ArticlePDF Available

Glucose promotes pancreatic islet β-cell survival through a PI 3-kinase/Akt-signaling pathway

Authors:

Abstract and Figures

The concentration of glucose in plasma is an important determinant of pancreatic beta-cell mass, whereas the relative contributions of hypertrophy, proliferation, and cell survival to this process are unclear. Glucose results in depolarization and subsequent calcium influx into islet beta-cells. Because depolarization and calcium (Ca(2+)) influx promote survival of neuronal cells, we hypothesized that glucose might alter survival of islet beta-cells through a similar mechanism. In the present studies, cultured mouse islet beta-cells showed a threefold decrease in apoptosis under conditions of 15 mM glucose compared with 2 mM glucose (P < 0.05). MIN6 insulinoma cells incubated in 25 mM glucose for 24 h showed a threefold decrease in apoptosis compared with cells in 5 mM glucose (1.7 +/- 0.2 vs. 6.3 +/- 1%, respectively, P < 0.001). High glucose (25 mM) enhanced survival-required depolarization and Ca(2+) influx and was blocked by phosphatidylinositol (PI) 3-kinase inhibitors. Glucose activation of the protein kinase Akt was demonstrated in both insulinoma cells and cultured mouse islets by means of an antibody specific for Ser(473) phospho-Akt and by an in vitro Akt kinase assay. Akt phosphorylation was dependent on PI 3-kinase but not on MAPK. Transfection of insulinoma cells with an Akt kinase-dead plasmid (Akt-K179M) resulted in loss of glucose-mediated protection, whereas transfection with a constitutively active Akt enhanced survival in glucose-deprived insulinoma cells. The results of these studies defined a novel pathway for glucose-mediated activation of a PI 3-kinase/Akt survival-signaling pathway in islet beta-cells. This pathway may provide important targets for therapeutic intervention.
Content may be subject to copyright.
Glucose promotes pancreatic islet -cell survival
through a PI 3-kinase/Akt-signaling pathway
SHANTHI SRINIVASAN,
1
ERNESTO BERNAL-MIZRACHI,
2
MITSURU OHSUGI,
2
AND MARSHALL ALAN PERMUTT
2
Divisions of
1
Gastroenterology and
2
Endocrinology, Diabetes and Metabolism, Department of
Internal Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
Received 29 April 2002; accepted in final form 17 June 2002
Srinivasan, Shanthi, Ernesto Bernal-Mizrachi, Mit-
suru Ohsugi, and Marshall Alan Permutt. Glucose pro-
motes pancreatic islet -cell survival through a PI 3-kinase/
Akt-signaling pathway. Am J Physiol Endocrinol Metab 283:
E784–E793, 2002. First published June 25, 2002; 10.1152/
ajpendo.00177.2002.—The concentration of glucose in
plasma is an important determinant of pancreatic -cell
mass, whereas the relative contributions of hypertrophy,
proliferation, and cell survival to this process are unclear.
Glucose results in depolarization and subsequent calcium
influx into islet -cells. Because depolarization and calcium
(Ca
2
) influx promote survival of neuronal cells, we hypoth-
esized that glucose might alter survival of islet -cells
through a similar mechanism. In the present studies, cul-
tured mouse islet -cells showed a threefold decrease in
apoptosis under conditions of 15 mM glucose compared with
2 mM glucose (P0.05). MIN6 insulinoma cells incubated in
25 mM glucose for 24 h showed a threefold decrease in
apoptosis compared with cells in 5 mM glucose (1.7 0.2 vs.
6.3 1%, respectively, P0.001). High glucose (25 mM)
enhanced survival-required depolarization and Ca
2
influx
and was blocked by phosphatidylinositol (PI) 3-kinase inhib-
itors. Glucose activation of the protein kinase Akt was dem-
onstrated in both insulinoma cells and cultured mouse islets
by means of an antibody specific for Ser
473
phospho-Akt and
by an in vitro Akt kinase assay. Akt phosphorylation was
dependent on PI 3-kinase but not on MAPK. Transfection of
insulinoma cells with an Akt kinase-dead plasmid (Akt-
K179M) resulted in loss of glucose-mediated protection,
whereas transfection with a constitutively active Akt en-
hanced survival in glucose-deprived insulinoma cells. The
results of these studies defined a novel pathway for glucose-
mediated activation of a PI 3-kinase/Akt survival-signaling
pathway in islet -cells. This pathway may provide impor-
tant targets for therapeutic intervention.
phosphatidylinositol 3-kinase; apoptosis; islet -cell mass;
depolarization
THE MASS OF INSULIN-PRODUCING islet -cells is deter-
mined by the combined rates of proliferation of existing
cells, by neogenesis, and by cell death (5). For example,
animals that are insulin resistant develop compensa-
tory hypertrophy and hyperplasia. Under such condi-
tions, growth factors and glucose are thought to be
important determinants of islet -cell mass and func-
tion (15). The importance of glucose in these responses
has been suggested by observing that animals sub-
jected to infusions of glucose over several days devel-
oped increased -cell mass (51). Similarly, glucose
treatment of insulinoma cells in culture resulted in
-cell hyperplasia (24). Pancreatic islet -cells are
unique in their responses to physiological changes in
glucose, converting metabolic energy into electrical ac-
tivity (26). Glucose results in closure of the ATP-acti-
vated potassium (K
ATP
) channels, leading to depolar-
ization of -cells and an influx of cytosolic calcium. The
importance of islet -cell depolarization in proper -cell
generation and proliferation was recently demon-
strated by disruption of the L-type calcium channel
1D
subunit in mice (44). Knockout of this calcium channel
subunit resulted in a decrease in the number and size
of the islets due to a decrease in -cell generation.
These results highlighted the significance of depolar-
ization/Ca
2
signaling for normal -cell development.
Sustained Ca
2
overload in neurons is associated
with enhanced apoptosis, yet neurons deprived of de-
polarization and Ca
2
also do not survive (53). This has
been demonstrated by the fact that promotion of sur-
vival of several types of neurons by depolarization can
be prevented by dihydropyridine calcium channel an-
tagonists (10, 17, 18, 34). Neurons and islet -cells
share many biochemical and molecular mechanisms.
We hypothesized that glucose might affect islet -cell
mass through similar mechanisms. Like neurons,
chronic depolarization and Ca
2
overload could result
in apoptosis (42, 49); yet previous studies suggested
that glucose may enhance islet -cell viability. Isolated
rat islet -cells cultured in 10 mM glucose for 1 wk
exhibited marked enhancement of survival relative to
those in 3 mM glucose (23). Another study reported
increasing apoptosis when islet -cells were incubated
in 12 mM glucose for 40 h; yet interestingly, this
study also showed a 80% reduction in apoptosis when
-cells were incubated with glucose increasing from 2
to 12 mM (14). These studies provide preliminary evi-
dence that glucose in the 10–12 mM range, perhaps
Address for reprint requests and other correspondence: S. Srini-
vasan, Washington Univ. School of Medicine, Campus Box 8127, 660
S. Euclid Ave., St. Louis, MO 63110 (E-mail: ssriniva@im.wustl.edu).
The costs of publication of this article were defrayed in part by the
payment of page charges. The article must therefore be hereby
marked ‘‘advertisement’’ in accordance with 18 U.S.C. Section 1734
solely to indicate this fact.
Am J Physiol Endocrinol Metab 283: E784–E793, 2002.
First published June 25, 2002; 10.1152/ajpendo.00177.2002.
0193-1849/02 $5.00 Copyright ©2002 the American Physiological Society http://www.ajpendo.orgE784
by 10.220.33.4 on September 9, 2016http://ajpendo.physiology.org/Downloaded from
through activation of depolarization, may enhance sur-
vival of -cells by mechanisms similar to those ob-
served in neuronal cells.
The purpose of the present study was to attempt to
conrm previous observations on the survival-enhanc-
ing effects of glucose on islet -cells in culture and, if
so, evaluate potential mechanisms. Using cultures of
mouse islets and rodent insulinoma cell lines previ-
ously shown to secrete insulin in response to physio-
logical changes in glucose concentrations (225 mM)
(26), we determined that there were signicant effects
of glucose at 15 mM on survival relative to those in
glucose at 5 mM during 24- to 96-h incubations. To
elucidate the mechanisms of glucose-mediated sur-
vival, we examined signal transduction pathways that
might be involved. The results suggested that glucose
promotes pancreatic islet -cell survival through depo-
larization and subsequent Ca
2
activation of a phos-
phatidylinositol (PI) 3-kinase/Akt survival-signaling
pathway.
EXPERIMENTAL PROCEDURES
Antibodies
We used the following antibodies: Akt, phospho-Akt, and
cleaved caspase-3 antibodies (New England Biolabs); hemag-
glutinin (HA) antibody (Santa Cruz Biotechnologies); Alexa
488 goat anti-rabbit IgG antibody, and Alexa 594 goat anti-
mouse IgG antibody (Molecular Probes); CY3-conjugated Af-
nipure donkey anti-guinea pig antibody (Jackson Immu-
noresearch Lab), and anti-insulin antibody (Biogenex).
Cell Culture
MIN6 cells were cultured in Dulbeccos minimal essential
medium (DMEM) containing 15% FCS and 25 mM glucose
(passage no. 2040). INS-1 cells were cultured in RPMI
containing 10% FCS and 11.2 mM glucose and PC12 cells in
DMEM containing 10% FCS, 5% goat serum, and 25 mM
glucose. For detection of apoptosis, cells were plated at
1,000,000 per coverslip, and experiments were performed
24 h after plating at 7080% conuence. The protocol for
glucose experiments involved washing cells with phosphate-
buffered saline (PBS), followed by incubation for 2496 h
with 240 mM glucose and other reagents such as mannitol,
diazoxide (DZ), PD-98059, wortmannin, LY-294002, and ni-
fedipine.
Isolation of mouse islets was accomplished by collagenase
digestion and Ficoll centrifugation, followed by hand picking
as described (39). Islets were cultured for 24 h in RPMI with
10% serum and 11.2 mM glucose.
Detection of Apoptosis
The TdT-mediated dUTP nick end labeling (TUNEL) tech-
nique was used to detect DNA strand breaks formed during
apoptosis (19). Cells on coverslips were xed with 4% para-
formaldehyde for 45 min at room temperature and then
permeabalized with 1% Triton X-100. After a rinse with PBS,
cells were incubated with uorescein isothiocyanate (FITC)-
labeled dUTP in the presence of enzyme TdT for1hat37°C.
Coverslips were mounted on glass slides in mounting me-
dium containing the counterstain propidium iodide (2.5 g/
ml) and visualized using a uorescent microscope. Five hun-
dred cells were scored (in triplicate) in a blinded fashion to
determine the percentage of TUNEL-positive cells. In RE-
SULTS,nrepresents the number of independent experiments.
Activated caspase-3 was detected by immunohistochemis-
try using an antibody (1:100) to the cleaved caspase-3 (17
kDa) fragment as described (16). Alexa 488 goat anti-rabbit
IgG antibody (1:200) was used as a secondary antibody. Five
hundred cells were scored in a blinded fashion to determine
the percentage of cleaved caspase-3-positive cells by use of a
uorescent microscope.
For detection of apoptosis in cultured islet cells, the islets
were isolated and cultured for 24 h in different glucose
concentrations (2 and 15 mM). After 24 h of incubation, the
islets were dispersed into single cells with dispase (46) and
were spun down with a Cytospin machine onto a slide. Cells
were xed with 4% paraformaldehyde for 45 min at room
temperature and then permeabalized with 1% Triton X-100.
After a rinse with PBS, cells were incubated with FITC-
labeled dUTP in the presence of enzyme TdT for1hat37°C.
After a wash with PBS, cells were incubated with guinea pig
anti-insulin antibody (1:200) for 1 h followed by a 30-min
wash with PBS; nally, cells were incubated with secondary
antibody (CY3-conjugated donkey anti-guinea pig antibody)
for 1 h. After a 30-min wash with PBS, cells were covered in
mounting medium and visualized with a uorescent micro-
scope. No propidium iodide counterstain was used in the
TUNEL method. Staining for insulin as described identied
-cells. Five hundred -cells were scored in a blinded fashion
to determine the percentage of FITC-positive (TUNEL-posi-
tive) -cells.
Detection of Necrosis
MIN6 cells were treated with 5 and 25 mM glucose for
24 h, and necrosis was assessed by trypan blue exclusion.
Five hundred cells were counted in a blinded fashion, and the
number of trypan blue-positive cells determined the percent-
age of necrosis (2).
Western Blot Analysis
MIN6 cells. MIN6 cells were plated in 100-mm dishes at a
seeding density of 5,000,000 cells per dish and cultured in
DMEM for 4 days. At 80% conuence, cells were preincu-
bated in the absence of serum and with 5 mM glucose for
14 h, followed by no serum and no glucose for 4 h. Kinase
inhibitors were added 1 h before the addition of glucose. After
incubation with glucose (5 or 25 mM) for various time inter-
vals, cells were lysed and assayed on an acrylamide gel (10%)
for phospho-Akt as described (1). Protein (40 g) was loaded
per lane. Phospho-Akt was assessed by use of an antibody to
Akt phosphorylated at Ser
473
. The blot was stripped and
reprobed for total Akt.
Isolated islets. Isolated islets were cultured overnight in
complete medium (11.2 mM glucose and 10% FCS). Cells
were incubated in Krebs buffer for 1 h followed by exposure
to 2 or 5 mM glucose for 45 min. After incubation with
glucose (2 and 15 mM) for various time intervals, cells were
lysed and assayed on an acrylamide gel (10%) for phospho-
Akt and total Akt.
Akt Kinase Activity
Measurement of Akt kinase activity was performed using a
kit (Cell Signaling Technology, Beverly, MA). MIN6 cells
were stimulated as described and lysed in cell lysis buffer.
Cell lysates containing 300 g of total protein were immuno-
precipitated with immobilized Akt monoclonal antibody
slurry with gentle rocking for3hat4°C. After two washes
E785GLUCOSE ACTIVATES SURVIVAL IN -CELLS
AJP-Endocrinol Metab VOL 283 OCTOBER 2002 www.ajpendo.org
by 10.220.33.4 on September 9, 2016http://ajpendo.physiology.org/Downloaded from
with cell lysis buffer followed by two washes with kinase
buffer (25 mM Tris, pH 7.5, 5 mM -glycerophosphate, 2 mM
DTT, 0.1 mM Na
3
VO
4
, 10 mM MgCl
2
), the immunoprecipi-
tates were resuspended in 40 l of kinase buffer added with
200 M ATP and 1 g of GSK-3 fusion protein. Samples were
incubated for 30 min at 30°C. The reaction was terminated
with 20 lof3SDS loading buffer, and then samples were
analyzed by immunoblotting with the phospho-GSK-3/
(Ser
21/9
) antibody (38).
Transient Transfection and Detection of Akt
by Immunofluorescence
Akt constructs were a gift of Dr. Philip Stahl (Washington
University School of Medicine). Plasmids containing three
HA-tagged Akt constructs were used: wild-type Akt (Akt-
WT), kinase-dead Akt (Akt-K179M), and a constitutively
active form of Akt (Akt-CA) (33). Transfection efciency was
5%, determined by immunohistochemical detection of the HA
tags of the Akt constructs with an antibody to HA (1:200).
Cells were transfected with 1 g of DNA per dish with the
use of lipofectamine plus reagent under serum-free condi-
tions for 6 h, and then the cells were replenished with
complete medium for 24 h. Next, the cells were cultured for
24 h in different concentrations of glucose. The cells were
xed and assayed for transfection and apoptosis. TUNEL
staining was performed rst, followed by staining for the HA
tag of Akt. No propidium iodide counterstain was used in the
TUNEL method. The TUNEL-positive cells appeared green
and the HA tag appeared red due to the ALEXA 594 second-
ary antibody. Apoptosis in transfected cells was detected by
scoring yellow cells due to overlap of red and green uores-
cence.
Statistical Analysis
Statistical analysis was performed using a two-tailed Stu-
dentst-test. A Pvalue 0.05 was considered signicant.
RESULTS
Effects of Glucose on Survival of Insulinoma Cells
and Cultured Islet Cells
To assess whether glucose affected survival of islet
-cells, apoptosis was measured by the TUNEL method
in cultures of isolated mouse pancreatic islets and
insulinoma cells incubated in various glucose concen-
trations. The effects of glucose were assessed on sur-
vival of primary mouse islet -cells maintained in
culture for 24 h. -Cells were identied using a stain-
ing protocol for insulin in conjunction with the TUNEL
method, as described in EXPERIMENTAL PROCEDURES.
Mouse islet -cells cultured in the presence of 15 mM
glucose had a threefold reduction in apoptosis com-
pared with those cultured in 2 mM glucose [29 9.2 vs.
75 8.9% apoptosis (n4 independent experiments),
P0.03]. In Fig. 1A, the percentage of apoptosis is
shown relative to apoptosis in 15 mM glucose (2 mM
glucose: 258 30.68 vs. 15 mM glucose: 100 31.72,
P0.03). MIN6 insulinoma cells cultured in 5 mM
glucose over 24 h showed 6.7 1% TUNEL-positive
cells at the end of the incubation (n13). In contrast,
cells incubated in 25 mM glucose exhibited a threefold
decrease in TUNEL-positive cells [1.7 0.2% (n11),
P0.001]. In Fig. 1B, the percentage of apoptosis is
shown relative to apoptosis in 5 mM glucose (5 mM
glucose: 370 61.76 vs. 25 mM glucose: 100 17.54,
P0.001). A representative photograph of MIN6 cells
is shown to illustrate TUNEL-positive yellow uores-
cent cells (Fig. 1B). A similar effect of glucose on
apoptosis was also observed in another rodent insulin-
oma cell line, INS-1 [10 mM glucose: 2.69 0.4% (n
6) vs. 2 mM glucose: 6.02 0.4% (n6), P0.001].
Measurement of apoptosis by the TUNEL method
was conrmed by detecting apoptosis by means of im-
munohistochemical detection of cleaved caspase-3 (17-
kDa fragment). Activation of caspase-3, one of the key
executioners of apoptosis, requires its cleavage into
two fragments (17 and 12 kDa). As shown in Fig. 1C,
MIN6 cells cultured for 24 h under conditions of 5 mM
glucose showed a 294.87 54.48% increase in apopto-
sis by cleaved caspase-3 staining compared with those
cultured in 25 mM glucose [100 14.86% (n6), P
0.001].
MIN6 cells are typically cultured in 25 mM glucose
(26). To determine whether the effects of lowering
glucose were due to differences in osmolarity, experi-
ments were performed with the addition of mannitol.
To maintain osmolarity, cells were incubated in 5 mM
glucose plus 20 mM mannitol. This treatment did not
change the percentage of apoptosis seen under condi-
tions of 5 mM glucose [%apoptosis; 25 mM glucose:
1.7 0.2 (n11); 5 mM glucose: 6.7 1(n13); 5
mM glucose 20 mM mannitol: 6.3 1.2 (n6), P
0.05 vs. 5 mM glucose]. To decrease the rates of prolif-
eration to reduce this as a variable while assessing
apoptosis, experiments were performed in the presence
of low serum (1%). Even in the presence of low serum
concentrations, a three- to fourfold decrease in apopto-
sis was seen in cells cultured in the presence of 25 mM
glucose compared with those in 5 mM glucose [2.8
0.29% (n3) vs. 7.63 2.2% (n3), P0.05]. Min6
cells express predominantly GLUT2, a high-K
m
glucose
transporter (26, 27). To determine whether the protec-
tive effect of 25 mM glucose was a property of cells with
high-K
m
glucose transporters and glucokinase, the
same experiments were performed in a neuronal cell
line, PC12. After incubation for 24 h in 5 or 25 mM
glucose, no difference in apoptosis was observed [5 mM
glucose: 0.26 0.11% (n6), 25 mM glucose: 0.56
0.07% (n6), P0.05]. This result suggested that the
correlation between physiological levels of glucose and
the regulation of apoptosis might be unique to cells
with high-K
m
glucose transporters and hexokinases
like islet -cells.
Effect of Glucose on Necrosis in Insulinoma Cells
Cell survival can be determined by apoptosis or ne-
crosis. MIN6 insulinoma cells were treated with glu-
cose (5 or 25 mM) for 24 h. There was no difference in
necrosis noted in cells cultured in 5 vs. 25 mM glucose
[%necrotic cells, 5 mM: 2.1 0.3 (n4); 25 mM: 2.3
0.5 (n6), P0.7].
E786 GLUCOSE ACTIVATES SURVIVAL IN -CELLS
AJP-Endocrinol Metab VOL 283 OCTOBER 2002 www.ajpendo.org
by 10.220.33.4 on September 9, 2016http://ajpendo.physiology.org/Downloaded from
Dose Dependency and Time Course of Glucose-
Regulated Islet
-Cell Survival
In nonislet -cells, absence of glucose induces apop-
tosis, whereas the protective effect on survival is max-
imal at 2 mM (29). In contrast, in islet -cells, be-
cause of the high-K
m
, low-afnity glucose transporter
and hexokinase activities, the glucose effect on sur-
vival would be predicted to have a K
m
in the 515 mM
range (52). After 24 h of incubation, inhibition of apop-
tosis was present at 15 mM glucose and was not sig-
nicantly altered by glucose concentrations up to 40
mM [%apoptosis at various glucose concentrations: 0
mM: 4.64 1.1; 5 mM: 3.08 1.1; 10 mM: 3.04 1.1;
15 mM: 2.43 0.07; 25 mM: 2.08 0.6; 40 mM: 2.33
0.55 (n4 in each category), P0.05 between 0 and
15 mM]. These results indicate that glucose regula-
tion of islet -cell apoptosis occurs over a physiologi-
cally relevant glucose concentration range (515 mM).
To assess the time course of the glucose effect on
apoptosis, cells were examined at 24-h intervals for up
to 96 h of incubation. The rate of apoptosis increased
with time in both 5 and 25 mM glucose, whereas at
each time point cells incubated in 25 mM glucose had a
three- to fourfold reduced rate of apoptosis relative to
those in 5 mM glucose [%apoptosis at 24 h at 5 mM
glucose: 6.3 1(n13) and 25 mM glucose: 1.7 0.3
(n11), P0.001; at 48 h in 5 mM glucose: 4.16 0.3
(n8) and 25 mM glucose: 0.7 0.3 (n8), P0.001;
at 72 h in 5 mM glucose: 15.99 4(n6) and 25 mM
glucose: 0.9 0.3 (n4), P0.0001; and at 96 h at 5
mM glucose: 34.4 2.4 (n6) and 25 mM glucose:
13.6 4.3 (n6), P0.05]. The data in Fig. 2
represent the aforementioned results expressed as a
percentage of apoptosis at 24 h of incubation in the
presence of 25 mM glucose.
Fig. 1. Effect of glucose on survival in islet -cells and insulinoma
cells. A: apoptosis in mouse islets. Isolated mouse islets were treated
with 2 or 15 mM glucose for 24 h, and apoptosis was assessed in
-cells by the TdT-mediated dUTP nick end labeling (TUNEL)
method as described in EXPERIMENTAL PROCEDURES. Apoptosis was
higher in the presence of 2 mM glucose (258.62 30.68%) compared
with 15 mM glucose (100 31.72%). Results represent means SE;
*P0.05. B: MIN6 insulinoma cells were treated with glucose for
24 h at the indicated concentrations and assessed for apoptosis using
the TUNEL method as described in EXPERIMENTAL PROCEDURES. Apop-
tosis was higher in the presence of 5 mM glucose (370 61.76%)
compared with 25 mM glucose (100 17.64%). Results represent
means SE; **P0.001. A representative photograph to illustrate
TUNEL-positive yellow (stained with FITC and propidium iodide)
uorescent cells is shown. C: caspase-3 activity assay. MIN6 insu-
linoma cells were treated with glucose for 24 h at the indicated
concentrations and assessed for cleaved caspase-3 staining as de-
scribed in EXPERIMENTAL PROCEDURES. The cleaved caspase-3-positive
cells were higher in the presence of 5 mM glucose (294 54.48%)
compared with 25 mM glucose (100 16.66%). Results represent
means SE; **P0.001. Representative photograph of immuno-
histochemical detection of cleaved caspase-3 positive cell is shown.
The cleaved caspase-3-positive cells appear green due to Alexa 488-
conjugated secondary antibody.
E787GLUCOSE ACTIVATES SURVIVAL IN -CELLS
AJP-Endocrinol Metab VOL 283 OCTOBER 2002 www.ajpendo.org
by 10.220.33.4 on September 9, 2016http://ajpendo.physiology.org/Downloaded from
Glucose-Regulated Survival is Dependent on RNA
Synthesis, Depolarization, and Ca
2
Inux
Having established a model system where reproduc-
ible effects of glucose on -cell survival could be ob-
served, we next sought mechanisms. In neuronal cells,
apoptosis induced by growth factor withdrawal is de-
pendent on RNA synthesis (43). Therefore, MIN6 cells
were treated for 24 h with actinomycin D (1 g/ml) to
inhibit RNA synthesis in the presence of 5 or 25 mM
glucose. This treatment completely attenuated the en-
hanced apoptosis seen in 5 mM glucose [5 mM glucose:
6.3 1% (n13); 5 mM glucose actinomycin D:
2.06 0.6% (n5), P0.01]. No change in apoptosis
was seen when actinomycin D was added under condi-
tions of 25 mM glucose [25 mM glucose: 1.7 0.2%
(n11); 25 mM glucose actinomycin D: 1.11 0.3%
(n6), P0.05 vs. 25 mM glucose]. In Fig. 3A, these
data are represented as percentages relative to apop-
tosis at 24 h in the presence of 25 mM glucose. These
results suggested that the increased rate of apoptosis
observed in the presence of 5 mM glucose is dependent
Fig. 3. Glucose-regulated islet -cell survival is dependent on RNA
synthesis, depolarization, and Ca
2
inux. A: effect of actinomycin
D. MIN6 cells were treated for 24 h at the indicated glucose concen-
trations in the presence or absence of actinomycin D (1 g/ml) to
inhibit RNA synthesis, and apoptosis was assessed by the TUNEL
method. Low glucose-induced apoptosis was attenuated in the pres-
ence of actinomycin D (%apoptosis relative to 25 mM glucose; 5 mM
glucose: 370 61%; 5 mM glucose actinomycin D: 121 35%; 25
mM glucose: 100 17%; 25 mM glucose actinomycin D: 65 17%).
Results represent means SE; **P0.001 (n.s., not signicant, P
0.05.) B: effect of diazoxide. MIN6 cells were treated for 24 h at the
indicated glucose concentrations in the presence or absence of dia-
zoxide, and apoptosis was assessed by the TUNEL method. The 25
mM glucose suppression of apoptosis was lost in the presence of
diazoxide (%apoptosis relative to 25 mM glucose; 5 mM glucose:
370 61; 5 mM glucose diazoxide: 345 35; 25 mM glucose: 100
17; 25 mM glucose diazoxide: 267 58). Results represent
means SE; **P0.001. C: effect of nifedipine. MIN6 cells were
treated for 24 h with glucose at the indicated concentrations in the
presence or absence of nifedipine, and apoptosis was assessed by the
TUNEL method. Results are all given relative to those in 25 mM
glucose and represent means SE; ***P0.0001.
Fig. 2. Time course of glucose-regulated survival in insulinoma cells.
MIN6 cells were treated with glucose at the indicated concentrations
and time points. Apoptosis was assessed using the TUNEL method
as described in EXPERIMENTAL PROCEDURES. Percent apoptosis relative
to apoptosis at 24 h in 25 mM glucose; 24 h at 5 mM glucose: 370
61 (n13) and 25 mM glucose: 100 17 (n11), P0.001; at 48 h
in 5 mM glucose: 244 17 (n8) and 25 mM glucose: 41 17 (n
8), P0.001; at 72 h in 5 mM glucose: 940 235 (n6) and 25 mM
glucose: 52 17 (n4), P0.0001; and 96 h at 5 mM glucose:
2,020 141 (n6) and 25 mM glucose: 880 31 (n6), P0.05.
Results represent means SE; *P0.05, **P0.001, ***P
0.0001.
E788 GLUCOSE ACTIVATES SURVIVAL IN -CELLS
AJP-Endocrinol Metab VOL 283 OCTOBER 2002 www.ajpendo.org
by 10.220.33.4 on September 9, 2016http://ajpendo.physiology.org/Downloaded from
on synthesis of new RNA, whereas the protective effect
of 25 mM glucose is not.
To determine whether the glucose effect on survival
required membrane depolarization, experiments were
performed using DZ, an agent that activates K
ATP
channels and blocks glucose-mediated depolarization
of the -cell (50). The protective effect conferred by 25
mM glucose was lost in the presence of DZ (0.6 mM),
suggesting that depolarization is essential for glucose-
mediated protection of the -cell [25 mM glucose: 1.7
0.3% (n11); 25 mM glucose DZ: 4.54 0.75% (n
6), P0.001 vs. 25 mM glucose; 5 mM glucose 6.3
1% (n13), P0.001 vs. 25 mM glucose; 5 mM
glucose DZ: 5.88 0.6% (n6), P0.05 vs. 5 mM
glucose]. In Fig. 3B, these data are represented as the
percentage of apoptosis relative to that seen at 24 h in
the presence of 25 mM glucose.
To determine whether Ca
2
inux was required for
glucose inhibition of apoptosis, cells were incubated in
the presence of the L-type Ca
2
channel blocker nifed-
ipine (10 M). As shown in Fig. 3C, the protective effect
conferred by 25 mM glucose was lost in the presence of
nifedipine, indicating that glucose-dependent Ca
2
in-
ux is essential for the glucose-mediated protection of
the -cell at 25 mM glucose [%apoptosis relative to
apoptosis in 25 mM glucose; 25 mM glucose: 102 15
(n3); 25 mM glucose nifedipine: 996 161 (n6),
P0.0001 vs. 25 mM glucose]. Addition of nifedipine
to cells incubated in 5 mM glucose resulted in an
almost threefold increase in apoptosis [%apoptosis rel-
ative to apoptosis in 25 mM glucose; 5 mM glucose:
343 55 (n3), P0.0001; 5 mM glucose nifedi-
pine: 942 136 (n6), P0.0001]. Thus it appears
that Ca
2
inux following glucose-induced depolariza-
tion is playing a critical role in survival and that
perhaps the further increase in survival in 25 mM is
due to the enhanced Ca
2
inux known to occur with
depolarization.
Signal Transduction Pathways Involved in Glucose-
Regulated Survival
Having determined that glucose-induced depolariza-
tion and Ca
2
inux are important mediators of sur-
vival, the signal transduction pathways involved in
this process were examined. In islet -cells, glucose-
mediated depolarization and Ca
2
inux have previ-
ously been shown to result in activation of several
serine-threonine kinases, including MAP kinases and
PI-3 kinase, and these have been shown to be involved
in the survival of other cells (22, 31). MIN6 cells were
cultured in either 5 or 25 mM glucose in the presence
or absence of the MAPK inhibitor PD-98059 (50 M) or
the PI 3-kinase inhibitors wortmannin (200 nM) or
LY-294002 (50 M). Kinase inhibitors were added 1 h
before the change in the glucose concentration of the
medium. As shown in Fig. 4, the MAPK inhibitor did
not alter the protective effect of 25 mM glucose on
apoptosis [%change in apoptosis relative to apoptosis
in 25 mM; 25 mM: 101 10 (n6); 25 mM
PD-98059: 123 1(n6), P0.5; Fig. 4]. However,
both PI 3-kinase inhibitors decreased the protective
effect of 25 mM glucose, indicating this effect to be PI
3-kinase dependent [%change in apoptosis relative to
apoptosis in 25 mM; 25 mM: 101 10 (n6); 25 mM
wortmannin: 311 29 (n3), P0.001; 25 mM
LY-294002: 286 73 (n6), P0.008; Fig. 4]. No
signicant change in apoptosis was noted when kinase
inhibitors were added to cells cultured in 5 mM glucose
compared with apoptosis in 5 mM glucose alone (data
not shown).
A major downstream target of activated PI 3-kinase
is the serine-threonine kinase Akt, and activation of
this enzyme by phosphorylation of Ser
473
has been
associated with inhibition of apoptosis (13, 32). To
determine whether glucose-regulated survival ob-
served in MIN6 cells was associated with glucose acti-
vation of Akt, cells were treated with glucose as indi-
cated, and protein extracts were immunoblotted with
an antibody specic for Ser
473
phospho-Akt (57 kDa) as
a measure of Akt activation (54). At 120 min after
addition of 25 mM glucose, there was marked Ser
473
phosphorylation of Akt (n4; Fig. 5A), whereas addi-
tion of 5 mM glucose showed no phosphorylation at
similar time points (data not shown). In separate ex-
periments, it was shown that the activation of Akt by
glucose was rst observed at 15 min and peaked at 120
min. Under these conditions, there was no change in
Akt protein detected by an antibody to total Akt. Ad-
dition of kinase inhibitors indicated that glucose acti-
vation of Akt was PI 3-kinase dependent and MAPK
independent (n3; Fig. 5B).
To determine whether similar treatment of primary
cultures of isolated mouse islets would also result in
Ser
473
phosphorylation of Akt, cells were incubated in
2 or 15 mM glucose for 45 min and immunoblotted with
the antibody specic for Ser
473
phospho-Akt (57 kDa).
As shown in Fig. 5C, although phospho-Akt was de-
tected in cells incubated in 2 mM glucose, similar
treatment with 15 mM glucose resulted in a threefold
increase (3.05 0.23, P0.01) in phospho-Akt. The
Fig. 4. Signal transduction pathways involved in glucose-regulated
islet -cell survival. MIN6 cells were cultured for 24 h at the indi-
cated glucose concentrations in the presence or absence of the indi-
cated inhibitors, and apoptosis was assessed by the TUNEL method.
Results represent means SE; **P0.001.
E789GLUCOSE ACTIVATES SURVIVAL IN -CELLS
AJP-Endocrinol Metab VOL 283 OCTOBER 2002 www.ajpendo.org
by 10.220.33.4 on September 9, 2016http://ajpendo.physiology.org/Downloaded from
software Scion Image was used to determine the inten-
sity of bands on the Western blots.
To conrm that Ser
473
phosphorylation of Akt was
associated with Akt kinase activity, we measured the
Akt kinase activity under similar conditions. In this
assay, immunoprecipitates of Akt are incubated with a
substrate GSK-3 fusion protein in the presence of ATP
and kinase buffer. Akt phosphorylates GSK-3, which is
then detected by Western blot analysis using a phos-
pho-GSK-3/antibody. MIN6 cells were incubated in
25 mM glucose overnight and then removed from me-
dium and replaced with Krebs buffer containing glu-
cose or potassium chloride as indicated. After 15 min of
treatment, protein was extracted and Akt kinase activ-
ity measured with a GSK-3 substrate. As shown in Fig.
6, exposure of cells to 3 mM glucose resulted in a
minimal increase in kinase activity, whereas exposure
to 25 mM glucose for 15 min resulted in a marked
increase in Akt kinase activity similar to that seen
with depolarization with 50 mM potassium chloride
(Fig. 6).
Akt Activity is Both Necessary and Sufcient
for Glucose-Mediated Survival
The previous observations demonstrated that glu-
cose-mediated reduction of apoptosis in insulinoma
cells was associated with PI 3-kinase-dependent acti-
vation of Akt. To attempt to demonstrate a causal
relationship between Akt activation and inhibition of
apoptosis, MIN6 cells were transfected with cytomeg-
alovirus promoter-based expression vectors encoding
either 1) wild-type HA-tagged Akt (Akt-WT), 2) a cat-
alytically inactive mutant (kinase dead) form of HA-
tagged Akt (Akt-K179M) with dominant-inhibitory ac-
tivity toward wild-type Akt kinase activity, or 3)a
constitutively active HA-tagged Akt mutant (Akt-CA)
that lacks amino acids 4129 and that has a 14-amino
acid src myristoylation signal on the amino terminus of
Akt that targets Akt to the membrane (33). Because
our rate of transfection was 5%, as determined by
immunohistochemistry using an antibody to HA, we
used immunohistochemical staining techniques to
identify transfected cells and study apoptosis in these
cells. This method has been used successfully in the
neuronal literature (13). The constitutively active Akt
targets Akt to the membrane, demonstrated by an
antibody to the HA tag (data not shown). Figure 7
demonstrates the percentage of apoptosis relative to
wild-type Akt-transfected cells cultured in 25 mM glu-
cose. The cells transfected with the kinase-dead plas-
mid Akt-K179M had increased rates of apoptosis
[246 57% increase vs. Akt-WT, 25 mM glucose (n
8), P0.05; Fig. 7]. Transfections with constitutively
active Akt plasmids in 25 mM glucose reduced the rate
of apoptosis [31 7.3% vs. Akt-WT in 25 mM glucose
(n8), P0.05; Fig. 7]. Transfections with constitu-
tively active Akt plasmids in 5 mM glucose reduced the
rate of apoptosis [31 7.3% reduction vs. Akt-WT in 5
mM glucose (n8), P0.05].
Fig. 6. Glucose activates Akt-kinase activity in insulinoma cells.
Min6 cells were cultured overnight in 25 mM glucose and 15% FCS.
Cells were then preincubated in Krebs buffer for 1 h (i.e., 0 time
point) and then treated with Krebs buffer containing glucose or KCl
as indicated. Akt kinase activity was measured as indicated in
EXPERIMENTAL PROCEDURES with a GSK-3 substrate at the indicated
times.
Fig. 5. Glucose activates Akt in insulinoma cells and isolated islets.
A: assessment of Ser
473
phospho-Akt (P-Akt). Cells were preincu-
bated in the absence of serum and with 5 mM glucose for 14 h,
followed by no serum and no glucose for 4 h and then treatment with
25 mM glucose for the indicated time. Lysates were prepared as
described in EXPERIMENTAL PROCEDURES. Protein (40 g) was loaded
in each lane and assayed on an acrylamide gel (10%). Activation of
Akt by glucose and total Akt was assessed by use of antibodies
specic for Ser
473
phospho-Akt and Akt, respectively. B: Akt activa-
tion is phosphatidylinositol (PI) 3-kinase dependent. Cells were
treated and assessed as described in A. Kinase inhibitors were added
1 h before addition of 25 mM glucose. The effect of glucose on
phospho-Akt and total Akt in the presence or absence of MAPK
inhibitor PD-98059 (50 M) or PI-3 kinase inhibitor wortmannin
(200 nM) was assessed. C: Akt activation in primary mouse islets.
Isolated mouse islets were preincubated in Krebs buffer for1hand
then treated with glucose for the indicated times and concentrations.
Lysates were prepared as described in EXPERIMENTAL PROCEDURES.
Protein (75 g) was loaded in each lane and assayed on an acryl-
amide gel (10%). Activation of Akt by glucose and total Akt was
assessed using antibodies specic for Ser
473
phospho-Akt and Akt.
E790 GLUCOSE ACTIVATES SURVIVAL IN -CELLS
AJP-Endocrinol Metab VOL 283 OCTOBER 2002 www.ajpendo.org
by 10.220.33.4 on September 9, 2016http://ajpendo.physiology.org/Downloaded from
DISCUSSION
The results of the present study provide evidence for
a glucose-mediated PI-3-kinase/Akt-dependent sur-
vival pathway in pancreatic islet -cells. We have dem-
onstrated glucose-mediated activation of Akt in iso-
lated mouse islets and in the MIN6 insulinoma cell
line. The results of this study conrm previous obser-
vations that glucose activates Akt in insulinoma cells
(8). Our ndings contribute to the similarities in prop-
erties of -cells and neurons, since activation of Akt in
neuronal cells has been shown to enhance survival and
to be induced by neurotransmitter-mediated depolar-
ization, Ca
2
inux, and PI 3-kinase activation (13). In
neurons, survival mediated both by growth factors
such as insulin and by depolarization through neuro-
transmitters seems to be converging on this common
survival pathway (56). Certainly, the effects of tar-
geted disruption of the insulin receptor (35) and insulin
receptor substrates (36, 57) in islet -cells, along with
the present studies, suggest that the depolarization
and growth factor survival pathways may be playing a
role in -cell survival, thus affecting its mass and
function.
The novel nding in this study is the nutritional
regulation of a survival pathway in islet -cells
through activation of Akt. The importance of the role of
glucose and Akt in regulating -cell mass is seen in the
transgenic mice expressing constitutively active Akt in
islet -cells. Expression of constitutively active Akt
linked to an insulin gene promoter in transgenic mice
resulted in increased islet -cell mass by altering -cell
size and number (4, 9, 55). These mice are also resis-
tant to streptozotocin-induced diabetes. Although in-
creased -cell mass was shown to be accounted for, at
least in part, by increased proliferation, further exper-
iments will be needed to determine whether these
animals have decreased islet -cell apoptosis as well.
Thus Akt activation may be of importance in both islet
-cell proliferation and survival.
Although the present studies demonstrated the ef-
fects of glucose and Ca
2
inux on the short-term
survival of -cells, they do not address the question of
reduced -cell survival that may be associated with
chronic hyperglycemia and excessive Ca
2
inux (28,
45). As in neurons, the effects of prolonged sustained
hyperglycemia on islet -cells may result in Ca
2
ex-
cess and subsequent apoptosis. Some studies have re-
ported increased apoptosis in rodent islets incubated in
glucose at 20 mM for periods as short as 24 days.
However, we did not observe increased apoptosis in
Min6 cells cultured in 40 mM glucose for 3 days. The
effects of prolonged hyperglycemia on -cells need to be
further addressed.
In the present studies, the increased rate of apopto-
sis at 5 mM glucose was inhibited by addition of acti-
nomycin D, suggesting the requirement for transcrip-
tion of proapoptotic proteins. This is similar to the
growth factor withdrawal-induced apoptosis in neu-
rons that is also dependent on transcription of new
RNA (41). The effects of high glucose on survival in
MIN6 cells, however, appeared to be independent of
RNA synthesis. This suggests that glucose-induced de-
polarization and Ca
2
inux are rapidly activating a
signal transduction pathway mediating survival, per-
haps through activation of a serine-threonine kinase.
In fact, the results of the present studies in which
glucose rapidly activates Akt are consistent with these
data. Certain possible mechanisms for this could be
through the well known effect of Akt activation on
inhibition of proapoptotic transcription factors of the
forkhead family (40) or inactivation of BAD (12). The
downstream targets of activated Akt and their possible
effects on -cell survival can now be assessed.
The results of the present studies do not rule out the
possibility that the glucose/depolarization/Ca
2
-medi-
ated survival pathway noted in these experiments is
dependent on insulin secretion, perhaps acting in an
autocrine/paracrine fashion. Insulin has been shown to
inhibit apoptosis in a number of cells (3, 20), and
treatment of MIN6 cells with 10
7
M insulin did
result in increased survival (S. Srivivasan, unpub-
lished observations). Recent studies have suggested
that glucose regulation of insulin and L-pyruvate ki-
nase gene transcription are mediated through glucose-
stimulated insulin secretion in an autocrine/paracrine
fashion (11). Our results are consistent with this hy-
pothesis, and this important question needs to be ex-
amined by further studies.
What could the possible signicance of glucose-me-
diated survival be to the normal physiology of islet
-cells? Glucose regulation of islet -cell survival may
play a critical role in the neonatal and postnatal pe-
riod, a time during which the rates of -cell apoptosis
are high and thus inuence the ultimate islet -cell
mass in the adult (21, 48). Epidemiological studies
correlate low birth weight and fetal malnutrition with
the onset of non-insulin-dependent diabetes later in
life (47). In animal models, maternal protein restric-
Fig. 7. Akt activity is both necessary and sufcient for glucose-
mediated survival. MIN6 cells were transfected with 1 g of either
wild-type Akt (Akt-WT), a catalytically inactive mutant (kinase
dead) form of Akt (Akt-K179M), or a constitutively active mutant
(Akt-CA) that targets Akt to the membrane and cultured in 25 mM
glucose. Apoptosis was assessed by the TUNEL method in trans-
fected cells at 24 h. Results are expressed as percent apoptosis
relative to Akt-WT cultured in 25 mM glucose and represent
means SE; *P0.05, ***P0.0001.
E791GLUCOSE ACTIVATES SURVIVAL IN -CELLS
AJP-Endocrinol Metab VOL 283 OCTOBER 2002 www.ajpendo.org
by 10.220.33.4 on September 9, 2016http://ajpendo.physiology.org/Downloaded from
tion imposed during fetal life and suckling resulted in
lower serum glucose levels and a reduction in -cell
mass in the offspring (6, 37). Examples of animal
models where hypoglycemia is associated with apopto-
sis have been reported (7, 42). Studies in rats trans-
planted with insulinoma cells have demonstrated in-
creased apoptosis in the endogenous islet -cells in the
animals transplanted with insulinomas. These ani-
mals have hyperinsulinemia and hypoglycemia. There
wasa6713% reduction in islet cell volume in
insulinoma-transplanted animals, with a reduction in
-cell size (7). In the -cell-specic Kir6.2-dominant-
negative transgenic mouse, the neonatal mice develop
hypoglycemia (42). These mice have enhanced -cell
apoptosis at 2 wk of age. Subsequent to this, these mice
develop hyperglycemia, believed to be secondary to a
reduction in -cell mass. These results suggest that
glucose may be important not only in the regulation of
insulin synthesis and secretion but also in the survival
of the islet -cell. Increased -cell apoptosis has been
noted in patients with hyperinsulinism-induced hypo-
glycemia compared with age-matched controls (30).
The results of the present study dene a molecular
mechanism for glucose regulation of an islet -cell
survival pathway that could have important conse-
quences for understanding the etiology of diabetes and
that may suggest new means of therapeutic interven-
tion as well.
We thank Dr. Philip D. Stahl for the Akt constructs; Dr. J.
Milbrandt for the PC12 cells; Drs. Eugene M. Johnson, Burton Wice,
and David Holtzman for their helpful advice; and Gary Skolnick for
preparation of the manuscript. We also acknowledge Michael Shor-
nick for technical support.
This work was supported in part by National Institute of Diabetes
and Digestive and Kidney Diseases Grant DK-16746 (M. A. Per-
mutt), a Howard Hughes Medical Institute Biomedical Research
grant (S. Srinivasan), and the Diabetes Research and Training
Center at the Washington University School of Medicine.
REFERENCES
1. Alessi DR, Andjelkovic M, Caudwell B, Cron P, Morrice N,
and Hemmings BA. Mechanism of activation of protein kinase
B by insulin and IGF-1. EMBO J 15: 65416551, 1996.
2. Altman SA, Randers L, and Rao G. Comparison of trypan
blue exclusion and uorometric assays for mammalian cell via-
bility determination. Biotech Prog 9: 671674, 1993.
3. Barber AJ, Nakamura M, Wolpert EB, Reiter CE, Siegel
GM, Antonetti DA, and Gardner TW. Insulin rescues retinal
neurons from apoptosis by a phosphatidylinositol 3-kinase/Akt-
mediated mechanism that reduces the activation of caspase-3.
J Biol Chem 276: 3281432821, 2001.
4. Bernal-Mizrachi E, Wen W, Stahlhut S, Welling C, and
Permutt MA. Islet cell expression of constitutively active
Akt1/PKB induces striking hypertrophy, hyperplasia, and hyper-
insulinemia. J Clin Invest 108: 16311638, 2001.
5. Bernard C, Berthault MF, Saulnier C, and Ktorza A. Neo-
genesis vs. apoptosis as main components of pancreatic beta cell
mass changes in glucose-infused normal and mildly diabetic
adult rats. FASEB J 13: 11951205, 1999.
6. Bertin E, Gangnerau MN, Bailbe D, and Portha B. Glucose
metabolism and -cell mass in adult offspring of rats protein
and/or energy restricted during the last week of pregnancy. Am J
Physiol Endocrinol Metab 277: E11E17, 1999.
7. Blume N, Skouv J, Larsson LI, Holst JJ, and Madsen OD.
Potent inhibitory effects of transplantable rat glucagonomas and
insulinomas on the respective endogenous islet cells are associ-
ated with pancreatic apoptosis. J Clin Invest 96: 22272235,
1995.
8. Buteau J, Foisy S, Rhodes CJ, Carpenter L, Biden TJ, and
Prentki M. Protein kinase C activation mediates glucagon-like
peptide-1-induced pancreatic -cell proliferation. Diabetes 50:
22372243, 2001.
9. Cho H, Mu J, Kim JK, Thorvaldsen JL, Chu Q, Crenshaw
ERI, Kaestner KH, Bartolomei MS, Shulman GI, and Birn-
baum MJ. Insulin resistance and a diabetes mellitus-like syn-
drome in mice lacking the protein kinase Akt2 (PKBB). Science
292: 17281731, 2001.
10. Collins F and Lile JD. The role of dihydropyridine-sensitive
voltage-gated calcium channels in potassium-mediated neuronal
survival. Brain Res 502: 99108, 1989.
11. Da Silva XG, Varadi A, and Rutter GA. Regulation of gene
expression by glucose in pancreatic cells (MIN6) via insulin
secretion and activation of phosphatidylinositol 3-kinase. J Biol
Chem 275: 3626936277, 2000.
12. Datta SR, Dudek H, Tao X, Masters S, Fu HA, Gotoh Y, and
Greenberg ME. Akt phosphorylation of BAD couples survival
signals to the cell-intrinsic death machinery. Cell 91: 231241,
1997.
13. Dudek H, Datta SR, Franke TF, Birnbaum MJ, Yao R,
Cooper GM, Segal RA, Kaplan DR, and Greenberg ME.
Regulation of neuronal survival by the serine-threonine protein
kinase Akt. Science 275: 661665, 1997.
14. Efanova IB, Zaitsev SV, Zhivotovsky B, Kohler M, Efendic
S, Orrenius S, and Berggren PO. Glucose and tolbutamide
induce apoptosis in pancreatic beta-cells. A process dependent on
intracellular Ca2concentration. J Biol Chem 273: 33501
33507, 1998.
15. Efendic S, Kindmark H, and Berggren PO. Mechanisms
involved in the regulation of the insulin secretory process. J In-
tern Med Suppl 735: 922, 1991.
16. Fernandes-Alnemri T, Litwack G, and Alnemri ES. CPP32,
a novel human apoptotic protein with homology to Caenorhab-
ditis elegans cell death protein Ced-3 and mammalian interleu-
kin-1 beta-converting enzyme. J Biol Chem 269: 3076130764,
1994.
17. Fox AP, Nowycky MS, and Tsien RW. Kinetic and pharma-
cological properties distinguishing three types of calcium cur-
rents in chick sensory neurons. J Physiol 394: 149172, 1987.
18. Gallo V, Kingsbury A, Balazs R, and Jorgensen OS. The
role of depolarization in the survival and differentiation of cer-
ebellar granule cells in culture. J Neurosci 7: 22032213, 1987.
19. Gavrieli Y, Sherman Y, and Ben-Sasson SA. Identication of
programmed cell death in situ via specic labeling of nuclear
DNA fragmentation. J Cell Biol 119: 493501, 1992.
20. Goetze S, Blaschke F, Stawowy P, Bruemmer D, Spencer
C, Graf K, Grafe M, Law RE, and Fleck E. TNFalpha inhibits
insulins antiapoptotic signaling in vascular smooth muscle cells.
Biochem Biophys Res Commun 287: 662670, 2001.
21. Hanke J. Apoptosis and occurrence of Bcl-2, Bak, Bax, Fas and
FasL in the developing and adult rat endocrine pancreas. Anat
Embryol (Berl) 202: 303312, 2000.
22. Harvey J, McKay NG, Walker KS, Van der Kaay J, Downes
CP, and Ashford ML. Essential role of phosphoinositide 3-ki-
nase in leptin-induced K(ATP) channel activation in the rat
CRI-G1 insulinoma cell line. J Biol Chem 275: 46604669, 2000.
23. Hoorens A, Van de Casteele M, Kloppel G, and Pipeleers
D. Glucose promotes survival of rat pancreatic beta cells by
activating synthesis of proteins which suppress a constitutive
apoptotic program. J Clin Invest 98: 15681574, 1996.
24. Hugl SR, White MF, and Rhodes CJ. Insulin-like growth
factor I (IGF-I)-stimulated pancreatic beta-cell growth is glu-
cose-dependent. Synergistic activation of insulin receptor sub-
strate-mediated signal transduction pathways by glucose and
IGF-I in INS-1 cells. J Biol Chem 273: 1777117779, 1998.
25. Inagaki N, Gonoi T, Clement JPI, Manga N, Inazawa J,
Gonzalez G, Aguilar-Bryan L, Seino S, and Bryan J. Recon-
stitution of I
KATP
: an inward rectier subunit plus the sulfonyl-
urea receptor. Science 270: 11661170, 1995.
26. Ishihara H, Asano T, Tsukuda K, Katagiri H, Inukai K,
Anai M, Kikuchi M, Yazaki Y, Miyazaki JI, and Oka Y.
E792 GLUCOSE ACTIVATES SURVIVAL IN -CELLS
AJP-Endocrinol Metab VOL 283 OCTOBER 2002 www.ajpendo.org
by 10.220.33.4 on September 9, 2016http://ajpendo.physiology.org/Downloaded from
Pancreatic beta cell line MIN6 exhibits characteristics of glucose
metabolism and glucose-stimulated insulin secretion similar to
those of normal islets. Diabetologia 36: 11391145, 1993.
27. Ishihara H, Asano T, Tsukuda K, Katagiri H, Inukai K,
Anai M, Kikuchi M, Yazaki Y, Miyazaki JI, and Oka Y.
Overexpression of hexokinase 1 but not GLUT1 glucose trans-
porter alters concentration dependence of glucose-stimulated
insulin secretion in pancreatic -cell line MIN6. J Biol Chem
269: 30813089, 1994.
28. Jonas JC, Sharma A, Hasenkamp W, Ilkova H, Patane G,
Laybutt R, Bonner-Weir S, and Weir GC. Chronic hypergly-
cemia triggers loss of pancreatic beta cell differentiation in an
animal model of diabetes. J Biol Chem 274: 1411214121, 1999.
29. Kan O, Baldwin SA, and Whetton AD. Apoptosis is regulated
by the rate of glucose transport in an interleukin 3 dependent
cell line. J Exp Med 180: 917923, 1994.
30. Kassem SA, Ariel I, Thornton PS, Scheimberg I, and Gla-
ser B. Beta-cell proliferation and apoptosis in the developing
normal human pancreas and in hyperinsulinism of infancy.
Diabetes 49: 13251333, 2000.
31. Khoo S and Cobb MH. Activation of mitogen-activating protein
kinase by glucose is not required for insulin secretion. Proc Natl
Acad Sci USA 94: 55995604, 1997.
32. Khwaja A. Akt is more than just a Bad kinase. Nature 401:
3334, 1999.
33. Kohn AD, Takeuchi F, and Roth RA. Akt, a pleckstrin ho-
mology domain containing kinase, is activated primarily by
phosphorylation. J Biol Chem 271: 2192021926, 1996.
34. Koike T, Martin DP, and Johnson EMJ. Role of Ca
2
chan-
nels in the ability of membrane depolarization to prevent neu-
ronal death induced by trophic-factor deprivation: evidence that
levels of internal Ca
2
determine nerve growth factor depen-
dence of sympathetic ganglion cells. Proc Natl Acad Sci USA 86:
64216425, 1989.
35. Kulkarni RN, Bruning JC, Winnay JN, Postic C, Magnu-
son MA, and Kahn CR. Tissue-specic knockout of the insulin
receptor in pancreatic beta cells creates an insulin secretory
defect similar to that in type 2 diabetes. Cell 96: 329339, 1999.
36. Kulkarni RN, Winnay JN, Daniels M, Bruning JC, Flier
SN, Hanahan D, and Kahn CR. Altered function of insulin
receptor substrate-1-decient mouse islets and cultured beta-cell
lines. J Clin Invest 104: R69R75, 1999.
37. Latorraca MQ, Carneiro EM, Boschero AC, and Mello MA.
Protein deciency during pregnancy and lactation impairs glu-
cose-induced insulin secretion but increases the sensitivity to
insulin in weaned rats. Br J Nutr 80: 291297, 1998.
38. Livingstone C, Patel G, and Jones N. ATF-2 contains a
phosphorylation-dependent transcriptional activation domain.
EMBO J 14: 17851797, 1995.
39. McDaniel ML, Colca JR, Kotagal N, and Lacy PE. A sub-
cellular fractionation approach for studying insulin release
mechanisms and calcium metabolism in islets of Langerhans.
Methods Enzymol 98: 182200, 1983.
40. Medema RH, Kops GJ, Bos JL, and Burgering BM. AFX-
like forkhead transcription factors mediate cell-cycle regulation
by Ras and PKB through p27kip1. Nature 404: 782787, 2000.
41. Mesner PW, Winters TR, and Green SH. Nerve growth factor
withdrawal-induced cell death in neuronal PC12 cells resembles
that in sympathetic neurons. J Cell Biol 119: 16691680, 1992.
42. Miki T, Tashiro F, Iwanaga T, Nagashima K, Yoshitomi H,
Aihara H, Nitta Y, Gonoi T, Inagaki N, Miyazaki J, and
Seino S. Abnormalities of pancreatic islets by targeted expres-
sion of a dominant-negative KATP channel. Proc Natl Acad Sci
USA 94: 1196911973, 1997.
43. Miller TM and Johnson EMJ. Metabolic and genetic analyses
of apoptosis in potassium/serum-deprived rat cerebellar granule
cells. J Neurosci 16: 74877495, 1996.
44. Namkung Y, Skrypnyk N, Jeong M-J, Lee T, Lee M-S, Kim
H-L, Chin H, Suh P-H, Kim S-S, and Shin H-S. Requirement
for the L-type Ca
2
channel alpha1D subunit in postnatal pan-
creatic cell generation. J Clin Invest 108: 10151022, 2001.
45. Olson LK, Redmon JB, Towle HC, and Robertson RP.
Chronic exposure of HIT cells to high glucose concentrations
paradoxically decreases insulin gene transcription and alters
binding of insulin gene regulatory protein. J Clin Invest 92:
514519, 1993.
46. Ono J, Takaki R, and Fukuma M. Preparation of single cells
from pancreatic islets of adult rat by the use of dispase. Endo-
crinol Jpn 24: 265270, 1977.
47. Poulsen P, Vaag AA, Kyvik KO, Moller JD, and Beck-
Nielsen H. Low birth weight is associated with NIDDM in
discordant monozygotic and dizygotic twin pairs. Diabetologia
40: 439446, 1997.
48. Scaglia L, Smith FE, and Bonner-Weir S. Apoptosis contrib-
utes to the involution of beta cell mass in the post partum rat
pancreas. Endocrinology 136: 54615468, 1995.
49. Seino S, Iwanaga T, Nagashima K, and Miki T. Diverse roles
of K(ATP) channels learned from Kir6.2 genetically engineered
mice. Diabetes 49: 311318, 2000.
50. Shyng SL, Ferrigni T, and Nichols CG. Regulationof KATP
channel activity by diazoxide and by MgADP: distinct functions
of the two nucleotide binding folds of the sulfonylurea receptor.
J Gen Physiol 110: 655664, 1997.
51. Steil GM, Trivedi N, Jonas JC, Hasenkamp WM, Sharma
A, Bonner-Weir S, and Weir GC. Adaptation of -cell mass to
substrate oversupply: enhanced function with normal gene ex-
pression. Am J Physiol Endocrinol Metab 280: E788E796,
2001.
52. Sweet IR and Matschinsky FM. Are there kinetic advantages
of GLUT2 in pancreatic glucose sensing? Diabetologia 40: 112
119, 1997.
53. Toescu EC. Apoptosis and cell death in neuronal cells: where
does Ca2t in? Cell Calcium 24: 387403, 1998.
54. Toker A and Newton AC. Akt/protein kinase B is regulated by
autophosphorylation at the hypothetical PDK-2 site. J Biol
Chem 275: 82718274, 2000.
55. Tuttle RL, Gill NS, Pugh W, Lee JP, Koeberlein B, Furth
EE, Polonsky KS, Naji A, and Birnbaum MJ. Regulation of
pancreatic beta-cell growth and survival by the serine/threonine
protein kinase Akt1/PKBalpha. Nat Med 7: 11331137, 2001.
56. Vaillant AR, Mazzoni I, Tudan C, Boudreau M, Kaplan DR,
and Miller FD. Depolarization and neurotrophins converge on
the phosphatidylinositol 3-kinase-Akt pathway to synergistically
regulate neuronal survival. J Cell Biol 146: 955966, 1999.
57. Withers DJ, Gutierrez JS, Towery H, Burks DJ, Ren J-M,
Previs S, Zhang Y, Bernal D, Pons S, Shulman GI, Bonner-
Weir S, and White MF. Disruption of IRS-2 causes type 2
diabetes in mice. Nature 391: 900903, 1998.
E793GLUCOSE ACTIVATES SURVIVAL IN -CELLS
AJP-Endocrinol Metab VOL 283 OCTOBER 2002 www.ajpendo.org
by 10.220.33.4 on September 9, 2016http://ajpendo.physiology.org/Downloaded from
... Under normal physiology, glucose uptake by β-cells raises the cytosolic Ca 2+ that induces exocytosis of insulin vesicles and increases cell survival. However, the exact role of SERCA2b/ALN on cytosolic Ca 2+ needs further investigation in T2D pathophysiological conditions, as persistent hyperglycemia and stimulation of Ca 2+ signaling pathways are detrimental to β-cells [46][47][48]. ...
... Previous studies identified different pathways responsible for inducing apoptosis in β-cells in severe T2D [60][61][62]. In contrast, other studies found that the culture of mouse islet β-cells and MIN6 insulinoma cells reduced the rate of apoptosis at high glucose concentration compared to low-glucose concentration [33,47]. Furthermore, a study presents evidence that glucose induces the β-cells survival through activating the PI3-kinase/Akt-signaling pathway [47]. ...
... In contrast, other studies found that the culture of mouse islet β-cells and MIN6 insulinoma cells reduced the rate of apoptosis at high glucose concentration compared to low-glucose concentration [33,47]. Furthermore, a study presents evidence that glucose induces the β-cells survival through activating the PI3-kinase/Akt-signaling pathway [47]. Thus, accumulating evidence suggests that activation of anti-apoptosis and apoptosis pathways during the progression of IR to severe T2D. ...
Article
Full-text available
Type 2 diabetes (T2D) is a major global health problem often caused by the inability of pancreatic islets to compensate for the high insulin demand due to apoptosis. However, the complex mechanisms underlying the activation of apoptosis and its counter process, anti-apoptosis, during T2D remain unclear. In this study, we employed bioinformatics and systems biology approaches to understand the anti-apoptosis-associated gene expression and the biological network in the pancreatic islets of T2D mice. First, gene expression data from four peripheral tissues (islets, liver, muscle and adipose) were used to identify differentially expressed genes (DEGs) in T2D compared to non-T2D mouse strains. Our comparative analysis revealed that Gm2036 is upregulated across all four tissues in T2D and is functionally associated with increased cytosolic Ca²⁺ levels, which may alter the signal transduction pathways controlling metabolic processes. Next, our study focused on islets and performed functional enrichment analysis, which revealed that upregulated genes are significantly associated with sucrose and fructose metabolic processes, as well as negative regulation of neuron apoptosis. Using the Ingenuity Pathway Analysis (IPA) tool of QIAGEN, gene regulatory networks and their biological effects were analyzed, which revealed that glucose is associated with the underlying change in gene expression in the islets of T2D; and an activated gene regulatory network—containing upregulated CCK, ATF3, JUNB, NR4A1, GAST and downregulated DPP4—is possibly inhibiting apoptosis of islets and β-cells in T2D. Our computational-based study has identified a putative regulatory network that may facilitate the survival of pancreatic islets in T2D; however, further validation in a larger sample size is needed. Our results provide valuable insights into the underlying mechanisms of T2D and may offer potential targets for developing more efficacious treatments.
... Gene an insight on the molecular mechanism of islet functionality is pivotal in disease prevention and transplantation [25][26][27][28][29]. Hence it was interesting to initiate a study to assess the basal levels of islet functionality indices such as, GLUT receptor, Ca2+, glucose kinase and Insulin expression immediately after the purification of islets are done. ...
... Kir channels are known to be modulated by intracellular ATP levels, which are regulated by nutrient availability. In pancreatic beta-cells, glucose metabolism activates the PI3K and Akt signaling pathway [25], which, in turn, phosphorylates and inhibits Kir6.2 subunits of KATP channels [22]. This inhibition leads to the closure of KATP channels and subsequent depolarization of the membrane potential, triggering insulin secretion in pancreatic β-cells by glucose metabolism that in turn increases intracellular ATP levels [15]. ...
Article
Inwardly rectifying potassium (Kir) channels play a key role in maintaining the resting membrane potential and supporting potassium homeostasis. There are many variants of Kir channels, which are usually tetramers in which the main subunit has two trans-membrane helices attached to two N- and C-terminal cytoplasmic tails with a pore-forming loop in between that contains the selectivity filter. These channels have domains that are strongly modulated by molecules present in nutrients found in different diets, such as phosphoinositols, polyamines and Mg2+. These molecules can impact these channels directly or indirectly, either allosterically by modulation of enzymes or via the regulation of channel expression. A particular type of these channels is coupled to cell metabolism and inhibited by ATP (KATP channels, essential for insulin release and for the pathogenesis of metabolic diseases like diabetes mellitus). Genomic changes in Kir channels have a significant impact on metabolism, such as conditioning the nutrients and electrolytes that an individual can take. Thus, the nutrigenomics of ion channels is an important emerging field in which we are attempting to understand how nutrients and diets can affect the activity and expression of ion channels and how genomic changes in such channels may be the basis for pathological conditions that limit nutrition and electrolyte intake. In this contribution we briefly review Kir channels, discuss their nutrigenomics, characterize how different components in the diet affect their function and expression, and suggest how their genomic changes lead to pathological phenotypes that affect diet and electrolyte intake.
... In cultured mouse islet beta cells and MIN6 insulinoma cells, a three-fold decrease in apoptosis was found in the presence of high glucose concentrations compared to low glucose concentrations. Blocking of the L-type Ca 2+ channel with nifidepine reversed the protective effects of high glucose, suggesting the requirement of Ca 2+ influx for beta cell survival at high glucose concentrations [116]. A recent publication studied the effects of prolonged hyperinsulinemia exposure on beta cell survival. ...
Article
Full-text available
Diabetes mellitus is a chronic disease affecting over 500 million adults globally and is mainly categorized as type 1 diabetes mellitus (T1DM), where pancreatic beta cells are destroyed, and type 2 diabetes mellitus (T2DM), characterized by beta cell dysfunction. This review highlights the importance of the divalent cation calcium (Ca2+) and its associated signaling pathways in the proper functioning of beta cells and underlines the effects of Ca2+ dysfunction on beta cell function and its implications for the onset of diabetes. Great interest and promise are held by human pluripotent stem cell (hPSC) technology to generate functional pancreatic beta cells from diabetic patient-derived stem cells to replace the dysfunctional cells, thereby compensating for insulin deficiency and reducing the comorbidities of the disease and its associated financial and social burden on the patient and society. Beta-like cells generated by most current differentiation protocols have blunted functionality compared to their adult human counterparts. The Ca2+ dynamics in stem cell-derived beta-like cells and adult beta cells are summarized in this review, revealing the importance of proper Ca2+ homeostasis in beta-cell function. Consequently, the importance of targeting Ca2+ function in differentiation protocols is suggested to improve current strategies to use hPSCs to generate mature and functional beta-like cells with a comparable glucose-stimulated insulin secretion (GSIS) profile to adult beta cells.
... La protéine Akt, également nommée PKB, est une sérine/thréonine kinase impliquée dans de nombreux mécanismes intracellulaires tels que le métabolisme des nutriments, la croissance et la survie cellulaire (Song et al., 2005). Parmi les activateurs canoniques majeurs d'Akt dans la cellule bêta, on retrouve notamment la PI3K (PhosphoInositide 3 Kinase) activée par certains facteurs de croissance, par le glucose, mais aussi par l'insuline (De Meyts, 2016;Manning and Toker, 2017;Srinivasan et al., 2002). Une fois activée, la PI3K catalyse la conversion du Phosphatidyl-Inositol 4,5-bisphosphate (PIP2) en Phosphatidyl-Inositol 3,4,5trisphosphate (PIP3) au niveau de la membrane plasmique (De Meyts, 2016). ...
Thesis
Full-text available
En 2019, la Fédération Internationale du Diabète a révélé que près de 500 millions de personnes étaient atteintes de diabète dans le monde. On estime que cette incidence atteindra les 700 millions de personnes en 2045. Outre, l’aspect financier de la prise en charge, le diabète est un véritable enjeu de santé publique. En effet, l’environnement hyperglycémique délétère associé au diabète est à l’origine de graves complications pouvant altérer le fonctionnement de nombreux organes tels que le cœur, le cerveau ou encore le rein. La résistance à l’insuline associée à la détérioration de la sécrétion d’insuline et à la perte de la masse cellulaire bêta pancréatique constituent les principales caractéristiques du diabète de type 2. Ainsi, afin d’améliorer la prise en charge des patients diabétiques, l’identification d’une approche thérapeutique maîtrisée permettant de protéger la masse cellulaire bêta et de favoriser la sécrétion d’insuline uniquement en réponse au glucose et ce, sans effets secondaires, apparaît idéale.Les précédents travaux du laboratoire ont identifié le PE endogène et ses dérivés synthétiques la Spadine et la Mini-Spadine comme des inhibiteurs sélectifs des canaux potassiques TREK-1 au fort potentiel antidépresseur et impliqués dans la sécrétion de sérotonine, la prolifération et la survie neuronale. Au niveau périphérique, la Spadine a été décrite in vitro et in vivo comme un peptide à l’effet incrétine comparable à celui de l’exendine-4, un antidiabétique couramment utilisé en clinique. Ainsi, à la suite de cette étude et par analogie aux effets protecteurs observés sur le neurone, nous avons émis l’hypothèse que le PE et ses dérivés pouvaient avoir un rôle bénéfique dans les mécanismes de survie de la cellule bêta pancréatique.Dans ce manuscrit, nous démontrons que le PE endogène et ses dérivés protègent les cellules bêta de l’apoptose induite par la présence chronique de l’interleukine pro-inflammatoire et diabétogène IL-1β, ainsi que d’un choc toxique aigu induit par la staurosporine. De plus, l’analyse des mécanismes intracellulaires révèle que ces peptides provoquent une augmentation de la concentration en calcium intracellulaire, activent les voies prolifératives et de survie ERK et Akt, et maintiennent l’activité du facteur transcriptionnel CREB dans un environnement délétère via un mécanisme dépendant des calmodulines kinases.Ainsi, ces travaux de thèse montrent que le PE et ses dérivés synthétiques protègent la cellule bêta pancréatique et initient des processus cellulaires vertueux par une voie de signalisation originale indépendante de la PKA, où le potentiel de membrane et le calcium occupent un rôle crucial. Ces données suggèrent le PE endogène et ses dérivés synthétiques comme une nouvelle classe de peptides protecteurs des cellules bêta pancréatiques.
... ERK5 est également connue pour protéger les cellules β en empêchant la mise en place de l'apoptose médiée par le stress du réticulum(Nam et al. 2017). De plus, outre l'activation des MAPKs, le glucose permet l'activation d'Akt entrainant l'augmentation de l'expression de PDX-1 favorisant la prolifération et donc l'augmentation de la masse des cellules β(Srinivasan et al. 2002 ;Babu et al. 2007 ;Jara et al. 2020). D'autre part, le glucose agit sur les GKAs (GlucoKinase Activators) qui facilitent la prolifération cellulaire, réduisent l'apoptose tout en modulant négativement l'expression de gènes relatifs au stress du RE tels que Chop (C/EBPhomologous protein) ou Bax (Bcl2-associated X protein)(Porat et al. 2011 ;Oyadomari et Mori Voies de régulation de la masse fonctionnelle des cellules β pancréatiques par le glucose.2004 ...
Thesis
Une petite molécule inhibitrice de la MAP3 kinase TPL2 améliore la dysfonction et la mort des cellules bêta induite par des nutriments.Introduction : Les preuves suggèrent que les stress métaboliques induits par les nutriments, tel que l'hyperglycémie chronique, établissent un lien entre le diabète de type 2 et le dysfonctionnement et la mort des cellules bêta. Si la MAP3 kinase 8, Tumor progression locus 2 (TPL2), joue un rôle important dans la mort des cellules bêta; après une exposition aux cytokines, son implication dans la dysfonction et la mort induite par hyperglycémie chronique est inconnue.Méthode : La sécrétion d’insuline est mesurée par HTRF. Les expressions protéiques (TPL2, caspase 3 clivée et JNK1/2 (marqueurs de mort), sont évaluées par Western Blotting. L’expression d’ARNm d’Icer, est mesurée par RT-qPCR.Résultats : Nous avons montré que TPL2 est surexprimée dans les îlots de rats GK et de souris db/db. L’hyperglycémie chronique augmente l’expression de TPL2 dans les cellules bêta; et les îlots humains. Le traitement des souris db/db avec une petite molécule inhibitrice de TPL2 entraîne une réduction de la progression du diabète avec une diminution de l'hyperglycémie et de l'hyperinsulinémie à jeun, une meilleure tolérance au glucose, sans affecter la sensibilité à l'insuline. L'inhibition de TPL2 améliore les effets néfastes de l’hyperglycémie sur la sécrétion d’insuline induite par le glucose, réduit l’apoptose des cellules bêta, et empêche la régulation positive de l’ARNm d’Icer, un répresseur transcriptionnel de l'insuline et des gènes clés des protéines d’exocytose, et de l’activation des kinases c-Jun N-terminales 1/2 (JNK1/2). L'association d'un agoniste du récepteur du GLP-1 (GLP-1RA, exendine-4) avec l'inhibiteur de TPL2 a totalement empêché l'apoptose induite par l'hyperglycémie.Conclusion : Nos résultats identifient l’inhibition de TPL2, seule ou en association avec un GLP-1RA, comme une nouvelle stratégie thérapeutique potentielle prometteuse pour prévenir le dysfonctionnement et l’apoptose des cellules bêta et pour ralentir le développement du DT2.
Article
Scope Consuming goat milk is known to benefit high‐fat diet‐fed and streptozocin (STZ)‐induced diabetic rats, but the underlying mechanisms are unknown. This study is conducted to investigate the metabolic effects of a goat milk diet (a form of goat milk powder) on glucose homeostasis and pancreatic conditions in a mouse model of Type 2 diabetes mellitus (T2DM) induced by STZ. Methods and results T2DM mice are fed with a goat‐milk‐based diet containing 10.3% w/w goat milk powder for 10 weeks for investigating the in vivo effects; a β‐cell line MIN6 cells are used to test the in vitro effects of digested goat milk (DGM). Goat milk diet improves the deleterious effects of STZ on fasting glucose levels and glucose tolerance, accelerates pancreatic structure recovery, and alters blood metabolites in mice. Based on the significant differences observed in metabolites, the key pathways, metabolite regulatory enzymes, metabolite molecular modules, and biochemical reactions are identified as critical integrated pathways. DGM promotes the cell activity, glucose transportation, and AKT activation in cultured STZ‐treated MIN6 cells in vitro. Conclusions Goat milk diet improves glucose homeostasis and pancreatic conditions of T2DM mice, in association with improved blood metabolite profiles and activation of pancreatic AKT pathway.
Article
Aberrant AKT activation occurs in a number of cancers, metabolic syndrome, and immune disorders, making it an important target for the treatment of many diseases. To monitor spatial and temporal AKT activity in a live setting, we generated an Akt-FRET biosensor mouse that allows longitudinal assessment of AKT activity using intravital imaging in conjunction with image stabilization and optical window technology. We demonstrate the sensitivity of the Akt-FRET biosensor mouse using various cancer models and verify its suitability to monitor response to drug targeting in spheroid and organotypic models. We also show that the dynamics of AKT activation can be monitored in real time in diverse tissues, including in individual islets of the pancreas, in the brown and white adipose tissue, and in the skeletal muscle. Thus, the Akt-FRET biosensor mouse provides an important tool to study AKT dynamics in live tissue contexts and has broad preclinical applications.
Article
Bile acids have received increasing attention over the past years as their multiple alternative roles became clearer. Tauroursodeoxycholic Acid (TUDCA) in specific has generated special interest due to its ability to promote pancreatic survival and function, as well as reduce endoplasmic reticulum stress. However, there are few studies explaining the molecular mechanisms behind TUDCA's beneficial actions on pancreatic beta cells. In this review, we decided to review the literature in order to craft a primer for researchers on what is known about TUDCA's receptors and the molecular pathways involved in this bile acid's function in the endocrine pancreas. We review the studies that focused on G protein-coupled bile acid receptor (TGR5), Sphingosine-1-phosphate receptor 2 (S1PR2) and α5β1 Integrin function in pancreatic cells. Our hope is to provide a basis for future studies to expand upon, especially considering the current lack of studies focusing on the importance of these receptors, either through TUDCA signaling or other signaling molecules.
Article
Full-text available
In this report, we have examined the mechanisms whereby neurotrophins and neural activity coordinately regulate neuronal survival, focussing on sympathetic neurons, which require target-derived NGF and neural activity for survival during development. When sympathetic neurons were maintained in suboptimal concentrations of NGF, coincident depolarization with concentrations of KCl that on their own had no survival effect, synergistically enhanced survival. Biochemical analysis revealed that depolarization was sufficient to activate a Ras-phosphatidylinositol 3-kinase–Akt pathway (Ras–PI3-kinase–Akt), and function-blocking experiments using recombinant adenovirus indicated that this pathway was essential for ∼50% of depolarization-mediated neuronal survival. At concentrations of NGF and KCl that promoted synergistic survival, these two stimuli converged to promote increased PI3-kinase–dependent Akt phosphorylation. This convergent PI3-kinase–Akt pathway was essential for synergistic survival. In contrast, inhibition of calcium/calmodulin-dependent protein kinase II revealed that, while this molecule was essential for depolarization-induced survival, it had no role in KCl- induced Akt phosphorylation, nor was it important for synergistic survival by NGF and KCl. Thus, NGF and depolarization together mediate survival of sympathetic neurons via intracellular convergence on a Ras–PI3-kinase–Akt pathway. This convergent regulation of Akt may provide a general mechanism for coordinating the effects of growth factors and neural activity on neuronal survival throughout the nervous system.
Article
Full-text available
Glucagon-like peptide-1 (GLP-1), an insulinotropic and, glucoincretin hormone, is a potentially important therapeutic agent in the treatment of diabetes. We previously provided evidence that GLP-1 induces pancreatic beta-cell growth nonadditively with glucose in a phosphatidylinositol-3 kinase (PI-3K)-dependent manner. In the present study, we investigated the downstream effectors of PI-3K to determine the precise signal transduction pathways that mediate the action of GLP-1 on beta-cell proliferation. GLP-1 increased extracellular signal-related kinase 1/2, p38 mitogen-activated protein kinase. (MAPK), and protein kinase B activities nonadditively with glucose in pancreatic beta (INS 832/13) cells. GLP-1 also caused nuclear translocation of the atypical protein kinase C aPKC isoform in INS as well as in dissociated normal rat beta-cells as shown by immunolocalization and Western immunoblotting analysis. Tritiated thymidine incorporation measurements showed that the p38 MAPK inhibitor SB203580 suppressed GLP-1-induced beta-cell proliferation. Further investigation was performed using isoform-specific pseudosubstrates of classical (alpha, beta, and gamma) or zeta aPKC isoforms. The PKC zeta pseudosubstrate suppressed the proliferative action of GLP-1, whereas the inhibitor of classical PKC isoforms had no effect. Overexpression of a kinase-dead PKC zeta acting as a dominant negative protein suppressed GLP-1 -induced proliferation. In addition, ectopic expression of a constitutively active PKC zeta mutant stimulated tritiated thymidine incorporation to the same extent as GLP-1, and the glucoincretin had no growth-promoting action under this condition. The data indicate that GLP-1-induced activation of PKC zeta is implicated in the beta -cell proliferative signal of the insulinotropic hormone. The results are consistent with a model in which GLP-1-induced PI-3K activation results in PKC zeta translocation to the nucleus, which may play a role in the pleiotropic effects (DNA synthesis, metabolic enzymes, and insulin gene expression) of the glucoineretin.
Article
Full-text available
Insulin receptor substrate-1 (IRS-1) is pivotal in mediating the actions of insulin and growth factors in most tissues of the body, but its role in insulin-producing beta islet cells is unclear. Freshly isolated islets from IRS-1 knockout mice and SV40-transformed IRS-1-deficient beta-cell lines exhibit marked insulin secretory defects in response to glucose and arginine. Furthermore, insulin expression is reduced by about 2-fold in the IRS-1-null islets and beta-cell lines, and this defect can be partially restored by transfecting the cells with IRS-1. These data provide evidence for an important role of IRS-1 in islet function and provide a novel functional link between the insulin signaling and insulin secretion pathways. This article may have been published online in advance of the print edition. The date of publication is available from the JCI website, http://www.jci.org.
Article
The ATF‐2 transcription factor can mediate adenovirus E1A‐inducible transcriptional activation. Deletion analysis has indicated that the N‐terminal region of ATF‐2 is essential for this response. Furthermore, the N‐terminus can activate transcription in the absence of E1A when fused to a heterologous DNA binding domain. However, in the intact protein this activation domain is masked. In this report we show that residues in the N‐terminus required for activation are also required for mediating E1A stimulation. In particular two threonine residues at positions 69 and 71 are essential. These residues are phosphorylated in vivo and can be efficiently phosphorylated in vitro by the JNK/SAPK subgroup of the MAPK family. ATF‐2 can bind to a UV‐inducible kinase through a region in the N‐terminus that is distinct from the sites of phosphorylation; this binding region is both necessary for phosphorylation by JNK/SAPK in vitro and for transcriptional activation in vivo. The activity of the N‐terminus is stimulated by UV irradiation which stimulates the signalling pathway leading to JNK/SAPK. Finally, although ATF‐2 binds to the E1A protein, the N‐terminal activation domain is not required for this interaction. The results show that ATF‐2, like other members of the ATF/CREB family of DNA binding proteins is regulated by specific signalling pathways.
Article
An association between low birth weight and later impaired glucose tolerance was recently demonstrated in several human populations. Although fetal malnutrition is probably involved, the biological bases of such a relationship are not yet clear, and animal studies on the matter are scarce. The present study was aimed to identify, in adult (8-wk) female offspring, the effects of reduced protein and/or energy intake strictly limited to the last week of pregnancy. Thus we have tested three protocols of gestational malnutrition: a low-protein isocaloric diet (5 instead of 15%), with pair feeding to the mothers receiving the control diet; a restricted diet (50% of the control diet); and a low-protein restricted diet (50% of low-protein diet). Only the low-protein diet protocols, independent of total energy intake, led to a lower birth weight. The adult offspring female rats in the three deprived groups exhibited no decrease in body weight and no major impairment in glucose tolerance, glucose utilization, or glucose production (basal state and hyperinsulinemic clamp studies). However, pancreatic insulin content and beta-cell mass were significantly decreased in the low-protein isocaloric diet group compared with the two energy-restricted groups. Such impairment of beta-cell mass development induced by protein deficiency limited to the last part of intrauterine life could represent a situation predisposing to impaired glucose tolerance.
Article
Previous studies have shown that in neuronal cells the developmental phenomenon of programmed cell death is an active process, requiring synthesis of both RNA and protein. This presumably reflects a requirement for novel gene products to effect cell death. It is shown here that the death of nerve growth factor-deprived neuronal PC12 cells occurs at the same rate as that of rat sympathetic neurons and, like rat sympathetic neurons, involves new transcription and translation. In nerve growth factor-deprived neuronal PC12 cells, a decline in metabolic activity, assessed by uptake of [3H]2-deoxyglucose, precedes the decline in cell number, assessed by counts of trypan blue-excluding cells. Both declines are prevented by actinomycin D and anisomycin. In contrast, the death of nonneuronal (chromaffin-like) PC12 cells is not inhibited by transcription or translation inhibitors and thus does not require new protein synthesis. DNA fragmentation by internucleosomal cleavage does not appear to be a consistent or significant aspect of cell death in sympathetic neurons, neuronal PC12 cells, or nonneuronal PC12 cells, notwithstanding that the putative nuclease inhibitor aurintricarboxylic acid protects sympathetic neurons, as well as neuronal and nonneuronal PC12 cells, from death induced by trophic factor removal. Both phenotypic classes of PC12 cells respond to aurintricarboxylic acid with similar dose-response characteristics. Our results indicate that programmed cell death in neuronal PC12 cells, but not in nonneuronal PC12 cells, resembles programmed cell death in sympathetic neurons in significant mechanistic aspects: time course, role of new protein synthesis, and lack of a significant degree of DNA fragmentation.
Article
Pancreatic β cells are the source of insulin, which directly lowers blood glucose levels in the body. Our analyses of α1D gene-knockout (α1D-/-) mice show that the L-type calcium channel, α1D, is required for proper β cell generation in the postnatal pancreas. Knockout mice were characteristically slightly smaller than their littermates and exhibited hypoinsulinemia and glucose intolerance. However, isolated α1D-/- islets persisted in glucose sensing and insulin secretion, with compensatory overexpression of another L-type channel gene, α1C. Histologically, newborn α1D-/- mice had an equivalent number of islets to wild-type mice. In contrast, adult α1D-/- mice showed a decrease in the number and size of islets, compared with littermate wild-type mice due to a decrease in β cell generation. TUNEL staining showed that there was no increase in cell death in α1D-/- islets, and a 5-bromo-2′ deoxyuridine-labeling (BrdU-labeling) assay illustrated significant reduction in the proliferation rate of β cells in α1D-/- islets.
Article
Programmed cell death (PCD) plays a key role in developmental biology and in maintenance of the steady state in continuously renewing tissues. Currently, its existence is inferred mainly from gel electrophoresis of a pooled DNA extract as PCD was shown to be associated with DNA fragmentation. Based on this observation, we describe here the development of a method for the in situ visualization of PCD at the single-cell level, while preserving tissue architecture. Conventional histological sections, pretreated with protease, were nick end labeled with biotinylated poly dU, introduced by terminal deoxy-transferase, and then stained using avidin-conjugated peroxidase. The reaction is specific, only nuclei located at positions where PCD is expected are stained. The initial screening includes: small and large intestine, epidermis, lymphoid tissues, ovary, and other organs. A detailed analysis revealed that the process is initiated at the nuclear periphery, it is relatively short (1-3 h from initiation to cell elimination) and that PCD appears in tissues in clusters. The extent of tissue-PCD revealed by this method is considerably greater than apoptosis detected by nuclear morphology, and thus opens the way for a variety of studies.
Article
The physiological performance of an organ depends on an interplay between changes in cellular function and organ size, determined by cell growth, proliferation and death. Nowhere is this more evident than in the endocrine pancreas, where disturbances in function or mass result in severe disease. Recently, the insulin signal-transduction pathway has been implicated in both the regulation of hormone secretion from β cells in mammals as well as the determination of cell and organ size in Drosophila melanogaster. A prominent mediator of the actions of insulin and insulin-like growth factor 1 (IGF-1) is the 3′-phosphoinositide–dependent protein kinase Akt, also known as protein kinase B (PKB). Here we report that overexpression of active Akt1 in the mouse β cell substantially affects compartment size and function. There was a significant increase in both β-cell size and total islet mass, accompanied by improved glucose tolerance and complete resistance to experimental diabetes.
Article
Tumor necrosis factor α (TNFα) interferes with insulin signaling in adipose tissue and may promote insulin resistance. Insulin resistance is associated with vascular injury, but little is known about the interaction of TNFα and insulin in the vasculature. By activating the Insulin receptor (IR) → IRS-1 → phosphatidylinositol-3-kinase (PI3K) → Akt-pathway, insulin protects vascular smooth muscle cells (VSMC) from undergoing apoptosis. We therefore investigated the effect of TNFα on insulin's antiapoptotic signaling in rat aortic VSMC. Insulin induced rapid tyrosine-phosphorylation of the IR and IRS-1 and caused a 2.8-fold increase of IRS-1-bound PI3K. TNFα had no effect on insulin-induced tyrosine-phosphorylation of IR or IRS-1, but inhibited insulin-stimulated IRS-1/PI3K-association by 84%. Insulin-induced phosphorylation of Akt downstream of PI3K was inhibited by TNFα in a similar pattern. We next examined the effect of TNFα on insulin's protective actions on H2O2-induced apoptosis. Insulin alone prevented 72.8% of H2O2-induced apoptosis, which was significantly inhibited by TNFα. TNFα alone did not induce apoptosis. In contrast, TNFα had no effect on PDGF-induced antiapoptotic signal transduction via Akt. Thus, TNFα selectively interferes with insulin's antiapoptotic signaling in VSMC by inhibiting the association of IRS-1/PI3K and the downstream activation of Akt.