ArticlePDF Available

Glucose Inhibition of Glucagon Secretion From Rat α-Cells Is Mediated by GABA Released From Neighboring β-Cells

Authors:

Abstract and Figures

gamma-Aminobutyric acid (GABA) has been proposed to function as a paracrine signaling molecule in islets of Langerhans. We have shown that rat beta-cells release GABA by Ca(2+)-dependent exocytosis of synaptic-like microvesicles. Here we demonstrate that GABA thus released can diffuse over sufficient distances within the islet interstitium to activate GABA(A) receptors in neighboring cells. Confocal immunocytochemistry revealed the presence of GABA(A) receptors in glucagon-secreting alpha-cells but not in beta- and delta-cells. RT-PCR analysis detected transcripts of alpha(1) and alpha(4) as well as beta(1-3) GABA(A) receptor subunits in purified alpha-cells but not in beta-cells. In whole-cell voltage-clamp recordings, exogenous application of GABA activated Cl(-) currents in alpha-cells. The GABA(A) receptor antagonist SR95531 was used to investigate the effects of endogenous GABA (released from beta-cells) on pancreatic islet hormone secretion. The antagonist increased glucagon secretion at 1 mmol/l glucose twofold and completely abolished the inhibitory action of 20 mmol/l glucose on glucagon release. Basal and glucose-stimulated secretion of insulin and somatostatin were unaffected by SR95531. The L-type Ca(2+) channel blocker isradipine evoked a paradoxical stimulation of glucagon secretion. This effect was not observed in the presence of SR95531, and we therefore conclude that isradipine stimulates glucagon secretion by inhibition of GABA release.
Content may be subject to copyright.
Glucose Inhibition of Glucagon Secretion From Rat
-Cells Is Mediated by GABA Released From
Neighboring -Cells
Anna Wendt,
1
Bryndis Birnir,
1
Karsten Buschard,
2
Jesper Gromada,
3
Albert Salehi,
1
Sabine Sewing,
3
Patrik Rorsman,
1
and Matthias Braun
1
-Aminobutyric acid (GABA) has been proposed to
function as a paracrine signaling molecule in islets of
Langerhans. We have shown that rat -cells release
GABA by Ca
2
-dependent exocytosis of synaptic-like
microvesicles. Here we demonstrate that GABA thus
released can diffuse over sufficient distances within the
islet interstitium to activate GABA
A
receptors in neigh-
boring cells. Confocal immunocytochemistry revealed
the presence of GABA
A
receptors in glucagon-secreting
-cells but not in - and -cells. RT-PCR analysis de-
tected transcripts of
1
and
4
as well as
1–3
GABA
A
receptor subunits in purified -cells but not in -cells.
In whole-cell voltage-clamp recordings, exogenous ap-
plication of GABA activated Cl
currents in -cells. The
GABA
A
receptor antagonist SR95531 was used to inves-
tigate the effects of endogenous GABA (released from
-cells) on pancreatic islet hormone secretion. The
antagonist increased glucagon secretion at 1 mmol/l
glucose twofold and completely abolished the inhibitory
action of 20 mmol/l glucose on glucagon release. Basal
and glucose-stimulated secretion of insulin and soma-
tostatin were unaffected by SR95531. The L-type Ca
2
channel blocker isradipine evoked a paradoxical stimu-
lation of glucagon secretion. This effect was not ob-
served in the presence of SR95531, and we therefore
conclude that isradipine stimulates glucagon secretion
by inhibition of GABA release. Diabetes 53:1038 –1045,
2004
I
n the mammalian central nervous system (CNS),
-aminobutyric acid (GABA) is the most important
inhibitory neurotransmitter. Three types of GABA
receptors have been identified: GABA
A
and GABA
C
receptors are ligand-gated Cl
channels, while GABA
B
receptors are G protein coupled (1). In the CNS, applica-
tion of GABA reduces excitability by a combination of
GABA
A
and GABA
B
receptor activation, leading to mem-
brane repolarization, reduced Ca
2
influx, and suppres-
sion of neurotransmitter release.
Outside the CNS, GABA and GAD, the enzyme cata-
lyzing the formation of the neurotransmitter, are present
at high levels in the pancreatic -cells (2,3). GAD65 is the
most abundant, if not the only, isoform of GAD in human
islets (4) and has been implicated in the etiology of
autoimmune type 1 diabetes (5). At the time of diagnosis,
80% of type 1 diabetic patients have autoantibodies
against GAD65 (6). The physiological roles of GABA and
GAD65 in pancreatic islets are unknown. GABA can, via
the formation of succinic semialdehyde and succinic acid,
be introduced into the tricarboxylic acid cycle and has
therefore been suggested to serve as an energy source
within the -cell (7). Others have hypothesized that GABA,
by analogy to its role in the CNS, functions as a paracrine
signaling molecule that conveys messages from the -cells
to surrounding cells in the islet (7–9).
In the -cell, GAD65 and GABA are associated with
synaptic-like microvesicles (SLMVs), which are distinct
from the insulin-containing large dense-core vesicles
(LDCVs) (10,11). SLMVs are smaller in diameter (90 nm)
than LDCVs (300 nm) (12) and accumulate GABA by
active transport (11). We have developed a patch clamp–
based technique to investigate GABA secretion from single
-cells with high temporal resolution. This technique in-
volves the expression of GABA
A
receptors at a high
density in the -cell using adenoviral vectors. We have
thus been able to demonstrate that GABA is released by
voltage- and Ca
2
-dependent exocytosis of SLMVs from
-cells (12). In this study we have focused on the influence
of endogenously released GABA on islet hormone secre-
tion. We demonstrate that GABA, following its exocytotic
release, travels over sufficient distances to activate GABA
receptors in neighboring cells, that functional GABA
A
receptors are present in -cells, and that GABA receptor
antagonism selectively affects glucagon release.
RESEARCH DESIGN AND METHODS
Islet preparation. Pancreatic islets and single islet cells were prepared from
Sprague Dawley or Wistar rats as described previously (13). In experiments
involving detection of vesicular GABA release (Fig. 1BD), cells were coin-
fected with adenoviruses expressing the
1
and
1
subunits of the GABA
A
receptor (12) and cultured for 24 h. All experimental procedures involving
animals were approved by the ethical committees in Lund, the city of
Hamburg, and the University of Copenhagen.
Electrophysiology. The electrophysiological measurements were conducted
in the standard whole-cell configuration. -Cells were identified on the basis
of their small size (4 pF as compared with 4pFfor-cells) and the
From the
1
Department of Physiological Sciences, Lund University, Lund,
Sweden;
2
Bartholin Instituttet, Kommunehospitalet, Copenhagen, Denmark;
and
3
Lilly Research Laboratories, Hamburg, Germany.
Address correspondence and reprint requests to Matthias Braun, Depart-
ment of Physiological Sciences, BMC B11, SE-221 84 Lund, Sweden. E-mail:
matthias.braun@mphy.lu.se.
Received for publication 18 November 2003 and accepted in revised form 19
January 2004.
CNS, central nervous system; FACS, fluorescence-activated cell sorter;
GABA, -aminobutyric acid; LDCV, large dense-core vesicle; SLMV, synaptic-
like microvesicle.
© 2004 by the American Diabetes Association.
1038 DIABETES, VOL. 53, APRIL 2004
inactivation properties of their voltage-gated Na
channels (14,15). The
experiments were performed either at 32°C (Fig. 1) or at room temperature
(Fig. 4).
Solutions. The standard extracellular solution consisted of (in mmol/l) 138
NaCl, 5.6 KCl, 2.6 CaCl
2
, 1.2 MgCl
2
, 5 HEPES (pH 7.4 with NaOH), and 15
glucose. In the experiment shown in FIg. 1BC, pentobarbital (0.05) and
forskolin (2 mol/l) were included to increase the amplitude of the GABA-
activated Cl
currents (16) and to elevate intracellular cAMP levels to
stimulate GABA release (12), respectively. When the cells were depolarized
with 50 mmol/l KCl (Fig. 1), the concentration of NaCl was correspondingly
decreased. For the electrophysiological recordings of endogenous GABA
A
receptor activity (Fig. 4), 20 TEA-Cl was substituted for an equal concentra-
tion of NaCl. The pipette solution (intracellular solution) used for all mea-
surements was composed of (in mmol/l) 125 CsCl, 30 CsOH, 10 EGTA, 1
MgCl
2
, 5 HEPES (pH 7.15 with HCl), and 3 Mg-ATP. With this intracellular
solution, activation of Cl
channels will result in Cl
efux at negative
membrane potentials, thus giving rise to an inward current (downward
deection). In the experiments displayed in Fig. 1D, 9 mmol/l CaCl
2
was added
to the above intracellular solution to increase the intracellular free concen-
tration of Ca
2
to 3 mol/l.
Immunouorescence. Intact rat islets were xed with the pH-shift/formal-
dehyde method and permeabilized with 5% Triton X-100. Normal donkey
serum at 5% was used to block unspecic binding. The islets were coincubated
with antibodies directed against insulin (1:200 dilution; Eurodiagnostica,
Malmo¨ , Sweden), glucagon (1:200; Dako, A
¨
lvsjo¨ , Sweden), somatostatin
(1:200; Biogenesis, Poole, U.K.), and the
2/3
subunits of the GABA
A
receptor
(1:100, clone 62-3G1; Upstate Biotechnology, Lake Placid, NY) at 4°C over-
night. Cy2-, Cy3-, and Cy5-conjugated secondary antibodies (Jackson Immu-
noResearch Laboratories, West Grove, PA) were used to detect the labeled
sites. Immunouorescence was then detected using a confocal microscope
(Zeiss 510; Zeiss, Go¨ ttingen, Germany).
RT-PCR. Total RNA was extracted from uorescence-activated cell sorter
(FACS)-puried (17) rat - and -cells using RNeasy Mini Kit (Qiagen, Hilden,
Germany) and used for reverse transcription using random hexamer primers
and Superscript II (Invitrogen, Karlsruhe, Germany). Total RNA from human
brain (Clontech, Heidelberg, Germany) was used as positive control. The
predicted sizes for the bands in the positive control were
1
363 bp,
2
407 bp,
3
505 bp,
4
246 bp,
5
589 bp,
6
177 bp,
1
270 bp,
2
342 bp, and
3
379 bp;
the s and s correspond to previously identied GABA
A
receptor subunits.
PCR amplications were performed under standard conditions running 40
cycles. Primer sequences are available on request (sewing@lilly.com).
Western blot. Islet homogenate was separated by SDS-PAGE on 8% acryl-
amide gels and transferred onto polyvinylidine uoride membranes. The
membranes were incubated overnight with the anti-GABA
A
2/3
antibody (2
g/ml, clone 62-3G1; Upstate Biotechnology). After 1 h incubation with
horseradish-peroxidase coupled secondary antibody (1:20,000 dilution), the
blots were developed by using SuperSignal West Pico Chemiluminescent
Substrate (Pierce, Cheshire, U.K.) and visualized on X-ray lms.
Hormone release measurements. Insulin, glucagon, and somatostatin re-
lease was determined by radioimmunoassay as described elsewhere (18,19).
Briey, batches of 8 10 islets were preincubated in 1 ml of Krebs-Ringer
buffer (KRB) supplemented with 1 mmol/l glucose for 30 min followed by 1 h
incubation in 1 ml Krebs-Ringer buffer containing 1 or 20 mmol/l glucose. The
specic GABA
A
receptor antagonist SR95531 (10 mol/l; Sigma, St. Louis, MO)
or the specicCa
2
channel antagonists isradipine (2.5 mol/l; kindly
provided by J. Striessing, Innsbruck, Austria), SNX482 (0.1 mol/l; Peptids
International, Louisville, KY), and -conotoxin GVIA (0.1 mol/l; Alomone
Labs, Jerusalem, Israel) were included as indicated. At the end of the
incubation, duplicate aliquots (25100 l) of the medium were removed and
frozen pending the radioimmunoassay.
Statistical analysis.Data are given as means SE. Statistical signicances
were evaluated using Students t test.
RESULTS
Intercellular GABA signaling between pancreatic is-
let cells. We have previously demonstrated that GABA
FIG. 1. Ability of GABA to function as a paracrine signaling molecule. A: Schematic picture showing the principles of the experiments. Membrane
currents are recorded from a cell voltage clamped at 70 mV and dialyzed with 10 mmol/l EGTA. Exocytosis from surrounding cells is induced
by depolarization with elevated K
. B: Whole-cell recording from a voltage-clamped cell in a cluster of rat islet cells infected with adenovirus
expressing GABA
A
1
1
receptors. Addition of K
(50 mmol/l) is indicated by the horizontal bar. Inward current transients triggered by GABA
release are indicated by arrows (n 4). C: Examples of current transients from B on an extended time base. D: Examples of current transients
due to exocytosis of GABA-containing SLMVs, recorded from an isolated cell. Exocytosis was elicited by intracellular dialysis with 3 mol/l free
Ca
2
.
A. WENDT AND ASSOCIATES
DIABETES, VOL. 53, APRIL 2004 1039
can be released from rat -cells by Ca
2
-dependent exo
-
cytosis of SLMVs (12). Here we investigate whether GABA
released from one cell can activate GABA receptors in
neighboring cells, a requirement for GABA to function as a
paracrine regulator. To this end, patch-clamp recordings
were applied to cells within small islet cell clusters in-
fected with adenovirus expressing GABA
A
receptor Cl
channels. Secretion was stimulated by adding 50 mmol/l
K
to the extracellular medium. This leads to membrane
depolarization, opening of voltage-gated Ca
2
channels,
and exocytosis in the unclamped cells. The membrane
potential of the patch-clamped cell was kept at 70 mV
throughout the experiment, which is too negative for Ca
2
channel activation and voltage-dependent GABA release
(12). Furthermore, intracellular free Ca
2
was clamped to
nonexocytotic levels by including 10 mmol/l EGTA in the
pipette-lling solution dialyzing the cell interior. Any re-
corded GABA-activated Cl
currents must accordingly
result from GABA being released by the surrounding
unclamped cells as illustrated schematically in Fig. 1A.
Figure 1B shows the holding current before and during
stimulation with high extracellular K
. Before stimulation,
the holding current was stable. Following addition of 50
mmol/l K
to the extracellular medium, a series of nine
fast current transients were observed (arrows). Examples
of these current transients are shown in Fig. 1C on an
expanded time base. Figure 1D shows examples of GABA-
activated Cl
currents recorded in a single cell in which
exocytosis was stimulated by dialyzing the interior of the
voltage-clamped cell with a Ca
2
-EGTA buffer containing
3 mol/l free Ca
2
instead of the Ca
2
-free medium. The
events shown in Fig. 1CD show a great resemblance. We
analyzed the activation of the current spikes and their
durations by measuring the t
10 90%
(i.e., the time it takes
for the current to increase from 10 to 90% of its maximal
amplitude) and half-width (the time the current exceeds
the half-maximal amplitude). Mean values for t
10 90%
and
the half-width of 15 2 and 53 5ms(n 9),
respectively, were obtained. These values are similar to
those recorded from -cells in which exocytosis was
stimulated by intracellular application of 3 mol/l Ca
2
(t
10 90%
12.6 0.7 ms, half-width 30 1 ms [12]). We
conclude that GABA released from one cell can diffuse to
neighboring cells and that the concentration of the neuro-
transmitter remains sufciently high to activate GABA
A
receptors in these cells.
GABA
A
receptor subunits are expressed in -cells
but not in -or-cells. Ionotropic GABA
A
receptors
have been reported to be endogenously expressed in rat
(20) and human (21) pancreatic islets. We used RT-PCR to
determine the expression pattern of endogenous GABA
A
receptors in FACS-puried - and -cells from rat, utilizing
specic primers for the
1
-
6
and the
1
-
3
subunits of the
GABA
A
receptor. The
1
,
4
,
1
,
2
, and
3
subunits were
expressed in rat pancreatic -cells, whereas none of the
subunits investigated were detected in -cells (Fig. 2). The
identity of all PCR products was conrmed by sequencing.
We veried that the GABA
A
receptor transcripts detected
with RT-PCR translated into protein by performing West-
ern blot on rat islet homogenate. As shown in Fig. 3A,an
antibody directed against the
2/3
subunits detected a
protein with a molecular weight of 60 kDa, close to the
FIG. 2. RT-PCR on puried -and -cells with specic primers against
different GABA
A
receptor subunits. Total RNA from FACS-puried -
and -cells was reverse transcribed into cDNA, and PCR analysis was
performed with specic primers for the
1 6
and
13
subunits (). In
the negative control reaction (), reverse transcriptase was omitted.
Total RNA from human brain was reverse transcribed into cDNA and
used as a positive control. Molecular standards are shown to the left.
FIG. 3. Western blot and confocal immunocytochemistry with a GABA
A
receptorspecic antibody. A: Western blot on rat islet homogenate (150
islets) using a specic antibody against the
2/3
subunits of the GABA
A
receptor. B and C: Confocal immunocytochemistry of isolated rat islets
detecting the
2/3
subunits of the GABA
A
receptor, insulin, and glucagon or somatostatin and overlay as indicated. Scale bars: 10 m.
GABAergic SIGNALING IN ISLETS
1040 DIABETES, VOL. 53, APRIL 2004
expected size (57 kDa) (22). Confocal immunocytochem-
istry using the same antibody revealed the presence of
GABA
A
receptors in the glucagon-containing -cells (Fig.
3B), whereas no
2/3
immunoreactivity was observed in
-cells (Fig. 3B C)or-cells (Fig. 3C).
Electrophysiological measurements of endogenous
GABA
A
receptors. We next applied whole-cell patch-
clamp measurements to conrm the presence of func-
tional GABA
A
receptors in -cells. Application of 1 mmol/l
GABA to functionally identied (see
RESEARCH DESIGN AND
METHODS
and below) -cells elicited inward Cl
currents
(Fig. 4A). In this particular experiment, the amplitude of
the current was unusually large, reaching a peak ampli-
tude of approximately 600 pA. The amplitude of the
GABA-evoked Cl
current was extremely variable and
ranged between 20 pA and 600 pA, with an average of
138 93 pA (n 6). The current elicited by GABA was
blocked by the specic GABA
A
receptor antagonist
SR95531 (Fig. 4B). As expected for -cells (23), the
capacitance of the cells containing GABA-activated cur-
rents averaged 3.0 0.3 pA (n 6), signicantly less than
the 4.7 0.2 pF (n 31) obtained for -cells (P 0.001).
No inward Cl
currents could be elicited in -cells (not
shown).
Effect of endogenous GABA release on pancreatic
hormone release. -Cells are electrically active (24) and
secrete glucagon in response to membrane depolarizations
and opening of voltage-dependent Ca
2
channels (17).
Because GABA
A
receptors are present in -cells, it is
expected that exocytosis of GABA-containing SLMVs from
-cells will hyperpolarize the -cells with resultant sup-
pression of action-potential ring and glucagon secretion.
This is not easily studied using electrophysiological tech-
niques, but by using the GABA
A
receptor antagonist
SR95531, some insight into the signicance of the intrinsic
GABAergic signaling in the islets can nevertheless be
obtained. Pancreatic hormone secretion was measured in
isolated intact rat pancreatic islets at 1 and 20 mmol/l
glucose in the absence and presence of 10 mol/l SR95531.
It should be noted that by using an antagonist, we explore
the role of endogenous GABA and no exogenous GABA
was applied in these experiments. As seen in Table 1,
including SR95531 in the extracellular medium had no
effect on the release of insulin or somatostatin, irrespec-
tive of the glucose concentration. By contrast, glucagon
secretion at 1 and 20 mmol/l glucose was stimulated
approximately two- to threefold in the presence of the
antagonist. Importantly, whereas glucagon secretion was
reduced by 50% under control conditions when raising
glucose to 20 mmol/l, this effect was abolished in the
presence of SR95531.
Ca
2
channel antagonists reveal paracrine interac
-
tions between islet cells. Table 2 summarizes the effects
of the L-type Ca
2
channel blocker isradipine (2.5 mol/l),
the N-type Ca
2
channel antagonist -conotoxin GVIA
(100 nmol/l), and the R-type Ca
2
channel blocker SNX482
(100 nmol/l) on insulin, glucagon, and somatostatin secre-
tion at 1 and 20 mmol/l glucose in isolated rat pancreatic
islets. Isradipine had no effect on insulin, somatostatin, or
glucagon release measured at 1 mmol/l glucose. Glucose-
induced insulin secretion was inhibited by 80% by the
L-type channel blocker, whereas there was no signicant
effect on somatostatin release. By contrast, SNX482 failed
to affect insulin secretion, but produced an 50% reduc-
tion in glucose-stimulated somatostatin secretion. SNX482
likewise lacked effect on glucagon release measured at 1
mmol/l glucose.
In the presence of 1 mmol/l glucose, -conotoxin GVIA
(0.1 mol/l) inhibited glucagon secretion by 50%, similar
to the inhibition produced by raising glucose to 20 mmol/l.
Importantly, neither isradipine nor SNX482 affected gluca-
gon secretion at 1 mmol/l glucose. Addition of isradipine in
the presence of 20 mmol/l glucose resulted in a paradox-
ical stimulation of glucagon release. Figure 5 summarizes
glucagon release measured at 20 mmol/l glucose under
control conditions and in the presence of 2.5 mol/l
isradipine, 0.1 mol/l SNX482, and 10 mol/l of the GABA
A
receptor antagonist SR95531. These data have been nor-
malized to glucagon secretion measured at 1 mmol/l
glucose. This analysis reveals that, whereas glucagon
secretion in the presence of SR95531 and 20 mmol/l
glucose exceeds that observed at 1 mmol/l glucose, the
release of the hormone measured in the presence of a
combination of 20 mmol/l glucose and 2.5 mol/l isradip-
ine was 80% of that observed at 1 mmol/l glucose. We
propose that isradipine stimulates glucagon release by
FIG. 4. Electrophysiological measurements of endogenous GABA
A
receptors. Cl
current elicited by application of 1 mmol/l GABA to an
-cell under control conditions (A) and in presence of the GABA
A
receptor antagonist SR95531 (B).
TABLE 1
Effect of the GABA
A
receptor antagonist SR95531 on hormone release from isolated rat islets
Glucose (mmol/l)
Insulin (ng islet
1
h
1
)
Somatostatin
(pmol islet
1
h
1
)
Glucagon (pg islet
1
h
1
)
1 20 1 20 1 20
Control 0.37 0.03 6.17 0.3 5.05 0.5 24.48 1.1 27.18 2.1 15.31 1.2
10 mol/l SR95531 0.32 0.06 6.01 0.2 5.50 0.2 26.1 1.6 46.72 3.8* 51.04 4.0*
Data are means SE of 10 experiments. *P 0.001 vs. control; NS vs. 10 mol/l SR95531 at 1 mmol/l glucose.
A. WENDT AND ASSOCIATES
DIABETES, VOL. 53, APRIL 2004 1041
inhibiting the release of a paracrine regulator from the
-cell. Given the observation that isradipine was ineffec-
tive when added to islets already exposed to SR95531, it
appears that the compound mediating the effect is GABA.
DISCUSSION
We have recently developed an electrophysiological assay
based on the overexpression of GABA
A
receptor Cl
channels that allows us to detect exocytosis of single
GABA-containing vesicles from -cells (12). In this study
we have applied the assay to clusters of islet cells (Fig.
1A). As the experimental conditions were designed to
prevent any exocytosis from the patch-clamped cell, the
Cl
current transients recorded must result from GABA
exocytosis from neighboring cells. The experiment there-
fore suggests that GABA is indeed capable of traveling in
the interstitium between islets cells. Close inspection of
the observed current spikes revealed that they rose with
the same velocity as those recorded in single cells, where
the current transients result from GABA being released
from the same cell. This indicates that the receptor cell
must be sitting very close to the cell in which GABA
release occurred. Indeed, electron microscopy reveals that
the interstitial space between neighboring islet cells is
very small (25), which facilitates paracrine regulation.
GABA
A
receptors are heteromultimeric channels typi
-
cally composed of two , two , and a varying third
subunit. In RT-PCR experiments performed on FACS-
puried islet cells, we found that rat -cells express the
1
and
4
subunit as well as
13
(Fig. 2). Although RT-PCR
is not a quantitative method, the intensity of the band for
the
4
subunit suggests that this is the predominant
subunit in -cells. Receptor combinations containing the
4
subunit are found extrasynaptically in hippocampus
and thalamus (26) and characteristically exhibit a higher
afnity for GABA (27). This argues that release of GABA
may activate GABA
A
receptors even beyond the immediate
vicinity of the release sites, which facilitates GABAergic
signaling within the islet. The experiment shown in Fig. 1B
was performed in cells transfected with
1
1
receptors,
which have a lower GABA sensitivity than receptors
containing the
4
subunit (11 mol/l [28] vs. 0.52.6 mol/l
[27]). The extent of intercellular GABAergic signaling
might accordingly be even stronger than suggested by the
present data.
To date, most functional experiments on GABAergic
signaling in pancreatic islets have been conducted using
application of exogenous GABA (29,30) or agonists such
as muscimol (7). GABA
A
receptor Cl
channels inactivate
(i.e., the channels enter a nonconducting state) within
seconds when exposed to GABA concentrations of 30
mol/l (31). Thus, in studies using application of high
concentrations of GABA to affect hormone release, a
current could ow through the channels only for a brief
period immediately after the addition of the agonist.
Accordingly, during most of the experiment, both mem-
brane potential and secretion are likely to be unaffected. A
way around this problem is to use an antagonist against
the GABA
A
receptor instead and study the inuence of
endogenous GABA on pancreatic hormone release. How-
ever, bicuculline, which has been used to this end in the
past has promiscuous pharmacological properties and
inhibits, for example, Ca
2
-activated K
channels (32) in
addition to the GABA
A
receptors, which may lead to
unspecic effects (M.B., A.W., unpublished data). In this
study we have instead used the selective antagonist
SR95531 and thereby found that, at least in rat islets, only
glucagon secretion is modulated by GABA
A
receptor acti
-
vation. This observation is corroborated by the nding that
GABA
A
receptors are only detected in pancreatic -cells.
The fact that neither somatostatin nor insulin secretion
were affected by the antagonist, although the electrophys-
iology of - and -cells is similar to that of the -cells,
conrms that the action of this compound is selective.
Surprisingly, addition of SR95531 stimulated glucagon
secretion measured at 1 mmol/l glucose. This might indi-
cate that GABA is present at biological concentrations
FIG. 5. Effect of the Ca
2
channel blockers SNX482 and isradipine and
the GABA
A
receptor antagonist SR95531 on glucagon secretion from
isolated rat islets. Glucagon secretion was measured in the presence of
20 mmol/l glucose alone or after including 0.1 mol/l SNX482, 2.5
mol/l isradipine, or 10 mol/l SR95531 as indicated. Data are normal-
ized to glucagon release at 1 mmol/l glucose and are means SE of
8 10 experiments.
TABLE 2
Effect of Ca
2
channel antagonists on hormone release from rat islets
Glucose (mmol/l)
Insulin (ng islet
1
h
1
)
Somatostatin
(pmol islet
1
h
1
)
Glucagon (pg islet
1
h
1
)
1 20120 1 20
Control 0.49 0.04 10.39 0.7 4.30 0.6 17.82 2.3 36.85 2.3 19.79 1.2
2.5 mol/l isradipine 0.47 0.05 1.72 0.2* 3.65 0.5 14.06 1.3 34.10 2.5 28.98 3.6§
0.1 mol/l SNX482 0.42 0.04 8.87 0.9 2.92 0.3 9.37 1.3 39.75 3.9 23.91 3.3
0.1 mol/l -conotoxin GVIA 0.44 0.04 9.40 0.6 3.44 0.5 14.13 2.6 17.03 2.9* 16.06 1.1
Data are means SE of 8 experiments. Hormone release was measured at 1 and 20 mmol/l glucose in the absence and presence of isradipine,
SNX482, and -conoxotin GVIA as indicated. Statistical signicance is evaluated against the control at the same glucose concentration. *P
0.001; P 0.01; NS; §P 0.05.
GABAergic SIGNALING IN ISLETS
1042 DIABETES, VOL. 53, APRIL 2004
already at low glucose concentrations. Indeed, it has been
estimated that 25% of the total -cell content of GABA (21
pmol per 1,000 -cells) is released per hour regardless of
the glucose concentration (33). This corresponds to a net
release rate of 0.4 amol of GABA, equivalent to approxi-
mately one GABA-containing SLMV, per second and cell.
Our studies on exocytotic GABA release performed on
single cells did not provide evidence for this relatively high
basal release rate (12). However, additional signaling
cascades may be operative in intact islets. For example, it
has been proposed that GABA release from the -cells is
controlled by glutamate, cosecreted with glucagon from
the -cells, rather than by glucose directly (25). It should
be pointed out, however, that even millimolar amounts of
glutamate have no detectable effect of GABA release in our
hands (12). Alternatively, basal GABA release may reect
passive leakage mediated by an uncharacterized trans-
porter. The relative contributions of basal and stimulated
release to GABAergic signaling remain to be elucidated.
In this study we demonstrate that hormone release from
-, -, and -cells depends differentially on Ca
2
inux
through different types of Ca
2
channels. Thus glucose-
induced insulin secretion is almost completely blocked by
the L-type Ca
2
channel blocker isradipine. Glucose-in
-
duced somatostatin release is principally due to SNX482-
sensitive R-type Ca
2
channels, while glucagon secretion
is dependent on -conotoxinsensitive N-type Ca
2
chan
-
nels (Table 2). Interestingly, isradipine stimulated gluca-
gon secretion in the presence of a maximally inhibitory
concentration of glucose, although this Ca
2
channel
antagonist had no effect when release of the hormone was
stimulated by hypoglycemia. This observation is easiest to
explain if glucagon secretion was suppressed at high
glucose concentrations by a compound released from the
-cells. Theoretically, any substance released from the
-cell could exert such an inhibitory action; possible
candidates include insulin itself, zinc, ATP, islet amyloid
polypeptide, etc. (34). Indeed, both insulin (35) and zinc
(36) have been suggested to suppress glucagon release. It
is pertinent that isradipine had no additional stimulatory
effect in the presence of SR95531. The data therefore
suggest that a signicant part of the inhibitory action of
glucose in isolated rat islets is mediated by GABAergic
mechanisms. They also argue that exocytosis of GABA-
containing SLMVs, like insulin-containing LDCVs, depends
on Ca
2
entry via L-type Ca
2
channels. This could either
reect a requirement of such channels for glucose-induced
electrical activity (37) or exocytosis of SLMVs being
directly coupled to Ca
2
entry via these channels. Our
single-cell measurements of GABA release using -cells
infected with GABA
A
receptors have clearly established
that exocytosis of SLMVs is controlled by Ca
2
entry via
voltage-gated Ca
2
channels (12), but the molecular iden
-
tity of the channel remains to be determined. However,
given that L-type Ca
2
channels account for the largest
part (60%) of the whole-cell Ca
2
current with little
(10%) contribution by both R- and N-type Ca
2
channels
(A.W., M.B., P.R., unpublished results), it seems reason-
able to conclude that the L-type Ca
2
channels are indeed
important for exocytosis of the GABA-containing SLMVs.
Figure 6 summarizes schematically the molecular play-
ers and nature of interactions involved. Glucose-induced
electrical activity of the -cell leads to opening of voltage-
gated L-type Ca
2
channels with subsequent Ca
2
inux
and Ca
2
-dependent exocytosis of both insulin-containing
LDCVs and GABA-containing SLMVs. GABA thus released
diffuses in the narrow space between the release site and
the neighboring -cells, where it activates GABA
A
receptor
Cl
channels. The rate of release is one vesicle per second
in the absence of glucose but increases 5- to 10-fold during
electrical activity. Each event elicits a rapidly activating
current that lasts 100 ms. Because the -cells have a high
input resistance (few active channels), the activation of
the GABA
A
receptor Cl
channels will clamp the -cell
membrane potential to the Cl
equilibrium potential (E
Cl
).
Depending on the intracellular Cl
concentration, opening
of Cl
channels will suppress electrical activity by either
hyperpolarizing or depolarizing the -cell. In the former
case, the membrane potential becomes too negative for
action potential generation. In the latter case, the mem-
brane potential becomes so positive that the ion currents
involved in action-potential generation undergo voltage-
dependent steady-state inactivation. The reduction in ac-
tion-potential ring leads to reduced activation of N-type
Ca
2
channels and reduced exocytosis of the glucagon-
containing secretory granules in the -cell. In addition to
N-type Ca
2
channels, -cells are also equipped with
L-type Ca
2
channels (17) that open during depolarization
and contribute to the increases in cytoplasmic free Ca
2
concentration (38,39). However, these channels contribute
little to exocytosis under basal conditions (17,40), but
their importance increases dramatically under certain
physiological situations. For example, L-type Ca
2
chan
-
nels account for 80% of glucagon secretion under experi-
mental conditions associated with enhanced protein
kinase A activity (17).
We acknowledge that paracrine regulation by GABA
may not be the only mechanism inuencing glucagon
FIG. 6. Model for GABAergic regula-
tion of glucagon secretion. See text
for details. [Ca
2
]
i
, cytoplasmic free
Ca
2
concentration; GABA
R
, GABA
A
receptor Cl
channels; VGLC, voltage-
gated L-type Ca
2
channels; VGNC,
voltage-gated N-type Ca
2
channels;
, membrane potential; 8, an increase
or decrease in ion concentration or
membrane potential (depolarization/
hyperpolarization).
A. WENDT AND ASSOCIATES
DIABETES, VOL. 53, APRIL 2004 1043
release. Release of Zn
2
from the -cells has recently been
proposed to suppress glucagon release in neighboring
-cells in rat islets (36). Studies using a novel somatostatin
receptor antagonist have shown that somatostatin inhibits
glucagon secretion in rat islets (41). We point out that
these mechanisms are not mutually exclusive, and it is
indeed possible that all these mechanisms operate in
parallel in -cells in situ. The nding that SR95531 com-
pletely reverses the inhibitory effect of glucose on gluca-
gon secretion strongly suggests, however, that GABAergic
signaling plays an important role in the metabolic control
of glucagon secretion in intact rat pancreatic islets. In
addition, pancreatic -cells contain metabotropic GABA
B
receptors (42), thus providing yet another GABAergic
regulatory mechanism within the islet. This aspect will be
addressed at length elsewhere (M.B., A.W., K.B., A.S., S.S.,
J.G., P.R., unpublished observations). Interestingly, mouse
islets contain much less GABA (2), and GABAergic mech-
anisms such as those we discuss here are consequently
likely to be less important in such islets. This argues that
other mechanisms regulate glucagon secretion in mouse
islets (see for example ref. 43). It is now essential to
establish whether human -cells also are equipped with
GABA
A
receptor Cl
channels and, if so, to clarify their
role in the control of glucagon secretion. Circumstantial
evidence in support of an important role for GABAergic
mechanisms in human islets comes from the nding that
these islets contain high levels of both GABA and GAD65.
ACKNOWLEDGMENTS
This study was supported by the Swedish Research Coun-
cil (Medicine, grant no. 8647), the Juvenile Diabetes Re-
search Foundation, the Swedish Diabetes Association, the
NovoNordisk Foundation, and the European Commission
(projects Growbeta and Neuronal Ca
2
-channels). We
thank Kristina Borglid and Britt-Marie Nilsson for excel-
lent technical assistance.
REFERENCES
1. Chebib M, Johnston GA: The ABCof GABA receptors: a brief review. Clin
Exp Pharmacol Physiol 26:937940, 1999
2. Michalik M, Nelson J, Erecinska M: GABA production in rat islets of
Langerhans. Diabetes 42:1506 1513, 1993
3. Taniguchi H, Okada Y, Seguchi H, Shimada C, Seki M, Tsutou A, Baba S:
High concentration of -aminobutyric acid in pancreatic -cells. Diabetes
28:629 633, 1979
4. Petersen JS, Russel S, Marshall MO, Kofod H, Buschard K, Cambon N,
Karlsen AE, Boel E, Hagopian WA, Hejnaes KR, Moody A, Dryberg T,
Lernmark A, Madsen O, Michelson BK: Differential expression of glutamic
acid decarboxylase in rat and human islets. Diabetes 42:484 495, 1993
5. Lernmark AF: Immunology of Insulin-Dependent Diabetes Mellitus. Ox-
ford, Blackwell Science, 1997
6. Schmidli RS, Colman PG, Bonifacio E, Bottazzo GF, Harrison LC: High
level of concordance between assays for glutamic acid decarboxylase
antibodies: the First International Glutamic Acid Decarboxylase Antibody
Workshop. Diabetes 43:10051009, 1994
7. Sorenson RL, Garry DG, Brelje TC: Structural and functional consider-
ations of GABA in islets of Langerhans: -cells and nerves. Diabetes
40:13651374, 1991
8. Shi Y, Kanaani J, Menard-Rose V, Ma YH, Chang PY, Hanahan D, Tobin A,
Grodsky G, Baekkeskov S: Increased expression of GAD65 and GABA in
pancreatic -cells impairs rst-phase insulin secretion. Am J Physiol
Endocrinol Metab 279:E684 E694, 2000
9. Rorsman P, Berggren PO, Bokvist K, Ericson H, Mo¨ hler H, O
¨
stenson CG,
Smith PA: Glucose-inhibition of glucagon secretion involves activation of
GABA
A
-receptor chloride channels. Nature 341:233236, 1989
10. Reetz A, Solimena M, Matteoli M, Folli F, Takei K, De Camilli P: GABA and
pancreatic -cells: colocalization of glutamic acid decarboxylase (GAD)
and GABA with synaptic-like microvesicles suggests their role in GABA
storage and secretion. EMBO J 10:12751284, 1991
11. Thomas-Reetz A, Hell JW, During MJ, Walch-Solimena C, Jahn R, De
Camilli P: A -aminobutyric acid transporter driven by a proton pump is
present in synaptic-like microvesicles of pancreatic -cells. Proc Natl Acad
SciUSA90:53175321, 1993
12. Braun M, Wendt A, Birnir B, Broman J, Eliasson L, Galvanovskis J,
Gromada J, Mulder H, Rorsman P: Regulated exocytosis of GABA-
containing synaptic-like microvesicles in pancreatic -cells. J Gen Physiol.
In press
13. Olofsson CS, Go¨ pel SO, Barg S, Galvanovskis J, Ma X, Salehi A, Rorsman
P, Eliasson L: Fast insulin secretion reects exocytosis of docked granules
in mouse pancreatic -cells. Pflugers Arch 444:4351, 2002
14. Go¨ pel S, Kanno T, Barg S, Galvanovskis J, Rorsman P: Voltage-gated and
resting membrane currents recorded from -cells in intact mouse pancre-
atic islets. J Physiol 521:717728, 1999
15. Hiriart M, Matteson DR: Na channels and two types of Ca channels in rat
pancreatic -cells identied with the reverse hemolytic plaque assay.
J Gen Physiol 91:617 639, 1988
16. Birnir B, Tierney ML, Dalziel JE, Cox GB, Gage PW: A structural determi-
nant of desensitization and allosteric regulation by pentobarbitone of the
GABA
A
receptor. J Membr Biol 155:157166, 1997
17. Gromada J, Bokvist K, Ding WG, Barg S, Buschard K, Renstro¨ mE,
Rorsman P: Adrenaline stimulates glucagon secretion in pancreatic -cells
by increasing the Ca
2
current and the number of granules close to the
L-type Ca
2
channels. J Gen Physiol 110:217228, 1997
18. Vonen B, Florholmen J, Giaever AK, Burhol P: Radio-immunoassay of
somatostatin from isolated rat pancreatic islets. Scand J Clin Lab Invest
49:135138, 1989
19. Salehi A, Chen D, Hakanson R, Nordin G, Lundquist I: Gastrectomy induces
impaired insulin and glucagon secretion: evidence for a gastro-insular axis
in mice. J Physiol 514:579 591, 1999
20. Borboni P, Porzio O, Fusco A, Sesti G, Lauro R, Marlier LN: Molecular and
cellular characterization of the GABA
A
receptor in the rat pancreas. Mol
Cell Endocrinol 103:157163, 1994
21. Yang W, Reyes AA, Lan NC: Identication of the GABA
A
receptor subtype
mRNA in human pancreatic tissue. FEBS Lett 346:257262, 1994
22. Li M, De Blas AL: Coexistence of two -subunit isoforms in the same
-aminobutyric acid type A receptor. J Biol Chem 272:16564 16569, 1997
23. Pipeleers DG, int Veld PA, Van de Winkel M, Maes E, Schuit FC, Gepts W:
A new in vitro model for the study of pancreatic and cells. Endocri-
nology 117:806 816, 1985
24. Rorsman P, Hellman B: Voltage-activated currents in guinea pig pancreatic
2
cells: evidence for Ca
2
-dependent action potentials. J Gen Physiol
91:223242, 1988
25. Hayashi M, Yamada H, Uehara S, Morimoto R, Muroyama A, Yatsushiro S,
Takeda J, Yamamoto A, Moriyama Y: Secretory granule-mediated co-
secretion of L-glutamate and glucagon triggers glutamatergic signal trans-
mission in islets of Langerhans. J Biol Chem 278:1966 1974, 2003
26. Mody I: Distinguishing between GABA
A
receptors responsible for tonic
and phasic conductances. Neurochem Res 26:907913, 2001
27. Brown N, Kerby J, Bonnert TP, Whiting PJ, Wafford KA: Pharmacological
characterization of a novel cell line expressing human
4
3
GABA
A
receptors. Br J Pharmacol 136:965974, 2002
28. Birnir B, Tierney ML, Pillai NP, Cox GB, Gage PW: Rapid desensitization of
1
1
GABA
A
receptors expressed in Sf9 cells under optimized conditions.
J Membr Biol 148:193202, 1995
29. Gu XH, Kurose T, Kato S, Masuda K, Tsuda K, Ishida H, Seino Y:
Suppressive effect of GABA on insulin secretion from the pancreatic
-cells in the rat. Life Sci 52:687 694, 1993
30. Gilon P, Bertrand G, Loubatieres-Mariani MM, Remacle C, Henquin JC: The
inuence of -aminobutyric acid on hormone release by the mouse and rat
endocrine pancreas. Endocrinology 129:25212529, 1991
31. Frosch MP, Lipton SA, Dichter MA: Desensitization of GABA-activated
currents and channels in cultured cortical neurons. J Neurosci 12:3042
3053, 1992
32. Seutin V, Johnson SW: Recent advances in the pharmacology of quaternary
salts of bicuculline. Trends Pharmacol Sci 20:268 270, 1999
33. Smismans A, Schuit F, Pipeleers D: Nutrient regulation of -aminobutyric
acid release from islet -cells. Diabetologia 40:14111415, 1997
34. Rorsman P, Renstro¨ m E: Insulin granule dynamics in pancreatic beta cells.
Diabetologia 46:1029 1045, 2003
35. Greenbaum CJ, Havel PJ, Taborsky GJ, Jr, Klaff LJ: Intra-islet insulin
permits glucose to directly suppress pancreatic -cell function. J Clin
Invest 88:767773, 1991
GABAergic SIGNALING IN ISLETS
1044 DIABETES, VOL. 53, APRIL 2004
36. Ishihara H, Maechler P, Gjinovci A, Herrera PL, Wollheim CB: Islet -cell
secretion determines glucagon release from neighbouring -cells. Nat Cell
Biol 5:330 335, 2003
37. Antunes CM, Salgado AP, Rosario LM, Santos RM: Differential patterns of
glucose-induced electrical activity and intracellular calcium responses in
single mouse and rat pancreatic islets. Diabetes 49:2028 2038, 2000
38. Berts A, Gylfe E, Hellman B: Ca
2
oscillations in pancreatic islet cells
secreting glucagon and somatostatin. Biochem Biophys Res Commun
208:644 649, 1995
39. Berts A, Gylfe E, Hellman B: Cytoplasmic Ca
2
in glucagon-producing
pancreatic -cells exposed to carbachol and agents affecting Na
uxes.
Endocrine 6:79 83, 1997
40. Gromada J, Hoy M, Buschard K, Salehi A, Rorsman P: Somatostatin
inhibits exocytosis in rat pancreatic -cells by G
i2
-dependent activation of
calcineurin and depriming of secretory granules. J Physiol 535:519 532,
2001
41. Cejvan K, Coy DH, Efendic S: Intra-islet somatostatin regulates glucagon
release via type 2 somatostatin receptors in rats. Diabetes 52:1176 1181,
2003
42. Brice NL, Varadi A, Ashcroft SJ, Molnar E: Metabotropic glutamate
and GABA
B
receptors contribute to the modulation of glucose-stimu
-
lated insulin secretion in pancreatic -cells. Diabetologia 45:242252,
2002
43. Go¨ pel SO, Kanno T, Barg S, Weng XG, Gromada J, Rorsman P: Regulation
of glucagon release in mouse -cells by K
ATP
channels and inactivation of
TTX-sensitive Na
channels. J Physiol 528:509 520, 2000
A. WENDT AND ASSOCIATES
DIABETES, VOL. 53, APRIL 2004 1045
... This effect contrasts with the known inhibitory action of GABA in the CNS and is due to the differential expression of chloride cation cotransporters in the two tissues [7]. γ-Aminobutyric acid also has an inhibitory effect on the secretion of glucagon from pancreatic alpha cells via the GABA A receptor and insulin acts in a paracrine manner to upregulate the expression of these receptors on the alpha cell surface [8,9]. Glucagon works counter to insulin when conditions necessitate an increase in blood glucose by stimulating gluconeogenic pathways. ...
... Although it was not the expected result, the overall reduction in plasma GABA during heat stress could be indicative of an increase in receptor populations and, thus, greater uptake by the pancreas and other GABA-responsive tissues. Previous work demonstrated that GABA A receptor subunits are not found in substantial quantities on the surface of beta cells [8], which could suggest that GABA does not have much impact on the release of insulin from the pancreas at physiological concentrations. If heat stress causes an increase in receptor numbers within the pancreas, the increased sensitivity to available GABA could be a mechanism driving the increase in insulin during heat stress. ...
... The relationship between blood glucose and plasma GABA observed in this study is of particular interest going forward. While the precise relationship between GABA and pancreatic beta cells is somewhat unclear, there is substantial unified evidence that GABA A receptors exist on the surface of alpha cells and allow GABA to mediate an inhibitory effect on glucagon release [8,9,42]. Additionally, Xu and co-workers [9] provided evidence that insulin induces GABA A receptor translocation to the alpha cell membrane and that this is the mechanism by which insulin can suppress glucagon release. ...
Article
Full-text available
Heat-stressed lactating dairy cattle exhibit unique metabolic symptoms, many of which are undoubtedly involved in heat-induced subfertility. Because of its known systemic effects, we hypothesized that γ-aminobutyric acid (GABA) participates in the regulation of insulin and progesterone during heat stress. Multiparous lactating Holstein cows (n = 6) were studied during four experimental periods: (1) thermoneutral (TN; d 1–5), (2) TN + hyperinsulinemic–hypoglycemic clamp (d 6–10), (3) heat stress (HS; d 16–20), and (4) HS + euglycemic clamp (d 21–25). Blood samples were collected once daily via coccygeal venipuncture into heparinized evacuated tubes. Analysis of GABA concentrations from all four treatment periods yielded no differences. In direct comparison to TN concentrations, plasma GABA tended to decrease during the HS period (16.57 ± 2.64 vs. 13.87 ± 2.28 ng/mL, respectively, p = 0.06). Both milk production and plasma insulin were moderately correlated with plasma GABA (r = 0.35, p < 0.01; r = −0.32, p < 0.01). Plasma progesterone was correlated with plasma GABA concentrations during TN but not HS periods. These results are the first to indicate that peripheral GABA could be involved in the regulation of factors known to affect production and reproduction during heat stress. More research is needed to determine its precise role(s).
... This hyperpolarization deviates from the direction in which the action potential occurs, thereby inhibiting cell activity. On the contrary, when positively charged ions from the extracellular fluid enter the cells, the cells will depolarize and thus get excited (Wendt et al. 2004). After a meal, the elevated blood glucose level stimulates β-cells to produce insulin, which in turn activates protein kinase B (Akt) within the cells (Rahman et al. 2021). ...
... Additionally, GABA produced by β-cells diffuses to the surface of α-cells through GABA transporters (GAT) and activates GABA A R on the cell membranes. This results in an influx of Cl − , causing hyperpolarization of α-cells (Wendt et al. 2004 Hypothetical mechanism of GaBa action on α-and β-cells. GaBa increases the influx of Ca 2+ in β-cells and depolarizes the cell membranes, which may activate Ca 2+ -dependent Pi3K/aKt signaling pathway followed by inducing the secretion of insulin and the biosynthesis GaBa in β-cells. ...
... The GABA A R is a receptor expressed on the surface of the α-cell membranes, and its activation is associated with the Cl − channel on the cell membrane surface. Studies have found that GABA produced by β-cells can diffuse to the surface of α-cells, thereby activating the GABA A R on their surface and subsequently inhibiting glucagon secretion (Wendt et al. 2004). In a study by Wendt et al. (2004), islet cells were treated with SR95531 (a GABA A R antagonist), resulting in a 2-3 fold increase in glucagon secretion compared to the cells without SR95531 treatment. ...
Article
Diabetes is a metabolic disease due to impaired or defective insulin secretion and is considered one of the most serious chronic diseases worldwide. Gamma-aminobutyric acid (GABA) is a naturally occurring non-protein amino acid commonly present in a wide range of foods. A number of studies documented that GABA has good anti-diabetic potential. This review summarized the available dietary sources of GABA as well as animal and human studies on the anti-diabetic properties of GABA, while also discussing the underlying mechanisms. GABA may modulate diabetes through various pathways such as inhibiting the activities of α-amylase and α-glucosidase, promoting β-cell proliferation, stimulating insulin secretion from β-cells, inhibiting glucagon secretion from α-cells, improving insulin resistance and glucose tolerance, and increasing antioxidant and anti-inflammatory activities. However, further mechanistic studies on animals and human are needed to confirm the therapeutic effects of GABA against diabetes.
... This effect 2 contrasts with the known inhibitory action of GABA in the CNS and is due to the differential expression of chloride cation cotransporters in the two tissues [7]. γ-Aminobutyric acid also has an inhibitory effect on the secretion of glucagon from pancreatic alpha cells via the GABAA receptor and insulin acts in a paracrine manner to upregulate the expression of these receptors on the alpha cell surface [8][9]. Glucagon works counter to insulin when conditions necessitate an increase in blood glucose by stimulating gluconeogenic pathways. ...
... The relationship between blood glucose and plasma GABA observed in this study is of particular interest going forward. While the precise relationship between GABA and pancreatic beta cells is somewhat unclear, there is substantial unified evidence that GABAA receptors exist on the surface of alpha cells and allow GABA to mediate an inhibitory effect on glucagon release [8][9]42]. Additionally, Xu and co-workers [9] provided evidence that insulin induces GABAA receptor translocation to the alpha cell membrane and that this is the mechanism by which insulin can suppress glucagon release. ...
Preprint
Full-text available
Heat-stressed lactating dairy cattle exhibit unique metabolic symptoms; many of which are undoubtedly involved in heat-induced subfertility. Because of its known systemic effects, we hypothesized that γ-aminobutyric acid (GABA) participates in the regulation of insulin and progesterone during heat stress. Multiparous lactating Holstein cows (n=6) were studied during four experimental periods: 1) thermoneutral (TN; d 1-5), 2) TN + hyperinsulinemic-hypoglycemic clamp (d 6-10), 3) heat stress (HS; d 16-20), and 4) HS + euglycemic clamp (d 21-25). Blood samples were collected once daily via coccygeal venipuncture into heparinized evacuated tubes. Analysis of GABA concentrations from all four treatment periods yielded no differences. In direct comparison to TN concentrations, plasma GABA tended to be decreased during HS (16.57±2.64 vs 13.87±2.28 ng/ml, respectively, P = 0.06). Both milk production and plasma insulin were moderately correlated with plasma GABA (r=0.35, P<0.01; r = -0.32, P<0.01). Plasma progesterone was correlated with plasma GABA concentrations during TN but not HS. These results are the first to indicate that peripheral GABA could be involved in regulating aspects of production and reproduction during heat stress. More research is needed to determine its precise role(s).
... Glucose is an inhibitor of glucagon secretion that regulates α-cell activity through both paracrine and intrinsic mechanisms. At elevated glucose concentrations, β-cell activation suppress glucagon secretion by releasing several paracrine factors that include insulin, GABA, zinc, and serotonin [21,[24][25][26][27]. Insulin, despite reducing α-cell cAMP immunofluorescence [21], was later found to reduce glucagon secretion independently of cAMP [28,29], while UCN3 that is coreleased with insulin suppresses α-cell cAMP by stimulating somatostatin release from δ-cells [21,30,31]. ...
Preprint
Full-text available
Objective Pancreatic islets are nutrient sensors that regulate organismal blood glucose homeostasis. Glucagon release from the pancreatic α-cell is important under fasted, fed, and hypoglycemic conditions, yet metabolic regulation of α-cells remains poorly understood. Here, we identified a previously unexplored role for physiological levels of leucine, which is classically regarded as a β-cell fuel, in the intrinsic regulation of α-cell glucagon release. Methods GcgCre ERT :CAMPER and GcgCre ERT :GCaMP6s mice were generated to perform dynamic, high-throughput functional measurements of α-cell cAMP and Ca ²⁺ within the intact islet. Islet perifusion assays were used for simultaneous, time-resolved measurements of glucagon and insulin release from mouse and human islets. The effects of leucine were compared with glucose and the mitochondrial fuels 2-aminobicyclo(2,2,1)heptane-2-carboxylic acid (BCH, non-metabolized leucine analog that activates glutamate dehydrogenase), α-ketoisocaproate (KIC, leucine metabolite), and methyl-succinate (complex II fuel). CYN154806 (Sstr2 antagonist), diazoxide (K ATP activator, which prevents Ca ²⁺ -dependent exocytosis from α, β, and δ-cells), and dispersed α-cells were used to inhibit islet paracrine signaling and identify α-cell intrinsic effects. Results Mimicking the effect of glucose, leucine strongly suppressed amino acid-stimulated glucagon secretion. Mechanistically, leucine dose-dependently reduced α-cell cAMP at physiological concentrations, with an IC 50 of 57, 440, and 1162 μM at 2, 6, and 10 mM glucose, without affecting α-cell Ca ²⁺ . Leucine also reduced α-cell cAMP in islets treated with Sstr2 antagonist or diazoxide, as well as dispersed α-cells, indicating an α-cell intrinsic effect. The effect of leucine was matched by KIC and the glutamate dehydrogenase activator BCH, but not methyl-succinate, indicating a dependence on mitochondrial anaplerosis. Glucose, which stimulates anaplerosis via pyruvate carboxylase, had the same suppressive effect on α-cell cAMP but with lower potency. Similarly to mouse islets, leucine suppressed glucagon secretion from human islets under hypoglycemic conditions. Conclusions These findings highlight an important role for physiological levels of leucine in the metabolic regulation of α-cell cAMP and glucagon secretion. Leucine functions primarily through an α-cell intrinsic effect that is dependent on glutamate dehydrogenase, in addition to the well-established α-cell regulation by β/δ-cell paracrine signaling. Our results suggest that mitochondrial anaplerosis-cataplerosis facilitates the glucagonostatic effect of both leucine and glucose, which cooperatively suppress α-cell tone by reducing cAMP. Graphical Abstract Highlights Leucine inhibits glucagon secretion from mouse and human islets Leucine suppresses α-cell cAMP via both direct and paracrine effects Anaplerosis via glutamate dehydrogenase is sufficient to suppress α-cell cAMP Leucine suppresses α-cell cAMP and glucagon secretion more potently than glucose
... So far, the most accepted hypothesis seems to be autocrine regulation via the release (or co-secretion with insulin) of GABA through the stimulation of either GABA-A or GABA-B receptors [8,9] (See Figures 1 and 2 of reference [7] for a schematic and immunostaining images of GAD65 and GABA in islets and B-cells). Additionally, a possible mechanism involving paracrine regulation of glucagon secretion by GABA, which is co-secreted with insulin, through the activation of GABAA receptors of α-cells has been postulated [10]. However, in this paper, we limit our review to experimental data on the participation of intracellular GABA in islet metabolism. ...
Article
Full-text available
The stimulus-secretion coupling of a glucose-induced release is generally attributed to the metabolism of the hexose in the β-cells in the glycolytic pathway and the citric acid cycle. Glucose metabolism generates an increased cytosolic concentration of ATP and of the ATP/ADP ratio that closes the ATP-dependent K+-channel at the plasma membrane. The resultant depolarization of the β-cells opens voltage-dependent Ca2+-channels at the plasma membrane that triggers the exocytosis of insulin secretory granules. The secretory response is biphasic with a first and transient peak followed by a sustained phase. The first phase is reproduced by a depolarization of the β-cells with high extracellular KCl maintaining the KATP-channels open with diazoxide (triggering phase); the sustained phase (amplifying phase) depends on the participation of metabolic signals that remain to be determined. Our group has been investigating for several years the participation of the β-cell GABA metabolism in the stimulation of insulin secretion by three different secretagogues (glucose, a mixture of L-leucine plus L-glutamine, and some branched chain alpha-ketoacids, BCKAs). They stimulate a biphasic secretion of insulin accompanied by a strong suppression of the intracellular islet content of gamma-aminobutyric acid (GABA). As the islet GABA release simultaneously decreased, it was concluded that this resulted from an increased GABA shunt metabolism. The entrance of GABA into the shunt is catalyzed by GABA transaminase (GABAT) that transfers an amino group between GABA and alpha-ketoglutarate, resulting in succinic acid semialdehyde (SSA) and L-glutamate. SSA is oxidized to succinic acid that is further oxidized in the citric acid cycle. Inhibitors of GABAT (gamma-vinyl GABA, gabaculine) or glutamic acid decarboxylating activity (GAD), allylglycine, partially suppress the secretory response as well as GABA metabolism and islet ATP content and the ATP/ADP ratio. It is concluded that the GABA shunt metabolism contributes together with the own metabolism of metabolic secretagogues to increase islet mitochondrial oxidative phosphorylation. These experimental findings emphasize that the GABA shunt metabolism is a previously unrecognized anaplerotic mitochondrial pathway feeding the citric acid cycle with a β-cell endogenous substrate. It is therefore a postulated alternative to the proposed mitochondrial cataplerotic pathway(s) responsible for the amplification phase of insulin secretion. It is concluded the new postulated alternative suggests a possible new mechanism of β-cell degradation in type 2 (perhaps also in type 1) diabetes.
... However, abnormal glucagon secretion is sometimes observed in diabetes mellitus 24 , and its regulatory mechanism is not fully understood. Glucagon is not only regulated by α-cell-intrinsic glucose sensing but is precisely controlled by the other regulatory mechanisms, including various nutrients, autonomic nerves, the endocrine system, and also the paracrine β-cell effects of insulin [25][26][27] , GABA 28,29 , and Zn 2+24,30 in the islets. Especially, a paracrine effect between β-cell GLS2/insulin and α-cell glucagon might exist. ...
Article
Full-text available
Glutaminase 2 (GLS2), a master regulator of glutaminolysis that is induced by p53 and converts glutamine to glutamate, is abundant in the liver but also exists in pancreatic β-cells. However, the roles of GLS2 in islets associated with glucose metabolism are unknown, presenting a critical issue. To investigate the roles of GLS2 in pancreatic β-cells in vivo, we generated β-cell-specific Gls2 conditional knockout mice (Gls2 CKO), examined their glucose homeostasis, and validated the findings using a human islet single-cell analysis database. GLS2 expression markedly increased along with p53 in β-cells from control (RIP-Cre) mice fed a high-fat diet. Furthermore, Gls2 CKO exhibited significant diabetes mellitus with gluconeogenesis and insulin resistance when fed a high-fat diet. Despite marked hyperglycaemia, impaired insulin secretion and paradoxical glucagon elevation were observed in high-fat diet-fed Gls2 CKO mice. GLS2 silencing in the pancreatic β-cell line MIN6 revealed downregulation of insulin secretion and intracellular ATP levels, which were closely related to glucose-stimulated insulin secretion. Additionally, analysis of single-cell RNA-sequencing data from human pancreatic islet cells also revealed that GLS2 expression was elevated in β-cells from diabetic donors compared to nondiabetic donors. Consistent with the results of Gls2 CKO, downregulated GLS2 expression in human pancreatic β-cells from diabetic donors was associated with significantly lower insulin gene expression as well as lower expression of members of the insulin secretion pathway, including ATPase and several molecules that signal to insulin secretory granules, in β-cells but higher glucagon gene expression in α-cells. Although the exact mechanism by which β-cell-specific GLS2 regulates insulin and glucagon requires further study, our data indicate that GLS2 in pancreatic β-cells maintains glucose homeostasis under the condition of hyperglycaemia.
Article
Glucagon's ability to promote hepatic glucose production has been known for over a century with initial observations touting this hormone as a diabetogenic agent. However, glucagon receptor agonism (when balanced with an incretin, including glucagon-like peptide 1 (GLP-1) to dampen glucose excursions) is now being developed as a promising therapeutic target in the treatment of metabolic diseases, like metabolic dysfunction-associated steatotic disease/metabolic dysfunction-associated steatohepatitis (MASLD/MASH), and may also have benefit for obesity and chronic kidney disease. Conventionally regarded as the opposing tag-team partner of the anabolic mediator insulin, glucagon is gradually emerging as more than just a 'catabolic hormone'. Glucagon action on glucose homeostasis within the liver has been well characterized. However, growing evidence, in part thanks to new and sensitive 'omics' technologies, has implicated glucagon as more than just a 'glucose liberator'. Elucidation of glucagon's capacity to increase fatty acid oxidation while attenuating endogenous lipid synthesis speaks to the dichotomous nature of the hormone. Furthermore, glucagon action is not limited to just glucose homeostasis and lipid metabolism, as traditionally reported. Glucagon plays key regulatory roles in hepatic amino acid and ketone body metabolism, as well as mitochondrial turnover and function, indicating broader glucagon signaling consequences for metabolic homeostasis mediated by the liver. Here we examine the broadening role of glucagon signaling within the hepatocyte and question the current dogma, to appreciate glucagon as more than just that 'catabolic hormone'.
Article
Long lagging behind insulin, glucagon research has caught up in large part thanks to technological breakthroughs. Here we review how the field was propelled by the development of novel techniques and approaches. The glucagon radioimmunoassay and islet isolation are methods that now seem trivial, but for decades they were crucial in defining the biology of the pancreatic alpha cell and the role of glucagon in glucose homeostasis. More recently, mouse models have become the main workhorse of this research effort, if not of biomedical research in general. The mouse model allowed detailed mechanistic studies that are revealing alpha cell functions beyond its canonical glucoregulatory role. A recent profusion of gene expression and transcription regulation studies is providing new vistas into what constitutes alpha cell identity. In particular, the combination of transcriptomic techniques with functional recordings promises to move molecular guesswork into real time physiology. The challenge right now is not to get enamored with these powerful techniques and to make sure that the research continues to be transformative and paradigm shifting. We should imagine a future in which the biology of the alpha cell will be studied at single cell resolution, non-invasively, and in real time in the human body.
Article
Full-text available
Glucagon is the principal glucose-elevating hormone that forms the first-line defence against hypoglycaemia. Along with insulin, glucagon also plays a key role in maintaining systemic glucose homeostasis. The cells that secrete glucagon, pancreatic α-cells, are electrically excitable cells and use electrical activity to couple its hormone secretion to changes in ambient glucose levels. Exactly how glucose regulates α-cells has been a topic of debate for decades but it is clear that electrical signals generated by the cells play an important role in glucagon secretory response. Decades of studies have already revealed the key players involved in the generation of these electrical signals and possible mechanisms controlling them to tune glucagon release. This has offered the opportunity to fully understand the enigmatic α-cell physiology. In this review, we describe the current knowledge on cellular electrophysiology and factors regulating excitability, glucose sensing, and glucagon secretion. We also discuss α-cell pathophysiology and the perspective of addressing glucagon secretory defects in diabetes for developing better diabetes treatment, which bears the hope of eliminating hypoglycaemia as a clinical problem in diabetes care.
Article
Full-text available
Application of GABA to rat neocortical neurons maintained in cell culture produced a response that declined over several seconds, even in the continued presence of agonist. The decrement could be attributed to both a redistribution of Cl- and a true decline in GABA-induced membrane conductance, or desensitization. The extent and rate of desensitization were dose dependent in a manner similar to the dose dependence of the GABA-induced current, but were not related to the absolute magnitude of the current or to the charge transfer. Bicuculline slowed desensitization while diazepam enhanced the rate of desensitization, consistent with a localization of desensitization to the agonist-receptor binding site. When measured in the whole-cell recording mode, desensitization was voltage dependent, becoming much slower as the membrane was depolarized. Changes in extracellular or intracellular [Ca2+] did not appear to grossly affect the desensitization process or its voltage dependence. GABA-activated single channels, recorded in the outside-out configuration, also desensitized in the continued presence of agonist. However, desensitization differed from that seen in the same neurons in the whole-cell mode. Desensitization was considerably more rapid and did not show any voltage sensitivity. Moreover, single-channel responses often failed to recover after only a few exposures to agonist. Desensitization of GABA responses may play a role in the regulation of cortical inhibition, especially under conditions of intense excitatory and inhibitory synaptic activation.
Article
Full-text available
Inhibition of pancreatic glucagon secretion during hyperglycemia could be mediated by (a) glucose, (b) insulin, (c) somatostatin, or (d) glucose in conjunction with insulin. To determine the role of these factors in the mediation of glucagon suppression, we injected alloxan while clamping the arterial supply of the pancreatic splenic lobe of dogs, thus inducing insulin deficiency localized to the ventral lobe and avoiding hyperglycemia. Ventral lobe insulin, glucagon, and somatostatin outputs were then measured in response to a stepped IV glucose infusion. In control dogs glucagon suppression occurred at a glucose level of 150 mg/dl and somatostatin output increased at glucose greater than 250 mg/dl. In alloxan-treated dogs glucagon output was not suppressed nor did somatostatin output increase. We concluded that insulin was required in the mediation of glucagon suppression and somatostatin stimulation. Subsequently, we infused insulin at high rates directly into the artery that supplied the beta cell-deficient lobe in six alloxan-treated dogs. Insulin infusion alone did not cause suppression of glucagon or stimulation of somatostatin; however, insulin repletion during glucose infusions did restore the ability of hyperglycemia to suppress glucagon and stimulate somatostatin. We conclude that intra-islet insulin permits glucose to suppress glucagon secretion and stimulate somatostatin during hyperglycemia.
Article
Full-text available
GABA, a major inhibitory neurotransmitter of the brain, is also present at high concentration in pancreatic islets. Current evidence suggests that within islets GABA is secreted from beta-cells and regulates the function of mantle cells (alpha- and delta-cells). In the nervous system GABA is stored in, and secreted from, synaptic vesicles. The mechanism of GABA secretion from beta-cells remains to be elucidated. Recently the existence of synaptic-like microvesicles has been demonstrated in some peptide-secreting endocrine cells. The function of these vesicles is so far unknown. The proposed paracrine action of GABA in pancreatic islets makes beta-cells a useful model system to explore the possibility that synaptic-like microvesicles, like synaptic vesicles, are involved in the storage and release of non-peptide neurotransmitters. We report here the presence of synaptic-like microvesicles in beta-cells and in beta-cells. Some beta-cells in culture were found to extend neurite-like processes. When these were present, synaptic-like microvesicles were particularly concentrated in their distal portions. The GABA synthesizing enzyme, glutamic acid decarboxylase (GAD), was found to be localized around synaptic-like microvesicles. This was similar to the localization of GAD around synaptic vesicles in GABA-secreting neurons. GABA immunoreactivity was found to be concentrated in regions of beta-cells which were enriched in synaptic-like microvesicles. These findings suggest that in beta-cells synaptic-like microvesicles are storage organelles for GABA and support the hypothesis that storage of non-peptide signal molecules destined for secretion might be a general feature of synaptic-like microvesicles of endocrine cells.
Article
γ-Aminobutyric acid (GABA), a prominent inhibitory neurotransmitter, is present in high concentrations in β-cells of islets of Langerhans. The GABA shunt enzymes, glutamate decarboxylase (GAD) and GABA transaminase (GABA-T), have also been localized in islet β-cells. With the recent demonstration that the 64,000-Mr antigen associated with insulin-dependent diabetes mellitus is GAD, there isincreased interest in understanding the role of GABA in islet function. Only a small component of β-cell GABA is contained in insulin secretory granules, making it unlikely that GABA, co-released with insulin, is physiologically significant. Our immunohistochemical study of GABA in β-cells of intact islets indicates that GABA is associated with a vesicular compartment distinctly different from insulin secretory granules. Whether this compartment represents a releasable pool of GABA has yet to be determined. GAD in β-cells is associated with a vesicular compartment, similar to the GABA vesicles. In addition, GAD is found in a unique extensive tubular cisternal complex (GADcomplex). It islikely that the GABA-GAD vesicles are derived from this GAD-containing complex. Physiological studies on the effect of extracellular GABA on islet hormonal secretion have had variable results. Effects of GABA on insulin, glucagon, and somatostatin secretion have been proposed. The most compelling evidence for GABA regulation of islet hormone secretion comes from studies on somatostatin secretion, where it has an inhibitory effect. We present new evidence demonstrating the presence of GABAergic nerve cell bodies at the periphery of islets with numerous GABA-containing processes extending into the islet mantle. This close association between GABAergic neurons and islet α- and δ-cells strongly suggests that GABA inhibition of somatostatin and glucagon secretionis mediated by these neurons. Intracellular β-cell GABAA and its metabolismmay have a role in β-cell function. New evidence indicates that GABA shunt activity isinvolved in regulation of insulin secretion. In addition, GABA or its metabolites may regulate proinsulin synthesis. These new observations provide insight into the complex nature of GABAergic neurons and β-cell GABA in regulation of islet function.
Article
a1 and ß1 subunits of human GABAA receptors were expressed in Sf9 cells using the Sf9-baculovirus system. Better expression was obtained by manipulating the system. Cell growth phase at the time of infection determined the practical range of virus titre, the period postinfection during which cells were useful for signal detection and the maximal current obtained. Cells in the early exponential phase were relatively insensitive to multiplicity of infection (MOI) whereas cells in the midto late-exponential phase were highly dependent on MOI and they responded with the largest Cl-} current generated by GABA. Channels activated by GABA were chloride-selective. Half the maximum peak whole-cell current was obtained with 11 µm GABA. The time course of Cl-} currents activated by saturating GABA concentrations in cells infected with a1ß1recombinant viruses was examined employing a rapid perfusion system which allowed whole-cell solution exchange in less than 1 msec. The current decay could be fitted by 3 to 4 exponentials for the first 8 sec. The initial fast current decrease had a time constant of about 23 msec. No voltage dependence of time constants was detected but the whole-cell IV relation showed outward rectification. Currents were depressed by bicuculline, penicillin and picrotoxin and potentiated by pentobarbitone.
Article
• Measurements of cell capacitance were used to investigate the molecular mechanisms by which somatostatin inhibits Ca2+-induced exocytosis in single rat glucagon-secreting pancreatic α-cells. • Somatostatin decreased the exocytotic responses elicited by voltage-clamp depolarisations by 80 % in the presence of cyclic AMP-elevating agents such as isoprenaline and forskolin. Inhibition was time dependent and half-maximal within 22 s. • The inhibitory action of somatostatin was concentration dependent with an IC50 of 68 nmand prevented by pretreatment of the cells with pertussis toxin. The latter effect was mimicked by intracellular dialysis with specific antibodies to Gi1/2 and by antisense oligonucleotides against G proteins of the subtype Gi2. • Somatostatin lacked inhibitory action when applied in the absence of forskolin or in the presence of the L-type Ca2+ channel blocker nifedipine. The size of the -conotoxin-sensitive and forskolin-independent component of exocytosis was limited to 60 fF. By contrast, somatostatin abolished L-type Ca2+ channel-dependent exocytosis in α-cells exposed to forskolin. The magnitude of the latter pool amounted to 230 fF. • The inhibitory effect of somatostatin on exocytosis was mediated by activation of the serine/threonine protein phosphatase calcineurin and was prevented by pretreatment with cyclosporin A and deltamethrin or intracellularly applied calcineurin autoinhibitory peptide. Experiments using the stable ATP analogue AMP-PCP indicate that somatostatin acts by depriming of granules. • We propose that somatostatin receptors associate with L-type Ca2+ channels and couple to Gi2 proteins leading to a localised activation of calcineurin and depriming of secretory granules situated close to the L-type Ca2+ channels.
Article
Certain aspects of radio-immunoassay of somatostatin from isolated rat pancreatic islets are described. Somatostatin-14, and not somatostatin-28, is secreted from isolated rat pancreatic islets. Less somatostatin secretion is measured per islet owing to purity of tracer in the radio-immunoassay. Theophylline apparently cross-reacts with somatostatin in the assay described, and this has to be taken into consideration when studying somatostatin release induced by theophylline in isolated islets.
Article
The gamma-aminobutyric acid (GABA) concentration of pancreatic islets in rats treated with streptozotocin (STZ) and of human insulinoma tissue was studied. Seven hours after the administration of 65 mg/kg body weight of STZ, a distinct increase in serum insulin concentration and at the same time a decrease in blood glucose level were seen. Twenty-four hours after the injection of STZ, however, the level of serum insulin decreased much, whereas that of blood glucose increased considerably. On the other hand, the GABA concentration of the islet was reduced dramatically to about one-tenth the control level after both 7 and 24 h. The histologic investigations of the islets revealed the destruction of B cells but no changes in A and D cells 7 and 24 h after the treatment of STZ. Nerve fibers and nerve endings in the islets were preserved intact all through the study. The GABA and insulin contents of the two cases of human insulinoma were determined. One insulinoma, which was compactly occupied with B cells according to its histologic features, contained a high concentration of GABA. The other tumor, having a rather sparse distribution of B cells in it as compared with the former case, possessed a lower concentration of GABA, but it was still high compared with that of its surrounding tissues. The present observations indicate that a large amount of GABA is available in the B cells of the pancreatic islets.
Article
The present study was aimed at localizing gamma-aminobutyric acid (GABA) and its enzyme of synthesis, glutamic acid decarboxylase (GAD), in the mouse pancreas by immunocytochemical methods. The influence of GABA on hormone release was also studied with normal mouse and rat islets and the isolated perfused rat pancreas. Particular attention was paid to glucagon release to test a recent hypothesis suggesting that GABA mediates the still unexplained glucose-induced inhibition of glucagon release. GABA and GAD were identified only in islet cells and never in the exocrine tissue. Exogenous GABA, baclofen (agonist of GABAB receptors), muscimol (agonist of GABAA receptors), or bicuculline (antagonist of GABAA receptors) did not affect insulin and somatostatin release by isolated mouse or rat islets. GABA was also without effect on glucose-induced electrical activity in mouse B-cells. Glucagon secretion by mouse islets was only slightly inhibited (approximately 20%) by GABA. Since muscimol had a similar effect, and baclofen was ineffective, the inhibition by GABA probably involves GABAA receptor activation. Bicuculline, however, did not antagonize the inhibitory effects of GABA and muscimol, probably because the antagonist alone also decreased glucagon secretion. In contrast to GABA, low (3 mM) and high (20 mM) concentrations of glucose strongly inhibited (approximately 50-65%) glucagon release; this inhibition was not prevented by bicuculline. Similar results were obtained with the perfused rat pancreas; muscimol slightly inhibited glucagon release under various conditions, and bicuculline did not reverse the strong inhibition produced by 16.7 mM glucose. In conclusion, GABA does not affect insulin and somatostatin secretion, but inhibits A-cells, probably by acting on GABAA receptors. It is unlikely, however, that this small inhibitory effect can account for the inhibition of glucagon release produced by glucose.
Article
gamma-Aminobutyric acid (GABA), a prominent inhibitory neurotransmitter, is present in high concentrations in beta-cells of islets of Langerhans. The GABA shunt enzymes, glutamate decarboxylase (GAD) and GABA transaminase (GABA-T), have also been localized in islet beta-cells. With the recent demonstration that the 64,000-M, antigen associated with insulin-dependent diabetes mellitus is GAD, there is increased interest in understanding the role of GABA in islet function. Only a small component of beta-cell GABA is contained in insulin secretory granules, making it unlikely that GABA, coreleased with insulin, is physiologically significant. Our immunohistochemical study of GABA in beta-cells of intact islets indicates that GABA is associated with a vesicular compartment distinctly different from insulin secretory granules. Whether this compartment represents a releasable pool of GABA has yet to be determined. GAD in beta-cells is associated with a vesicular compartment, similar to the GABA vesicles. In addition, GAD is found in a unique extensive tubular cisternal complex (GAD complex). It is likely that the GABA-GAD vesicles are derived from this GAD-containing complex. Physiological studies on the effect of extracellular GABA on islet hormonal secretion have had variable results. Effects of GABA on insulin, glucagon, and somatostatin secretion have been proposed. The most compelling evidence for GABA regulation of islet hormone secretion comes from studies on somatostatin secretion, where it has an inhibitory effect. We present new evidence demonstrating the presence of GABAergic nerve cell bodies at the periphery of islets with numerous GABA-containing processes extending into the islet mantle. This close association between GABAergic neurons and islet alpha- and delta-cells strongly suggests that GABA inhibition of somatostatin and glucagon secretion is mediated by these neurons. Intracellular beta-cell GABAA and its metabolism may have a role in beta-cell function. New evidence indicates that GABA shunt activity is involved in regulation of insulin secretion. In addition, GABA or its metabolites may regulate proinsulin synthesis. These new observations provide insight into the complex nature of GABAergic neurons and beta-cell GABA in regulation of islet function.