate/pharmacology
10.
J Histochem Cytochem ; 63(8): 575-91, 2015 08.
Article in English | MEDLINE | ID: mdl-26216135

ABSTRACT

Islet non-ß-cells, the α- δ- and pancreatic polypeptide cells (PP-cells), are important components of islet architecture and intercellular communication. In α-cells, glucagon is found in electron-dense granules; granule exocytosis is calcium-dependent via P/Q-type Ca(2+)-channels, which may be clustered at designated cell membrane sites. Somatostatin-containing δ-cells are neuron-like, creating a network for intra-islet communication. Somatostatin 1-28 and 1-14 have a short bioactive half-life, suggesting inhibitory action via paracrine signaling. PP-cells are the most infrequent islet cell type. The embryologically separate ventral pancreas anlage contains PP-rich islets that are morphologically diffuse and α-cell deficient. Tissue samples taken from the head region are unlikely to be representative of the whole pancreas. PP has anorexic effects on gastro-intestinal function and alters insulin and glucagon secretion. Islet architecture is disrupted in rodent diabetic models, diabetic primates and human Type 1 and Type 2 diabetes, with an increased α-cell population and relocation of non-ß-cells to central areas of the islet. In diabetes, the transdifferentiation of non-ß-cells, with changes in hormone content, suggests plasticity of islet cells but cellular function may be compromised. Understanding how diabetes-related disordered islet structure influences intra-islet cellular communication could clarify how non-ß-cells contribute to the control of islet function.


Subject(s)
Islets of Langerhans/anatomy & histology , Islets of Langerhans/cytology , Animals , Humans , Islets of Langerhans/metabolism , Islets of Langerhans/ultrastructure , Microscopy, Electron
11.
Hypertension ; 66(1): 190-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25916722

ABSTRACT

Elevated B-type natriuretic peptide (BNP) regulates cGMP-phosphodiesterase activity. Its elevation is regarded as an early compensatory response to cardiac failure where it can facilitate sympathovagal balance and cardiorenal homeostasis. However, recent reports suggest a paradoxical proadrenergic action of BNP. Because phosphodiesterase activity is altered in cardiovascular disease, we tested the hypothesis that BNP might lose its efficacy by minimizing the action of cGMP on downstream pathways coupled to neurotransmission. BNP decreased norepinephrine release from atrial preparations in response to field stimulation and also significantly reduced the heart rate responses to sympathetic nerve stimulation in vitro. Using electrophysiological recording and fluorescence imaging, BNP also reduced the depolarization evoked calcium current and intracellular calcium transient in isolated cardiac sympathetic neurons. Pharmacological manipulations suggested that the reduction in the calcium transient was regulated by a cGMP/protein kinase G pathway. Fluorescence resonance energy transfer measurements for cAMP, and an immunoassay for cGMP, showed that BNP increased cGMP, but not cAMP. In addition, overexpression of phosphodiesterase 2A after adenoviral gene transfer markedly decreased BNP stimulation of cGMP and abrogated the BNP responses to the calcium current, intracellular calcium transient, and neurotransmitter release. These effects were reversed on inhibition of phosphodiesterase 2A. Moreover, phosphodiesterase 2A activity was significantly elevated in stellate neurons from the prohypertensive rat compared with the normotensive control. Our data suggest that abnormally high levels of phosphodiesterase 2A may provide a brake against the inhibitory action of BNP on sympathetic transmission.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 2/physiology , Heart Conduction System/enzymology , Hypertension/enzymology , Natriuretic Peptide, Brain/pharmacology , Sympathetic Nervous System/drug effects , Animals , Calcium Signaling/drug effects , Cells, Cultured , Cyclic GMP/physiology , Cyclic GMP-Dependent Protein Kinases/physiology , Cyclic Nucleotide Phosphodiesterases, Type 2/genetics , Heart Conduction System/drug effects , Heart Conduction System/physiology , Heart Rate , Hypertension/genetics , Hypertension/physiopathology , Isatin/pharmacology , Male , Natriuretic Peptide, Brain/physiology , Neurons/enzymology , Neurons/physiology , Rats , Rats, Sprague-Dawley , Receptors, Atrial Natriuretic Factor/drug effects , Receptors, Atrial Natriuretic Factor/physiology , Recombinant Fusion Proteins/metabolism , Second Messenger Systems/drug effects , Stellate Ganglion/cytology , Stellate Ganglion/drug effects , Stellate Ganglion/physiology , Sympathetic Nervous System/physiology , Synaptic Transmission/physiology
12.
Mol Cell Biol ; 35(6): 914-27, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25561468

ABSTRACT

The type 1 diabetes autoantigen ICA512/IA-2/RPTPN is a receptor protein tyrosine phosphatase of the insulin secretory granules (SGs) which regulates the size of granule stores, possibly via cleavage/signaling of its cytosolic tail. The role of its extracellular region remains unknown. Structural studies indicated that ß2- or ß4-strands in the mature ectodomain (ME ICA512) form dimers in vitro. Here we show that ME ICA512 prompts proICA512 dimerization in the endoplasmic reticulum. Perturbation of ME ICA512 ß2-strand N-glycosylation upon S508A replacement allows for proICA512 dimerization, O-glycosylation, targeting to granules, and conversion, which are instead precluded upon G553D replacement in the ME ICA512 ß4-strand. S508A/G553D and N506A/G553D double mutants dimerize but remain in the endoplasmic reticulum. Removal of the N-terminal fragment (ICA512-NTF) preceding ME ICA512 allows an ICA512-ΔNTF G553D mutant to exit the endoplasmic reticulum, and ICA512-ΔNTF is constitutively delivered to the cell surface. The signal for SG sorting is located within the NTF RESP18 homology domain (RESP18-HD), whereas soluble NTF is retained in the endoplasmic reticulum. Hence, we propose that the ME ICA512 ß2-strand fosters proICA512 dimerization until NTF prevents N506 glycosylation. Removal of this constraint allows for proICA512 ß4-strand-induced dimerization, exit from the endoplasmic reticulum, O-glycosylation, and RESP18-HD-mediated targeting to granules.


Subject(s)
Cytoplasmic Granules/metabolism , Endoplasmic Reticulum/metabolism , Insulin/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 8/metabolism , Secretory Vesicles/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Cytosol/metabolism , Dimerization , Glycosylation , Islets of Langerhans/metabolism , Molecular Sequence Data , Protein Structure, Tertiary , Rats
13.
Biomolecules ; 2(2): 269-81, 2012 May 24.
Article in English | MEDLINE | ID: mdl-24970137

ABSTRACT

The secretion of insulin by pancreatic islet ß-cells plays a pivotal role in glucose homeostasis and diabetes. Recent work suggests an important role for SUMOylation in the control of insulin secretion from ß-cells. In this paper we discuss mechanisms whereby (de)SUMOylation may control insulin release by modulating ß-cell function at one or more key points; and particularly through the acute and reversible regulation of the exocytotic machinery. Furthermore, we postulate that the SUMO-specific protease SENP1 is an important mediator of insulin exocytosis in response to NADPH, a metabolic secretory signal and major determinant of ß-cell redox state. Dialysis of mouse ß-cells with NADPH efficiently amplifies ß-cell exocytosis even when extracellular glucose is low; an effect that is lost upon knockdown of SENP1. Conversely, over-expression of SENP1 itself augments ß-cell exocytosis in a redox-dependent manner. Taken together, we suggest that (de)SUMOylation represents an important mechanism that acutely regulates insulin secretion and that SENP1 can act as an amplifier of insulin exocytosis.

SELECTION OF CITATIONS
SEARCH DETAIL
...