Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Am J Physiol Endocrinol Metab 295: E751–E761, 2008.

First published July 22, 2008; doi:10.1152/ajpendo.90295.2008. Review

Insulin as a physiological modulator of glucagon secretion


Pritpal Bansal1 and Qinghua Wang1,2,3
1
Department of Physiology, 2Department of Medicine, and 3Division of Endocrinology and Metabolism, Li Ka-Shing
Knowledge Institute, St. Michael’s Hospital, University of Toronto, Toronto, Ontario, Canada
Submitted 15 March 2008; accepted in final form 15 July 2008

Bansal P, Wang Q. Insulin as a physiological modulator of glucagon secretion.


Am J Physiol Endocrinol Metab 295: E751–E761, 2008. First published July 22,
2008; doi:10.1152/ajpendo.90295.2008.—Glucose homeostasis is regulated pri-
marily by the opposing actions of insulin and glucagon, hormones that are secreted
by pancreatic islets from ␤-cells and ␣-cells, respectively. Insulin secretion is
increased in response to elevated blood glucose to maintain normoglycemia by
stimulating glucose transport in muscle and adipocytes and reducing glucose
production by inhibiting gluconeogenesis in the liver. Whereas glucagon secretion
is suppressed by hyperglycemia, it is stimulated during hypoglycemia, promoting
hepatic glucose production and ultimately raising blood glucose levels. Diabetic
hyperglycemia occurs as the result of insufficient insulin secretion from the ␤-cells
and/or lack of insulin action due to peripheral insulin resistance. Remarkably,
excessive secretion of glucagon from the ␣-cells is also a major contributor to the
development of diabetic hyperglycemia. Insulin is a physiological suppressor of
glucagon secretion; however, at the cellular and molecular levels, how intraislet
insulin exerts its suppressive effect on the ␣-cells is not very clear. Although the
inhibitory effect of insulin on glucagon gene expression is an important means to
regulate glucagon secretion, recent studies suggest that the underlying mechanisms
of the intraislet insulin on suppression of glucagon secretion involve the modulation
of KATP channel activity and the activation of the GABA-GABAA receptor system.
Nevertheless, regulation of glucagon secretion is multifactorial and yet to be fully
understood.
diabetes; hyperglycemia; ␥-aminobutyric acid; adenosine 5⬘-triphosphate-sensitive
K⫹ channel

INSULIN AND GLUCAGON ARE PRIMARY ENDOCRINE FACTORS respon- Both the insulin-producing ␤-cells and the glucagon-secret-
sible for regulating the blood glucose levels (146). During ing ␣-cells are electrically excitable; however, each cell type
periods of hyperglycemia (i.e., after meal ingestion), whereas features a unique set of ion channels (72, 89). Unlike in ␤-cells,
insulin secretion is increased, glucagon secretion is reduced. ␣-cells are electrically silent in the presence of insulin-stimu-
Insulin promotes anabolism through facilitation of glucose latory glucose concentrations (56). Regulation of glucagon
transport in skeletal muscle and adipose tissue and stimulation secretion is mediated by electrical machinery comprised of ion
of hepatic glycogen synthesis (41, 124). Glucagon protects the channels and is modulated by glucose and paracrine factors
body against hypoglycemia to maintain an adequate level of (Fig. 1). The ␣-cells contain a large tetrodotoxin (TTX)-
blood glucose. This procedure is important particularly during sensitive Na⫹ current that inactivates at intermediate voltages
fasting, because glucose is the primary fuel source of the (56, 119) and are equipped with two types, low-voltage acti-
central nervous system (146). vated (T-type) (56, 88) and high-voltage activated (L- or
N-type) (11, 56, 88, 89, 95, 104, 156), of voltage-gated Ca2⫹
Glucagon Production and Secretion channels. T-type Ca2⫹ channels are partly responsible for
initiation of the depolarization cascade that is required to elicit
Glucagon is a 29-amino acid peptide synthesized and se- exocytosis (56, 72). Activated by the mild depolarization
creted by pancreatic ␣-cells, which comprise ⬃20 –30% of all caused by activation of the T-type Ca2⫹ channel, the TTX-
islet cells (7, 23). The human proglucagon gene is a 9.4-kb sensitive Na⫹ channel continues the depolarization cascade
sequence located on chromosome 2 (85), and translation of this (56) to trigger the influx of Ca2⫹ ions via either the L-type (56)
gene in the pancreas, gut, and brain creates a precursor peptide or N-type Ca2⫹ channel (55, 59, 95, 104), although in some
(85) that is differentially processed by a set of prohormone cases blockade of the L-type Ca2⫹ channel has no effect on
covertases, depending on the cell type (70). In the ␣-cell, ␣-cell secretion (55, 95, 104). In comparison, pancreatic
proglucagon is cleaved into glucagon by prohormone conver- ␤-cells also express TTX-sensitive Na⫹ channels (20, 37, 54,
tase 2 (120). 149), but they are half-maximally inactivated at a more nega-
tive voltage (Vh ⫽ ⫺100 mV) (54, 89) compared with TTX-
Address for reprint requests and other correspondence: Q. Wang, Dept. of
sensitive Na⫹ channels in ␣-cells (Vh ⫽ ⫺47 mV) (56, 89).
Medicine, Univ. of Toronto, St. Michael’s Hospital, 30 Bond St., Rm. 7005, Mouse ␤-cells lack low-voltage-activated Ca2⫹ channels (89) and
Toronto, ON, Canada M5B 1W8 (e-mail: qinghua.wang@utoronto.ca). possess only L-, P/Q-, N-, and R-type Ca2⫹ channels (149),
http://www.ajpendo.org 0193-1849/08 $8.00 Copyright © 2008 the American Physiological Society E751
Downloaded from journals.physiology.org/journal/ajpendo (177.016.070.107) on December 14, 2022.
Review
E752 INSULIN ACTION ON ␣-CELL SECRETION

Fig. 1. Electrical machinery of glucagon secretion in mouse ␣-cells. Glucagon secretion requires the initiation of a full depolarization cascade, which begins
when T-type Ca2⫹ channels are activated and depolarize the cell to an intermediate membrane potential. The tetrodotoxin (TTX)-sensitive Na⫹ channels then
open and allow an influx of Na⫹ ions to further depolarize the ␣-cell, leading to activation of the L- or N-type Ca2⫹ channels and generation of sustained Ca2⫹
influx that triggers the glucagon granule exocytosis. The cell membrane is repolarized by the opening of voltage-gated delayed rectifier K⫹ channels (KDR), and
the depolarization cascade recurs (56, 95). The ␣-cell ATP-sensitive K⫹ (KATP) channel activity within a “narrow window” is critical for allowing the operation
of this machinery; at low glucose concentrations, a subpopulation of the total number of KATP channels present on the ␣-cell is open and sets the membrane
potential to ⫺60 mV, causing activation of the T-type Ca2⫹ channels and subsequent depolarization cascades (56, 95). An elevation in glucose concentration
and a rise in the intracellular ATP/ADP ratio results in a closure of the KATP channels (56, 61, 95) that depolarizes the ␣-cell membrane potential beyond the
narrow window, causing voltage inactivation of ion channels involved in the depolarization cascade. Of note, in the rat ␣-cells the ATP/ADP ratio does not
increase in high glucose conditions (35), whereas in isolated ␣-cells glucose-induced closure of KATP channels stimulates glucagon release (42, 104). Therefore,
during hyperglycemia the paracrine modulators (i.e., insulin, Zn2⫹, and GABA) appear to be predominant in suppressing glucagon secretion presumably through
either direct interference of the KATP channel activity (42, 61, 75, 87, 132) or the ␣-cell membrane potential (118, 153, 157). [Ca2⫹]i, intracellular free Ca2⫹
concentration.

whereas a recent study using human ␤-cells found that they Kir6.2 (pore-forming) subunits (17, 138) yet were thought to
lack N- or R-type Ca2⫹ channels and express the T-type be operated differently in the ␣-cells (61). Interestingly, it has
isoform (20). Rat ␤-cells express T- and L-type Ca2⫹ been demonstrated by Olsen et al. (104) that the glucagon
channels (6). secretion pathway in isolated rat ␣-cells mimics the stimulus-
Both ␣-cells and ␤-cells possess ATP-sensitive K⫹ (KATP) secretion mechanism in ␤-cells. In these studies the authors
channels, which are weakly inwardly rectifying K⫹ channels found that prolonged treatment of 20 mM glucose causes
that are gated by the intracellular levels of adenosine triphos- closure of KATP channels and stimulates glucagon exocytosis
phate (ATP) (121). The KATP channels set the resting mem- (104). This effect is glycolysis dependent and mimicked by
brane potential of the cell (17, 56, 72) and thus link the treatment with the KATP channel blocker tolbutamide (104).
metabolic state of a cell to its electrophysiological activity. In Similar results were obtained by Franklin et al. (42). Although
the ␤-cells, a postprandially increased blood glucose concen- one must be cautious when interpreting results from experi-
tration enhances glucose metabolism (i.e., increases the ments on isolated ␣-cells because they are removed from any
[ATP]i) (137). As a result, the KATP channels close, leading to potential paracrine regulation from neighboring ␤-cells or
membrane depolarization and subsequent activation of voltage- somatostatin-secreting ␦-cells, these findings suggest that glu-
gated Ca2⫹ channels (137). The increased Ca2⫹ entry triggers cose metabolism and increased ATP production in ␣-cells
insulin release (137). Both the ␣-cell and ␤-cell KATP channels leads to KATP channel closure, ␣-cell membrane depolariza-
are comprised of the SUR1 (sulphonylurea receptor) (138) and tion, and glucagon secretion (104). However, a biphasic effect
AJP-Endocrinol Metab • VOL 295 • OCTOBER 2008 • www.ajpendo.org
Downloaded from journals.physiology.org/journal/ajpendo (177.016.070.107) on December 14, 2022.
Review
INSULIN ACTION ON ␣-CELL SECRETION E753
of KATP channel activity on glucagon secretion has recently els could be restored to the basal levels within 60 min during
been demonstrated in isolated mouse islets, because low doses hypoglycemia induced by insulin injection (50). The clear
of tolbutamide stimulate glucagon secretion (95) while inhib- counterregulatory mechanism functioning in the lack of gluca-
iting glucagon secretion at high concentrations (61, 95). These gon action has yet to be determined; however, a remarkable
findings suggest that a certain degree (i.e., narrow window) of increase in basal cAMP signaling in Gcgr⫺/⫺ liver and en-
␣-cell KATP channel activity sets the ␣-cell membrane poten- hanced responsiveness to epinephrine in the Gcgr⫺/⫺ hepato-
tial within a range that is critical for the operation of the cytes and white adipose tissues might be, in part, responsible
machinery of glucagon secretion (Fig. 1). Nevertheless, the for this compensation (50). The presence of marked ␣-cell
effects of modulating ␣-cell KATP channels are complex and hyperplasia in Gcgr⫺/⫺ islets also suggests that glucagon
yet to be fully understood. It is clear that the ␣-cell must be signaling is required for the preservation of normal islet cell
subject to a regulatory mechanism other than the direct action growth and function (50, 131). These islets also exhibit im-
of glucose (i.e., paracrine or intraislet) to ensure that glucagon paired insulin secretion when treated with insulin secreto-
secretion is inhibited during hyperglycemia. gogues such as arginine and carbachol, indicating that loss of
glucagon signaling may impact the insulin secretion pathway
Physiological Role of Glucagon (131). Interestingly, whole body insulin sensitivity in Gcgr⫺/⫺
Glucagon exerts a variety of biological actions via the mice was enhanced as indicated by hyperinsulinemic euglyce-
activation of the glucagon receptor, which is a member of a mic clamp studies (131), which may represent a compensatory
subfamily of G protein-coupled receptors. Glucagon receptors mechanism for a reduction in ␤-cell function. Furthermore, one
are present in the islet ␤-cells and a wide array of tissues, such compensatory pathway could be the elevation of gluca-
including the liver, kidney, adipose tissue, brain, adrenal gland, gon-like peptide 1 (GLP-1) signaling as a consequence of
duodenum, and heart (24). Glucagon has been shown to mod- increased production of the hormone by the ␣-cells in the face
ulate heart muscle contractility (108), ghrelin secretion (5), and of whole body glucagon resistance (31, 50). GLP-1 is an
gastrointestinal motility (102), among other actions (Table 1). insulinotropic hormone that exerts a variety of biological
Most importantly, glucagon protects against hypoglycemia by actions in mammals, including simulation of insulin secretion,
stimulating net hepatic glucose production through promotion promotion of ␤-cell growth, improvement of peripheral insulin
of glycogenolysis and gluconeogenesis and simultaneous inhi- sensitivity, and moderation of body weight gain (39). It is thus
bition of glycolysis and glycogenolysis (70, 146). not surprising that the elevated levels of GLP-1 may explain
Generation of mouse models featuring a whole body knock- why Gcgr⫺/⫺ mice are protected against streptozotocin-in-
out of the glucagon receptor gene (Gcgr⫺/⫺) has advanced our duced ␤-cell destruction and high-fat diet-induced obesity (31).
understanding of glucagon signaling and its roles in the regu- However, it is not clear whether the improved action of insulin
lation of glucose homeostasis. For instance, Gcgr⫺/⫺ mice on glucose clearance in Gcgr⫺/⫺ mice is a result of direct
display reduced fasting blood glucose levels and improved glucagon action or indirect GLP-1 action on the insulin-
glucose tolerance compared with the wild-type mice (50, 106). sensitive tissues. It is important to note that the remarkable
Interestingly, despite lacking glucagon action, these mice still elevation of glucagon levels in the Gcgr⫺/⫺ mice might have
retain their counterregulatory capacity since their glucose lev- further facilitated the GLP-1 signaling, because glucagon can,
although with reduced affinity and efficacy, bind and activate
the GLP-1 receptor (32).
Table 1. Effects of glucagon signaling in various organs
Receptor Hyperglucagonemia Causes Diabetic Hyperglycemia
Location Effect Ref. No.
Hyperglucagonemia has been established as a major contrib-
Liver Stimulates glycogenolysis and glyconeogenesis 70 utor to the development of diabetic hyperglycemia (128, 143).
Inhibits glycolysis and glycogenesis 70
Adrenal gland Inhibits ACTH-stimulated cortisol production 98
A recent study using mice that were administered glucagon via
Kidney Stimulates bicarbonate secretion 101 osmotic minipump for 28 days showed that chronic hyperglu-
Reduces urinary acidification 34 cagonemia caused elevated blood glucose levels, impaired
Increases glomerular mesangial cell proliferation 90 glucose tolerance, and glomerular abnormalities (91), symp-
Promotes natriuresis 80
Stimulates K⫹ excretion 3
toms that are representative of early-stage type 2 diabetes.
Increases glomerular filtration rate 2 Humans with type 2 diabetes have day-long plasma glucagon
Adipose tissue Stimulates brown fat thermogenesis 15 concentrations that are higher than normal (116), and this
Stimulates brown fat cell growth 14 hyperglucagonemia causes inappropriate elevation of hepatic
Duodenum Inhibits gastrointestinal motility 102 glucose output due to enhanced gluconeogenesis (48) and
Promotes relaxation of Sphincter of Oddi 26
Stomach Stimulates ghrelin secretion 73 glycogenolysis (48, 127). Interestingly, clinical studies have
Heart Increases heart rate and myocardial contractility 108 demonstrated that challenge with glucagon results only in a
and improves atrioventricular conduction transient and small rise in the glucose and insulin levels of
Brain Suppresses ghrelin secretion 5 healthy individuals (117). However, in insulin-withdrawn dia-
Stimulates somatostatin release from 129
hypothalamus
betes patients, the glycemic response to hyperglucagonemia is
Stimulates neuronal ketone metabolism 76 five to 15 times greater than in nondiabetic controls (128).
Suppresses feeding 67 These findings suggest that the action of glucagon is diabeto-
Stimulates sympathetic nervous activity 83 genic only under the conditions of insulin deficiency (117, 128,
␤-Cell Promotes glucose-stimulated insulin secretion 66 143). Studies in human subjects with type 1 diabetes, a disease
␣-Cell Stimulates ␣-cell exocytosis 94
characterized by insufficient insulin production due to autoim-
AJP-Endocrinol Metab • VOL 295 • OCTOBER 2008 • www.ajpendo.org
Downloaded from journals.physiology.org/journal/ajpendo (177.016.070.107) on December 14, 2022.
Review
E754 INSULIN ACTION ON ␣-CELL SECRETION

mune destruction of the ␤-cells, have demonstrated that hyper- ated by the PI 3-kinase-/Akt-dependent signaling pathway.
glucagonemia could develop even after an oral glucose chal- Although results derived from clonal cell lines may not nec-
lenge (1, 145). However, administration of exogenous insulin essarily be representative of actual ␣-cell physiology, insulin
in these patients is able to suppress excessive glucagon secre- also inhibits glucagon secretion in isolated rat islets treated
tion induced by hyperglycemia (136) or other stimuli such as with pyruvate, an intermediate of glycolysis that selectively
arginine (51). Therefore, the insulin action is essential to exert stimulates glucagon secretion (69). The role of intraislet insulin
the suppressive effect of glucose on ␣-cells since, in its in suppressing glucagon secretion from the ␣-cells has been
absence, glucose is unable to suppress glucagon release in vivo illustrated further in studies using isolated rat and human islets
(58, 64). These studies provide an explanation underlying the and islet hormonal RIAs (157). In these studies, challenging
observation of unsuppressed glucagon secretion in diabetes islets with 20 mM glucose results in insulin secretion associ-
despite the presence of hyperglycemia, presumably due to ated with suppressed glucagon secretion; however, blockade of
relatively insufficient insulin levels in the internal environment insulin signaling by the PI 3-kinase inhibitor wortmannin pre-
of the islet or as a result of ␣-cell insulin resistance (68, vents glucose from suppressing glucagon secretion, whereas the
91, 141). glucose-stimulated insulin secretion is not affected, indicating that
the insulin-signaling pathway mediates the glucose-induced glu-
Intraislet Insulin Action on Glucagon Secretion cagon secretion in the islets (157). Studies in rat pancreata have
shown that glucose-induced suppression of glucagon secretion is
Actions of insulin are initiated by binding of the hormone to blocked in the presence of insulin-neutralizing antibodies (96),
its receptor at the cell surface in insulin-responsive tissues, further supporting the physiological role of insulin in mediating
which activates a signaling complex through recruitment of glucose effect on the ␣-cells. These findings may provide a
adaptor molecules, including the insulin receptor substrate mechanism underlying the well-characterized push-pull phenom-
family. Upon tyrosine phosphorylation, these proteins interact enon between the secretion of insulin and glucagon in response to
with signaling molecules through their SH2 domains, which glucose challenge (144) (Fig. 2). Thus, it is hypothesized that
results in the activation of diverse signaling pathways, includ- defects in the intraislet insulin-signaling pathway may contribute
ing phosphatidylinositol (PI) 3-kinase/Akt signaling and to the development of diabetic hyperglucagonemia and hypergly-
MAPK activation. These pathways act in a coordinated manner cemia, because ␣-cells with insulin resistance imposed by chronic
to regulate glucose transport, protein and lipid synthesis, and treatment with high glucose and insulin displayed reduced insulin
mitogenic responses (12). responsiveness, including blunted insulin-stimulated Akt phos-
Within the islets, the ␣-cells are major targets of insulin phorylation and insulin-suppressed glucagon secretion in the
action. Studies on islet microvasculature have suggested that ␣-cells (157). Therefore, it is conceivable that the postprandial
the ␣-cells lie downstream from the ␤-cells and are therefore hyperglucagonemia in type 2 diabetes may be due to the loss of
presumably bathed by high concentrations of insulin (18). It is intraislet postprandial suppression of glucagon secretion driven by
thought that the islet is arranged such that a core of ␤-cells is insulin (144). This hypothesis has recently been verified by
surrounded by a thin layer of ␣- and ␦-cells (7, 18, 25, 84, 105). in vivo studies using pigs treated with or without alloxan (79,
The directionality of microvascular perfusion within the islet 100), a chemical that selectively destroys ␤-cells and causes
has been investigated in perfused rat (125), dog (134), rhesus diabetic hyperglycemia. Postprandial insulin secretion induced a
monkey (135), and human (133) pancreas by infusing insulin- suppression of glucagon secretion via inhibition of glucagon pulse
neutralizing antibodies through the arterial and venous systems mass (100); on the contrary, the postprandial insulin-driven sup-
of the organ, representing anterograde and retrograde perfu- pression of glucagon secretion was lost in the pigs treated with
sion, respectively. The perfusate was then analyzed for pan- alloxan, which caused a reduction of ␤-cell mass and a deficit in
creatic hormone secretions, using radioimmunoassays to deter- postprandial insulin secretion (100). Despite this, it has been
mine the directional effects of abolishing insulin signaling on shown that glucose can inhibit glucagon secretion at concentra-
␣- and ␦-cell secretion. These “passive immunization” exper- tions below that required to elicit insulin secretion (95, 104, 123),
iments demonstrated a ␤ 3 ␣ 3 ␦ directionality of islet blood indicating that glucose may directly inhibit glucagon secretion in
flow and strongly suggested that intraislet insulin regulates the ␣-cells.
hormone secretion from ␣-cells. Hypoglycemia is a serious acute complication in type 1
Consistent with an important role for insulin in the ␣-cell, diabetic patients undergoing intensive insulin therapy (33).
insulin receptor and the insulin-signaling molecules are ex- Defects in glucose counterregulation have been attributed to
pressed highly in pancreatic ␣-cells (13, 71, 77, 107, 147) at tonic inhibition of ␣-cell activity due to intraislet hyperinsu-
levels that are similar to those in the hepatocytes (42). The linemia (9), which prevents a decrement in intraislet insulin
insulin receptor plays a role in modulating ␣-cell function, as during hypoglycemia that would normally act as the switch for
siRNA-mediated “knockdown” of the insulin receptor abol- ␣-cells to secrete glucagon (114). This “switch-off” hypothesis
ishes glucose-regulated glucagon secretion from the ␣-cells has been supported by studies in rats rendered diabetic via
(36). Action of insulin on suppression of glucagon secretion treatment with the ␤-cell-selective toxin streptozotocin (159)
has been demonstrated directly using cultured clonal glucagon- as well as by using isolated rat and human islets (65) and a rat
secreting ␣-cell lines, including ␣TC6 and In-R1-G9 cells (71, model of spontaneous type 1 diabetes (161). These studies
77, 157). These studies have demonstrated that insulin-induced demonstrated that cessation of exogenous insulin administra-
suppression of glucagon secretion could be blocked by the PI tion prior to induction of hypoglycemia produces a glucagon
3-kinase inhibitor wortmannin (71, 157) or ablation of Akt secretory response that would otherwise be absent during
kinase activity using dominant negative approaches (157), continuous insulin infusion (65, 159, 161). A clincal study in
suggesting that insulin-suppressed glucagon secretion is medi- nondiabetic humans attributed a 30% decrease in the counter-
AJP-Endocrinol Metab • VOL 295 • OCTOBER 2008 • www.ajpendo.org
Downloaded from journals.physiology.org/journal/ajpendo (177.016.070.107) on December 14, 2022.
Review
INSULIN ACTION ON ␣-CELL SECRETION E755

Fig. 2. The reciprocal response of insulin (䊐) and


glucagon (E) to changes in glucose concentration in
the perfused dog pancreas (144). Figure is reproduced
from the article by Dr. R. H. Unger (144) with
permission from the author.

regulatory glucagon response to hypoglycemia after somatosta- through phosphorylation of the Kir6.2 subunit, leading to a
tin infusion was administered to abolish any decrement in decrease in ATP sensitivity (92). It is interesting to note that
intraislet insulin during hypoglycemia (57), indicating that ERK2 is a component in the Ras-MAPK pathway that is
there may be other mechanisms involved in triggering gluca- activated by insulin (139). Since the Ras-MAPK pathway is PI
gon secretion in humans. However, the specificity of the 3-kinase independent, it is plausible that insulin modulates
switch-off hypothesis has become broad, since a recent study KATP channel function via PI 3-kinase-dependent and/or -in-
has suggested that a decrease in the intraislet concentration of dependent pathways.
Zn2⫹ and not insulin is the switch required to trigger glucagon Treatment of isolated rat ␣-cells with insulin can also tran-
secretion during hypoglycemia in rats (160). The effects of siently enhance KATP-mediated currents (42), suggesting that
Zn2⫹ on ␣-cell function are discussed later in this review, but insulin-mediated suppression of glucagon secretion from the
these results highlight the importance of ␤-cell secretory prod- ␣-cells could occur via modulation of KATP currents. Interest-
ucts on ␣-cell glucagon secretion. ingly, this effect is not blocked by wortmannin (42), which
suggests the potential involvement of nonclassical PI 3-kinases
Intraislet Insulin Modulates ␣-Cell KATP Channels
in glucagon secretory competence. Studies in transgenic mice
Insulin’s action as a negative regulator of glucagon secretion expressing green fluorescent protein under the control of the
may involve diverse machineries, including modulating the mouse insulin promoter, which facilitates differentiation be-
␣-cell KATP channel activity. As mentioned above, glucagon tween ␤-cells and non-␤-cells within the pancreatic islet (89),
secretion requires ␣-cell depolarization to trigger the release of have characterized the KATP channels in ␣-cells to be five
glucagon-containing granules. Conversely, hyperpolarization times more sensitive to intracellular ATP-mediated channel
of the ␣-cell as a consequence of KATP channel activation can inhibition than ␤-cell KATP channels, although the densities of
block glucagon secretion (61). Insulin has been shown to KATP channels found in both cell types are equal (89). In
activate KATP channels in hypothalamic neurons in rats (132) isolated ␣-cells from mice expressing insulin promoter-green
and isolated mouse ␤-cells (75), resulting in membrane hyper- fluorescent protein, insulin markedly reduces the sensitivity of
polarization and cessation of electrical activity (75, 132). KATP channels to ATP in a wortmannin-sensitive fashion (87).
Interestingly, a recent study showed that, in isolated mouse Interestingly, although glucose can dose-dependently modulate
␤-cells, glucose could directly enhance KATP channel conduc- the intracellular [ATP] in the ␤-cells, glucose has no effect on
tance by upregulating KATP channel plasma membrane local- (35) or increases the intracellular [ATP] less efficiently in
ization (158). Furthermore, it has also been demonstrated that ␣-cells compared with ␤-cells (69). Therefore, it is conceivable
activation of ERK2 enhanced the activity of KATP channels in that glucose-induced suppression of glucagon secretion could
HEK-293 cells transiently transfected with the KATP channels occur through insulin-mediated desensitization of ␣-cell KATP
AJP-Endocrinol Metab • VOL 295 • OCTOBER 2008 • www.ajpendo.org
Downloaded from journals.physiology.org/journal/ajpendo (177.016.070.107) on December 14, 2022.
Review
E756 INSULIN ACTION ON ␣-CELL SECRETION

channels to ATP inhibition, increasing channel activity and plasticity (30, 152). Increasing cell surface abundance of
causing the ␣-cell to become hyperpolarized. It is of great GABAARs in pancreatic ␣-cells, as a result of direct modula-
interest to further determine how insulin precisely reduces the tion by glucose or indirectly by insulin, in response to in-
␣-cell KATP channel sensitivity and its downstream cascades. creased glucose concentrations may be a major mechanism
underlying glucose-induced suppression of glucagon secretion.
Intraislet Insulin Promotes GABAergic Inhibition This conception is in accord with a recent study using a
of Glucagon Secretion well-designed in situ patch-clamp recording technique that
illustrates that GABA released from the ␤-cells could diffuse
␥-Aminobutyric acid (GABA) is a major inhibitory neu- across the islet interstitium to activate GABAARs in neighbor-
rotransmitter in the mammalian central nervous system and ing cells (153). It is interesting to note that, in vitro, high
is synthesized from glutamic acid by glutamic acid decar- glucose concentrations stimulate glucagon secretion from iso-
boxylase (82). Three types of the GABA receptor (GABAR) lated rat ␣-cells via the glucose-sensing pathway as used by the
have been identified: type A GABAR (GABAAR), a hetero- ␤-cells (104), further emphasizing the crucial importance of
pentameric ligand-gated Cl⫺ channel that is antagonized by islet paracrine signaling in the regulation of glucagon release in
the compound bicuculline (93); GABABR, a heterodimeric vivo. The intraislet insulin pathway and these modulators
G protein-coupled receptor (19, 28); and GABACR, which is appear to constitute precise regulatory networks that operate to
also a ligand-gated Cl⫺ channel; however, it is bicuculline
regulate glucagon release and, hence, to control glucose ho-
insensitive, homopentameric, and active at lower concentra-
meostasis. Given that in diabetic individuals, insulin deficiency
tions of GABA than GABAAR (28). Activation of GABAAR
is associated with elevated glucagon levels, which rapidly
results in membrane hyperpolarization as a consequence of
return to normal with insulin treatment, it is possible that
an inward Cl⫺ flux (78). Although GABAergic signaling is
defects in this pathway may contribute to diabetic hyperglu-
an essential regulatory component of neural excitability
accounting for 20 –30% of all synaptic signaling in the cagonemia and subsequent hyperglycemia. This pathway may
mammalian brain (154), GABA also modulates the endo- also partly explain that type 1 diabetic hypoglycemia that
crine function of the pancreas (43, 74). Pancreatic ␤-cells occurs in insulin-treated patients may be due to a lack of
contain high concentrations of GABA (21, 46, 140) and suppression by endogenous insulin that may potentially render
glutamic acid decarboxylase (45, 53). GABA is localized in the ␣-cells hypersensitive to the suppression induced by exog-
both synaptic-like microvesicles (21) and the insulin-con- enous insulin (9).
taining large dense core vesicles (22, 44) within the ␤-cells.
The functional GABAARs are expressed in both ␣-cells (63, Insulin Suppresses Glucagon Gene Expression
118) and ␤-cells (38).
Transcriptional regulation of the glucagon gene by insulin
The GABA-GABAAR system has been suggested to nega-
appears to be an important way to regulate glucagon pro-
tively regulate glucagon secretion (118, 153). GABA admin-
duction and secretion. The inhibitory effects of insulin on
istered to isolated mouse and rat islets, as well as to perfused
glucagon biosynthesis are mediated through a downregula-
rat pancreas, can inhibit the release of glucagon (52), whereas
inhibition of the GABAAR by bicuculline (47, 118) or ton of proglucagon gene expression (29, 109, 110) that
SR95531 (153) prevents GABA-mediated suppression of glu- requires activation of PI 3-kinase and Akt (126). The insulin
cagon secretion from the ␣-cells but does not affect the insulin response element that mediates insulin effects on glucagon
secretion (157). However, although controversial (47), the gene transcription is located in the proximal promoter re-
release of GABA from the ␤-cells does not appear to be gion of the proglucagon gene (111), and several proteins
stimulated by glucose (130, 151, 155), or the increase in have been identified to act in trans with this region to
GABA release by high glucose may be too small to detect facilitate insulin-induced transcriptional alterations (49,
(118). However, the failure to detect an increase in GABA 111, 126). Furthermore, it has recently been identified that
release from the ␤-cells in response to increased glucose FoxO1, a member of the forkhead transcription factor fam-
concentrations does not exclude the possibility that there is an ily, is required for the regulation of proglucagon gene
increase in the responsiveness of GABAARs on ␣-cells. Evi- expression by insulin by direct binding of FoxO1 to a
dence supporting this idea is the finding that GABAARs can be forkhead consensus binding site in the preproglucagon gene
phosphorylated directly by Akt, the important insulin-signaling promoter region (99). In vitro studies in clonal glucagon-
molecule, resulting in translocation of the receptors from secreting cells have demonstrated that insulin induced nu-
intracellular pools to the cell surface (152). Furthermore, a clear exclusion of FoxO1 and a subsequent reduction in
recent study also suggested that, at postreceptor levels, glucose proglucagon gene transcription. Experiments employing di-
can modulate GABAAR activity through increasing cell sur- rect-site mutagenesis and RNA interference have also dem-
face abundance of the GABAARs (8). Consistent with the role onstrated that diminishment of FoxO1 binding eliminated
of insulin in modulating GABAergic signaling in the central transcriptional regulation by glucose or insulin, whereas
nervous system (148, 150, 152), insulin enhances inhibitory FoxO1 silencing abolished the acute regulation by insulin,
GABA currents in clonal glucagon-secreting ␣-cells and iso- but not glucose, of glucagon secretion. (99). Therefore,
lated rat ␣-cells upon phosphorylation of the ␤-subunit of these studies further support that intraislet insulin is a key
GABAAR and plasma membrane recruitment of the GABAARs modulator of ␣-cell function. However, this transcriptional
by insulin (157). The rapid regulation of neurotransmitter regulation may be more critical for long-term maintenance
receptor numbers in the postsynaptic domain by direct receptor of ␣-cell glucagon content instead of for short-term re-
phosphorylation is an important means of modulating synaptic sponses to changes in blood glucose levels.
AJP-Endocrinol Metab • VOL 295 • OCTOBER 2008 • www.ajpendo.org
Downloaded from journals.physiology.org/journal/ajpendo (177.016.070.107) on December 14, 2022.
Review
INSULIN ACTION ON ␣-CELL SECRETION E757
Regulation of Glucagon Secretion is Multifactoral inhibit glucagon secretion by binding directly to two histidine
residues on the SUR1 subunit (10) of the KATP channel and
Although the intraislet insulin-mediated suppression of gluca- enhancing their activity (42, 112), as it has been reported to
gon secretion plays an important role in modulating glucagon
enhance KATP channel-mediated current in clonal rat ␤-cells (16);
secretion via mechanisms involving protein-protein interactions,
however, studies in mouse ␣-cells have shown no effect of Zn2⫹
signal transduction, receptor translocation, and transcriptional reg-
on KATP channels (62, 87). These findings potentially explain why
ulation (Fig. 3), it should be noted that factors other than intraislet
insulin, such as Zn2⫹, may also influence glucagon secretion from ZnCl2 has no effect on intracellular Ca2⫹ oscillations or glucagon
␣-cells. Insulin molecules are complexed with Zn2⫹ ions to secretion in isolated mouse ␣-cells in the absence of glucose
permit stable storage of insulin within large dense core vesicles (115), since mouse ␣-cells express fewer KATP channels than rat
(81). Zn2⫹ is coreleased with insulin from the ␤-cells (113) and ␣-cells (11, 115).
may be an important inhibitor of glucagon secretion (42, 69). Furthermore, there are a number of reported regulators/
Upon liberation, Zn2⫹ is transported into the ␣-cells via Ca2⫹ modulators that may participate in the process of glucagon
channels and Zn2⫹ transporters (62). Zn2⫹ has been shown to secretion. Among them are sympathetic and parasympathetic
inhibit pyruvate-stimulated glucagon secretion in perfused rat nerves (4), somatostatin (27), ghrelin (122), L-glutamate (142),
pancreas (69), reduce the release of glucagon in isolated ␣-cells and GLP-1 (97). However, L-glutamate (40, 86) and GLP-1
when incubated in the absence of glucose (42), and inhibit (103) are also known to stimulate insulin secretion, which
glucagon release from ␣TC6 cells as well as mouse islets at low suggests that, in vivo, the effects of these molecules on the
glucose concentrations (62). Also, a recent study in rats has found ␣-cells may be indirectly caused by raising intraislet insulin
that a decrement in intraislet Zn2⫹ is required for a normal concentrations. For a thorough and up-to-date review of the
glucagon secretory response to hypoglycemia (160). Zn2⫹ may effects these substances have on ␣-cells, refer to Ref. 60.

Fig. 3. Proposed mechanisms of intraislet insulin suppression of glucagon secretion. Insulin secreted from ␤-cells acts in a paracrine manner to activate the
insulin-signaling cascade in ␣-cells. This has 3 main consequences: 1) activation of KATP channels, 2) enhancement of type A GABA receptor (GABAAR)-
mediated GABA current, and 3) inhibition of proglucagon gene transcription. First, insulin decreases the ␣-cell KATP channel sensitivity to ATP inhibition in
a phosphatidylinositol 3-kinase-dependent manner, causing an increase in the efflux of K⫹ ions from the ␣-cell and hyperpolarizing the ␣-cell membrane
potential. The resultant cessation of action potentials prevents an increase in cytosolic [Ca2⫹] and inhibits glucagon secretion. Second, activation of the insulin
receptor (IR) promotes membrane translocation of GABAAR and enhances GABA-mediated Cl⫺ influx. The ␣-cell membrane potential becomes hyperpolarized,
and glucagon secretion is suppressed. Finally, insulin inhibits proglucagon gene transcription, possibly representing a long-term mechanism for regulating ␣-cell
function. Other modulators of ␣-cell glucagon secretion, including Zn2⫹ and somatostatin, are not shown.

AJP-Endocrinol Metab • VOL 295 • OCTOBER 2008 • www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo (177.016.070.107) on December 14, 2022.
Review
E758 INSULIN ACTION ON ␣-CELL SECRETION

Conclusion 17. Bokvist K, Olsen HL, Høy M, Gotfredsen CF, Holmes WF, Buschard
K, Rorsman P, Gromada J. Characterisation of sulphonylurea and
As a counterregulatory hormone for insulin, glucagon plays ATP-regulated K⫹ channels in rat pancreatic A-cells. Pflugers Arch 438:
a critical role in maintaining glucose homeostasis. Glucagon 428 – 436, 1999.
18. Bonner-Weir S, Orci L. New perspectives on the microvasculature of
increases hepatic glucose output by enhancing glycogenolysis the islets of Langerhans in the rat. Diabetes 31: 883– 889, 1982.
and gluconeogenesis in the liver. In diabetic subjects, abnormal 19. Bowery NG. GABAB receptor: a site of therapeutic benefit. Curr Opin
secretion of not only insulin but also glucagon leads to hyper- Pharmacol 6: 37– 43, 2006.
glucagonemia and altered insulin-to-glucagon ratios, which 20. Braun M, Ramracheya R, Bengtsson M, Zhang Q, Karanauskaite J,
predominantly contribute to diabetic hyperglycemia. Given Partridge C, Johnson PR, Rorsman P. Voltage-gated ion channels in
human pancreatic beta-cells: electrophysiological characterization and
that insulin is a physiological suppressor of glucagon secretion, role in insulin secretion. Diabetes 57: 1618 –1628, 2008.
understanding the precise molecular mechanism of glucagon 21. Braun M, Wendt A, Birnir B, Broman J, Eliasson L, Galvanovskis J,
secretion by intraislet insulin may facilitate our effort to de- Gromada J, Mulder H, Rorsman P. Regulated exocytosis of GABA-
velop strategies to combat diabetic hyperglycemia and to containing synaptic-like microvesicles in pancreatic beta-cells. J Gen
Physiol 123: 191–204, 2004.
prevent hypoglycemia in insulin-treated patients. 22. Braun M, Wendt A, Karanauskaite J, Galvanovskis J, Clark A,
ACKNOWLEDGMENTS MacDonald PE, Rorsman P. Corelease and differential exit via the
fusion pore of GABA, serotonin, and ATP from LDCV in rat pancreatic
We thank L. Ding, J. Zhang, and Dr. K. Chen for the credit in artwork. beta cells. J Gen Physiol 129: 221–231, 2007.
23. Brissova M, Fowler MJ, Nicholson WE, Chu A, Hirshberg B, Harlan
GRANTS
DM, Powers AC. Assessment of human pancreatic islet architecture and
Researchers in Dr. Wang’s laboratory are supported by grants from the composition by laser scanning confocal microscopy. J Histochem Cyto-
Canadian Institute for Health Research (CIHR), Canadian Diabetes Associa- chem 53: 1087–1097, 2005.
tion (CDA), and Juvenile Diabetes Research Foundation. Q. Wang was a CDA 24. Burcelin R, Li J, Charron MJ. Cloning and sequence analysis of the
Scholar and is presently supported by the New Investigator Program from murine glucagon receptor-encoding gene. Gene 164: 305–310, 1995.
CIHR. 25. Cabrera O, Berman DM, Kenyon NS, Ricordi C, Berggren PO,
Caicedo A. The unique cytoarchitecture of human pancreatic islets has
REFERENCES implications for islet cell function. Proc Natl Acad Sci USA 103:
1. Aguilar-Parada E, Eisentraut AM, Unger RH. Pancreatic glucagon 2334 –2339, 2006.
secretion in normal and diabetic subjects. Am J Med Sci 257: 415– 419, 26. Carr-Locke DL, Gregg JA, Aoki TT. Effects of exogenous glucagon
1969. on pancreatic and biliary ductal and sphincteric pressures in man dem-
2. Ahloulay M, Dechaux M, Hassler C, Bouby N, Bankir L. Cyclic AMP onstrated by endoscopic manometry and correlation with plasma gluca-
is a hepatorenal link influencing natriuresis and contributing to glucagon- gon. Dig Dis Sci 28: 312–320, 1983.
induced hyperfiltration in rats. J Clin Invest 98: 2251–2258, 1996. 27. Cejvan K, Coy DH, Efendic S. Intra-islet somatostatin regulates gluca-
3. Ahloulay M, Dechaux M, Laborde K, Bankir L. Influence of glucagon gon release via type 2 somatostatin receptors in rats. Diabetes 52:
on GFR and on urea and electrolyte excretion: direct and indirect effects. 1176 –1181, 2003.
Am J Physiol Renal Fluid Electrolyte Physiol 269: F225–F235, 1995. 28. Chebib M, Johnston GA. The ‘ABC’ of GABA receptors: a brief
4. Ahren B. Autonomic regulation of islet hormone secretion—implica- review. Clin Exp Pharmacol Physiol 26: 937–940, 1999.
tions for health and disease. Diabetologia 43: 393– 410, 2000. 29. Chen L, Komiya I, Inman L, McCorkle K, Alam T, Unger RH.
5. Arafat AM, Perschel FH, Otto B, Weickert MO, Rochlitz H, Schofl Molecular and cellular responses of islets during perturbations of glucose
C, Spranger J, Mohlig M, Pfeiffer AF. Glucagon suppression of ghrelin homeostasis determined by in situ hybridization histochemistry. Proc
secretion is exerted at hypothalamus-pituitary level. J Clin Endocrinol Natl Acad Sci USA 86: 1367–1371, 1989.
Metab 91: 3528 –3533, 2006. 30. Collingridge GL, Isaac JT, Wang YT. Receptor trafficking and syn-
6. Ashcroft FM, Rorsman P. Electrophysiology of the pancreatic beta- aptic plasticity. Nat Rev Neurosci 5: 952–962, 2004.
cell. Prog Biophys Mol Biol 54: 87–143, 1989. 31. Conarello SL, Jiang G, Mu J, Li Z, Woods J, Zycband E, Ronan J,
7. Baetens D, Malaisse-Lagae F, Perrelet A, Orci L. Endocrine pancreas: Liu F, Roy RS, Zhu L, Charron MJ, Zhang BB. Glucagon receptor
three-dimensional reconstruction shows two types of islets of langerhans. knockout mice are resistant to diet-induced obesity and streptozotocin-
Science 206: 1323–1325, 1979. mediated beta cell loss and hyperglycaemia. Diabetologia 50: 142–150,
8. Bailey SJ, Ravier MA, Rutter GA. Glucose-dependent regulation of 2007.
gamma-aminobutyric acid (GABA A) receptor expression in mouse 32. Creutzfeldt W, Ebert R. The Enteroinsular Axis. In: The Pancreas:
pancreatic islet alpha-cells. Diabetes 56: 320 –327, 2007. Biology, Pathobiology, and Disease, edited by Go VLW, DiMagno EP,
9. Banarer S, McGregor VP, Cryer PE. Intraislet hyperinsulinemia pre- Gardner JD, Lebenthal E, Reber HA, and Scheele GA. New York:
vents the glucagon response to hypoglycemia despite an intact autonomic Raven, 1993, chapt. 40, p. 769 –788.
response. Diabetes 51: 958 –965, 2002. 33. Cryer PE, Fisher JN, Shamoon H. Hypoglycemia. Diabetes Care 17:
10. Bancila V, Cens T, Monnier D, Chanson F, Faure C, Dunant Y, Bloc 734 –755, 1994.
A. Two SUR1-specific histidine residues mandatory for zinc-induced 34. Delahousse M, Mercier O, Bichara M, Paillard M, Leviel F, Assan R.
activation of the rat KATP channel. J Biol Chem 280: 8793– 8799, 2005. Glucagon inhibits urinary acidification in the rat. Am J Physiol Renal
11. Barg S, Galvanovskis J, Gopel SO, Rorsman P, Eliasson L. Tight Fluid Electrolyte Physiol 254: F762–F769, 1988.
coupling between electrical activity and exocytosis in mouse glucagon- 35. Detimary P, Dejonghe S, Ling Z, Pipeleers D, Schuit F, Henquin JC.
secreting alpha-cells. Diabetes 49: 1500 –1510, 2000. The changes in adenine nucleotides measured in glucose-stimulated
12. Bevan P. Insulin signalling. J Cell Sci 114: 1429 –1430, 2001. rodent islets occur in beta cells but not in alpha cells and are also
13. Bhathena SJ, Oie HK, Gazdar AF, Voyles NR, Wilkins SD, Recant L. observed in human islets. J Biol Chem 273: 33905–33908, 1998.
Insulin, glucagon, and somatostatin receptors on cultured cells and clones 36. Diao J, Asghar Z, Chan CB, Wheeler MB. Glucose-regulated glucagon
from rat islet cell tumor. Diabetes 31: 521–531, 1982. secretion requires insulin receptor expression in pancreatic alpha-cells.
14. Billington CJ, Bartness TJ, Briggs J, Levine AS, Morley JE. Gluca- J Biol Chem 280: 33487–33496, 2005.
gon stimulation of brown adipose tissue growth and thermogenesis. Am J 37. Donatsch P, Lowe DA, Richardson BP, Taylor P. The functional
Physiol Regul Integr Comp Physiol 252: R160 –R165, 1987. significance of sodium channels in pancreatic beta-cell membranes.
15. Billington CJ, Briggs JE, Link JG, Levine AS. Glucagon in physio- J Physiol 267: 357–376, 1977.
logical concentrations stimulates brown fat thermogenesis in vivo. Am J 38. Dong H, Kumar M, Zhang Y, Gyulkhandanyan A, Xiang YY, Ye B,
Physiol Regul Integr Comp Physiol 261: R501–R507, 1991. Perrella J, Hyder A, Zhang N, Wheeler M, Lu WY, Wang Q.
16. Bloc A, Cens T, Cruz H, Dunant Y. Zinc-induced changes in ionic Gamma-aminobutyric acid up- and downregulates insulin secretion from
currents of clonal rat pancreatic ␤-cells: activation of ATP-sensitive K⫹ beta cells in concert with changes in glucose concentration. Diabetologia
channels. J Physiol 529: 723–734, 2000. 49: 697–705, 2006.

AJP-Endocrinol Metab • VOL 295 • OCTOBER 2008 • www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo (177.016.070.107) on December 14, 2022.
Review
INSULIN ACTION ON ␣-CELL SECRETION E759
39. Drucker DJ. The biology of incretin hormones. Cell Metab 3: 153–165, 59. Gromada J, Bokvist K, Ding WG, Barg S, Buschard K, Renstrom E,
2006. Rorsman P. Adrenaline stimulates glucagon secretion in pancreatic
40. Eto K, Yamashita T, Hirose K, Tsubamoto Y, Ainscow EK, Rutter A-cells by increasing the Ca2⫹ current and the number of granules close
GA, Kimura S, Noda M, Iino M, Kadowaki T. Glucose metabolism to the L-type Ca2⫹ channels. J Gen Physiol 110: 217–228, 1997.
and glutamate analog acutely alkalinize pH of insulin secretory vesicles 60. Gromada J, Franklin I, Wollheim CB. Alpha-cells of the endocrine
of pancreatic ␤-cells. Am J Physiol Endocrinol Metab 285: E262–E271, pancreas: 35 years of research but the enigma remains. Endocr Rev 28:
2003. 84 –116, 2007.
41. Ferrannini E, Galvan AQ, Gastaldelli A, Camastra S, Sironi AM, 61. Gromada J, Ma X, Hoy M, Bokvist K, Salehi A, Berggren PO,
Toschi E, Baldi S, Frascerra S, Monzani F, Antonelli A, Nannipieri Rorsman P. ATP-sensitive K⫹ channel-dependent regulation of gluca-
M, Mari A, Seghieri G, Natali A. Insulin: new roles for an ancient gon release and electrical activity by glucose in wild-type and SUR1⫺/⫺
hormone. Eur J Clin Invest 29: 842– 852, 1999. mouse alpha-cells. Diabetes 53, Suppl 3: S181–S189, 2004.
42. Franklin I, Gromada J, Gjinovci A, Theander S, Wollheim CB. 62. Gyulkhandanyan AV, Lu H, Lee SC, Bhattacharjee A, Wijesekara
Beta-cell secretory products activate alpha-cell ATP-dependent potas- N, Fox JE, MacDonald PE, Chimienti F, Dai FF, Wheeler MB.
sium channels to inhibit glucagon release. Diabetes 54: 1808 –1815, Investigation of transport mechanisms and regulation of intracellular
2005. Zn2⫹ in pancreatic alpha-cells. J Biol Chem 283: 10184 –10197, 2008.
43. Franklin IK, Wollheim CB. GABA in the endocrine pancreas: its 63. Hales TG, Tyndale RF. Few cell lines with GABAA mRNAs have
putative role as an islet cell paracrine-signalling molecule. J Gen Physiol
functional receptors. J Neurosci 14: 5429 –5436, 1994.
123: 185–190, 2004.
64. Hamaguchi T, Fukushima H, Uehara M, Wada S, Shirotani T,
44. Gammelsaeter R, Froyland M, Aragon C, Danbolt NC, Fortin D,
Kishikawa H, Ichinose K, Yamaguchi K, Shichiri M. Abnormal
Storm-Mathisen J, Davanger S, Gundersen V. Glycine, GABA and
glucagon response to arginine and its normalization in obese hyperinsu-
their transporters in pancreatic islets of Langerhans: evidence for a
linaemic patients with glucose intolerance: importance of insulin action
paracrine transmitter interplay. J Cell Sci 117: 3749 –3758, 2004.
45. Garry DJ, Appel NM, Garry MG, Sorenson RL. Cellular and subcel- on pancreatic alpha cells. Diabetologia 34: 801– 806, 1991.
lular immunolocalization of L-glutamate decarboxylase in rat pancreatic 65. Hope KM, Tran PO, Zhou H, Oseid E, LeRoy E, Robertson RP.
islets. J Histochem Cytochem 36: 573–580, 1988. Regulation of alpha-cell function by the beta-cell in isolated human and
46. Garry DJ, Sorenson RL, Elde RP, Maley BE, Madsen A. Immuno- rat islets deprived of glucose: the “switch-off” hypothesis. Diabetes 53:
histochemical colocalization of GABA and insulin in beta-cells of rat 1488 –1495, 2004.
islet. Diabetes 35: 1090 –1095, 1986. 66. Huypens P, Ling Z, Pipeleers D, Schuit F. Glucagon receptors on
47. Gaskins HR, Baldeon ME, Selassie L, Beverly JL. Glucose modulates human islet cells contribute to glucose competence of insulin release.
gamma-aminobutyric acid release from the pancreatic beta TC6 cell line. Diabetologia 43: 1012–1019, 2000.
J Biol Chem 270: 30286 –30289, 1995. 67. Inokuchi A, Oomura Y, Nishimura H. Effect of intracerebroventricu-
48. Gastaldelli A, Baldi S, Pettiti M, Toschi E, Camastra S, Natali A, larly infused glucagon on feeding behavior. Physiol Behav 33: 397– 400,
Landau BR, Ferrannini E. Influence of obesity and type 2 diabetes on 1984.
gluconeogenesis and glucose output in humans: a quantitative study. 68. Ipp E. Impaired glucose tolerance: the irrepressible alpha-cell? Diabetes
Diabetes 49: 1367–1373, 2000. Care 23: 569 –570, 2000.
49. Gauthier BR, Schwitzgebel VM, Zaiko M, Mamin A, Ritz-Laser B, 69. Ishihara H, Maechler P, Gjinovci A, Herrera PL, Wollheim CB. Islet
Philippe J. Hepatic nuclear factor-3 (HNF-3 or Foxa2) regulates gluca- beta-cell secretion determines glucagon release from neighbouring alpha-
gon gene transcription by binding to the G1 and G2 promoter elements. cells. Nat Cell Biol 5: 330 –335, 2003.
Mol Endocrinol 16: 170 –183, 2002. 70. Jiang G, Zhang BB. Glucagon and regulation of glucose metabolism.
50. Gelling RW, Du XQ, Dichmann DS, Romer J, Huang H, Cui L, Obici Am J Physiol Endocrinol Metab 284: E671–E678, 2003.
S, Tang B, Holst JJ, Fledelius C, Johansen PB, Rossetti L, Jelicks 71. Kaneko K, Shirotani T, Araki E, Matsumoto K, Taguchi T, Moto-
LA, Serup P, Nishimura E, Charron MJ. Lower blood glucose, shima H, Yoshizato K, Kishikawa H, Shichiri M. Insulin inhibits
hyperglucagonemia, and pancreatic alpha cell hyperplasia in glucagon glucagon secretion by the activation of PI3-kinase in In-R1-G9 cells.
receptor knockout mice. Proc Natl Acad Sci USA 100: 1438 –1443, 2003. Diabetes Res Clin Pract 44: 83–92, 1999.
51. Gerich JE, Tsalikian E, Lorenzi M, Schneider V, Bohannon NV, 72. Kanno T, Gopel SO, Rorsman P, Wakui M. Cellular function in
Gustafson G, Karam JH. Normalization of fasting hyperglucagonemia multicellular system for hormone-secretion: electrophysiological aspect
and excessive glucagon responses to intravenous arginine in human of studies on alpha-, beta- and delta-cells of the pancreatic islet. Neurosci
diabetes mellitus by prolonged infusion of insulin. J Clin Endocrinol Res 42: 79 –90, 2002.
Metab 41: 1178 –1180, 1975. 73. Katayama T, Shimamoto S, Oda H, Nakahara K, Kangawa K,
52. Gilon P, Bertrand G, Loubatieres-Mariani MM, Remacle C, Hen- Murakami N. Glucagon receptor expression and glucagon stimulation of
quin JC. The influence of gamma-aminobutyric acid on hormone release ghrelin secretion in rat stomach. Biochem Biophys Res Commun 357:
by the mouse and rat endocrine pancreas. Endocrinology 129: 2521– 865– 870, 2007.
2529, 1991. 74. Kawai, Unger RH. Effects of gamma-aminobutyric acid on insulin,
53. Gilon P, Tappaz M, Remacle C. Localization of GAD-like immunore-
glucagon, and somatostatin release from isolated perfused dog pancreas.
activity in the pancreas and stomach of the rat and mouse. Histochemistry
Endocrinology 113: 111–113, 1983.
96: 355–365, 1991.
75. Khan FA, Goforth PB, Zhang M, Satin LS. Insulin activates ATP-
54. Gopel S, Kanno T, Barg S, Galvanovskis J, Rorsman P. Voltage-gated
sensitive K(⫹) channels in pancreatic beta-cells through a phosphatidyl-
and resting membrane currents recorded from B-cells in intact mouse
pancreatic islets. J Physiol 521: 717–728, 1999. inositol 3-kinase-dependent pathway. Diabetes 50: 2192–2198, 2001.
55. Gopel S, Zhang Q, Eliasson L, Ma XS, Galvanovskis J, Kanno T, 76. Kirsch JR, D’Alecy LG. Glucagon stimulates ketone utilization by rat
Salehi A, Rorsman P. Capacitance measurements of exocytosis in brain slices. Stroke 15: 324 –328, 1984.
mouse pancreatic alpha-, beta- and delta-cells within intact islets of 77. Kisanuki K, Kishikawa H, Araki E, Shirotani T, Uehara M, Isami S,
Langerhans. J Physiol 556: 711–726, 2004. Ura S, Jinnouchi H, Miyamura N, Shichiri M. Expression of insulin
56. Gopel SO, Kanno T, Barg S, Weng XG, Gromada J, Rorsman P. receptor on clonal pancreatic alpha cells and its possible role for insulin-
Regulation of glucagon release in mouse alpha-cells by KATP channels stimulated negative regulation of glucagon secretion. Diabetologia 38:
and inactivation of TTX-sensitive Na⫹ channels. J Physiol 528: 509 – 422– 429, 1995.
520, 2000. 78. Kittler JT, Moss SJ. Modulation of GABAA receptor activity by
57. Gosmanov NR, Szoke E, Israelian Z, Smith T, Cryer PE, Gerich JE, phosphorylation and receptor trafficking: implications for the efficacy of
Meyer C. Role of the decrement in intraislet insulin for the glucagon synaptic inhibition. Curr Opin Neurobiol 13: 341–347, 2003.
response to hypoglycemia in humans. Diabetes Care 28: 1124 –1131, 79. Kjems LL, Kirby BM, Welsh EM, Veldhuis JD, Straume M, McIn-
2005. tyre SS, Yang D, Lefebvre P, Butler PC. Decrease in beta-cell mass
58. Greenbaum CJ, Havel PJ, Taborsky GJ Jr, Klaff LJ. Intra-islet leads to impaired pulsatile insulin secretion, reduced postprandial hepatic
insulin permits glucose to directly suppress pancreatic A cell function. insulin clearance, and relative hyperglucagonemia in the minipig. Dia-
J Clin Invest 88: 767–773, 1991. betes 50: 2001–2012, 2001.

AJP-Endocrinol Metab • VOL 295 • OCTOBER 2008 • www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo (177.016.070.107) on December 14, 2022.
Review
E760 INSULIN ACTION ON ␣-CELL SECRETION

80. Kolanowski J, Salvador G, Desmecht P, Henquin JC, Crabbe J. tubule, and collecting ducts in the rat. Am J Physiol Renal Fluid
Influence of glucagon on natriuresis and glucose-induced sodium reten- Electrolyte Physiol 257: F1003–F1014, 1989.
tion in the fasting obese subject. Eur J Clin Invest 7: 167–175, 1977. 102. Mochiki E, Suzuki H, Takenoshita S, Nagamachi Y, Kuwano H,
81. Kristiansen LH, Rungby J, Sondergaard LG, Stoltenberg M, Dan- Mizumoto A, Itoh Z. Mechanism of inhibitory effect of glucagon on
scher G. Autometallography allows ultrastructural monitoring of zinc in gastrointestinal motility and cause of side effects of glucagon. J Gastro-
the endocrine pancreas. Histochem Cell Biol 115: 125–129, 2001. enterol 33: 835– 841, 1998.
82. Krnjevic K. How does a little acronym become a big transmitter? 103. Mojsov S, Weir GC, Habener JF. Insulinotropin: glucagon-like peptide
Biochem Pharmacol 68: 1549 –1555, 2004. I (7–37) co-encoded in the glucagon gene is a potent stimulator of insulin
83. Krzeski R, Czyzyk-Krzeska MF, Trzebski A, Millhorn DE. Centrally release in the perfused rat pancreas. J Clin Invest 79: 616 – 619, 1987.
administered glucagon stimulates sympathetic nerve activity in rat. Brain 104. Olsen HL, Theander S, Bokvist K, Buschard K, Wollheim CB,
Res 504: 297–300, 1989. Gromada J. Glucose stimulates glucagon release in single rat alpha-cells
84. Ku SK, Lee HS, Lee JH. An immunohistochemical study on the by mechanisms that mirror the stimulus-secretion coupling in beta-cells.
pancreatic endocrine cells of the C57BL/6 mouse. J Vet Sci 3: 327–333, Endocrinology 146: 4861– 4870, 2005.
2002. 105. Orci L, Unger RH. Functional subdivision of islets of Langerhans and
85. Lefebvre PJ. Glucagon and its family revisited. Diabetes Care 18: possible role of D cells. Lancet 2: 1243–1244, 1975.
715–730, 1995. 106. Parker JC, Andrews KM, Allen MR, Stock JL, McNeish JD. Glyce-
86. Lehtihet M, Honkanen RE, Sjoholm A. Glutamate inhibits protein mic control in mice with targeted disruption of the glucagon receptor
phosphatases and promotes insulin exocytosis in pancreatic beta-cells. gene. Biochem Biophys Res Commun 290: 839 – 843, 2002.
Biochem Biophys Res Commun 328: 601– 607, 2005. 107. Patel YC, Amherdt M, Orci L. Quantitative electron microscopic
87. Leung YM, Ahmed I, Sheu L, Gao X, Hara M, Tsushima RG, autoradiography of insulin, glucagon, and somatostatin binding sites on
Diamant NE, Gaisano HY. Insulin regulates islet alpha-cell function by islets. Science 217: 1155–1156, 1982.
reducing KATP channel sensitivity to adenosine 5⬘-triphosphate inhibi- 108. Peterson CD, Leeder JS, Sterner S. Glucagon therapy for beta-blocker
tion. Endocrinology 147: 2155–2162, 2006. overdose. Drug Intell Clin Pharm 18: 394 –398, 1984.
88. Leung YM, Ahmed I, Sheu L, Tsushima RG, Diamant NE, Gaisano 109. Philippe J. Insulin regulation of the glucagon gene is mediated by an
HY. Two populations of pancreatic islet alpha-cells displaying distinct insulin-responsive DNA element. Proc Natl Acad Sci USA 88: 7224 –
Ca2⫹ channel properties. Biochem Biophys Res Commun 345: 340 –344, 7227, 1991.
2006. 110. Philippe J. Glucagon gene transcription is negatively regulated by
89. Leung YM, Ahmed I, Sheu L, Tsushima RG, Diamant NE, Hara M, insulin in a hamster islet cell line. J Clin Invest 84: 672– 677, 1989.
Gaisano HY. Electrophysiological characterization of pancreatic islet 111. Philippe J, Morel C, Cordier-Bussat M. Islet-specific proteins interact
cells in the mouse insulin promoter-green fluorescent protein mouse. with the insulin-response element of the glucagon gene. J Biol Chem 270:
Endocrinology 146: 4766 – 4775, 2005. 3039 –3045, 1995.
90. Li XC, Carretero OA, Shao Y, Zhuo JL. Glucagon receptor-mediated 112. Prost AL, Bloc A, Hussy N, Derand R, Vivaudou M. Zinc is both an
extracellular signal-regulated kinase 1/2 phosphorylation in rat mesangial intracellular and extracellular regulator of KATP channel function.
cells: role of protein kinase A and phospholipase C. Hypertension 47: J Physiol 559: 157–167, 2004.
580 –585, 2006. 113. Qian WJ, Aspinwall CA, Battiste MA, Kennedy RT. Detection of
91. Li XC, Liao TD, Zhuo JL. Long-term hyperglucagonaemia induces secretion from single pancreatic beta-cells using extracellular fluorogenic
early metabolic and renal phenotypes of Type 2 diabetes in mice. Clin Sci
reactions and confocal fluorescence microscopy. Anal Chem 72: 711–
(Lond) 114: 591– 601, 2007.
717, 2000.
92. Lin YF, Chai Y. Functional modulation of the ATP-sensitive potassium
114. Raju B, Cryer PE. Loss of the decrement in intraislet insulin plausibly
channel by extracellular signal-regulated kinase-mediated phosphoryla-
explains loss of the glucagon response to hypoglycemia in insulin-
tion. Neuroscience 152: 371–380, 2008.
deficient diabetes: documentation of the intraislet insulin hypothesis in
93. Luscher B, Keller CA. Regulation of GABAA receptor trafficking,
humans. Diabetes 54: 757–764, 2005.
channel activity, and functional plasticity of inhibitory synapses. Phar-
115. Ravier MA, Rutter GA. Glucose or insulin, but not zinc ions, inhibit
macol Ther 102: 195–221, 2004.
glucagon secretion from mouse pancreatic alpha-cells. Diabetes 54:
94. Ma X, Zhang Y, Gromada J, Sewing S, Berggren PO, Buschard K,
1789 –1797, 2005.
Salehi A, Vikman J, Rorsman P, Eliasson L. Glucagon stimulates
116. Reaven GM, Chen YD, Golay A, Swislocki AL, Jaspan JB. Docu-
exocytosis in mouse and rat pancreatic alpha-cells by binding to glucagon
receptors. Mol Endocrinol 19: 198 –212, 2005. mentation of hyperglucagonemia throughout the day in nonobese and
95. MacDonald PE, De Marinis YZ, Ramracheya R, Salehi A, Ma X, obese patients with noninsulin-dependent diabetes mellitus. J Clin En-
Johnson PR, Cox R, Eliasson L, Rorsman P. A K ATP channel- docrinol Metab 64: 106 –110, 1987.
dependent pathway within alpha cells regulates glucagon release from 117. Rizza R, Verdonk C, Miles J, Service FJ, Gerich J. Effect of inter-
both rodent and human islets of Langerhans. PLoS Biol 5: e143, 2007. mittent endogenous hyperglucagonemia on glucose homeostasis in nor-
96. Maruyama H, Hisatomi A, Orci L, Grodsky GM, Unger RH. Insulin mal and diabetic man. J Clin Invest 63: 1119 –1123, 1979.
within islets is a physiologic glucagon release inhibitor. J Clin Invest 74: 118. Rorsman P, Berggren PO, Bokvist K, Ericson H, Mohler H, Osten-
2296 –2299, 1984. son CG, Smith PA. Glucose-inhibition of glucagon secretion involves
97. Matsumura T, Itoh H, Watanabe N, Oda Y, Tanaka M, Namba M, activation of GABAA-receptor chloride channels. Nature 341: 233–236,
Kono N, Matsuyama T, Komatsu R, Matsuzawa Y. Glucagonlike 1989.
peptide-1(7–36)amide suppresses glucagon secretion and decreases cy- 119. Rorsman P, Hellman B. Voltage-activated currents in guinea pig pan-
clic AMP concentration in cultured In-R1-G9 cells. Biochem Biophys Res creatic alpha 2 cells. Evidence for Ca2⫹-dependent action potentials.
Commun 186: 503–508, 1992. J Gen Physiol 91: 223–242, 1988.
98. Mazzocchi G, Gottardo L, Aragona F, Albertin G, Nussdorfer GG. 120. Rouille Y, Kantengwa S, Irminger JC, Halban PA. Role of the
Glucagon inhibits ACTH-stimulated cortisol secretion from dispersed prohormone convertase PC3 in the processing of proglucagon to gluca-
human adrenocortical cells by activating unidentified receptors nega- gon-like peptide 1. J Biol Chem 272: 32810 –32816, 1997.
tively coupled with the adenylate cyclase cascade. Horm Metab Res 32: 121. Sakura H, Ammala C, Smith PA, Gribble FM, Ashcroft FM. Cloning
265–268, 2000. and functional expression of the cDNA encoding a novel ATP-sensitive
99. McKinnon CM, Ravier MA, Rutter GA. FoxO1 is required for the potassium channel subunit expressed in pancreatic beta-cells, brain, heart
regulation of preproglucagon gene expression by insulin in pancreatic and skeletal muscle. FEBS Lett 377: 338 –344, 1995.
alphaTC1–9 cells. J Biol Chem 281: 39358 –39369, 2006. 122. Salehi A, Dornonville de la Cour C, Håkanson R, Lundquist I. Effects
100. Meier JJ, Kjems LL, Veldhuis JD, Lefebvre P, Butler PC. Postpran- of ghrelin on insulin and glucagon secretion: a study of isolated pancre-
dial suppression of glucagon secretion depends on intact pulsatile insulin atic islets and intact mice. Regul Pept 118: 143–150, 2004.
secretion: further evidence for the intraislet insulin hypothesis. Diabetes 123. Salehi A, Vieira E, Gylfe E. Paradoxical stimulation of glucagon
55: 1051–1056, 2006. secretion by high glucose concentrations. Diabetes 55: 2318 –2323, 2006.
101. Mercier O, Bichara M, Delahousse M, Prigent A, Leviel F, Paillard 124. Saltiel AR, Kahn CR. Insulin signalling and the regulation of glucose
M. Effects of glucagon on H⫹-HCO⫺ 3 transport in Henle’s loop, distal and lipid metabolism. Nature 414: 799 – 806, 2001.

AJP-Endocrinol Metab • VOL 295 • OCTOBER 2008 • www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo (177.016.070.107) on December 14, 2022.
Review
INSULIN ACTION ON ␣-CELL SECRETION E761
125. Samols E, Stagner JI, Ewart RB, Marks V. The order of islet 146. Unger RH, Orci L. Physiology and pathophysiology of glucagon.
microvascular cellular perfusion is B—A—D in the perfused rat pan- Physiol Rev 56: 778 – 826, 1976.
creas. J Clin Invest 82: 350 –353, 1988. 147. Velloso LA, Carneiro EM, Crepaldi SC, Boschero AC, Saad MJ.
126. Schinner S, Barthel A, Dellas C, Grzeskowiak R, Sharma SK, Oetjen Glucose- and insulin-induced phosphorylation of the insulin receptor and
E, Blume R, Knepel W. Protein kinase B activity is sufficient to mimic its primary substrates IRS-1 and IRS-2 in rat pancreatic islets. FEBS Lett
the effect of insulin on glucagon gene transcription. J Biol Chem 280: 377: 353–357, 1995.
7369 –7376, 2005. 148. Vetiska SM, Ahmadian G, Ju W, Liu L, Wymann MP, Wang YT.
127. Shah P, Vella A, Basu A, Basu R, Schwenk WF, Rizza RA. Lack of GABAA receptor-associated phosphoinositide 3-kinase is required for
suppression of glucagon contributes to postprandial hyperglycemia in insulin-induced recruitment of postsynaptic GABAA receptors. Neuro-
subjects with type 2 diabetes mellitus. J Clin Endocrinol Metab 85: pharmacology 52: 146 –155, 2007.
4053– 4059, 2000.
149. Vignali S, Leiss V, Karl R, Hofmann F, Welling A. Characterization of
128. Sherwin RS, Fisher M, Hendler R, Felig P. Hyperglucagonemia and
voltage-dependent sodium and calcium channels in mouse pancreatic A-
blood glucose regulation in normal, obese and diabetic subjects. N Engl
J Med 294: 455– 461, 1976. and B-cells. J Physiol 572: 691–706, 2006.
129. Shimatsu A, Kato Y, Matsushita N, Ohta H, Kabayama Y, Imura H. 150. Wan Q, Xiong ZG, Man HY, Ackerley CA, Braunton J, Lu WY,
Glucagon-induced somatostatin release from perifused rat hypothalamus: Becker LE, MacDonald JF, Wang YT. Recruitment of functional
calcium dependency and effect of cysteamine treatment. Neurosci Lett GABA(A) receptors to postsynaptic domains by insulin. Nature 388:
37: 285–289, 1983. 686 – 690, 1997.
130. Smismans A, Schuit F, Pipeleers D. Nutrient regulation of gamma- 151. Wang C, Kerckhofs K, Van de Casteele M, Smolders I, Pipeleers D,
aminobutyric acid release from islet beta cells. Diabetologia 40: 1411– Ling Z. Glucose inhibits GABA release by pancreatic ␤-cells through an
1415, 1997. increase in GABA shunt activity. Am J Physiol Endocrinol Metab 290:
131. Sorensen H, Winzell MS, Brand CL, Fosgerau K, Gelling RW, E494 –E499, 2006.
Nishimura E, Ahren B. Glucagon receptor knockout mice display 152. Wang Q, Liu L, Pei L, Ju W, Ahmadian G, Lu J, Wang Y, Liu F,
increased insulin sensitivity and impaired beta-cell function. Diabetes 55: Wang YT. Control of synaptic strength, a novel function of Akt. Neuron
3463–3469, 2006. 38: 915–928, 2003.
132. Spanswick D, Smith MA, Mirshamsi S, Routh VH, Ashford ML. 153. Wendt A, Birnir B, Buschard K, Gromada J, Salehi A, Sewing S,
Insulin activates ATP-sensitive K⫹ channels in hypothalamic neurons of Rorsman P, Braun M. Glucose inhibition of glucagon secretion from rat
lean, but not obese rats. Nat Neurosci 3: 757–758, 2000. alpha-cells is mediated by GABA released from neighboring beta-cells.
133. Stagner JI, Samols E. The vascular order of islet cellular perfusion in Diabetes 53: 1038 –1045, 2004.
the human pancreas. Diabetes 41: 93–97, 1992. 154. Whiting PJ, Bonnert TP, McKernan RM, Farrar S, Le BB, Heavens
134. Stagner JI, Samols E, Bonner-Weir S. Beta—alpha— delta pancreatic RP, Smith DW, Hewson L, Rigby MR, Sirinathsinghji DJ, Thomp-
islet cellular perfusion in dogs. Diabetes 37: 1715–1721, 1988.
son SA, Wafford KA. Molecular and functional diversity of the expand-
135. Stagner JI, Samols E, Koerker DJ, Goodner CJ. Perfusion with
ing GABA-A receptor gene family. Ann NY Acad Sci 868: 645– 653,
anti-insulin gamma globulin indicates a B to A to D cellular perfusion
sequence in the pancreas of the rhesus monkey, Macaca mulatta. Pan- 1999.
creas 7: 26 –29, 1992. 155. Winnock F, Ling Z, De Proft R, Dejonghe S, Schuit F, Gorus F,
136. Starke A, Imamura T, Unger RH. Relationship of glucagon suppres- Pipeleers D. Correlation between GABA release from rat islet ␤-cells
sion by insulin and somatostatin to the ambient glucose concentration. and their metabolic state. Am J Physiol Endocrinol Metab 282: E937–
J Clin Invest 79: 20 –24, 1987. E942, 2002.
137. Straub SG, Sharp GW. Glucose-stimulated signaling pathways in 156. Xia F, Leung YM, Gaisano G, Gao X, Chen Y, Fox JE, Bhattacharjee
biphasic insulin secretion. Diabetes Metab Res Rev 18: 451– 463, 2002. A, Wheeler MB, Gaisano HY, Tsushima RG. Targeting of voltage-
138. Suzuki M, Fujikura K, Inagaki N, Seino S, Takata K. Localization of gated K⫹ and Ca2⫹ channels and soluble N-ethylmaleimide-sensitive
the ATP-sensitive K⫹ channel subunit Kir6.2 in mouse pancreas. Dia- factor attachment protein receptor proteins to cholesterol-rich lipid rafts
betes 46: 1440 –1444, 1997. in pancreatic alpha-cells: effects on glucagon stimulus-secretion cou-
139. Taniguchi CM, Emanuelli B, Kahn CR. Critical nodes in signalling pling. Endocrinology 148: 2157–2167, 2007.
pathways: insights into insulin action. Nat Rev Mol Cell Biol 7: 85–96, 157. Xu E, Kumar M, Zhang Y, Ju W, Obata T, Zhang N, Liu S, Wendt
2006. A, Deng S, Ebina Y, Wheeler MB, Braun M, Wang Q. Intra-islet
140. Taniguchi H, Okada Y, Seguchi H, Shimada C, Seki M, Tsutou A, insulin suppresses glucagon release via GABA-GABAA receptor system.
Baba S. High concentration of gamma-aminobutyric acid in pancreatic Cell Metab 3: 47–58, 2006.
beta cells. Diabetes 28: 629 – 633, 1979. 158. Yang SN, Wenna ND, Yu J, Yang G, Qiu H, Yu L, Juntti-Berggren
141. Tsuchiyama N, Takamura T, Ando H, Sakurai M, Shimizu A, Kato L, Kohler M, Berggren PO. Glucose recruits K(ATP) channels via
K, Kurita S, Kaneko S. Possible role of alpha-cell insulin resistance in non-insulin-containing dense-core granules. Cell Metab 6: 217–228,
exaggerated glucagon responses to arginine in type 2 diabetes. Diabetes
2007.
Care 30: 2583–2587, 2007.
159. Zhou H, Tran PO, Yang S, Zhang T, LeRoy E, Oseid E, Robertson
142. Uehara S, Muroyama A, Echigo N, Morimoto R, Otsuka M, Yatsus-
hiro S, Moriyama Y. Metabotropic glutamate receptor type 4 is involved RP. Regulation of alpha-cell function by the beta-cell during hypogly-
in autoinhibitory cascade for glucagon secretion by alpha-cells of islet of cemia in Wistar rats: the “switch-off” hypothesis. Diabetes 53: 1482–
Langerhans. Diabetes 53: 998 –1006, 2004. 1487, 2004.
143. Unger RH. Role of glucagon in the pathogenesis of diabetes: the status 160. Zhou H, Zhang T, Harmon JS, Bryan J, Robertson RP. Zinc, not
of the controversy. Metabolism 27: 1691–1709, 1978. insulin, regulates the rat alpha-cell response to hypoglycemia in vivo.
144. Unger RH. Glucagon physiology and pathophysiology in the light of Diabetes 56: 1107–1112, 2007.
new advances. Diabetologia 28: 574 –578, 1985. 161. Zhou H, Zhang T, Oseid E, Harmon J, Tonooka N, Robertson RP.
145. Unger RH, Aguilar-Parada E, Muller WA, Eisentraut AM. Studies of Reversal of defective glucagon responses to hypoglycemia in insulin-
pancreatic alpha cell function in normal and diabetic subjects. J Clin dependent autoimmune diabetic BB rats. Endocrinology 148: 2863–
Invest 49: 837– 848, 1970. 2869, 2007.

AJP-Endocrinol Metab • VOL 295 • OCTOBER 2008 • www.ajpendo.org


Downloaded from journals.physiology.org/journal/ajpendo (177.016.070.107) on December 14, 2022.

You might also like