Comprehensive Physiology Wiley Online Library

Electrophysiology of the β Cell and Mechanisms of Inhibition of Insulin Release

Full Article on Wiley Online Library



Abstract

The sections in this article are:

1 Ion Channels in Insulin‐Secreting Cells
1.1 Adenosine Triphosphate–Sensitive Potassium Channels
1.2 Extracellular Control of Adenosine Triphosphate‐Sensitive Potassium Channel Function
1.3 Therapeutic Manipulation by Modulators of Adenosine Triphosphate‐Sensitive Potassium Channels
1.4 Architecture of the β‐cell Adenosine Triphosphate‐Sensitive Potassium Channel
1.5 Calcium‐Selective Ion Channels
1.6 Voltage‐Gated Sodium Channels
1.7 Voltage‐Gated Potassium Channels
1.8 Voltage‐Independent Potassium Channels
1.9 Nonselective Cation Channels
1.10 Anion‐Selective Channels
2 Ionic Defects of β‐Cell Function
2.1 Persistent Hyperinsulinemic Hypoglycemia of Infancy
2.2 Altered Ionic Control of β Cells and Hypersecretion of Insulin
2.3 Correlation of Gene Defects in the Adenosine Triphosphate‐Sensitive Potassium Channel with Persistent Hyperinsulinemic Hypoglycemia of Infancy
2.4 Clinical Therapy for Persistent Hyperinsulinemic Hypoglycemia of Infancy
2.5 Implications for Diabetes Mellitus
3 Stimulus‐Secretion Coupling Mechanisms Other Than Depolarization
4 Novel Methods for the Measurement of Insulin Secretion
4.1 Capacitance
4.2 Amperometry and Voltametry
4.3 Calcium and Exocytosis
4.4 Cyclic Adenosine Monophosphate and Exocytosis
4.5 Effects of Phospholipases and Protein Kinases C and A
4.6 Sulfonylureas and Exocytosis
4.7 G Proteins and Exocytosis
4.8 Other Modulators of Exocytosis
4.9 Modeling Calcium‐, Cyclic, and Adenosine Monophosphate–, and Guanosine Triphosphate–Dependent Exocytosis
5 Molecular Mechanisms of Exocytosis in the β Cell
6 Receptor‐Mediated Inhibition of Insulin Release: Early and Late Effects
6.1 Involvement of G Proteins
6.2 Receptor–G Protein Interactions
7 Inhibitory Mechanisms
7.1 Adenosine Triphosphate–Sensitive Potassium Channel Activation and Membrane Repolarization
7.2 Calcium Channel Inhibition
7.3 Inhibition of Adenylate Cyclase
7.4 Inhibition at a Distal Site
8 G Protein–Target Interactions
8.1 Adenosine Triphosphate–Sensitive Potassium Channel
8.2 L‐Type Calcium Channels
8.3 Adenylate Cyclase
8.4 Distal Inhibitory Site
9 Other Possible Mechanisms
9.1 Inhibition of Glucose Metabolism
9.2 Inhibition of Fatty Acid Metabolism
9.3 Stimulation of Calcium–Adenosine Triphosphatase Activity
9.4 Cyclic Guanosine Monophosphate
9.5 Cytoskeleton
10 Summary of Inhibitory Mechanisms
Figure 1. Figure 1.

Function of the KATP channel in β cells. The model simplifies the fundamental role of KATP, channels in insulin‐secreting cells: control of the resting membrane potential by open KATP channels, while glucose‐dependent closure facilitates depolarization of the membrane and subsequent opening of voltage‐gated Ca2+ channels. The model details neither the function of other ion channels nor the action of glucose upon secretion by mechanisms independent of KATP, channel activity. Glut glucose transporter.

Figure 2. Figure 2.

Molecular architecture of the KATP channel in β cells. The model shows two subunits: sulfonylurea receptor (SUR1) which has a predicted topology of multiple membrane‐spanning domains and two nucleotide‐binding folds, and Kir6.2, a K+ channel. Subunit SUR1 has two glycosylation sites (both thought to be extracellularly disposed) and an extracellular N‐terminal sequence.

Figure 3. Figure 3.

Ion channels in β cells. The major ion channels characterized in insulin‐secreting cells: several K+‐selective channels, such as the ATP‐sensitive K+ channel (KATP), the Ca2+‐ and voltage‐gated K+ channel (KCa or Kmaxi), the delayed‐rectifier K+ channel (KV), the low‐conductance/receptor‐operated K+ channel (KI), and the outward‐rectifier K+ channel, (KA); Ca2+ selective L‐type and T‐type voltage‐gated channels; cation‐selective channels (Ca2+ store‐depleted and KNS); voltage‐gated Na+ channels; and C1 channels modulated by intracellular Ca2+ and cAMP. The KATP channels are shown as being critically important for linking glucose metabolism to the initiation of cell depolarization, which is then required for Ca2+‐dependent exocytosis. Δψ voltage‐gated.

Figure 4. Figure 4.

Ion channel records from intact normal and persistent hyperinsulinemic hypoglycemia of infancy (PHHI) human β cells. Experiments were made under exactly the same conditions; note the presence of KATP channels as upward deflections from the baseline in normal cells but not in β cells from the patient. Loss of KATP channel function is associated with the appearance of verapamil‐inhibited Ca2+ action potentials. Both records were made using cell‐attached patch‐clamp techniques (see cartoon) and are plotted, for clarity, on the same scale.

[From Kane et al. 138 with permission.]
Figure 5. Figure 5.

Isolated inside‐out patch records from normal and persistent hyperinsulinemic hypoglycemia of infancy (PHHI) β cells that confirm the absence of functional KATP channels in patient tissue. Both traces show the time course of events that typically follow inside‐out patch formation. In normal tissue, a KATP, current typically develops as ATP is lost from the inside face of the membrane. No currents are seen in PHHI β cells under exactly the same conditions.

[From Kane et al. 138 with permission.]
Figure 6. Figure 6.

“Spontaneous” action potentials in persistent hyperinsulinemic hypoglycemia of infancy (PHHI) β cells. Recording of these clusters of electrical responses occur in the absence of stimulatory concentrations of glucose.

[From Kane et al. 138 with permission.]
Figure 7. Figure 7.

Model to account for hypersecretion of insulin from persistent hyperinsulinemic hypoglycemia of infancy (PHHI) β cells. Loss of KATP channel function leads to spontaneously electrically active cells as the result of a depolarized cell membrane potential. Hyperactivity of Ca2+ channels is predicted to cause constant Ca2+ influx, leading to initiation of insulin release.

Figure 8. Figure 8.

Reported sites of familial persistent hyperinsulinemic hypoglycemia of infancy (PHHI) mutations in sulfonylurea receptor (SUR1) and Kir6.2 (insert) In SUR1, these mutations are clustered about the nucleotide‐binding folds (NBFs) and result in loss of Walker A/B consensus sequences. See text for further descriptions of mutations. Arrows indicate mutations in SUR1 NBF1 described by Thomas and colleagues 274 and the intron 32 mutation leading to a truncation of the SUR1 gene product.

Figure 9. Figure 9.

Organization of sulfonylurea receptor (SUR1) and the corresponding position of exons 1–39. TM1 and TM2, regions of putative “transmembrane” spanning domains (see also fig. 2). Nucleotide‐binding Fold 1 (NBF1) and NBF2 are encoded for by exons 16–21 and 34–37, respectively. Arrows indicate the positions of familial persistent hyperinsulinemic hypoglycemia of infancy mutations, which are collected around the NBF1 and NBF2, the principal sites of SUR1 regulation. Shaded arrowhead represents the intron 32 mutation.

Figure 10. Figure 10.

Model to account for how an ion channel‐based therapy for persistent hyperinsulinemic hypoglycemia of infancy (PHHI) might achieve normoglycemia. The strategy is directed toward termination of voltage‐gated Ca2+ influx. This could be brought about by direct inhibition of channels 172 or by hyperpolarizing the membrane away from the threshold for activation of Ca2+ channels. The latter scenario could result from direct agonists of K+ channels or receptor‐mediated events that involve K+ channel activation and/or Ca2+ channel inhibition. Glut, glucose transporter.

Figure 11. Figure 11.

Major pathways involved in stimulation of insulin release. Shown is the KATP channel‐dependent pathway in which increased blood glucose concentrations and consequent increased β‐cell metabolism result in a charge in the intracellular ATP: ADP ratio. This is thought to be a major contributory factor in the closure of ATP‐dependent K+ channels, depolarization of the β cell membrane and increased L‐type Ca2+ channel activity. Increased channel activity and accelerated Ca2+ influx result in elevation of intracellular Ca2+ and stimulated insulin release. Also shown are the important KATP channel‐independent pathways which augment the Ca2+‐stimulated insulin release of the KATP channel‐dependent pathway and the combined stimulation by protein kinases A (PKA) and C (PKC). Major potentiation of release results from hormonal and peptidergic activation of receptors positively linked to adenylate cyclase. Examples of such receptors are those for vasoactive intestinal peptide (VIP), pituitary adenylate cyclase‐activating peptide (PACAP), and glucagon‐like peptide (GLP‐1). Occupation of these receptors by hormones results in activation of adenylate cyclase, increased cAMP levels, and potentiation of release by two mechanisms: (1) activation of PKA and phosphorylation of the L‐type Ca2+ channel to increase Ca2+ entry and (2) phosphorylation at an as yet unknown distal site in stimulus‐secretion coupling to enhance stimulated release. A third potentiating mechanism is activation of receptors linked to phospholipase C, increased phosphoinositide hydrolysis, and increased production of inositol 1,4,5‐trisphosphate (IP3) and diacylglycerol (DAG). As is the case for adenylate cyclase stimulation, the two products of phospholipase C activity result in increased [Ca2+]i (by mobilization of intracellular Ca2+) and potentiation of insulin release by PKC activation and phosphorylation of another, as yet unknown distal site in stimulus‐secretion coupling. This figure has been drawn simply, to illustrate only the major pathways and sites at which the physiological inhibitors may act. CCK, cholecystokinin.

Figure 12. Figure 12.

Methodologies for capacitance studies. These can be carried out with cell‐attached patch experiments (A), standard whole‐cell experiments (B), or perforated patch whole‐cell experiments (C). D: Schematic representation of the change in cell membrane area during either exocytosis or endocytosis. This change is proportional to the cell capacitance.

Figure 13. Figure 13.

Experimental setup for capacitance measurements. A: Cell membrane of a secretory granule before (top) and after (bottom) exocytosis. The area of the plasma membrane increases due to incorporation of the granular membrane. B: Experimental design of capacitance measurements. To the left is the electrical equivalent of the cell (Gm, Cm) in series with the recording pipette (Gs). When the cell is stimulated with a varying sinusoidal command voltage (V), the resulting sinusoidal current will be phase‐shifted (Θ) relative to V due to the capacitative and conductive properties of the cell. The recorded current is fed into a computer and analyzed at two orthogonal vectors. By finding the correct phase angle (Theta;), the current representing the capacitative (ICm) current and the conductive (IG = IGs + IGm) current can be separated. Changes in cell capacitance (ΔCm) and conductance (ΔG) can therefore be detected as changes in ICm and IG, respectively.

Figure 14. Figure 14.

Dual recordings of insulin secretion from a single β cell A: Changes in capacitance induced by Ca2+ currents elicited by a train of voltage‐clamp depolarizations (200 ms, 1 Hz). B: Initiation of exocytosis is associated with a concomitant decrease in fluorescence of quinacrine, an agent used to fluorescently label the secretory granules of β cells.

Figure 15. Figure 15.

Regulation of exocytosis by local Ca2+ concentration in the vicinity of Ca2+ channels. A(i): When the Ca2+ channel opens, Ca2+ immediately reaches a high concentration in the active zone and exocytosis is initiated. A(ii): Upon closure of the Ca2+ channel, local Ca2+ transient and exocytosis stop though a fluorescence Ca2+ indicator may continue to report elevated intracellular Ca2+ signals for some time. B(i): In the absence of the Ca2+ chelator EGTA, Ca2+ influx through open Ca2+ channels both inactivates Ca2+ channels and initiates exocytosis, while in the presence of EGTA (ii) Ca2+ still inactivates the channel but, due to the diffusion path, exocytosis is abolished.

Figure 16. Figure 16.

cAMP potentiation of Ca2+‐stimulated insulin release. In these experiments, exocytosis is stimulated by the photorelease of cAMP (80 μM) from a caged precursor 6. Cells were infused with Ca2+–EGTA mixtures, yielding a final [Ca2+]i concentration of (A) O mM, (B) 60 nM, or (C) 2 μM. The same concentration of cAMP causes a more marked stimulation of exocytosis at a higher Ca2+ concentration.

Figure 17. Figure 17.

Exocytosis. Multiple components play a role in the late stages of stimulus‐secretion coupling and exocytosis. A (top): The unstimulated state is shown with the insulin‐containing granule and the vesicle‐associated membrane protein (VAMP)/synaptobrevin and synaptotagmin components. The rab3 protein thought to be involved in trafficking, NSF (N‐ethylmaleimide‐sensitive factor), and SNAPS (soluble NSF‐attachment proteins) are also shown. On the plasma membrane are syntaxin, Munc‐18, and SNAP‐25. For docking shown in A (bottom), the VAMP/synaptobrevin (the v‐SNARE) in the granule membrane binds to syntaxin and SNAP‐25 (the two making up the t‐SNARE) in the plasma membrane. As Munc‐18 inhibits the binding of the v‐ and t‐SNAREs, the binding, or “docking,” requires the prior dissociation of Munc‐18. Synaptotagmin association with the complex is also thought to be inhibitory, and dissociation of synaptotagmin from the complex occurs during “priming,” as shown in B (top). Subsequently, NSF and SNAP(s) complete the formation of the fusion complex that leads finally to exocytosis [B (bottom)]. SNAP‐25, synaptosome‐associated protein of Mr 25,000; Munc‐18, mammalian homologue of the Caenorhabditis elegans unc‐18 gene.

[Adapted from refs. 89, 236, 259, and 266, using only those components known to be present in the β cell and presumed to be involved in exocytosis.]
Figure 18. Figure 18.

The G‐protein cycle in the pancreatic β cell in relation to inhibitory ligands and effector G‐protein targets. Receptor activation by agonists such as norepinephrine, somatostatin, galanin, and prostaglandins induces them to bind and destabilize specific heterotrimeric G proteins (GαGDPβγ). Decreased affinity for GDP and increased affinity for GTP result in GDP–GTP exchange on the α subunit of the G protein and subsequent dissociation of the α and βγ subunits into their active forms. Actions of the α and βγ subunits on the target molecules (the channels, adenylate cyclase, and the distal mechanism) are responsible for inhibition of insulin release. These actions are terminated by the inherent GTPase activity of the α subunits, probably under the control; of GTPase‐activating proteins (GAPs) and/or the GAP activity of the targets. Hydrolysis of GTP allows reformation of the heterotrimeric (GDP‐bound) G protein. The cycle is complete when the G protein interacts again with an activated receptor.

Figure 19. Figure 19.

Sites of inhibition of insulin release in the β cell. The four major sites of inhibitory action: The KATP channel, which is activated by inhibitors to repolarize the β cell; the L‐type Ca2+ channel, which is closed as the β cell repolarizes and may be subject to a minor level of direct inhibition (both effects lead to a decrease in [Ca2+], and inhibition of secretion); adenylate cyclase, the activity of which is decreased by the inhibitors, resulting in a lowering of [Ca2+]i and loss of the potentiating effect of cAMP; and the distal site of inhibition, which, while powerful and well documented, is not yet understood. VIP, vasoactive intestinal peptide; PACAP, pituitary adenylate cyclase‐activating peptide; GLP‐1, glucagon‐like peptide 1.

Figure 20. Figure 20.

Multiple inhibitory agonists, multiple G‐protein mediators, and multiple effector systems which result in inhibition of insulin release. There are multiple inhibitors of insulin secretion, which may act simultaneously on the β cell. As there are five pertussis toxin‐sensitive G proteins that may mediate the inhibitory effects, the receptors share and compete according to relative amounts of receptors and individual G proteins, their affinities for them, and their subcellular location. Activated G proteins then seek out their several targets of inhibitory action. Shown are examples of inhibitory ligands, the three Gi and two Go proteins that may be present in β cells, the four well‐documented sites of inhibitory action (KATP channel, L‐type Ca2+ channel, adenylate cyclase, and the distal site), and other suggested sites of inhibitory action. CGRP, calcitonin gene–related peptide.



Figure 1.

Function of the KATP channel in β cells. The model simplifies the fundamental role of KATP, channels in insulin‐secreting cells: control of the resting membrane potential by open KATP channels, while glucose‐dependent closure facilitates depolarization of the membrane and subsequent opening of voltage‐gated Ca2+ channels. The model details neither the function of other ion channels nor the action of glucose upon secretion by mechanisms independent of KATP, channel activity. Glut glucose transporter.



Figure 2.

Molecular architecture of the KATP channel in β cells. The model shows two subunits: sulfonylurea receptor (SUR1) which has a predicted topology of multiple membrane‐spanning domains and two nucleotide‐binding folds, and Kir6.2, a K+ channel. Subunit SUR1 has two glycosylation sites (both thought to be extracellularly disposed) and an extracellular N‐terminal sequence.



Figure 3.

Ion channels in β cells. The major ion channels characterized in insulin‐secreting cells: several K+‐selective channels, such as the ATP‐sensitive K+ channel (KATP), the Ca2+‐ and voltage‐gated K+ channel (KCa or Kmaxi), the delayed‐rectifier K+ channel (KV), the low‐conductance/receptor‐operated K+ channel (KI), and the outward‐rectifier K+ channel, (KA); Ca2+ selective L‐type and T‐type voltage‐gated channels; cation‐selective channels (Ca2+ store‐depleted and KNS); voltage‐gated Na+ channels; and C1 channels modulated by intracellular Ca2+ and cAMP. The KATP channels are shown as being critically important for linking glucose metabolism to the initiation of cell depolarization, which is then required for Ca2+‐dependent exocytosis. Δψ voltage‐gated.



Figure 4.

Ion channel records from intact normal and persistent hyperinsulinemic hypoglycemia of infancy (PHHI) human β cells. Experiments were made under exactly the same conditions; note the presence of KATP channels as upward deflections from the baseline in normal cells but not in β cells from the patient. Loss of KATP channel function is associated with the appearance of verapamil‐inhibited Ca2+ action potentials. Both records were made using cell‐attached patch‐clamp techniques (see cartoon) and are plotted, for clarity, on the same scale.

[From Kane et al. 138 with permission.]


Figure 5.

Isolated inside‐out patch records from normal and persistent hyperinsulinemic hypoglycemia of infancy (PHHI) β cells that confirm the absence of functional KATP channels in patient tissue. Both traces show the time course of events that typically follow inside‐out patch formation. In normal tissue, a KATP, current typically develops as ATP is lost from the inside face of the membrane. No currents are seen in PHHI β cells under exactly the same conditions.

[From Kane et al. 138 with permission.]


Figure 6.

“Spontaneous” action potentials in persistent hyperinsulinemic hypoglycemia of infancy (PHHI) β cells. Recording of these clusters of electrical responses occur in the absence of stimulatory concentrations of glucose.

[From Kane et al. 138 with permission.]


Figure 7.

Model to account for hypersecretion of insulin from persistent hyperinsulinemic hypoglycemia of infancy (PHHI) β cells. Loss of KATP channel function leads to spontaneously electrically active cells as the result of a depolarized cell membrane potential. Hyperactivity of Ca2+ channels is predicted to cause constant Ca2+ influx, leading to initiation of insulin release.



Figure 8.

Reported sites of familial persistent hyperinsulinemic hypoglycemia of infancy (PHHI) mutations in sulfonylurea receptor (SUR1) and Kir6.2 (insert) In SUR1, these mutations are clustered about the nucleotide‐binding folds (NBFs) and result in loss of Walker A/B consensus sequences. See text for further descriptions of mutations. Arrows indicate mutations in SUR1 NBF1 described by Thomas and colleagues 274 and the intron 32 mutation leading to a truncation of the SUR1 gene product.



Figure 9.

Organization of sulfonylurea receptor (SUR1) and the corresponding position of exons 1–39. TM1 and TM2, regions of putative “transmembrane” spanning domains (see also fig. 2). Nucleotide‐binding Fold 1 (NBF1) and NBF2 are encoded for by exons 16–21 and 34–37, respectively. Arrows indicate the positions of familial persistent hyperinsulinemic hypoglycemia of infancy mutations, which are collected around the NBF1 and NBF2, the principal sites of SUR1 regulation. Shaded arrowhead represents the intron 32 mutation.



Figure 10.

Model to account for how an ion channel‐based therapy for persistent hyperinsulinemic hypoglycemia of infancy (PHHI) might achieve normoglycemia. The strategy is directed toward termination of voltage‐gated Ca2+ influx. This could be brought about by direct inhibition of channels 172 or by hyperpolarizing the membrane away from the threshold for activation of Ca2+ channels. The latter scenario could result from direct agonists of K+ channels or receptor‐mediated events that involve K+ channel activation and/or Ca2+ channel inhibition. Glut, glucose transporter.



Figure 11.

Major pathways involved in stimulation of insulin release. Shown is the KATP channel‐dependent pathway in which increased blood glucose concentrations and consequent increased β‐cell metabolism result in a charge in the intracellular ATP: ADP ratio. This is thought to be a major contributory factor in the closure of ATP‐dependent K+ channels, depolarization of the β cell membrane and increased L‐type Ca2+ channel activity. Increased channel activity and accelerated Ca2+ influx result in elevation of intracellular Ca2+ and stimulated insulin release. Also shown are the important KATP channel‐independent pathways which augment the Ca2+‐stimulated insulin release of the KATP channel‐dependent pathway and the combined stimulation by protein kinases A (PKA) and C (PKC). Major potentiation of release results from hormonal and peptidergic activation of receptors positively linked to adenylate cyclase. Examples of such receptors are those for vasoactive intestinal peptide (VIP), pituitary adenylate cyclase‐activating peptide (PACAP), and glucagon‐like peptide (GLP‐1). Occupation of these receptors by hormones results in activation of adenylate cyclase, increased cAMP levels, and potentiation of release by two mechanisms: (1) activation of PKA and phosphorylation of the L‐type Ca2+ channel to increase Ca2+ entry and (2) phosphorylation at an as yet unknown distal site in stimulus‐secretion coupling to enhance stimulated release. A third potentiating mechanism is activation of receptors linked to phospholipase C, increased phosphoinositide hydrolysis, and increased production of inositol 1,4,5‐trisphosphate (IP3) and diacylglycerol (DAG). As is the case for adenylate cyclase stimulation, the two products of phospholipase C activity result in increased [Ca2+]i (by mobilization of intracellular Ca2+) and potentiation of insulin release by PKC activation and phosphorylation of another, as yet unknown distal site in stimulus‐secretion coupling. This figure has been drawn simply, to illustrate only the major pathways and sites at which the physiological inhibitors may act. CCK, cholecystokinin.



Figure 12.

Methodologies for capacitance studies. These can be carried out with cell‐attached patch experiments (A), standard whole‐cell experiments (B), or perforated patch whole‐cell experiments (C). D: Schematic representation of the change in cell membrane area during either exocytosis or endocytosis. This change is proportional to the cell capacitance.



Figure 13.

Experimental setup for capacitance measurements. A: Cell membrane of a secretory granule before (top) and after (bottom) exocytosis. The area of the plasma membrane increases due to incorporation of the granular membrane. B: Experimental design of capacitance measurements. To the left is the electrical equivalent of the cell (Gm, Cm) in series with the recording pipette (Gs). When the cell is stimulated with a varying sinusoidal command voltage (V), the resulting sinusoidal current will be phase‐shifted (Θ) relative to V due to the capacitative and conductive properties of the cell. The recorded current is fed into a computer and analyzed at two orthogonal vectors. By finding the correct phase angle (Theta;), the current representing the capacitative (ICm) current and the conductive (IG = IGs + IGm) current can be separated. Changes in cell capacitance (ΔCm) and conductance (ΔG) can therefore be detected as changes in ICm and IG, respectively.



Figure 14.

Dual recordings of insulin secretion from a single β cell A: Changes in capacitance induced by Ca2+ currents elicited by a train of voltage‐clamp depolarizations (200 ms, 1 Hz). B: Initiation of exocytosis is associated with a concomitant decrease in fluorescence of quinacrine, an agent used to fluorescently label the secretory granules of β cells.



Figure 15.

Regulation of exocytosis by local Ca2+ concentration in the vicinity of Ca2+ channels. A(i): When the Ca2+ channel opens, Ca2+ immediately reaches a high concentration in the active zone and exocytosis is initiated. A(ii): Upon closure of the Ca2+ channel, local Ca2+ transient and exocytosis stop though a fluorescence Ca2+ indicator may continue to report elevated intracellular Ca2+ signals for some time. B(i): In the absence of the Ca2+ chelator EGTA, Ca2+ influx through open Ca2+ channels both inactivates Ca2+ channels and initiates exocytosis, while in the presence of EGTA (ii) Ca2+ still inactivates the channel but, due to the diffusion path, exocytosis is abolished.



Figure 16.

cAMP potentiation of Ca2+‐stimulated insulin release. In these experiments, exocytosis is stimulated by the photorelease of cAMP (80 μM) from a caged precursor 6. Cells were infused with Ca2+–EGTA mixtures, yielding a final [Ca2+]i concentration of (A) O mM, (B) 60 nM, or (C) 2 μM. The same concentration of cAMP causes a more marked stimulation of exocytosis at a higher Ca2+ concentration.



Figure 17.

Exocytosis. Multiple components play a role in the late stages of stimulus‐secretion coupling and exocytosis. A (top): The unstimulated state is shown with the insulin‐containing granule and the vesicle‐associated membrane protein (VAMP)/synaptobrevin and synaptotagmin components. The rab3 protein thought to be involved in trafficking, NSF (N‐ethylmaleimide‐sensitive factor), and SNAPS (soluble NSF‐attachment proteins) are also shown. On the plasma membrane are syntaxin, Munc‐18, and SNAP‐25. For docking shown in A (bottom), the VAMP/synaptobrevin (the v‐SNARE) in the granule membrane binds to syntaxin and SNAP‐25 (the two making up the t‐SNARE) in the plasma membrane. As Munc‐18 inhibits the binding of the v‐ and t‐SNAREs, the binding, or “docking,” requires the prior dissociation of Munc‐18. Synaptotagmin association with the complex is also thought to be inhibitory, and dissociation of synaptotagmin from the complex occurs during “priming,” as shown in B (top). Subsequently, NSF and SNAP(s) complete the formation of the fusion complex that leads finally to exocytosis [B (bottom)]. SNAP‐25, synaptosome‐associated protein of Mr 25,000; Munc‐18, mammalian homologue of the Caenorhabditis elegans unc‐18 gene.

[Adapted from refs. 89, 236, 259, and 266, using only those components known to be present in the β cell and presumed to be involved in exocytosis.]


Figure 18.

The G‐protein cycle in the pancreatic β cell in relation to inhibitory ligands and effector G‐protein targets. Receptor activation by agonists such as norepinephrine, somatostatin, galanin, and prostaglandins induces them to bind and destabilize specific heterotrimeric G proteins (GαGDPβγ). Decreased affinity for GDP and increased affinity for GTP result in GDP–GTP exchange on the α subunit of the G protein and subsequent dissociation of the α and βγ subunits into their active forms. Actions of the α and βγ subunits on the target molecules (the channels, adenylate cyclase, and the distal mechanism) are responsible for inhibition of insulin release. These actions are terminated by the inherent GTPase activity of the α subunits, probably under the control; of GTPase‐activating proteins (GAPs) and/or the GAP activity of the targets. Hydrolysis of GTP allows reformation of the heterotrimeric (GDP‐bound) G protein. The cycle is complete when the G protein interacts again with an activated receptor.



Figure 19.

Sites of inhibition of insulin release in the β cell. The four major sites of inhibitory action: The KATP channel, which is activated by inhibitors to repolarize the β cell; the L‐type Ca2+ channel, which is closed as the β cell repolarizes and may be subject to a minor level of direct inhibition (both effects lead to a decrease in [Ca2+], and inhibition of secretion); adenylate cyclase, the activity of which is decreased by the inhibitors, resulting in a lowering of [Ca2+]i and loss of the potentiating effect of cAMP; and the distal site of inhibition, which, while powerful and well documented, is not yet understood. VIP, vasoactive intestinal peptide; PACAP, pituitary adenylate cyclase‐activating peptide; GLP‐1, glucagon‐like peptide 1.



Figure 20.

Multiple inhibitory agonists, multiple G‐protein mediators, and multiple effector systems which result in inhibition of insulin release. There are multiple inhibitors of insulin secretion, which may act simultaneously on the β cell. As there are five pertussis toxin‐sensitive G proteins that may mediate the inhibitory effects, the receptors share and compete according to relative amounts of receptors and individual G proteins, their affinities for them, and their subcellular location. Activated G proteins then seek out their several targets of inhibitory action. Shown are examples of inhibitory ligands, the three Gi and two Go proteins that may be present in β cells, the four well‐documented sites of inhibitory action (KATP channel, L‐type Ca2+ channel, adenylate cyclase, and the distal site), and other suggested sites of inhibitory action. CGRP, calcitonin gene–related peptide.

References
 1. Abrahamsson, H., and E. Gylfe. Demonstration of a proton gradient across the insulin granule membrane. Acta Physiol. Scand. 109: 113–114, 1980.
 2. Aguilar‐Bryan, L., D. A., Nelson, Q. A. Vu, M. B. Humphrey, and A. E. Boyd, III.. Photoaffinity labeling and partial purification of the β‐cell sulfonylurea receptor using a novel, biologically active glyburide analogue. J. Biol Chem. 265: 8218–8224, 1990.
 3. Aguilar‐Bryan, L., C. G., Nichols, S. W. Wechsler, J. P. Clement, IV, A. E. Boyd, III, G. Gonzalez, H. Herrera‐Sosa, K. Nguy, J. Bryan, and D. A. Nelson. Cloning of the beta cell high‐affinity sulphonylurea receptor: a regulator of insulin secretion. Science 268: 423–426, 1995.
 4. Ahren, B., P.‐O., Berggren, K. Bokvist, And P. Rorsman. Does galanin inhibit insulin secretion by opening of the ATP‐regulated K+ channel in the beta‐cell? Peptides 10: 453–157, 1989.
 5. Amiranoff, B., A.‐M., Lorinet, I. Lagny‐Pourmir, and M. Laburthe. Mechanism of galanin‐inhibited insulin release. Occurrence of a pertussis‐toxin‐sensitive inhibition of adenylate cyclase. Eur. J. Biochem. 177: 147–152, 1989.
 6. Ämmälä, C., F. M., Ashcroft, and P. Rorsman. Calcium‐independent potentiation of insulin release by cyclic AMP in single β‐cells. Nature 363: 356–358, 1993.
 7. Ämmälä, C., P.‐O., Berggren, K. Bokvist, O. Larsson, and P. Rorsman. Demonstration of a novel apamin‐insensitive calcium‐activated K+ channel in mouse pancreatic B cells. Pflugers Arch. 422: 443–448, 1993.
 8. Ämmälä, C., L., Eliasson, K. Bokvist, P.‐O. Berggren, R. E. Honkanen, A. Sjöholm, and P. Rorsman. Activation of protein kinases and inhibition of protein phosphatases play a central role in the regulation of exocytosis in mouse pancreatic beta cells. Proc. Natl. Acad. Sci. U.S.A. 91: 4343–4347, 1994.
 9. Ämmälä, C., L., Eliasson, K. Bokvist, O. Larsson, F. M. Ashcroft, and P. Rorsman. Exocytosis elicited by action potentials and voltage‐clamp calcium currents in individual mouse pancreatic B‐cells. J. Physiol. (Lond.) 472: 665–688, 1993.
 10. Ämmälä, C., O., Larsson, P.‐O. Berggren, K. Bokvist, L. Juntti‐Berggren, H. Kindmark, and P. Rorsman. Inositol trisphosphate‐dependent periodic activation of a Ca2+ activated K+ conductance in glucose‐stimulated pancreatic β‐cells. Nature 353: 849–852, 1991.
 11. Ämmälä, C., A., Moorhouse, and F. M. Ashcroft. The sulphonylurea receptor confers diazoxide sensitivity on the inwardly rectifying K+ channel Kir6.2 expressed in human embryonic kidney cells. J. Physiol. (Lond.) 494: 709–714, 1996.
 12. Ämmälä, C., A., Moorhouse, F. Gribble, R. Ashfield, P. Proks, P. A. Smith, H. Sakura, B. Coles, S. J. H. Ashcroft, and F. M. Ashcroft. Promiscuous coupling between the sulphonylurea receptor and inwardly rectifying potassium channels. Nature 379: 545–548, 1996.
 13. Arkhammar, P., T., Nilsson, P. Rorsman, and P.‐O. Berggren. Inhibition of ATP‐sensitive K+ channels precedes depolarization‐induced increase in cytoplasmic free Ca2+ concentration in pancreatic β‐cells. J. Biol. Chem. 262: 5448–5454, 1987.
 14. Ashcroft, F., D. E., Harrison, and S. J. H. Ashcroft. Glucose induces closure of single potassium channels in isolated rat pancreatic islets. Nature 312: 446–448, 1984.
 15. Ashcroft, F. M., S. J. H., Ashchroft and D. E. Harrison. Properties of single potassium channels modulated by glucose in rat pancreatic β‐cells. J. Physiol. (Lond.) 400: 501–527, 1988.
 16. Ashcroft, F. M., M., Kakei, and R. P. Kelly. Rubidium and sodium permeability of the ATP‐sensitive K+ channel in single rat pancreatic β‐cells. J. Physiol. (Lond.) 408: 413–430, 1989.
 17. Ashcroft, F. M., and P. Rorsman. Electrophysiology of the pancreatic B‐cell. Prog. Biophys. Mol. Biol. 54: 87–143, 1989.
 18. Ashcroft, S. J. H., and F. M. Ashcroft. The sulfonylurea receptor. Biochim. Biophys. Acta 1175: 45–59, 1992.
 19. Atlas, D., and Y. Bumstein. Isolation of an endogenous clomdine‐displacing substance from rat brain. FEBS Lett. 170: 887–891, 1984.
 20. Aynsley‐Green, A.. Nesidioblastosis of the pancreas in infancy. Dev. Med. Child Neurol. 23: 372–79, 1981.
 21. Band, A. M., P. M., Jones, and S. L. Howell. The mechanism of arachidonic acid–induced insulin secretion from rat islets of Langerhans. Biochim. Biophys. Acta 1176: 64–68, 1993.
 22. Barnett, D. W., and S. Misler. Coupling of exocytosis to depolarization in rat pancreatic islet beta‐cells: effects of Ca2+, Sr2+ and Ba2+‐containing extracellular solutions. Pfulgers Arch. 430: 593–595, 1995.
 23. Barnett, D. W., D. M., Pressel, H. T. Chern, D. W. Scharp, and S. Misler. cAMP‐enhancing agents “permit” stimulus–secretion coupling in canine pancreatic islet beta‐cells. J. Membr. Biol. 138: 113–120, 1994.
 24. Barnett, D. W., D. M., Pressel, and S. Misler. Voltage‐dependent Na+ and Ca2+ currents in human pancreatic islet β‐cell: evidence for roles in the generation of action potentials and insulin secretion. Pflugers Arch. 431: 272–282, 1995.
 25. Bernadi, H., M., Fosset, and M. Lazdunski. Characterization, purification and affinity labeling of the brain 3H‐glibenclamidebinding protein, a putative neuronal ATP‐regulated K+ channel. Proc. Natl. Acad. Sci. U.S.A. 85: 9816–9820, 1988.
 26. Berridge, M. J., and R. F. Irvine. Inositol trisphosphate, a novel second messenger in cellular signal transduction. Nature 312: 315–321, 1984.
 27. Berrow, N. S., G., Milligan, and N. G. Morgan. Immunological characterization of tie guanine‐nucleotide binding proteins Gi and Go in rat islets of Langerhans. J. Mol. Endocrinol. 8: 103–108, 1992.
 28. Best, L.. Glucose and α‐ketoisocaproate induce transient inward currents in rat pancreatic β‐cells. Diahetologia 40: 1–6, 1997.
 29. Best, L, A. P., Yates, and S. Tomlinson. Stimulation of insulin secretion by glucose in the absence of diminished (86Rb+ permeability. Biochem. Pharmacol. 43: 2483–2485, 1992.
 30. Blachier, F., and W. J. Malaisse. Possible role of a GTP‐binding protein in the activation of phospholipase C by carbamylcholine in tumoral insulin‐producing cells. Res. Commun. Chem. Pathol. Pharmacol. 58: 237–255, 1987.
 31. Bokvist, K., C., Ämmälä, P.‐O. Berggren, P. Rorsman, and L. W˚ahlander. Alpha 2‐adrenergic stimulation stimulation does not inhibit L‐type calcium channels in mouse pancreatic β‐cells. Biosci. Rep. 11: 147–157, 1991.
 32. Bokvist, K., L., Eliasson, C. Ämmälä, E. Renström, and P. Rorsman. Co‐localization of L‐type Ca2+ channels and insulin‐containing secretory granules and its significance for the initiation of exocytosis in mouse pancreatic β‐cells. EMBOJ. 14: 50–57, 1995.
 33. Bokvist, K., P., Rorsman, and P. A. Smith. Effect of external tetraethylammonium ions and quinine on delayed rectifying K+ channels in mouse pancreatic β‐cells. J. Physiol. (Lond). 423: 311–325, 1990.
 34. Bordin, S., A. C., Boschero, E. M Carneiro, and I. Atwater. Ionic mechanisms involved in the regulation of insulin secretion by muscarinic agonists. J. Membr. Biol. 148: 177–184, 1995.
 35. Boyd, R. S., M. J., Duggan, C. C. Shone, and K. A. Foster. The effect of botulinum neurotoxins on the release of insulin from the insulinoma cell lines HIT‐15 and RINm5F. J. Biol. Chem. 270: 18216–18218, 1995.
 36. Bozem, M., M., Nenquin, and J. C. Henquin. The ionic, electrical and secretory effects of protein kinase C activation in mouse pancreatic β‐cells: studies with phorbol ester. Endocrinology 121: 1025–1033, 1987.
 37. Burgoyne, R. D.. Control of exocytosis in adrenal chromaffin cells. Biochim. Biophys. Acta 1071: 174–202, 1991.
 38. Byrom, W. D., A., Weil, T. J. Brown, and J. R. Bratty. BTS 67 5 82 improves 24H plasma glucose control and post‐prandial insulin secretion in NIDDM patients Diabetologia 39: A44, 1996.
 39. Cable, H. C., A. el‐Mansoury, and N. G. Morgan. Activation of alpha‐2–adrenoceptors results in an increase in F‐actin formation in HIT‐T15 pancreatic B‐cells. Biochem. J. 307: 169–74, 1995.
 40. Caldwell, K. K., C. L., Boyajian, and D. M. Cooper. The effects of Ca2+ and calmodulin on adenylyl cyclase activity in plasma membranes derived from neuronal and non‐neuronal cells. Cell Calcium 13: 107–121, 1992.
 41. Carter, B. D., and F. Medzihradsky. Go mediates the coupling of the u opioid receptor to adenylyl cyclase in cloned neural cell and brain. Proc. Natl. Acad. Sci. U.S.A. 90: 4062–4066, 1993.
 42. Chan, S. L. F., C. A., Brown, and N. G. Morgan. Stimulation of insulin secretion by the imidazoline α2‐adrenoceptor antagonist efaroxan is mediated by a novel, stereoselective, binding site. Eur. J. Pharmacol. 230: 375–378, 1993.
 43. Chan, S. L. F., M. J., Dunne, M. R. Stillings, and N. G. Morgan. Characterisation of the interactions of efaroxan, an α2‐adrenergic antagonist, with insulin‐secreting cells. Eur. J. Pharmacol. 204: 41–48, 1991.
 44. Chan, S. L. F., and N. G. Morgan. Stimulation of insulin secretion by efaroxan may involve interaction with potassium channels. Eur. J. Pharmacol. 176: 97–101, 1990.
 45. Chan, S. L. F., and N. G. Morgan. Clonidine‐displacing substance (CDS), an endogenous imidazoline receptor ligand, is a potent insulin secretagogue. Diabetohgia 39: A30, 1996.
 46. Chen, S., A., Ogawa, M. Ohneda, R. H. Unger, D. W. Foster, and J. D. McGarry. More direct evidence for a malonyl‐CoA‐carnitine palmitoyltransferase I interaction as a key event in pancreatic b‐cell signaling. Diabetes 43: 878–883, 1994.
 47. Chow, R. H., and L. von Rüden. Electrochemical detection of secretion from single cells In: Single‐Channel Recording (2nd ed.) edited by B. Sakmann and E. Neher, New York: Plenum, 1995.
 48. Civelek, V. N., J. T., Deeney, N. J. Shalosky, K. Tornheim, R. G. Hansford, M. Prentki, and B. E. Corkey. Regulation of pancreatic beta‐cell mitochondrial metabolism: influence of Ca2+, substrate and ADP. J. Biochem. 318: 615–621, 1996.
 49. Cook, D. L., and C. N. Hales. Intracellular ATP directly blocks K+ channels in pancreatic B‐cells. Nature 311: 271–273, 1984.
 50. Cook, D. L., M., Ikeuchi, and W. Y. Fujimoto. Lowering of pHi inhibits Ca2+‐activated K+‐channels in pancreatic β‐cells. Nature 311: 269–271, 1984.
 51. Cook, D. L., and E. Pare. Islet electrical pacemaker response to alpha‐adrenergic stimulation. Diabetes 31: 985–990, 1982.
 52. Cook, D. L., L. S., Satin, M. L. J. Ashford, and C. N. Hales. ATP‐sensitive K+ channels in pancreatic β‐cells. Diabetes 37: 495–498, 1988.
 53. Cook, D. L., L. S., Satin, and W. F. Hopkins. Pancreatic B cells are busting, but how? Trench Neurosci. 14: 411–414, 1991.
 54. Corkey, B. E., M. C., Glennon, K. S. Chen, J. T. Deeney, F. M. Matschinsky, and M. Prentki. A role for malonyl‐CoA in glucose‐stimulated insulin secretion from clonal pancreatic β‐cells. J. Biol. Chem. 264: 21608–21612, 1989.
 55. Cormont, M., Y. Le Marchand‐Brustel, E. Van Obberghen, A. M. Spiegel, and G. W. G. Sharp. Identification of G protein alpha‐subunits in RINm5F cells and their selective interaction with galanin receptor. Diabetes 40: 1170–1176, 1991.
 56. Dean, P. M.. Ultrastructural morphometry of the pancreatic β‐cell. Diabetohgia 9: 115–119, 1989.
 57. Dean, P. M., and E. K. Matthews. Electrical activity in pancreatic islet cells. Nature 219: 389–390, 1968.
 58. Degitiar, V. E., B., Wittig, G. Schultz, and K. Kalkbrenner. A specific G(o) heterotrimer couples somatostatin receptors to voltagegated calcium channels in RINm5F cells. FEBS Lett. 380: 137–141, 1996.
 59. De Mazancourt, P., P. K. Goldsmith, and L. S. Weinstein. Inhibition of adenylate cyclase activity by galanin in rat insulinoma cells is mediated by the G‐protein Gi3. Biochem. J. 303: 369–375, 1994.
 60. Detimary, P., J.‐C., Jonas, and J. C. Henquin. Possible links between glucose‐induced changes in the energy state of pancreatic B cells and insulin release. Unmasking by decreasing a stable pool of adenine nucleotides in mouse islets. J. Clin. Invest. 96: 1738–1745, 1995.
 61. De Weille, J. R., H. Schmid‐Antomarchi, M. Fosset, and M. Lazdunski. ATP‐sensitive K+ channels that are blocked by hypoglycaemia‐inducing sulphonylureas in insulin‐secreting cells are activated by galanin, a hyperglycemia‐inducing hormone. Proc. Natl. Acad. Sci. U.S.A. 85: 1312–1316, 1988.
 62. De Weille, J. R., H. Schmid‐Antomarchi, M. Fosset, and M. Lazdunski. Regulation of ATP‐sensitive K+ channel in insulinoma cells: activation by somatostatin and protein kinase C and the role of cAMP. Proc. Natl. Acad. Sci. U.S.A. 86: 2971–2975, 1989.
 63. Drews, G., A., Debuyser, and J.‐C. Henquin. Significance of the membrane repolarization and cyclic AMP changes in mouse pancreatic B‐cells for the inhibition of insulin release by galanin. Mol. Cell. Endocrinol. 105: 97–102, 1994.
 64. Drews, G., A., Debuyser, M. Nenquin, and J.‐C. Henquin. Galanin and epinephrine act on distinct receptors to inhibit insulin release by the same mechanisms including an increase in K+ permeability of the B‐cell membrane. Endocrinology, 126: 1646–1653, 1990.
 65. Drews, G., P., Detimary, and J.‐C. Henquin. Non‐additivity of adrenaline and galanin effects on +Rb efflux and membrane potential in mouse B‐cells suggests sharing of common targets. Bochim. Biophys. Acta 1175: 214–218, 1993.
 66. Drucker, D. J., J., Philippe, S. Mojsov, W. L. Chick, and J. F. Habener. Glucagon‐like peptide‐1 stimulates insulin gene expression and increases cyclic AMP levels in a rat islet cell line. Proc. Natl. Acad. Sci. U.S.A. 84: 3434–3438, 1987.
 67. Dunne, M. J.. Protein phosphorylation is required for diazoxide to open ATP‐sensitive potassium channels in insulin (RINm5F) secreting cells. FEBS Lett. 250: 262–266, 1989.
 68. Dunne, M. J.. Effects of pinacidil, RP 49356 and nicorandil on ATP‐sensitive potassium channels in insulin‐secreting cells. Br. J. Pharmacol. 99: 487–492, 1990.
 69. Dunne, M. J.. The physiology and pharmacology of ATP‐regulation potassium channels in insulin‐secreting cells. In: Potassium Channel Modulators: Pharmacological, Molecular and Clinical Aspects, edited by A. H. Weston and T. C. Hamilton. Oxford: Blackwell, 1992, p. 110–142.
 70. Dunne, M. J.. Phorbol myristate acetate and its action on ATP‐sensitive potassium channels in insulin‐secreting cells. Am. J. Physiol 267 (Cell Physiol. 36): C501–C506, 1994.
 71. Dunne, M. J., R. J., Aspinall, and O. H. Petersen. The effects of cromakalim BRL 34915 on ATP‐sensitive potassium channels in insulin‐secreting cells. Br. J. Pharmacol. 99: 169–175, 1990.
 72. Dunne, M. J., M. J., Bullett, G. Li, C. B. Wollheim, and O. H. Petersen. Galanin activates nucleotide‐dependent K+ channels in insulin‐secreting cells via a pertussis toxin‐sensitive G‐protein. EMBO J. 8: 412–420, 1989.
 73. Dunne, M. J., I., Findlay, and O. H. Petersen. The effects of pyridine nucelotides on the gating of ATP‐sensitive K+ channels in insulin‐secreting cells. J. Membr. Biol. 102: 205–216, 1988.
 74. Dunne, M. J., I. Findlay, O. H. Petersen, and C. B. Wollheim. ATP‐sensitive K+ channels in an insulin‐secreting cell‐line are inhibited by D‐glyceraldehyde and activated by membrane permeablization. J. Membr. Biol. 93: 271–275, 1986.
 75. Dunne, M. J., E. A., Harding, J. H. Jaggar, P. E. Squires, R. Liang, C. Kane, N. J. M. London, and R. L. F. James. Potassium channels, imidazolines and insulin‐secreting cells. Ann. NY Acad. Sci. 763: 243–262, 1995.
 76. Dunne, M. J., M. C., Illot, and O. II Petersen. Interactions of diazoxide, tolbutamide and ATP4 on nucleotide‐dependent K+ channels in an insulin‐secreting cell line. J. Metnbr. Biol. 99: 215–224, 1987.
 77. Dunne, M. J., C., Kane, R. M. Shepherd, J. A. Sanchez, R. F. L. James, P. R. V. Johnson, A. Aynsley‐Green, S. Lu, J. P. Clement, IV, K. J. Lindley, S. Seino, and L. Aguilar‐Bryan. Familial persistent hyperinsulinemic hypoglycaemia of infancy and mutations in the sulfonylurea receptor. N. Engl. J. Med. 336: 703–706, 1997.
 78. Dunne, M. J., and O. H. Petersen. Intracellular ADP activates K+ channels that can be inhibited by ATP. Pflugers Arch 407: 564–565, 1986.
 79. Dunne, M. J., and O. H. Petersen. Intracellular ADP activates K+ channels that are inhibited by ATP in an insulin‐secreting cell line. FEBS Lett. 208: 59–62, 1986.
 80. Dunne, M. J., and O. H. Petersen. Potassium selective ion channels in insulin‐secreting cells: physiology, pharmacology and their role in stimulus secretion coupling. Biochim. Biophys. Acta 1071: 67–82, 1991.
 81. Dunne, M. J., J. West‐Jordan, R. J. Abraham, R. T. H. Edwards, and O. H. Petersen. The gating of nucleotide dependent K+ channels in insulin‐secreting cells can be modulated by changes in the ratio of ATP4‐/ADP3‐ and by non‐hydrolysable analogues of ATP and ADP J. Membr. Biol. 104: 165–172, 1988.
 82. Easom, R. A., M., Land, J. R. Colca, J. H. Hughes, J. Turk, and M. McDaniel. Effects of insulin secretagogues on protein kinase C–catalyzed phosphorylation of an endogenous substrate in isolated pancreatic islets. J. Biol. Chem. 265: 14938–14946, 1990.
 83. Eddlestone, G. T., and P. M. Beigelman. Pancreatic beta‐cell electrical activity: the role of anions and the control of pH. Am. J. Physiol. 244 (Cell Physiol. 13): C188–C197, 1983.
 84. Eddlestone, G. T., M., Komatsu, L. Shen, and G. W. G. Sharp. Mastoparan increases the intracellular free calcium concentration in two insulin‐secreting cell lines by inhibition of ATP‐sensitive potassium channels. Mol. Pharmacol. 47: 787–797, 1995.
 85. Eliasson, L., P., Proks, C. Ämmälä, F. M. Ashcroft, K. Bokvist, E. Renström, P. Rorsman, and P. A. Smith. Endocytosis of secretory granules in mouse pancreatic β‐cells evoked by transient elevation of cytosolic calcium. J. Physiol. (Lond.) 493: 755–767, 1996.
 86. Eliasson, L., E., Renström, C. Ämmälä, P.‐O. Berggren, A. M. Bertorello, K. Bokvist, A. Chibalin, J. T. Deeney, P. R. Flatt, J. Gäbel, J. Gromada, O. Larsson, P. Lindström, C. J. Rhodes, and P. Rorsman. PKC‐dependent stimulation of exocytosis by sulfonylureas in pancreatic beta cells. Science 271: 813–815, 1996.
 87. Faivre, J.‐J., and I. Findlay. Effects of tolbutamide, glibenclamide and diazoxide upon action potentials recorded from rat ventricular muscle. Biochim. Biophys. Acta 984: 1–5, 1989.
 88. Fehmann, H. C., M., Janssen, and B. Goke. Interaction of glucagon‐like peptide‐1 and galanin in insulin (βTC‐1)‐and somatostatin (RIN T3)‐secreting cells and evidence that both peptides have no receptors on glucagon (INR1G9)‐secreting cells. Acta Diabetol. 32: 176–181, 1995.
 89. Ferro‐Novick, S., and R. Jahn. Vesicle fusion from yeast to man. Nature 370: 191–193, 1994.
 90. Findlay, I.. The effects of magnesium upon adenosine trisphosphate‐regulated potassium channel in a rat insulin‐secreting cell line. J. Physiol. (Lond.) 391: 611–629, 1987.
 91. Findlay, I., and M. J. Dunne. ATP maintains ATP‐inhibited K+ channels in an operational state. Pflugers Arch. 407: 238–240, 1986.
 92. Findlay, I., M. J., Dunne, and O. H. Petersen. ATP‐sensitive inward rectifier and voltage‐ and calcium‐activated K+ channels in cultured pancreatic islet cells. J. Membr. Biol. 88: 165–172, 1985.
 93. Findlay, I., M. J., Dunne, and O. H. Petersen. High conductance K+ channel in pancreatic islet cells can be activated and inactivated by internal calcium. J. Membr. Biol. 83: 169–175, 1985.
 94. Findlay, I., M. J., Dunne, S. Ullrich, C. B. Wollheim, and O. H. Petersen. Quinine inhibits Ca2+‐activated K+ channels in insulin‐secreting cells. FEBS Lett. 185: 4–8, 1985.
 95. Fosset, M., H. Schmid‐Antomarchi, J. R. de Weille, and M. Lazdunski. Somatostatin activates glibenclamide‐sensitive and ATP‐regulated K+ channels in insulinoma cells. FEBS Lett. 242: 94–96, 1988.
 96. Fridolf, T., and B. Ahren. Dual action of the neuropeptide galanin on the cytoplasmic free calcium concentration in RINm5F cells. Biochem. Biophys. Res. Commun. 191: 1224–1229, 1993.
 97. Gembal, M., P., Gilon, and J.‐C. Henquin. Evidence that glucose can control insulin release independently from its action on ATP‐sensitive K+ channels in mouse B cells. J. Clin. Invest. 89: 1288–1295, 1992.
 98. Gerhardt, M. A., and R. R. Neubig. Multiple Gi protein subtypes regulate a single effector mechanism. Mol. Pharmacol. 40: 707–711, 1991.
 99. Gilbey, S. G., J., Stephenson, D. J. O'Halloran, J. M. Burrin, and S. R. Bloom. High dose porcine galanin infusion and effect on intravenous glucose tolerance in humans. Diabetes 38: 1114–1116, 1989.
 100. Gillis, K. D.. Techniques for capacitance measurements In: Single‐Channel Recording (2nd ed.), edited by B. Sakmann and E. Neher. New York: Plenum, 1995.
 101. Gillis, K. D., and S. Misler. Single cell assay of exocytosis from pancreatic islet B cells. Pflugers Arch. 420: 121–123, 1992.
 102. Gillis, K. D., and S. Misler. Enhancers of cytosolic cAMP augment depolarization‐induced exocytosis from pancreatic B‐cells: evidence for effects distal to Ca2+ entry. Pflugers Arch. 424: 195–197, 1993.
 103. Gillis, K. D., R. Y., Pun, and S. Misler. Long‐term monitoring of depolarization‐induced exocytosis from adrenal medullary chromaffin cells and pancreatic islet B cells using “perforated patch recording.” Ann. NY Acad. Sci. 635: 464–467, 1991.
 104. Gillison, S. L., and G. W. G. Sharp. ADP ribosylation by cholera toxin identifies three G‐proteins that are activated by the galanin receptor. Studies with RINm5F cell membranes. Diabetes 43: 24–32, 1994.
 105. Gillison, S. L., S. G., Straub, and G. W. G Sharp. The α2 adrenergic receptor is more effective than the galanin receptor in activating G‐proteins in RINm5F β‐cell membranes. Diabetes 46: 401–407, 1997.
 106. Glaser, B., K. C., Chiu, R. Anker, A. Nestorowicz, H. Landau, H. Ben‐Bassat, Z. Sholmai, N. Kaiser, P. S. Thornton, C. A. Stanley, R. S. Spielman, K. Gogolin‐Ewens, E. Cerasi, L. Baker, J. Rice, H. Donis‐Keller, and M. A. Permutt. Familial hyperinsulinism maps to chromosome 11p14–15.1, 30 cM centromeric to the insulin gene. Nat. Genet. 7: 185–188, 1994.
 107. Grant, D. B., D. B., Dunger, and E. C. Burns. Long‐term treatment with diazoxide in childhood hyperinsulinism. Acta Endocrinol. Suppl. (Copenh.) 279: 304–345. 1986.
 108. Gronda, C. M., G. B., Diaz, J. P. Rossi, and J. J. Gagliardino. Correlation between Ca2+‐ATPase activity of rat islet cells and insulin secretion. Endocrinology 134: 221–225, 1992.
 109. Gulbenkian, A., L., Schobert, C. Nixon, and A. Tabachnick. Metabolic effects of pertussis sensitization in mice and rats. Endocrinology. 83: 885–892, 1968.
 110. Hellman, B., and I.‐B. Taljedal. Effects of sulphonylurea derivatives on pancreatic β‐cells. In. Handbook of Experimental Physiology, edited by A. Hasselblatt and F. Bruchhausen. Berlin: Springer‐Verlag, 1975, vol. XXXII, p. 175–194.
 111. Henquin, J. C.. D‐Glucose inhibits potassium efflux from pancreatic islet cells. Nature 271: 271–273, 1978.
 112. Henquin, J.‐C., A., Debuyser, G. Drews, and T. D. Plant. Regulation of K+ permeability and membrane potential in insulin‐secreting cells. In: Nutrient Regulation of Insulin Secretion, edited by P. R. Flatt. London: Portland: 1992, chapt. 9, p. 173–191.
 113. Henquin, J. C., and H. P. Meissner. Significance of ionic fluxes and changes in membrane potential for stimulus–secretion coupling in pancreatic β‐cells. Experientia 40: 1043–1054, 1984.
 114. Hermans, M. P., W., Schmeer, and J.‐C. Henquin. The permissive effect of glucose, tolbutamide and high K+ on arginine stimulation of insulin release in isolated mouse islets. Diabetologia 30: 659–665, 1987.
 115. Hermouet, S., J. J., Merendino, J. S. Gutkind, and A. M. Spiegel. Activating and inactivating mutations of the alpha‐subunit of Gi2 may have opposite effects on proliferation of NIH 3T3 cells. Proc. Natl. Acad. Sci. U.S.A. 88: 10455–10459, 1991.
 116. Hillaire‐Buys, D., R., Gross, M. Roye, G. Ribes, and M.‐M. Loubatières‐Mariani. Adrenergic inhibition of insulin secretion involves pertussis toxin–sensitive and insensitive mechanisms. Eur. J. Pharmacol. 218: 359–362, 1992.
 117. Hille, B.. Ionic Channels of Excitable Membranes (2nd ed.). Sunderland, MA: Sinauer 1992.
 118. Hiriart, M., and D. R. Matteson. Na channels and two types of Ca channels in rat pancreatic β‐cells identified with the reverse haemolytic plaque assay. J. Gen. Physiol. 91: 617–639, 1988.
 119. Homaidan, F. R., G. W. G., Sharp, and L. M. Nowak. Galanin inhibits a dihydropyridine‐sensitive Ca2+ current in the RINm5F cell line. Proc. Natl. Acad. Sci. U.S.A. 88: 8744–8748, 1991.
 120. Howell, S. L., and W. Montague. Adenylate cyclase activity in isolated rat islets of Langerhans: effects of agents which alter rates of insulin secretion. Biochim. Biophys. Acta 320: 44–52, 1973.
 121. Hsu, W. H., H. D. Xiang, A. S. Rajan, D. L. Kunze, A. E. Boyd, 3rd.. Somatostatin inhibits insulin secretion by a G‐protein‐mediated decrease in Ca2+ entry through voltage‐dependent Ca2+ channels in the beta cell. J. Biol. Chem. 266: 837–843, 1991.
 122. Huang, L., H., Shen, M. A. Atkinson, and R. T. Kennedy. Detection of exocytosis at individual pancreatic beta cells by amperometry at a chemically modified microelectrode. Proc. Natl. Acad. Sci. U.S.A. 92: 9608–9612, 1995.
 123. Hurst, R. D., and N. G. Morgan. Evidence for differential effects of noradrenaline and somatostatin on intracellular messenger systems in rat islets of Langerhans. J. Mol. Endocrinol. 4: 231–237, 1990.
 124. Hutton, J. C., E. J., Penn, and M. Peshavaria. Isolation and characterisation of insulin secretory granules from a rat islet cell tumour. Diabetologia 23: 365–373, 1982.
 125. Inagaki, N., T. Gonoi, J. P. Clement, IV, N. Namba, J. Inazawa, G. Gonzalez, L. Aguilar‐Bryan, S. Seino, and J. Bryan. Reconstitution of IKATP: an inward rectifier subunit plus the sulphonylurea receptor. Science 270: 1166–1170, 1995.
 126. Inagaki, N., T. Gonoi, J. P. Clement, IV, C.‐Z. Wang, L. Aguilar‐Bryan, J. Bryan, and S. Seino. A family of sulphonylurea receptors determines the pharmacological properties of ATP‐sensitive K+ channels. Neuron 16: 1011–1017, 1996.
 127. Inoue, H., J., Ferrer, M. Warren‐Perry, Y. Zhang, H. Millns, R. C. Turner, S. C. Elbein, C. L. Hampe, B. K. Suarez, N. Inagaki, S. Seino, and M. A. Permutt. Sequence variants in the pancreatic islet beta‐cell inwardly rectifying K+ channel Kir6.2 (Bir) gene: identification and lack of role in Caucasian patients with NIDDM. Diabetes 46: 502–507, 1997.
 128. Inoue, H., J., Ferrer, C. M. Welling, S. C. Elbein, M. Hoffman, R. Mayorga, M. Warran‐Perry, Y. Zhang, H. Millns, R. C. Turner, M. Province, J. Bryan, M. A. Permutt, and L. Aguilar‐Bryan. Sequence variants in the sulphonylurea receptor (SUR) gene are associated with NIDDM in Caucasians. Diabetes 45: 825–831, 1996.
 129. Isomoto, I., C., Kondon, M. Yamada, S. Matsumoto, O. Higashiguchi, Y. Horio, Y. Matsuzawa, and Y. Kurachi. A novel sulphonylurea receptor forms with BIR (Kir6.2) a smooth muscle type ATP‐sensitive K+ channel. J. Biol. Chem. 271: 24321–24324, 1996.
 130. Jacobsson, B., A., Bean, R. Scheller, L. Juntti‐Bergren, J. Deany, P.‐O. Berggren, and B. Meister. Identification of synaptic proteins and their isoform mRNAs in compartments of pancreatic endocrine cells. Proc. Natl. Acad. Sci. U.S.A. 91: 12487–12491, 1994.
 131. Jones, P. M., J. M., Fyles, S. J. Persaud, and S. L. Howell. Catecholamine inhibition of Ca2+‐induced insulin secretion from electrically permeabilized islets of Langerhans. FEBS Lett. 219: 139–44, 1987.
 132. Jones, P. M., F. M., Mann, S. J. Persaud, and C. P. D. Wheeler‐Jones. Mastoparan stimulates insulin secretion from pancreatic β‐cells by effects at a late stage in the secretory pathway. Mol. Cell. Endocrinol. 94: 97–103, 1993.
 133. Jones, P. M., D. M., Salmon, and S. L. Howell. Protein phosphorylation in electrically permeabilized islets of Langerhans. Effects of Ca2+, cyclic AMP, a phorbol ester and noradrenaline. Biochem. J. 254: 397–403, 1988.
 134. Jones, R. B., R. M., Shepherd, C. Kane, M. N. Hashmi, A. Moores, E. A. Harding, and M. J. Dunne. Modulation of KATP channels and cytosolic Ca2+ in isolated insulin‐secreting cells by BTS 67 5 82, a novel hypoglycaemia‐inducing agent. Diabetologia 39: A43, 1996.
 135. Joshi, C., and J. M. Fernandez. Capacitance measurements. An analysis of the phase detector technique used to study exocytosis and endocytosis. Biophys. J. 53: 885–892, 1988.
 136. Kalkbrenner, F., V. E., Degtiar, M. Schenker, S. Brendel, A. Zobel, J. Hescheler, B. Wittig, and G. Schultz. Subunit composition of G(o) proteins functionally coupling galanin receptors to voltage‐gated calcium channels. EMBO J. 14: 4728–4737, 1995.
 137. Kalman, K., J., Tseng‐Crank, I. D. Dukes, G. Chandy, M. E. Lancaster, R. H. Spencer, J. Aiyar, G. A Gutman, and K. G. Chandy. Heightened expression of a novel K+ shaker‐related channel Kvl.7 in diabetic β‐cells: potential role in impaired insulin secretion in diabetes. Biophys. J. 68: A268, 1995.
 138. Kane, C., R. M., Shepherd, P. E. Squires, P. R. V. Johnson, R. F. L. James, P. J. Milla, A. Aynsley‐Green, K. J. Lindley, and M. J. Dunne. Loss of functional KATP channels in β‐cells causes persistent hyperinsulinaemic hypoglycaemia of infancy. Nat. Med. 2: 1344–1347, 1996.
 139. Katada, U., and M. Ui. The effect of in vivo pre‐treatment of rats with a new protein purified from Bordetella pertussis on in vitro secretion of insulin: role of calcium. Endocrinology 1204: 1822–1827, 1979.
 140. Keahey, H. H., A. E. Boyd, III, and D. L. Kunze. Catecholamine modulation of calcium currents in clonal pancreatic beta‐cells. Am. J. Physiol. 257 (Cell Physiol. 26): C1171–C1176, 1989.
 141. Kennedy, R. T., L., Huang, M. A. Atkinson, and P. Dush. Amperometric monitoring of chemical secretions from individual pancreatic beta‐cells. Anal. Chem. 65: 1882–1887, 1993.
 142. Kieffer, T. J., R. S., Heller, C. A. Leech, G. G. Holz, and J. F. Habener. Leptin suppression of insulin secretion by the activation of ATP‐sensitive K+ channels in pancreatic beta‐cells. Diabetes 46: 1087–1093, 1997.
 143. Kinard, T. A., and L. S. Satin. An ATP‐sensitive C1‐ channel current that is activated by cell swelling, cAMP, and glyburide in insulin‐secreting cells. Diabetes 44: 1461–1466, 1995.
 144. Komatsu, M., T., Aizawa, N. Yokokawa, Y. Sato, N. Takasu, and T. Yamada. Mastoparan‐induced hormone release from rat pancreatic islets. Endocrinology, 130: 221–228, 1992.
 145. Komatsu, M., A. M., McDermott, S. L. Gillison, and G. W. G. Sharp. Mastoparan stimulates exocytosis at a Ca2+‐independentlate site in stimulus–secretion coupling. Studies with the RINm5F β‐cell line. J. Biol. Chem. 268: 23297–23306, 1993.
 146. Komatsu, M., A. M., McDermott, S. L. Gillison, and G. W. G. Sharp. Time course of action of pertussis toxin to block the inhibition of stimulated insulin release by norepinephrine. Endocrinology 136: 1857–1863, 1995.
 147. Komatsu, M., T., Schermerhorn, T. Aizawa, and G. W. G. Sharp. Glucose stimulation of insulin release in the absence of extracellular Ca2+ and in the absence of any rise in intracellular Ca2+ in rat pancreatic islets. Proc. Nat'l Acd. Sci. U.S.A. 92: 728–732, 1995.
 148. Komatsu, M., T., Schermerhorn, S. G. Straub, and G. W. G. Sharp. Pituitary adenylate cyclase–activating peptide, carbachol and glucose stimulate insulin release in the absence of an increase in intracellular Ca2+. Mol. Pharmacol. 50: 1047–1054, 1996.
 149. Konrad, R. J., Y. C., Jolly, C. Major, and B. A. Wolf. Inhibition of phospholipase A2 and insulin secretion in pancreatic islets. Biochim. Biophys. Acta 1135: 215–220, 1992.
 150. Konrad, R. J., R. A., Young, R. D. Record, R. M. Smith, P. Butkerait, D. Manning, L. Jarett, and B. A. Wolf. The heterotrimeric G‐protein Gi is localized to the insulin secretory granules of β‐cells and is involved in insulin exocytosis. J. Biol. Chem. 270: 12869–12876, 1995.
 151. Kowluru, A., and S. A. Metz. Stimulation by prostaglandin E2 of a high affinity GTPase in the secretory granules of normal rat and human pancreatic islets. Biochem. J. 15: 399–406, 1994.
 152. Kowluru, A., M. E., Rabiglia, K. E. Muse, and S. A. Metz. Subcellular localization and kinetic characterization of guanine nucleotide binding proteins in normal rat and human pancreatic islets and transformed cells. Biochim. Biaphys. Acta 1222: 348–359, 1994.
 153. Kozak, J. A., and D. E. Logothetis. A calcium‐dependent chloride current in the insulin‐secreting βTC‐3 cells. Pflugers Arch. 433: 679–690, 1997.
 154. Kozlowski, R. Z., and M. L. J. Ashford. ATP‐sensitive K+ channel run‐down is Mg2+ dependent. Proc. B. Soc. Lond. B Biol. Sci. 240: 397–410, 1990.
 155. Kukuljan, M., A. A., Goncalces, and I. Atwater. Charybdotoxin‐sensitive KCa‐channel is not involved in glucose‐induced electrical activity in pancreatic β‐cells. J. Membr. Biol. 119: 187–195, 1991.
 156. Lang, J., I., Nishimoto, R. Regazzi, C. Kiraly, U. Weller, and C. B. Wollheim. Direct control of exocytosis by receptor‐mediated activation of the heterotrimeric GTPases Gi and Go or by the expression of their active a‐subunits. EMBO J. 14: 3635–3644, 1995.
 157. Lankat‐Buttergereit, B., H. C., Fehman, B. J. Hering, R. G. Bretzel, and B. Goke. Expression of the ras‐related rab3a gene in human insulinomas and normal human pancreatic islets. Pancreas 9: 434–438, 1994.
 158. Larsson, O., J. T., Deeney, R. Branstrom, P.‐O. Berggren, and B. E. Corkey. Activation of the ATP‐sensitive K+ channel by long chain acyl‐CoA. A role in modulation of pancreatic beta‐cell glucose sensitivity. J. Biol. Chem. 271: 10623–10626, 1996.
 159. Larsson, O., H., Kindmark, R. Brandstrom, B. Fredholm, and P. O. Berggren. Oscillations in KATP channel activity promote oscillations in cytoplasmic free Ca2+ concentration in the pancreatic beta cell. Proc. Natl. Acad. Sci. U.S.A. 93: 5161–5165, 1996.
 160. Laychock, S. G.. Alpha 2‐adrenoceptor stimulation affects total glucose utilization in isolated islets of Langerhans. Mol. Pharmacol. 32: 241–248, 1987.
 161. Laychock, S. G.. Prostaglandin E2 and alpha 2 adrenoceptor agonists inhibit the pentose shunt in pancreatic islets. Arch. Biochem. Biophys. 269: 354–358, 1989.
 162. Laychock, S. G., and S. Bilgin. Alpha 2‐adrenoceptor agonists inhibition of pancreatic islet glucose utilization is mediated by an inhibitory guanine nucleotide regulatory protein. FEBS Lett. 218: 7–10, 1987.
 163. Laychock, S. G., and S. Bilgin. Calcium mobilization, prostaglandin E2 and α2‐adrenoceptor modulation of glucose utilization and insulin secretion in pancreatic islets. Biochem. Pharmacol. 38: 2511–2520, 1989.
 164. Lebrun, P., M.‐H., Antoine, R. Ouedraogo, M. J. Dunne, C. Kane, M. Hermann, A. Herchuelz, B. Marsereel, J. Delarge, P. de Tullio, and B. Pirotte. Activation of ATP‐dependent K+ channels and inhibition of insulin release; effect of BPDZ‐62. Pharmacol. Exp. Ter. 277: 156–162, 1996.
 165. Leech, C. A., G. C. Holz, IV, and J. F. Habener. Voltage‐independent calcium channels mediate slow oscillations of cytosolic calcium that are glucose dependent in pancreatic beta‐cells. Endocrinology. 135: 365–372, 1994.
 166. Leibowitz, G., and E. Cerasi. Sulphonylurea treatment of NIDDM patients with cardiovascular disease: a mixed blessing? Diabetologia 639: 503–514, 1996.
 167. Leibowitz, G., N., Weintrob, A. Pikarsky, Z. Josefsberg, H. Landau, B. Glaser, C. N. Hales, and E. Cerasi. Normal proinsulin processing despite beta‐cell dysfunction in persistent hyperinsulinaemic hypoglycaemia of infancy (nesidioblastosis). Diabetologia 11: 1338–1344, 1996.
 168. Li, G., D., Milani, M. J. Dunne, W.‐F. Pralong, J.‐M. Theler, O. H. Petersen, and C. B. Wollheim. Extracellular ATP causes Ca2+‐dependent and independent insulin secretion in RINm5F cells. J. Biol. Chem. 266: 3449–3457, 1991.
 169. Li, G., R., Regazzi, W. E. Balch, and C. B. Wollheim. Stimulation of insulin release from permeabilized cells by a synthetic peptide corresponding to the effector domain of the small GTP‐binding protein rab3. FEBS Lett. 327: 145–149, 1993.
 170. Li, G., S., Regunathan, C. J. Barrow, J. Eshraghi, R. Cooper, and D. J. Reis. Agmatine: an endogenous clonidine displacing substance in the brain. Science 263: 966–969, 1994.
 171. Lindau, M., and E. Neher. Patch‐clamp techniques for time‐resolved capacitance measurements in single cells. Pflugers Arch. 411: 137–146, 1988.
 172. Lindley, K. J., M. J., Dunne, C. Kane, R. M. Shepherd, P. E. Squires, R. F. L. James, P. R. V. Johnson, S. Eckhart, E. Wakeling, M. Dattani, P. J. Milla, and A. Aynsley‐Green. Ionic control of β‐cell function in nesidioblastosis. A possible therapeutic role for calcium channel blockade. Arch. Dis. Child. 74: 373–378, 1996.
 173. Lindskog, S., and B. Ahren. Studies on the mechanism by which galanin inhibits insulin secretion in islets. Eur. J. Pharmacol. 205: 21–27, 1991.
 174. Llinas, R., J. A., Gruner, M. Sugimori, T. L. McGuiness, and P. Greengard. Regulation by synapsin I and Ca2+‐calmodulin‐dependent protein kinase II of transmitter release in squid giant synapse. J. Physiol. (Lond.) 436: 257–282, 1991.
 175. Lundquist, I., B., Ahrén, R. H˚akansson, and F. Sundler. Quinacrine accumulation in pancreatic islet cells of rat and mouse: relationship to functional activity and effects on basal and stimulated insulin secretion. Diabetologia 28: 161–166, 1985.
 176. Martin, S. C., D. I., Yule, M. J. Dunne, D. V. Gallacher, and O. H. Petersen. Vasopressin directly closes ATP‐sensitive potassium channels evoking membrane depolarisation and an increase in the free intracellular Ca2+ concentration in insulin‐secreting cells. EMBO J. 8: 3595–3599, 1989.
 177. Mathias, P. C., L., Best, and W. J. Malaisse. Stimulation by glucose and carbamycholine of phospholipase C in pancreatic islets. Cell. Biochem. Funct. 3: 173–177, 1985.
 178. Matthews, E. K., and Y. Sakamoto. Electrical characteristics of pancreatic islet cells. J. Physiol. (Lond.) 246: 421–437, 1975.
 179. McClue, S. J., E., Selzer, M. Freissmuth, and G. Milligan. Gi3 does not contribute to the inhibition of adenylate cyclase when stimulation of an α2‐adrenergic receptor causes activation of both Gi2 and Gi3. Biochem. J. 284: 565–568, 1992.
 180. McDermott, A., and G. W. G. Sharp. Inhibition of insulin secretion; a fail‐safe system. Cell. Signal. 5: 229–234, 1993.
 181. McDermott, A. M., and G. W. G. Sharp. Gi‐2 and Gi‐3 proteins mediate the inhibition of adenylyl cyclase by galanin in the RINm5F cell. Diabetes 44: 453–459, 1995.
 182. McKenzie, F. R., and G. Milligan. δ‐Opioid‐receptor‐mediated inhibition of adenylate cyclase is transduced specifically by the guanine‐nucleotide‐binding protein Gi2. Biochem. J. 267: 391–398, 1990.
 183. Meltz, S. A.. Exogenous arachidonic acid promotes insulin release from intact or permeabilized rat islets by dual mechanisms: putative activation of Ca2+ mobilization and protein kinase C. Diabetes 37: 1453–1469, 1988.
 184. Misler S, L. C. Falke, K. Gillis, and M. L. McDaniel. A metabolite regulated potassium channel in rat pancreatic β‐cells. Proc. Natl. Acad. Sci. U.S.A. 83: 7119–7123, 1986.
 185. Mizuta, M., N., Inagald, Y. Nemoto, S. Matsukuru, M. Takahasi, and S. Seino. synaptotagmin III is a novel isoform of rat Synaptotagimin expressed in endocrine and neuronal cells. J. Biol. Chem. 169: 11675–11678, 1994.
 186. Mojsov, S., G. G., Weir, and J. F. Habener. Insulinotropin: glucagon‐like peptide I (7–37) co‐encoded in the glucagon gene is a potent stimulator of insulin release in the perfused rat pancreas. J. Clin. Invest. 79: 616–619, 1987.
 187. Morgan, N. G., S. L. F., Chan, C. A. Brown, and E. Tsoli. Characterization of the imidazoline binding site involved in regulation of insulin secretion. Ann. NY. Acad. Sci. 763: 361–366, 1995.
 188. Morgan, N. G., and W. Montague. Studies on the mechanism of inhibition of glucose‐stimulated insulin secretion in rat islets of Langerhans. Biochem. J. 226: 571–576, 1985.
 189. Neher, E., and A. Marty. Discrete changes in cell membrane capacitance observed under conditions of enhanced secretion in bovine adrenal chromaffin cells. Proc. Natl. Acad. Sci. U.S.A. 72: 6712–6716, 1982.
 190. Nestorowicz, A., K. P., Schoor, B. A. Wilson, B. Glaser, S. Seino, and M. A. Permutt. Identification of a nonsense mutation in the β‐cell inward rectifier gene, BIR, associated with familial hyperinsulinism [abstract]. J. Invest. Med. 44: p. 175, 1996.
 191. Nestorowicz, A., B. A., Wilson, K. P. Schoor, H. Inoue, B. Glaser, H. Landau, C. A. Stanley, P. S. Thornton, J. P. Clement, IV, J. Bryan, L. Aguilar‐Bryan, and M. A. Permutt. Mutations in the sulphonylurea receptor gene are associated with familial hyperinsulinism in Ashkenazi Jews. Hum. Mol. Genet. 5: 1813–1822, 1996.
 192. Nichols, C. G., S.‐L., Shyng, A. Nestorowicz, B. Glaser, J. P. Clement, IV, G. Gonzalez, L. Aguilar‐Bryan, M. A. Permutt, and J. Bryan. Adenosine diphosphate as an intracellular regulator of insulin secretion. Science 272: 1785–1787, 1996.
 193. Neuwenhuizen, A. G., S., Karlsson, T. Fridolf, and B. Ahren. Mechanisms underlying the insulinostatic effect of peptide YY in mouse pancreatic islets. Diabetologia 37: 871–878, 1994.
 194. Nilsson, T., P., Arkhammar, P. Rorsman, and P.‐O. Berggren. Suppression of insulin release by galanin and somatostatin is mediated by a G‐protein. An effect involving repolarization and reduction in cytoplasmic free Ca2+ concentration. J. Biol. Chem. 264: 973–980, 1989.
 195. Nilsson, T., V., Schultz, P. O. Berggren, B. E. Corkey, and K. Tornheim. Temporal patterns of changes in ATP/ADP ratio, glucose 6‐phosphate and cytoplasmic free Ca2+ in glucose‐stimulated pancreatic beta‐cells. Biochem. J. 314: 91–4, 1996.
 196. Niwa, K., I., Shibuya, and T. Kanno. Temperature dependence of processes proximal and distal to the glucose‐induced [Ca2+] rise in stimulus–secretion coupling in rat pancreatic islets. Biol. Signals 5: 30–43, 1996.
 197. Ohno‐Shosaku, T., B. J., Zunkler, and G. Trube. Dual effects of ATP on K+ currents in mouse pancreatic B‐cells. Pflugers Arch. 408: 133–138, 1987.
 198. Oiszewski, S., J. T., Deeny, G. T. Schupin, K. P. Williams, B. E. Corkey, and C. J. Rhodes. Rab3A effector domain peptides induce insulin exocytosis via a specific interaction with a cytosolic protein doublet. J. Biol. Chem. 269: 27987–27991, 1994.
 199. Östenson, C. G., J., Pigon, J. C. Doxey, and S. Efendic. Alpha 2‐adrenoceptor blockade does not enhance glucose‐induced insulin release in normal subjects or patients with non insulin‐dependent diabetes. Clin. Endocrinol. Metab. 67: 1054–1059, 1988.
 200. Ozanne, S. E., P. C., Guest, J. C. Hutton, and C. N. Hales. Intracellular localization and molecular heterogeneity of the sulphonylurea receptor in insulin‐secreting cells. Diabetologia 38: 277–282, 1995.
 201. Pace, C. S., M., Murphy, S. Conant, and P. E. Lacey. Somatostatin inhibition of glucose‐induced electrical activity in cultured rat islets. Am. J. Physiol. 223 (Cell Physiol): C164–C171, 1977.
 202. Pace, C. S., and J. S. Smith. The role of chemiosmotic lysis in the exocytotic release of insulin. Endocrinology 113: 964–969, 1983.
 203. Persaud, S. J., P. M., Jones, and S. L. Howell. Effects of Bordetella pertussis toxin on catecholamine inhibition of insulin release from intact and electrically permeabilized rat islets. Biochem. J. 258: 669–675, 1989.
 204. Petersen, O. H., and M. J. Dunne. Regulation of K+ channels plays a crucial role in control of insulin secretion. Pflugers Arch. 414: S115–S120, 1989.
 205. Petersen, O. H., and I. Findlay. Electrophysiology of the pancreas. Physiol. Rev. 67: 1054–1116, 1987.
 206. Petit, P., M., Manteghetti, and M. M. Loubatieres‐Mariani. Differential effects of purinergic and cholinergic activation on the hydrolysis of membrane polyphosphoinositides in rat pancreatic islets. Biochem. Pharmacol. 37: 1213–1217, 1988.
 207. Philipson, L. H., M. P., Rosenberg, A. Kuznetsov, M. E. Lancaster, J. F. Worley, III, M. W. Roe, and I. D. Dukes. Delayed rectifier K+ channel overexpression in transgenic islets and β‐cells associated with impaired glucose responsiveness. J. Biol. Chem. 269: 27787–27790, 1994.
 208. Pirotte, B., P. De Tullio, P. Lebrun, M. Antoine, J. Fontaine, B. Masereel, M. Schynts, L. Dupont, A. Herchuelz, and J. Delage. 3‐(Alkylamino)‐4H‐pryido[4,3‐e]‐1,2,4‐thiadiazine 1,1‐diazoxides as powerful inhibitors of insulin release from rat pancreatic β‐cells: a new class of potassium channel openers? J. Med. Chem. 36: 3211–3213, 1993.
 209. Plant, T. D.. Na+ currents in cultured mouse pancreatic B‐cells. Pflugers Arch. 411: 429–435, 1988.
 210. Powis, G., R., Bonjouklian, M. M. Berggren, A. Gallegos, R. Abraham, C. Ashendal, L. Zalkow, W. F. Matter, J. Dodge, G. Grindey, and C. J. Vlahos. Wortmannin, a potent and selective inhibitor of phosphatidylinositol‐3‐kinase. Cancer Res. 54: 2419–2423, 1994.
 211. Pralong, W. F., C., Bartley, and C. B. Wollheim. Single islet β‐cell stimulation by nutrients: relationship between pyridine nucleotides, cytosolic Ca2+ and secretion. EMBO J. 9: 53–60, 1990.
 212. Prentki, M., and B. E. Corkey. Are the beta‐cell signaling molecules malonyl‐CoA and cytosolic long‐chain acyl‐CoA implicated in multiple tissue defects of obesity and NIDDM? Diabetes 45: 273–283, 1996.
 213. Prentki, M., and F. Matschinsky. Ca2+, cAMP and phospholipid‐derived messengers in coupling mechanisms of insulin secretion. Physiol. Rev. 67: 1185–1248, 1987.
 214. Prentki, M., S., Vischer, M. C. Glennon, R. Regazzi, J. T. Deeney, and B. E. Corkev. Malonyl‐CoA and long chain acyl‐CoA esters as metabolic coupling factors in nutrient‐induced insulin secretion. J. Biol. Chem. 267: 5802–5810, 1992.
 215. Proks, P., and F. M. Aschcroft. Effects of divalent cations in exocytosis and endocytosis from single mouse pancreatic β‐cells. J. Physiol. (Lond.) 487: 465–477, 1995.
 216. Proks, P., L., Eliasson, C. Ämmälä, P. Rorsman, and F. M. Ashcroft. Ca2+ and GTP‐dependent exocytosis in mouse pancreatic β‐cells involves both common and distinct steps. J. Physiol. (Lond.) 496: 255–266, 1996.
 217. Proks, P., M., Takano, and F. M. Ashcroft. Effects of intracellular pH on ATP‐sensitive K+ channels in mouse pancreatic beta‐cells. J. Physiol. (Lond.) 475: 33–44, 1994.
 218. Rabinovitch, A., E. Cerasi, And G. W. E. Sharp. Adensoine 3′,5′‐monophosphate‐dependent and inhibitory effects of epinephrine on insulin release in rat pancreatic islets. Endocrinology 102: 1733–1740, 1978.
 219. Ramanadham, S., R. W., Gross, X. Han, and J. Turk. Inhibition of arachidonate release by secretagogue‐stimulated pancreatic islets suppresses both insulin secretion and the rise in β‐cell cytosolic calcium ion concentration. Biochemistry 32: 337–346, 1993.
 220. Reale, V., C. N., Hales, and M. L. J. Ashford. The effects of pyridine nucleotides on the activity of a calcium‐activated non‐selective cation channel in the rat insulinoma cell line, CRI‐G1. J. Memhr. Biol. 142: 299–307, 1994.
 221. Reale, V., C. N., Hales, and M. L. J. Ashford. Regulation of calcium‐activated non‐selective cation channel activity by cyclic nucleotides in the rat insulinoma cell line, CRI‐G1. J. Membr. Biol. 145: 267–278, 1995.
 222. Regazzi, R., C. B., Wollheim, J. Lang, J. M. Theler, O. Rossetto, C. Montecucco, K. Sadoul, U. Weller, M. Palmer, and B. Thorens. VAMP‐2 and cellubrevin are expressed in pancreatic β‐cells and are essential for Ca2+‐but not for GTP‐g‐S‐induced insulin secretion. EMBO J. 14: 2723–2730, 1995.
 223. Remaury, A., D., Larrouy, D. Daviaud, B. Rouot, and H. Paris. Coupling of β2‐adrenergic receptor to the inhibitory G‐protein Gi and adenylate cyclase in HT29 cells. Biochem. J. 292: 283–288, 1993.
 224. Renström, E., W. G., Ding, K. Bokvist, and P. Rorsman. Neurotransmitter induced inhibition of exocytosis in insulin secreting beta cells by activation of calcineurin. Neuron 17: 513–522, 1996.
 225. Renström, E., L., Eliasson, K. Bokvist, and P. Rorsman. Cooling inhibits exocytosis in single mouse pancreatic B cells by suppression of granule mobilization. J. Physiol. (Lond.) 494: 41–52, 1996.
 226. Ribalet, B., and S. Ciani. Characterization of the G protein coupling of a glucagon receptor to the K‐ATP channel in insulin‐secreting cells. J. Membr. Biol. 142: 395–408, 1994.
 227. Ribalet, B. and G. T., Eddlestone. Characterization of the G protein coupling of a somatostatin receptor to the KATP channel in insulin‐secreting mammalian HIT and RIN cell lines. J. Physiol. (Lond.) 485: 73–86, 1995.
 228. Ribalet, B., G. T., Eddlestone, and S. Ciani. Metabolic regulation of the K(ATP) and a K(MAXI) channel in the insulin‐secreting RINm5F cell. J. Gen. Physiol. 92: 219–237, 1988.
 229. Richardson, S. B., T., Laya, M. Gibson, N. Eyler, and M. Van Ooy. Somatostatin inhibits vasopressin‐stimulated phosphoinositide hydrolysis and influx of extracellular calcium in clonal hamster beta (HIT) cells. Biochem. J. 288: 847–851, 1992.
 230. Robertson, R. P., P., Tsai, S. A. Little, H. J. Zhang, and T. F. Walseth. Receptor‐mediated adenylate cyclase‐coupled mechanism for PGE2 inhibition of insulin secretion in HIT cells. Diabetes 36: 1047–1053, 1987.
 231. Roe, M. W., M. E., Lancaster, R. J. Mertz, J. F. Worley III, and I. D. Dukes. Voltage‐dependent intracellular calcium release from mouse islets stimulated by glucose. J. Biol. Chem. 268: 9953–9956, 1993.
 232. Ronner, P., T. J., Higgins, and G. A. Kimmich. Inhibition of ATP‐sensitive K+ channels in pancreatic β‐cells by non sulphonylurea drug linogliride. Diabetes 40: 885–892, 1991.
 233. Rorsman, P., K., Bokvist, C. Ämmälä, P. Arkhammar, P.‐O. Berggren, O. Larsson, and K. Wåhlander. Activation by adrenaline of a low conductance G protein‐dependent K+ channel in mouse pancreatic β‐cells. Nature 349: 77–79, 1991.
 234. Rorsman, P., and G. Trube. Calcium and delayed potassium currents in mouse β‐cells under voltage‐clamp conditions. J. Physiol. (Lond.) 371: 531–550, 1986.
 235. Rorsman, P., and G. Trube. Biophysics and physiology of ATP‐regulated K+ channel (KATP) channel. In. Potassium Channels, edited by N. S. Cook. New York: Wiley, 1990, p. 96–116.
 236. Rothman, J. E.. Mechanisms of intracellular protein transport. Nature 372: 55–63, 1994.
 237. Ripoll, C., F., Martin, J. M. Rovira, J. Pintor, M. T. Miras‐Portugal, and B. Soria. Diadenosine polyphosphates. A novel class of glucose‐induced intracellular messengers in the pancreatic beta‐cell. Diabetes 45: 1431–1434, 1996.
 238. Sadoul, K., J., Lang, C. Montecucco, U. Weller, R. Regazzi, S. Catsicas, C. B. Wollheim, and P. A. Halban. SNAP‐25 is expressed in islets of Langerhans and is involved in insulin release. J. Cell Biol. 128: 1019–1028, 1995.
 239. Sakura, H., C., Ämmälä, P. A. Smith, F. M. Gribble, and F. M. Ashcroft. Cloning and functional expression of the cDNA encoding a novel ATP‐sensitive potassium channel expressed in pancreatic B‐cells, brain, heart and skeletal muscle. FEBS Lett. 377: 338–344, 1995.
 240. Satin, L. S., S. J., Tavalin, and P. D Smolen. Inactivation of HIT cell Ca2+ current by a stimulated burst of Ca2+ action potentials. Biophys. J. 66: 141–148, 1994.
 241. Sato, Y., T., Aizawa, M. Komatsu, N. Okada, and T. Yamada. Dual functional role of membrane depolarization/Ca2+ influx in rat pancreatic β‐cell. Diabetes 41: 438–443, 1992.
 242. Schmidt, A., J., Hescheler, S. Offermanns, K. Spicher, K.‐D. Hinsch, F.‐J. Klinz, J. Codina, L. Birnbaumer, H. Gausepohl, R. Frank, G. Schultz, and W. Rosenthal. Involvement of pertussis toxin–sensitive G‐proteins in the hormonal inhibition of dihydropyridine‐sensitive Ca2+ currents in an insulin‐secreting cell line (RINm5F). J. Biol. Chem. 266: 18025–18033, 1991.
 243. Seaquist, E. R., M. B., Armstrong, T. W. Gettys, and T. F. Walseth. Somatostatin selectively couples to Goa in HIT‐T15 cells. Diabetes 44: 85–89, 1995.
 244. Seaquist, E. R., A. R., Neal, K. D. Shoger, T. F. Walseth, and R. P. Robertson. G‐proteins and hormonal inhibition of insulin secretion from HIT‐T15 cells and isolated rat islets. Diabetes 41: 1390–1399, 1992.
 245. Sehlin, J.. Interrelationship between chloride fluxes in pancreatic islets and insulin release. Am. J. Physiol. 235 (Endocrinol. Metab. Gastrointest. Physiol. 4): E501–508, 1978.
 246. Sehlin, J., and I.‐B. Täljedal. Glucose‐induced decrease in Rb+ permeability in pancreatic β‐cells. Nature 253: 635–636, 1975.
 247. Sener, A., P., Lebrun, F. Blachier, and W. J. Malaisse. Stimulus–secretion coupling of arginine‐induced insulin release. Biochem. Pharmacol. 38: 327–330, 1989.
 248. Sharp, G. W. G., Y. Le Marchand‐Brustel, T. Yada, L. L. Russo, C. R. Bliss, M. Cormont, L. Monge, and E. Van Obberghen. Galanin can inhibit insulin release by a mechanism other than membrane hyperpolarization or inhibition of adenylate cyclase. J. Biol. Chem. 264: 7302–7309, 1989.
 249. Sharp, G. W. G., D. E., Wiedenkeller, D. Kaelin, E. G. Siegel, and C. B. Wollheim. Stimulation of adenylate cyclase by Ca2+ and calmodulin in rat islets of Langerhans; explanation for the glucose‐induced increase in cyclic AMP levels. Diabetes 29: 74–77, 1980.
 250. Shepherd, R. M., M. N., Hashmi, C. Kane, P. E. Squires, and M. J. Dunne. Elevation of cytosolic calcium by imidazolines in mouse islets of Langerhans; implications for stimulus–response coupling of insulin release. Br. J. Pharmacol. 119: 911–916, 1996.
 251. Sher, E., A., Codignola, M. Rogers, and J. Richmond. Noradrenaline inhibition of Ca2+ channels and secretion in single patch clamped insulinoma cells. FEBS Lett. 385: 176–180, 1986.
 252. Simonds, W. F., P. K., Goldsmith, J. Codina, C. G. Unson, and A. M. Spiegel. Gi‐α2 mediates α2‐adrenergic inhibition of adenylyl cyclase in platelet membranes; in situ identification with Gα C‐terminal antibodies. Proc. Natl. Acad. Sci. U.S.A. 86: 7809–7813, 1989.
 253. Skoglund, G., C. R., Bliss, and G. W. G. Sharp. Galanin‐stimulated high‐affinity GTPase activity in plasma membranes from RINm5F cells. Diabetes 42: 74–79, 1993.
 254. Smith, P. A., F. M. Ashcroft, and C. Fewtrell. Permeation and gating properties of the L‐type calcium channel in mouse pancreatic β‐cells. J. Gen. Physiol. 101: 767–797, 1993.
 255. Smith, P. A., K. Bokvist, P. Arkhammar, P.‐O. Berggren, and P. Rorsman. Delayed rectifier and calcium‐activated K+ channels and their significance for action potential repolarization in mouse pancreatic β‐cells. J. Gen. Physiol. 95: 1041–1059, 1990.
 256. Smith, P. A., K., Bokvist, and P. Rorsman. Demonstration of Acurrents in pancreatic islet cells. Pflugers Arch. 413: 441–443, 1989.
 257. Smith, P. A., M. R., Duchen, and F. M. Ashcroft. A fluorimetric and amperometric study of calcium and secretion in isolated mouse pancreatic β‐cells. Pflugers Arch. 430: 808–818, 1995.
 258. Smith, P. A., P., Rorsman, and F. M. Ashcroft. Modulation of dihydropyridine‐sensitive Ca2+ channels by glucose in mouse pancreatic β‐cells. Nature 342: 550–553, 1990.
 259. Sollner, T., and J. E. Rothman. Neurotransmission: harnessing fusion machinery at the synapse. Trends Neurosci. 8: 344–347, 1994.
 260. Straub, S. G., and G. W. G. Sharp. A wortmannin‐sensitive signal transduction pathway is involved in the stimulation of insulin release by VIP and PACAP. J. Biol. Chem. 271: 1660–1668, 1996.
 261. Straub, S. G., and G. W. G. Sharp. Glucose‐dependent insulino‐tropic polypeptide stimulates insulin secretion via increased cyclic AMP and [Ca2+], and a wortmannin‐sensitive signaling pathway. Biochem. Biophys. Res. Commun. 224: 369–374, 1996.
 262. Straub, S. G., and G. W. G. Sharp. Mechanisms of action of VIP and PACAP in the stimulation of insulin release. Ann. NY Acad. Sci. 805: 607–612, 1996.
 263. Sturgess, N., M. L. J., Ashford, D. L. Cook, and C. N. Hales. The sulphonylurea receptor may be an ATP‐sensitive potassium channel. Lancet 2: 474–475, 1985.
 264. Sturgess, N. C., C. N., Hales, and M. L. J. Ashford. Inhibition of a calcium‐activated non‐selective cation channel in a rat insulinoma cell line, by adenine derivatives. FEBS Lett. 208: 397–400, 1986.
 265. Sturgess, N. C., C. N., Hales, and M. L. J. Ashford. Calcium and ATP regulate the activity of non‐selective cation channels in rat insulinoma cell line. Pflugers Arch. 409: 607–615, 1987.
 266. Südhof, T. C.. The synaptic vesicle cycle: a cascade of protein–protein interactions. Nature 375: 645–653, 1995.
 267. Szentivanyi, A., C. W., Fishel, and D. W. Talmadge. Adrenal mediation of histamine and serotonin hyperglycemia in normal mice and the absence of adrenal‐induced hyperglycemia in pertussis sensitized mice. J. Infect. Dis. 113: 86–91, 1963.
 268. Tallent, M., and T. Reisine. Gial selectively couples somatostatin receptors to adenylyl cyclase in pituitary‐derived AtT‐20 cells. Mol. Pharmacol. 41: 452–455, 1992.
 269. Tamagawa, T., and J. C. Henquin. Chloride modulation of insulin release, 86Rb+ efflux, and 45Ca2+ fluxes in rat islets stimulated by various secretagogues. Diabetes 32: 416–423, 1983.
 270. Tamagawa, T., H., Niki, and A. Niki. Insulin release independent of a rise in cytosolic free Ca2+ by forskolin and phorbol ester. FEBS Lett. 183: 430–432, 1985.
 271. Tamagawa, A., I., Niki, H. Niki, and A. Niki. Catecholamines inhibit insulin release independently of changes in cytosolic free Ca2+. Biomed Res. 6: 429–432, 1985.
 272. Tang, S., G. C., Yaney, and G. W. G. Sharp. Unusual response to carbachol in the RINm5F cell. Evidence for a “distal” site of action in stimulus–secretion coupling. Mol. Pharmacol 47: 863–870, 1995.
 273. Thomas, P. M., C. J., Cote, N. Wohlik, B. Haddad, P. M. Mathew, W. Rabl, L. Aguilar‐Bryan, R. F. Gagel, and J. Bryan. Mutations in the sulphonylurea receptor gene in familial hyperinsulinaemic hypoglycaemia of infancy. Science 268: 426–429, 1995.
 274. Thomas, P. M., N., Wohlik, E. Huang, U. Kuhnle, W. Rabl, R. F. Gagel, and C. J. Cote. Inactivation of the first nucleotide binding fold of the sulphonylurea receptor, and familial persistent hyperinsulinaemic hypoglycaemia of infancy. Am. J. Hum. Genet. 59: 510–518, 1996.
 275. Thomas, P. M., Y., Yuyang, and E. Lightner. Mutation of the pancreatic islet inward rectifier Kir6.2 also leads to familial persistent hyperinsulinaemic hypoglycaemia of infancy. Hum. Mol. Genet. 5: 1809–1812, 1996.
 276. Tucker, S. J., F. M., Gribble, C. Zhao, S. Trapp, and F. M. Ashcroft. Truncation of Kir6.2 produces ATP‐sensitive K+ channels in the absence of the sulphonylurea receptor. Nature 387: 179–183, 1997.
 277. UK Prospective Diabetes Study Group (UKPDS) 16. Overview of 6 years therapy of type II diabetes; a progressive disease. Diabetes 44: 1249–1258, 1995.
 278. Ullrich, S., M., Prentki, and C. B. Wollheim. Somatostatin inhibition of Ca2+‐induced insulin secretion in permeabilized HIT‐T15 cells. Biochem. J. 270: 273–276, 1990.
 279. Ullrich, S., and C. B. Wollheim. Galanin inhibits insulin secretion by direct interference with exocytosis. FEBS Lett. 247: 401–404, 1989.
 280. Vara, E., and J. Tamarit‐Rodriguez. Norepinephrine inhibits islet lipid metabolism, 45Ca2+ uptake, and insulin secretion. Am. J. Physiol. 257 (Endocrinol. Metab. 20): E923–E929, 1989.
 281. Vara, E., and J. Tamarit‐Rodriguez. Does cyclic guanosine monophosphate mediate noradrenaline‐induced inhibition of islet insulin secretion stimulated by glucose and palmitate? Biochem. J. 278: 243–248, 1991.
 282. Wheeler, M. B., L., Sheu, M. Ghai, A. Bouquillon, G. Grondin, U. Weller, A. R. Beaudoin, M. K. Bennett, W. S. Trimble, and H. Y. Gaisano. Characterization of SNARE protein expression in β cell lines and pancreatic islets. Endocrinology 137: 1340–1348, 1996.
 283. White, R. E., A., Schonbrunn, and D. L. Armstrong. Somatostatin stimulates Ca2+‐activated K+ channels through protein dephosphorylation. Nature 351: 570–573, 1991.
 284. Williams, B. A., P. A., Smith, K. Leow, S. Shimizu, D. W. Gray, and F. M. Ashcroft. Two types of potassium channel regulated by ATP in pancreatic β‐cells isolated from a type‐2 diabetic human. Pflugers Arch. 423: 265–273, 1993.
 285. Wolf, B. A., R. A., Eason, J. H. Hughes, M. L. McDaniel, and J. Urek. Secretagogue‐induced diacylglycerol accumulation in isolated pancreatic islets: mass spectrometric characterization of the fatty acyl content indicates multiple mechanisms of generation. Biochemistry 28: 4291–4301, 1989.
 286. Wolf, B. A., R. J., Konrad, and C. A. Alter. Role of phospholipases C, A2 and D in insulin secretion. In: Insulin Secretion and Pancreatic β‐Cell Research, edited by P. R. Flatt and S. Lenzen. 1994, vol. 39, p. 297–304.
 287. Wollheim, C. B., and T. J. Biden. Second messenger function of inositol 1,4,5‐trisphophate. J. Biol. Chem. 261: 8314–8319, 1986.
 288. Wollheim, C. B., M. J., Dunne, B. Peter‐Riesch, R. Bruzzone, T. Pozzan, and O. H. Petersen. Activators of protein kinase C depolarize insulin‐secreting cells by closing K+ channels. EMBO J. 7: 2443–2449, 1988.
 289. Wollheim, C. B., M., Kikuchi, A. E. Renold, and G. W. G. Sharp. Somatostatin‐ and epinephrine‐induced modifications of 45Ca2+ fluxes and insulin release in rat pancreatic islets maintained in tissue culture. J. Clin. Invest. 60: 1165–1173, 1977.
 290. Wollheim, C. B., and G. W. G. Sharp. Regulation of insulin release by calcium. Physiol. Rev. 61: 914–973, 1981.
 291. Worley III, J. F., M. S. McIntyre, B. Spencer, R. J. Mertz, M. W. Roe, and I. D. Dukes. Endoplasmic reticulum calcium store regulates membrane potential in mouse islet beta‐cells. J. Biol. Chem. 269: 14359–14362, 1994.
 292. Yada, T., M., Sakurada, K. Ihida, M. Nakata, F. Murata, A. Arimura, and M. Kikuchi. Pituitary adenylate cyclase activating polypeptide is an extraordinarily potent intra‐pancreatic regulator of insulin secretion from islet beta‐cells. J. Biol. Chem. 269: 1290–1293, 1994.
 293. Yada, T., M., Sakurada, M. Nakata, K. Ihida, K. Yaekura, S. Shioda, and M. Kikuchi. Current status of PACAP a regulator of insulin secretion in pancreatic islets. Ann. NY Acad. Sci. 805: 607–612, 1996.
 294. Yajima, M., K., Hosoda, Y. Kanbayashi, T. Nakamura, K. Nogimori, Y. Nakase, and M. Ui. Islets‐activating protein (IAP) in Bordetella pertussis that potentiates insulin secretory response of rats. Purification and characterization. J. Biochem. (Tokyo) 83: 295–303, 1978.
 295. Yamatani, Y., T., Chiba, S. Kadowaki, R. Hishikawa, A. Yamaguchi, T. Inui, T. Fujita, and S. Kawazu. Dual action of protein kinase C activation in the regulation of insulin release by muscarinic agonist from rat insulinoma cell line (RINr). Endocrinology 122: 2826–2832, 1988.
 296. Zaitsev, S. V., A. M., Efanov, I. B. Efanova, O. Larsson, C. G. Ostenson, G. Gold, P. O. Berggren, and S. Efendic. Imidazoline compounds stimulate insulin release by inhibition of K(ATP) channels and interaction with the exocytotic machinery. Diabetes 45: 1610–1618, 1996.
 297. Zawalich, W. S., and H. Rasmussen. Control of insulin secretion: a model involving calcium and cyclic AMP and diacylglycerol. Mol. Cell. Endocrinol. 70: 119–138, 1990.
 298. Zhou, Z., and S. Misler. Amperometric detection of quantal secretion from patch‐clamped rat pancreatic beta‐cells. J. Biol. Chem. 271: 270–277, 1996.
 299. Zigman, J. M., G. T., Westermark, J. LaMendola, E. Boel, and D. F. Steiner. Human Golfα: complementary deoxyribonucleic acid structure and expression in pancreatic islets and other tissues outside the olfactory neuroepithelium and central nervous system. Endocrinology 133: 2508–2514, 1993.
 300. Zigman, J. M., G. T., Westermark, J. LaMendola, and D. F. Steiner. Expression of cone transducin, Gzα, and other G‐protein α‐subunit messenger ribonucleic acids in pancreatic islets. Endocrinology 135: 31–37, 1994.
 301. Zunkler, B. J., S., Lenzen, K. Manner, U. Panten, and G. Trube. Concentration‐dependent effects of tolbutamide, meglitinide, glipizide, glibenclamide and diazoxide on ATP‐regulated K+ currents in pancreatic β‐cells. Naunyn Schmiedebergs Arch. Pharmacol. 337: 225–230, 1988.
 302. Zunkler, B. J., S., Lins, T. Ohno‐Shosaku, G. Trube, and U. Panten. Cytosolic ADP enhances the sensitivity to tolbutamide of ATP‐dependent K+ channels from pancreatic β‐cells. FEBS Lett. 239: 241–244, 1988.

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Mark J. Dunne, Carina Ämmälä, Susanne G. Straub, Geoffrey W. G. Sharp. Electrophysiology of the β Cell and Mechanisms of Inhibition of Insulin Release. Compr Physiol 2011, Supplement 21: Handbook of Physiology, The Endocrine System, The Endocrine Pancreas and Regulation of Metabolism: 79-123. First published in print 2001. doi: 10.1002/cphy.cp070204