ArticlePDF Available

A Protective Role for Heme Oxygenase1 in INS1 Cells and Rat Islets that are Exposed to High Glucose Conditions

Authors:

Abstract

Heme oxygenase-1 (HO-1) has been described as an inducible protein that is capable of cytoprotection via radical scavenging and the prevention of apoptosis. Chronic exposure to hyperglycemia can lead to cellular dysfunction that may become irreversible over time, and this process has been termed glucose toxicity. Yet little is known about the relation between glucose toxicity and HO-1 in the islets. The purposes of the present study were to determine whether prolonged exposure of pancreatic islets to a supraphysiologic glucose concentration disrupts the intracellular balance between reactive oxygen species (ROS) and HO-1, and so this causes defective insulin secretion; we also wanted to evaluate a protective role for HO-1 in pancreatic islets against high glucose levels. The intracellular peroxide levels of the pancreatic islets (INS-1 cell, rat islet) were increased in the high glucose media (30 mM glucose or 50 mM ribose). The HO-1 expression was induced in the INS-1 cells by the high glucose levels. Both the HO-1 expression and glucose stimulated insulin secretion (GSIS) was decreased simultaneously in the islets by treatment of the HO-1 antisense. The HO-1 was upregulated in the INS-1 cells by hemin, an inducer of HO-1. And, HO-1 upregulation induced by hemin reversed the GSIS in the islets at a high glucose condition. These results suggest HO-1 seems to mediate the protective response of pancreatic islets against the oxidative stress that is due to high glucose conditions.
INTRODUCTION
Antioxidant enzymes such as superoxide dismutase, cata-
lase, glutathione peroxidase, heat shock protein and heme
oxygenase-1 (HO-1) are induced by various stimuli, and they
represent a powerful endogenous protective mechanism that
act within the pancreatic islets against free radicals (1, 2).
HO-1 catalyzes the rate-limiting step in heme catabolism and
it generates CO and bilirubin, and bilirubin has been demon-
strated as a potent antioxidant (3). In addition, HO-1 has
been described as an inducible protein that is capable of cyto-
protection via radical scavenging and preventing apopotosis.
Glucose toxicity is defined as nonphysiological and poten-
tially irreversible cellular damage that results in defective
insulin gene expression, and this is caused by chronic expo-
sure to supraphysiologic glucose concentrations (4-9). The
beta cell in type 2 diabetes is also adversely affected by chronic
hyperglycemia and, in this sense, is also a target for secondary
complications. As hyperglycemia worsens, the beta cell steadi-
ly undergoes deterioration, secretes less and less insulin, and
becomes a participant in a downward spiral of loss of func-
tion (9). Recently, the overexpression of antioxidant gene
products has been induced in the islets, and along with using
antioxidant drugs, this helps to protect against oxidative stress
(10-14). However, the relation between glucose toxicity and
HO-1 in the islets is still not known completely.
Thus, we would like to determine whether prolonged expo-
sure of the pancreatic islets to a supraphysiologic glucose
con-
centration disrupts the intracellular balance between reactive
oxygen species (RO
S) and HO-1, and if this causes defective
insulin secretion; we also wanted to evaluate a protective role
for HO-1 against high glucose concentrations in the pancre-
atic islets.
MATERIALS AND METHODS
INS-1 cell culture
The INS-1 cells (15) were grown in 5% CO2-95% air at
37
in RPMI-1640 medium containing 11.1 mM pyru-
vate, 10 mM HEPES, 50 M 2-mercaptoethanol, 100 U
penicillin/mL and 100 g streptomycin/mL. The RPMI-1640
medium used in all the experiments contained the supple-
Kyu Chang Won, Jun Sung Moon,
Mi Jung Eun, Ji Sung Yoon,
Kyung Ah Chun
*
, Ihn Ho Cho
*
,
Yong Woon Kim
, Hyoung Woo Lee
Department of Internal Medicine, Department of
Nuclear Medicine
*
, Department of Physiology
,
College of Medicine, Yeungnam University, Daegu,
Korea
Address for correspondence
Kyu Chang Won, M.D.
Department of Internal Medicine, College of Medicine,
Yeungnam University, 317-1 Daemyung-dong,
Nam-gu, Daegu 705-717, Korea
Tel : +82.53-620-3846, Fax : +82.53-654-8386
E-mail : kcwon@med.yu.ac.kr
*This study was supported by a Grants of Yeungnam
University Medical Center.
418
J Korean Med Sci 2006; 21: 418-24
ISSN 1011-8934 Copyright
The Korean Academy
of Medical Sciences
A Protective Role for Heme Oxygenase-1 in INS-1 Cells and Rat
Islets that are Exposed to High Glucose Conditions
Heme oxygenase-1 (HO-1) has been described as an inducible protein that is capa-
ble of cytoprotection via radical scavenging and the prevention of apoptosis. Chronic
exposure to hyperglycemia can lead to cellular dysfunction that may become irre-
versible over time, and this process has been termed glucose toxicity. Yet little is
known about the relation between glucose toxicity and HO-1 in the islets. The pur-
poses of the present study were to determine whether prolonged exposure of pan-
creatic islets to a supraphysiologic glucose concentration disrupts the intracellular
balance between reactive oxygen species (ROS) and HO-1, and so this causes
defective insulin secretion; we also wanted to evaluate a protective role for HO-1
in pancreatic islets against high glucose levels. The intracellular peroxide levels of
the pancreatic islets (INS-1 cell, rat islet) were increased in the high glucose media
(30 mM glucose or 50 mM ribose). The HO-1 expression was induced in the INS-1
cells by the high glucose levels. Both the HO-1 expression and glucose stimulated
insulin secretion (GSIS) was decreased simultaneously in the islets by treatment
of the HO-1 antisense. The HO-1 was upregulated in the INS-1 cells by hemin, an
inducer of HO-1. And, HO-1 upregulation induced by hemin reversed the GSIS in
the islets at a high glucose condition. These results suggest HO-1 seems to medi-
ate the protective response of pancreatic islets against the oxidative stress that is
due to high glucose conditions.
Key Words : HMOX1 protein, human; HO-1; Glucose Toxicity; Oxidative Stress; Islets of Langerhans
Received : 5 August 2005
Accepted : 4 November 2005
Heme Oxygenase-1 Protects Glucose Toxicity in INS-1 Cells 419
ments described above. The cells were passaged weekly after
they has been detached with trypsin-EDTA. All the studies
were performed on INS-1 cells that were between passages
21 and 29.
Pancreatic islet isolation
The pancreata from male Wistar rats were infused with
10 mL of 1.5 mg/mL collagenase type XI (Sigma, St. Louis,
MO, U.S.A.)/1% fetal bovine serum/2 units/mL RQ1 DNase
(Promega, U.S.A.) solution in Medium 199 (Sigma). After
it was surgically excised, the pancreas was incubated in the
collagenase solution at 37
. The undigested tissue was re-
moved by using a 500 m screen,
and the recovered tissue
was washed twice with ice-cold Hanks
’ balanced salt solution
containing 0.1% bovine serum albumin; this was followed
by centrifugation at 250×g for 4 min. The islets in the pellet
were separated
by using a Histopaque-1077 gradient (Sigma),
and then islet
s were hand-picked and cultured in RPMI medi-
um 1640 containing 10% fetal bovine serum, 11.1 mM glu-
cose and penicillin/streptomycin/amphotericin B before the
experimentation.
In vitro induction of HO-1
Hemin (Sigma) was dissolved in 0.1N NaOH and it was
diluted 1:1 in phophate-buffered saline (PBS); the pH was
adjusted to 7.4 and the solution was sterilized by filtration.
The INS-1 cell or islets were incubated at 37
for 24 hr with
the selected concentration of hemin.
Evaluation of ROS with flow cytometry
The intracellular peroxide levels (16) were detected by flow
cytometric analysis with using an oxidation-sensitive fluo-
rescein-labeled dye, carboxylated dichlorodi-hydrofluores-
cein diacetate (carboxy-H2DCFDA, Molecular Probes, Carls-
bad, CA, U.S.A.). Upon oxidation by intracellular ROS, the
non-fluorescent dye is
converted into its fluorescent form.
The islets and INS-1 cells
were labeled with 100 M car-
boxy-H2DCFDA for 1 hr at 37
. Following the cell load-
ing of the dye, the islets were washed twice with PBS and
then put back into culture conditions for 2 hr. The islets and
INS-1 cells were then harvested, washed twice with PBS
and resuspended in trypsin-EDTA (0.25% trypsin, 2 mM
Na4-EDTA, Invitrogen) for 5 min at 37
. To disperse the
islets into a single cell suspension, the islets and INS-1 cells
were gently passed 20 times in and out of a 200-1,000- L
tip. The cells were then washed twice with ice-cold PBS. The
pellet was then resuspended in ice-cold PBS, and 2 g/mL
propidium iodide was added. The cells were analyzed using
a 488 nm argon laser EPICS XL-MCL flow cytometer that
was controlled by EXPO 32-ADC software (Beckman Coul-
ter, Fullerton, CA, U.S.A.). The ROS values were analyzed
using a histogram plot of carboxy-H2DCFDA (the log of the
fluorescence). The results were calculated as the fold differ-
ence from the untreated control cells.
Glucose stimulated insulin secretion (GSIS)
Static incubation of the islets in Krebs-Ringer buffer that
contained either non-stimulatory or stimulatory concentrations
of glucose was performed for 1 hr (4). The insulin levels in the
Krebs-Ringer buffer samples collected from the static incuba-
tions (5.6 mM and 30 mM glucose conditions) from the islets
and INS-1 cells by using a 95.5 ethanol:hydrochloric
acid solu-
tion were measured by using a sensitive rat insul
in radioimmu-
noassay kit (Linco Research Immunoassay, St. Charles, MO,
U.S.A.).
HO activity assay
The HO activity was assayed by preparing a cell homogenate
from the INS-1 cells that were treated with hemin. The cell
homogenate was incubated with 50 M heme, 2 mg/mL rat
liver cytosol (the source of the biliverdin reductase), 1 mM
MgCl2, 3 U of glucose 6-phosphate dehydrogenase, 1 mM
glucose 6-phosphate, and 2 mM NADP+in 0.5 mL of 0.1
M potassium phosphate buffer, (pH 7.4) for 30 min at 37
.
Placing the tubes on ice terminated the reaction, and the
bilirubin was extracted with chloroform as described by Wa-
gener et al. (2). The amount of bilirubin generated was deter-
mined by using a scanning spectrophotometer (Lambda 17
UV/VIS, Perkin-Elmer Cetus Instruments, Norwalk, CT,
U.S.A.) and this was defined as the difference between the
wavelengths 464 and 530 nm (the extinction coefficient for
bilirubin was 40/mM/cm). The HO activity was expressed
as the picomoles of bilirubin formed per milligram of INS-1
cell protein per hour.
HO-1 sense and antisense oligodeoxynucleotide (ODN)
treatment
For inhibition of HO-1, pretreated HO-1 antisense ODNs
(2.5 mg/mL) were transfected using a lipofection reagent to
the INS-1 cells (35). The INS-1 cells at 50% confluence were
washed three times with PBS and then they were cultured
in medium containing 10% Nu-serum, which lacks nuclease
activity (Collaborative Research Inc., Waltham, MA, U.S.A.),
for 5 hr before the addition of the ODNs. N-[-(2,3-dioleoy-
loxy)propyl]-N,N,N-trimethylammonium methylsulfate
(DOTAP; Boehringer Manngeim Diagnostics, Indianapolis,
IN, U.S.A.) was used as a vehicle for transfecting the cells
with the ODNs. The proportions used were 2 g ODN/1 g
DOTAP/mL of medium and the protocol used was described
by the manufacturer. The phosphorothioated oligonucleotides
derived from the rat HO-1 sequence were synthesized at the
Life technologies, U.S.A.. The sense/antisense ODNs for the
420 K.C. Won, J.S. Moon, M.J. Eun, et al.
HO-1 were directed against the flanking translation initia-
tion codon in the human HO-1 cDNA. The antisense se-
quence for the HO-1 was 5
-GCGCTCCATCGCGGG-3
,
whereas the sense sequence for the HO-1 was 5
-CCCGC-
GATGGAGCGC-3
and for HO-1 scramble, it was 5
-GG-
CCCTCTACGGGCG-3
. Each ODN was phospho-rothioat-
ed on the first three bases on the 3
end and then it was puri-
fied by HPLC. The cells were incubated for 24 hr with the
ODNs, and then the medium was replaced with fresh medi-
um containing 10% fetal bovine serum; the cells were then
incubated for 24 hr in the absence or presence of heme.
Western blot analyses
The pancreatic islets and the INS-1 cells are washed once
in ice-cold PBS buffer. The cells were lysed in buffer (140
mM NaCL, 10 mM Tris pH 7.4, 1 mM CaCl2, 1 mM MgCl2,
10% glycerol, 1% Nonidet P-40 and 1×complete protease
inhibitors [Roche, Indianapolis, ID, U.S.A.]) and then homo-
g
enized by sonication for 2 to 20 sec on ice. The cell homog
e-
nates are centrifuged at 12,000 rpm at 4
for 10 min to
pellet the insoluble material. The supernatant is used for
Western
blot analyses. The protein was determined by the
bicinchonic
acid (BCA) assay (Pierce, Rockford, IL, U.S.A.).
The protein was fractionated by SDS-polyacrylamide gel elec-
trophoresis and then it was electroblotted to a nitrocellulose
membrane. Nonspecific binding sites are blocked by nonfat
dry milk for 1 hr at room temperature. The blots are then
incubated with the specific primary antibodies against HO-1
at a dilution of 1:2,000-1:5,000 for 2 hr at room temperature;
this was followed by a 1 hr incubation period with the peroxi-
dase-labeled secondary antibody at a dilution of 1:10,000.
The protein bands are visualized by chemiluminescence with
using the NEN detection system.
Data analysis
All the values are expressed as means
±
SE. Analysis of
variance and Student’s t-test were used for the statistical anal-
ysis, and the differences between groups were considered to
be significant at pvalues <0.05.
RESULTS
Intracellular peroxide levels in the islets at the high glucose
condition
The INS-1 cells cultured for 3 days in glucose concentra-
tions ranging from 5.6 to 30 mM had progressively greater
peroxide levels with the higher glucose concentrations (Fig.
1A, p<0.05). The rat islets cultured for 3 days in 30 mM or
50 mM ribose had greater peroxide levels than that in the
rat islets cultured in 11.1 mM glucose (Fig. 2B, p<0.05). In
addition, the cells at higher glucose or ribose concentrations
displayed a decreased GSIS (p<0.05).
HO-1 was induced in the INS-1 cells by the high glucose
levels
The INS-1 cells were cultured for 3 days in 5.6 mM or 30
mM glucose concentrations. Compared with 5.6 mM glu-
cose, 30 mM glucose caused an increase of the HO-1 expres-
sion and activity in the INS-1 cells (Fig. 2, p<0.05).
HO-1 downregulation in the INS-1 cells by the HO-1 anti-
sense
After 3 days culture (5 hrs exposure of the ODN) of the
INS-1 cells at 5.6 mM or 30 mM glucose concentrations,
the intracellular peroxide level, the HO-1 expression and
the GSIS were measured. HO-1 and GSIS were decreased
90
10
0
10
1
10
2
10
3
INS-1 cell
5.6 mM
22.2 mM
30 mM
A
Events
36
010
0
10
1
10
2
10
3
10
4
Fluorescence Intensity
Rat islet cell
11.1 mM Glucose
30 mM Ribose
50 mM Ribose
B
Events
Fluorescence intensity
6
5
4
3
2
1
05.6 mM G 30 mM G
Intracellular peroxide level
*
p<0.05 vs. 5.6 mM G
Fig. 1. The effects of high glucose on the intracellular peroxide
level and Glucose stimulating insulin secretion (GSIS) in the INS-
1 cells and rat islets. (A) INS-1 cells were incubated at 5.6, 22.2
or 30 mM glucose for 3 days. INS-1 cells incubated at 30 mM glu-
cose increased levels of intracellular peroxides compared with
the 5.6 mM concentration of glucose. (B) Isolated rat islets were
incubated with 11.1 mM glucose or 30 mM ribose for 3 days. 30
mM ribose caused an increase of intracellular peroxide levels
compared with the 11.1 mM glucose. Each cell at the high glucose
or ribose concentrations showed decreased GSIS (p<0.05). Data
are means
±
SD from 3 separate experiments.
Insulin secretion ( U/mL)
1,200
1,000
800
600
400
200
0
5.6 mM G 30 mM G
GSIS
4 mM G
Fluorescence intensity
25
20
15
10
5
011.1 mM G 30 mM R
Intracellular peroxide level
*
p<0.05 vs. 11.1 mM G
16.7 mM G
Insulin secretion ( U/Islet)
1,000
800
600
400
200
0
11.1 mM G 30 mM G
GSIS
5.6 mM G
16.7 mM G
*
*
*
Heme Oxygenase-1 Protects Glucose Toxicity in INS-1 Cells 421
simultaneously by treatment of the HO-1 antisense (Fig. 3,
p<0.05), suggesting GSIS is associated with HO-1.
HO-1 upregulation in the islets by hemin
The INS-1 cells cultured for 3 days (with 1day pre-expo-
sure of the hemin) in hemin concentrations ranging from
0.1 mM to 10 mM had progressively greater HO-1 levels
with the higher hemin concentrations, and the cells had pro-
gressively smaller peroxide levels with the higher hemin con-
centrations (Fig. 4A, p<0.05). Similar results were also ob-
tained in the rat islets (Fig. 4B, p<0.05).
HO-1 upregulation induced by hemin increased GSIS in
the INS-1 cells at high glucose conditions
The GSIS in both the INS-1 cells and rat islets after 3 days
subculture with high glucose concentration was increased
in a dose-dependent manner by additional treatment of hemin
for 1 day, which was associated with HO-1 upregulation
induced by hemin (Fig. 5, p<0.05).
DISCUSSION
Glucose toxicity is defined as the nonphysiological and
potentially irreversible cellular damage that results in defec-
tive insulin gene expression, and this is caused by chronic
exposure to supraphysiologic glucose concentrations (4-9).
With using HIT-T15 cells, Robertson et al. (4) observed that
cells chronically cultured for 6 months in media containing
11.1 mM glucose, a concentration exceeding what is neces-
sary to elicit maximal insulin responses, caused a marked
loss of insulin mRNA, greatly diminished levels of the insulin
content and almost a complete disappearance of insulin secre-
tion. In contrast, the HIT-T15 cells of the same passage that
were serially cultured in media containing 0.8 mM glucose
for 6 months retained their insulin mRNA, insulin content
and their glucose-induced insulin secretion (4). The concept
of glucose autoxidation along with the consequent excess
generation of ROS in relation to diabetes mellitus has been
proposed as early as 1987 by Wolff and Dean (17). Hunt et
al. (18) demonstrated that glucose autoxidation produces
hydroxyl radicals and that the hydroxyl radical scavengers
protected against the glucose-induced fragmentation of pro-
tein. Earlier work by Grankvist et al. (19) demonstrated that
the pancreatic islets contained relatively small amounts of
Fig. 2. The HO-1 expression and activity after 3 days subculture
of the INS-1 cells. Compared with the 5.6 mM glucose concen-
tration, 30 mM glucose caused an increase in the HO-1 expres-
sion and activity in the INS-1 cells (p<0.05). Data are means
±
SD
from 3 separate experiments.
Glucose concentrations
5.6 mM 5.6 mM 5.6 mM 30 mM 30 mM 30 mM
6
5
4
3
2
1
05.6 mM G 30 mM G
*
HO-1 expression
6
5
4
3
2
1
05.6 mM G 30 mM G
*
HO-1 activity
p<0.05 vs. 5.6 mM G
nmol bilirubin/mg protein/hr Intensity
Fluorescence intensity
6
5
4
3
2
1
05.6 mM G 30 mM G Scramble Sense Antisense
HO-1 expresion
*
p<0.05 vs. 30 mM G
*
p<0.05 vs. 30 mM G
Fig. 3. The intracellular peroxide level, HO-1 expression and GSIS
after 3 days culture (5 hrs exposure to the ODNs) in the INS-1 cells.
HO-1 was downregulated in the INS-1 cells by the HO-1 antisense
ODNs (p<0.05). Data are means
±
SD from 3 separate experi-
ments.
*
6
5
4
3
2
1
05.6 mM G 30 mM G Scramble Sense Antisense
30 mM G
Intracellular peroxide level
*
30 mM G
Insulin secretion (mU/mL)
1,200
1,000
800
600
400
200
05.6 mM G 30 mM G Scramble Sense Antisense
4 mM G
16.7 mM G
*
422 K.C. Won, J.S. Moon, M.J. Eun, et al.
antioxidant enzymes such as Cu, Zn-superoxide dismutase,
Mn-superoxide dismutase, catalase and glutathione peroxi-
dase. Oliveira et al. (20) reported that superoxide dismutase
activity increases with the increasing glucose concentration.
These observations set the stage for the increased risk for ROS-
induced damage. We also found that the intracellular perox-
Fluorescense intensity
7
6
5
4
3
2
1
05.6 mM G 30 mM G 0.1 M 1 M 10 M Hemin
INS-1 cells
HO-1 expression
Fig. 4. The intracellular peroxide level and the HO-1 expression and activity after 3 days subculture (1 day pre-exposure of Hemin) in the
INS-1 cells (A) and rat islets (B). HO-1 was upregulated in the INS-1 cells and rat islets by Hemin (p<0.05). Data are means
±
SD from 3
separate experiments.
A
Fluorescense intensity
12
10
8
6
4
2
011.1 mM G 50 mM R 0.1 M 1 M 10 M Hemin
Rat Islets
B
*
nmol bilirubin/mg protein/hr
9
8
7
6
5
4
3
2
1
05.6 mM G 30 mM G 0.1 M 1 M 10 M Hemin
HO-1 activity
*
Fluorescense intensity
6
5
4
3
2
1
05.6 mM G 30 mM G 0.1 M 1 M 10 M Hemin
HO-1 activity
*
*
Fluorescense intensity
25
20
15
10
5
011.1 mM G 50 mM R 0.1 M 1 M 10 M Hemin
*
*
p<0.05 vs. 30 mM G Glucose
*
p<0.05 vs. 50 mM G Ribose
30 mM G 50 mM R
Insulin secretion (mU/mL)
1,000
800
600
400
200
05.6 mM G 30 mM G 0.1 M 1 M 10 M Hemin
30 mM G
A
*
4 mM G
16.7 mM G
*
p<0.05
INS-1 Cell
Insulin secretion (mU/mL)
1,000
800
600
400
200
011.1 mM G 50 mM R 0.1 M 1 M 10 M Hemin
50 mM R
B
*
5.6 mM G
16.7 mM G
*
p<0.05
Rat Islets
Fig. 5. GSIS after 3 days subculture (1 day pre-exposure of Hemin) in the INS-1 cells (A) and rat islets (B). Hemin induced HO-1 upregula-
tion and it reserved the GSIS in the islets at the high glucose condition (p<0.05). Data are means
±
SD from 3 separate experiments.
Heme Oxygenase-1 Protects Glucose Toxicity in INS-1 Cells 423
ide level was increased by a chronic exposure of high glucose
conditions in both the INS-1 cells as well as rat islets, which
was accompanying with a decreased GSIS (Fig. 1).
The heme oxygenases play critical roles in physiological
iron homeostasis, antioxidant defense, and, as has been shown
from the accumulating evidences, in the signaling pathways
that employ CO as a messenger (21). Three mammalian iso-
forms of HO have been identified: HO-1, an inducible en-
zyme that is most highly concentrated in the tissues that are
heavily involved in the catabolism of heme proteins (22);
HO-2, a non-inducible isoform that is present at the high-
est concentrations in the brain and testes, and it is thought
to be particularly involved in signaling pathways (23); and
HO-3, an isoform with low catalytic activity and an uncer-
tain physiological role (24). HO uses dioxygen and nicoti-
namide-adenine dinucleotide phosphate as cofactors, and
the resulting products of the reaction are carbon monoxide,
iron and biliverdin (21). Biliverdin is converted to bilirubin
by a ubiquitous cytosolic enzyme biliverdin reductase (22).
Both HO-2 and an inducible HO-1 have been identified in
rat pancreatic islets (23-25), as well as in other tissues (26).
Pancreatic islets respond to stress through the induction and
activation of several stress-activated proteins. Interleukin-1
(IL-1 ) induces an inflammatory response in pancreatic islets
that is characterized by increased levels of inducible nitric
oxide synthase (iNOS) and increased nitric oxide (NO)/nitrite
levels (27-30). IL-1 and heat shock protein increase the
expression of hsp70 (31, 32), as well as HO-1 (23, 33). The
protective effect of heat shock protein (HSP) on the islet cells
may be associated with the reduced lysis from NO, reactive
oxygen intermediates and streptozotocin (32); but the response
is nonspecific because many HSPs respond to these stimuli.
On the other hand, liposomal delivery of hsp70 into islet
cells protected the cells from the IL-1 effects on insulin
secretion (34), suggesting that heightened levels of specific
HSP can protect cells from the inhibitory effects of the
cytokine. Hemin induces the synthesis of HO-1 and so it
partly counteracts the IL-1 induced inhibition of aconitase
activity and glucose oxidation (33), and perhaps this occurs
through antioxidant mechanisms. However, hemin has also
been reported to increase insulin and glucagon secretion from
normal rat islets (24). Ye and Laychock (1) reported that HO,
which is also known as a heat shock protein (hsp 32), is an
enzyme that may protect cells by reducing the heme levels
that catalyze the oxygen radical reactions, and it appears to
be a protective agent for pancreatic islets against interleukin-
1 . Similarly, our study has shown that compared with 5.6
mM glucose, 30 mM glucose caused an induction of HO-1
expression and activity in the INS-1 cells (Fig. 2, p<0.05).
The HO-1 expression increases in response to heme and to
such stressors as UV radiation and oxidative stress, as well as
to endotoxin, hormones and heavy metals (26). HO-1 induc-
tion may protect cells by reducing the heme levels that cat-
alyze oxygen radical reactions and by elevating bilirubin,
which has antioxidant properties (21). Bilirubin inhibits the
autoxidation or peroxyl-radical-induced oxidation of the
unsaturated fatty acids, and it apparently does so through
its peroxyl radical-trapping antioxidant abilities (35-37). In
the present study, we hypothesized that HO-1 can protect
the suppression of GSIS resulted from glucose toxicity.
To evaluate our hypothesis, we measured GSIS after cul-
tured with high glucose conditions in both HO-1 downreg-
ulated state and upregulated state. We observed that the
GSIS was decreased by treatment of HO-1 antisense ODNs,
which was accompanying with a downregulation of HO-1
expression (Fig. 3, p<0.05). Moreover, the GSIS was increased
incompletely by hemin administration associating with the
upregulation of HO-1 expression and activity (Fig. 4, 5).
These results in this study supported our hypothesis. Thus,
our results suggest that HO-1 seems to mediate the protec-
tive responses of the pancreatic islets against the oxidative
stress that is due to high glucose conditions. Also, we sug-
gested that HO-1 is one of the targets for preserving islets
from glucose toxicity in diabetic state.
REFERENCES
1. Ye J, Laychock SG. A protective role for heme oxygenase expres-
sion in pancreatic islets exposed to interleukin-1 . Endocrinology
1998; 139: 4155-63.
2. Wagener FA, da Silva JL, Farley T, de Witte T, Kappas A, Abraham
NG. Differential effects of heme oxygenase isoforms on heme medi-
ation of endothelial intracellular adhesion molecule 1 expression. J
Pharmacol Exp Ther 1999; 291: 416-23.
3. Pileggi A, Molano RD, Berney T, Cattan P, Vizzardelli C, Oliver R,
Fraker C, Ricordi C, Pastori RL, Bach FH, Inverardi L. Heme oxy-
genase-1 induction in islet cells results in protection from apoptosis
and improved in vivo function after transplantation. Diabetes 2001;
50: 1983-91.
4. Robertson RP, Zhang HJ, Pyzdrowski KL, Walseth TF. Preservation
of insulin mRNA levels and insulin secretion in HIT cells by avoid-
ance
of chronic exposure to high glucose concentration. J Clin Inves
t
1992; 90: 320-5.
5. Won KC. Oxidative stress in pancreatic islet beta-cells exposed to
high glucose concentration. J Korean Diabetes Assoc 2004; 28: 250
-
54.
6. Harmon JS, Stein R, Robertson RP. Oxidative stress-mediated, post-
translational loss of MafA protein as a contributing mechanism to
loss of insulin gene expression in glucotoxic beta cells. J Biol Chem
2005; 280: 11107-13.
7. Sharma A, Olson LK, Robertson RP, Stein R. The reduction of insulin
gene transcription in HIT-T15 beta cells chronically exposed to high
glucose concentration is associated with the loss of RIPE3b1 and
STF-1 transcription factor expression. Mol Endocrinol 1995; 9: 1127
-
34.
8. Harmon JS, Tanaka Y, Olson LK, Robertson RP. Reconstitution of
glucotoxic HIT-T15 cells with somatostatin transcription factor-1
424 K.C. Won, J.S. Moon, M.J. Eun, et al.
partially restores insulin promoter activity. Diabetes 1998; 47: 900-4.
9. Robertson RP. Chronic oxidative stress as a central mechanism for
glucose toxicity in pancreatic islet beta cells in diabetes. J Biol Chem
2004; 279: 42351-4.
10. Tiedge M, Lortz S, Munday R, Lenzen S. Complementary action of
antioxidant enzymes in the protection of bioengineered insulin-pro-
ducing RINm5F cells against the toxicity of reactive oxygen species.
Diabetes 1998; 47: 1578-85.
11. Kralik PM, Xu B, Epstein PN. Catalase transfection decreases hydro-
gen
peroxide toxicity in a pancreatic beta cell line. Endocr Res 1998
;
24: 79-87.
12. Tiedge M, Lortz S, Munday R, Lenzen S. Protection against the co-
operative toxicity of nitric oxide and oxygen free radicals by overex-
pression of antioxidant enzymes in bioengineered insulin-producing
RINm5F cells. Diabetologia 1999; 42: 849-55.
13. Hohmeier HE, Thigpen A, Tran VV, Davis R, Newgard CB. Stable
expression of manganese superoxide dismutase (MnSOD) in insuli-
noma cells prevents IL-1 -induced cytotoxicity and reduces nitric
oxide production. J Clin Invest 1998; 101: 1811-20.
14. Moriscot C, Pattou F, Kerr-Conte J, Richard MJ, Lemarchand P,
Benhamou PY. Contribution of adenoviral-mediated superoxide
dismutase gene transfer to the reduction in nitric oxide-induced cy-
totoxicity on human islets and INS-1 insulin-secreting cells. Diabe-
tologia 2000; 43: 625-31.
15. Asfari M, Janjic D, Meda P, Li G, Halban PA, Wollheim CB. Estab-
lishment
of 2-mercaptoethanol-dependent differentiated insulin-secret-
in
g cell lines. Endocrinology 1992; 130: 167-78.
16. Tanaka Y, Tran PO, Harmon J, Robertson RP. A role for glutathione
peroxidase in protecting pancreatic beta cells against oxidative stress
in a model of glucose toxicity. Proc Natl Acad Sci USA 2002; 99:
12363-8.
17. Wolff SP, Dean RT. Glucose autoxidation and protein modification.
The potential role of autoxidative glycosylation in diabetes. Biochem
J 1987; 245: 243-50.
18. Hunt JV, Dean RT, Wolff SP. Hydroxyl radical production and autox-
idative glycosylation. Glucose autoxidation as the cause of protein
damage in the experimental glycation model of diabetes mellitus and
ageing. Biochem J 1988; 256: 205-12.
19. Grankvist K, Marklund SL, Taljedal IB. CuZn-superoxide dismutase,
Mn-superoxide dismutase, catalase and glutathione peroxidase in
pancreatic islets and other tissues in the mouse. Biochem J 1981;
199: 393-8.
20. Oliveira HR, Curi R, Carpinelli AR. Glucose induces an acute increase
of superoxide dismutase activity in incubated rat pancreatic islets.
Am J Physiol Cell Physiol 1999; 276: 507-10.
21. McDonagh AF. Is bilirubin good for you? Clin Perinatol 1990; 17:
359-69.
22. George JW, Nulk K, Weiss A, Bruss ML, Cornelius CE. Biliverdin
reductase activity in cattle, sheep, rabbits and rats. Int J Biochem
1989; 21: 477-81.
23. Helqvist S, Polla BS, Johannesen J, Nerup J. Heat shock protein induc-
tion in rat pancreatic islets by recombinant human interleukin 1 .
Diabetologia 1991; 34: 150-6.
24. Henningsson R, Alm P, Lundquist I. Occurrence and putative hor-
mone regulatory function of a constitutive heme oxygenase in rat
pancreatic islets. Am J Physiol Cell Physiol 1997; 273: 703-9.
25. Welsh N, Margulis B, Borg LA, Wiklund HJ, Saldeen J, Flodstrom
M, Mello M, Andersson A, Pipeleers DG, Hellerstrom C, Eizirid DL.
Differences in expression of heat-shock proteins and antioxidant
emzymes between human and rodent pancreatic islets: implications
for the pathogenesis of insulin-dependent diabetes mellitus. Mol Med
1995; 1: 806-20.
26. Abraham NG, Lin JH, Schwartzman ML, Levere RD, Shibahara S.
The physiological significance of heme oxygenase. Int J Biochem
1998; 20: 543-58.
27. Southern C, Schulster D, Green IC. Inhibition of insulin secretion
by interleukin-1 and tumor necrosis factor necrosis factor- via
an L-arginine-dependent nitric oxide generating mechanism. FEBS
Lett 1990; 276: 42-4.
28. Welsh N, Eizirik DL, Bendtzen K, Sandler S. Interleukin-1 -induced
nitric oxide production in isolated rat pancreatic islets requires gene
transcription and may lead to inhibition in isolated of the Krebs cycle
enzyme aconitase. Endocrinology 1991; 129: 3167-73.
29. Corbett JA, Wang JL, Hughes JH, Wolf BA, Sweetland MA, Lan-
caster JR Jr, McDaniel ML. Nitric oxide and cyclic GMP formation
induced by interleukin 1 in islets of Langerhans: evidence for a role
of nitric oxide in islet dysfunction. Biochem J 1992; 287: 229-35.
30. Corbett JA, Sweetland MA, Wang JL, Lancaster JR, McDaniel ML.
Nitric oxide mediates cytokine-induced inhibition of insulin secretion
by human islets of Langerhans. Proc Natl Acad Sci USA 1993; 90:
1731-5.
31. Eizirik DL, Welsh M, Strandell E, Welsh N, Sandler S. Interleukin-1
beta depletes insulin messenger ribonucleic acid and increases the
heat shock protein hsp 70 in mouse pancreatic islets without
impair-
ing the glucose metabolism. Endocrinology 1990; 127: 2290-7.
32. Bellmann K, Wenz A, Radons J, Burkhart V, Kleemann R, Kolb H.
Heat shock induces resistance in rat pancreatic islet cells against
nitric oxide, oxygen radicals and streptozotocin toxicity in vitro. J
Clin Invest 1995; 95: 2840-5.
33. Welsh N, Sandler S. Protective action by hemin against interleukin-
1 induced inhibition of rat pancreatic islet function. Mol Cell En-
docrinol 1994; 103: 109-14.
34. Margulis BA, Sandler S, Eizirik DL, Welsh N, Welsh N, Welsh M.
Liposomal delivery of purified heat shock protein hsp 70 into rat
pancreatic islets as protection against interleukin-1 -impaired -
cell function. Diabetes 1991; 40: 1418-22.
35. Polte T, Abate A, Dennery PA, Achroder H. Heme oxygenase-1 is a
cGMP-inducible endothelial protein and mediates the cytoprotective
action of nitric oxide. Arterioscler Thromb Vasc Biol 2000; 20: 1209-
15.
36. Farrera JA, Jauma A, Ribo JM, Peire MA, Parallada PP, Roques-
Choua S, Bienvenue E, Seta P. The antioxidant role of bile pigments
evaluated by chemical tests. Bioorg Med Chem 1994; 2: 181-5.
37. Choi MK, Alam J. Heme oxygenase-1: function, regulation and
implication of a novel stress-inducible protein in oxidant-induced
lung injury. Am J Respir Cell Mol Biol 1996; 15: 9-19.
... MSCs are a source of pro-angiogenic and anti-apoptotic cytokines, including VEGF, HGF, IL-6 and TGFβ1. TGFβ1 triggers the production of heat shock protein 32 (HSP32) and X-linked inhibitor of apoptosis protein (XIAP), which protect islets by supressing oxidative stress [117], inflammation and β-cell apoptosis [118,119]. MSCs can also differentiate into endothelial cells to facilitate peri-islet vascularisation. ...
Article
Full-text available
Islet transplantation represents an effective treatment for patients with type 1 diabetes mellitus (T1DM) and severe hypoglycaemia unawareness, capable of circumventing impaired counterregulatory pathways that no longer provide protection against low blood glucose levels. The additional beneficial effect of normalizing metabolic glycaemic control is the minimisation of further complications related to T1DM and insulin administration. However, patients require allogeneic islets from up to three donors, and the long-term insulin independence is inferior to that achieved with solid organ (whole pancreas) transplantation. This is likely due to the fragility of islets caused by the isolation process, innate immune responses following portal infusion, auto- and allo-immune-mediated destruction and β-cell exhaustion following transplantation. This review covers the specific challenges related to islet vulnerability and dysfunction that affect long-term cell survival following transplantation.
... Enhancement of this gene has shown to alter diabetes development and polymorphisms in the Hmox1 gene promotor are thought to increase the risk for the development of T2D [33]. Furthermore, upregulation of the Hmox1 gene was shown to protect rat β cells from oxidative stress [34]. NQO1, another antioxidative enzyme, has been known to protect β cells against oxidative stress, including the stressor STZ [35]. ...
Article
Full-text available
Oxidative stress and lipotoxicity effects on pancreatic β cells play a major role in the pathogenesis of type 2 diabetes (T2D). Flavonoids and antioxidants are under study for their cytoprotective effects and antidiabetic potential. In this study, we aimed to compare the protective effect of the Rooibos components aspalathin, isoorientin, 3-hydroxyphloretin (3-OH) and green Rooibos extract (GRT) itself, and exendin-4 and N-acetylcysteine (NAC) as reference molecules, against lipotoxicity and oxidative stress. The insulin-producing β cell line INS1E was exposed to hydrogen peroxide or streptozotocin (STZ) to induce oxidative stress, and palmitate to induce lipotoxicity. Cell viability was assessed by a MTS cell viability assay. Antioxidant response and antiapoptotic gene expression was performed by qRT-PCR. Glucose transporter 2 (GLUT 2) transporter inhibition was assessed through 2-NBDG uptake. GRT and the flavonoids aspalathin and 3-hydroxyphloretin offered significant protection against oxidative stress and lipotoxicity. GRT downregulated expression of pro-apoptotic genes Txnip and Ddit3 . The flavonoids aspalathin and 3-hydroxyphloretin also downregulated these genes and in addition upregulated expression of antioxidant response genes Hmox1 , Nqo1 and Sod1 . Isoorientin gave no cytoprotection. Cytoprotection by Rooibos components was significantly higher than by NAC or exendin-4. Rooibos components strongly protect INS1E β cells against diabetogenic stress. Cytoprotection was associated with the upregulation of antioxidant response genes of the NRF2/KEAP1 pathway or suppression of the TXN system. The Rooibos molecules offered better protection against these insults than exendin-4 and NAC, making them interesting candidates as β cell cytoprotectants for therapeutic or nutraceutical applications.
... It has been reported that hemin can reverse the inhibitory effect of pretreatment with high glucose on glucose-stimulated insulin secretion in INS-1 cells [28]. Here, we found that hemin affects insulin signaling, and alleviates palmitate-induced insulin resistance in cultured primary hepatocytes and HFD-induced insulin resistance. ...
Article
Full-text available
Hemin is a breakdown product of hemoglobin. It has been reported that the injection of hemin improves lipid metabolism and insulin sensitivity in various genetic models. However, the effect of hemin supplementation in food on lipid metabolism and insulin sensitivity is still unclear, and whether hemin directly affects cellular insulin sensitivity is yet to be elucidated. Here we show that hemin enhances insulin-induced phosphorylation of insulin receptors, Akt, Gsk3β, FoxO1 and cytoplasmic translocation of FoxO1 in cultured primary hepatocytes under insulin-resistant conditions. Furthermore, hemin diminishes the accumulation of triglyceride and increases in free fatty acid content in primary hepatocytes induced by palmitate. Oral administration of hemin decreases body weight, energy intake, blood glucose and triglyceride levels, and improves insulin and glucose tolerance as well as hepatic insulin signaling and hepatic steatosis in male mice fed a high-fat diet. In addition, hemin treatment decreases the mRNA and protein levels of some hepatic genes involved in lipogenic regulation, fatty acid synthesis and storage, and increases the mRNA level and enzyme activity of CPT1 involved in fatty acid oxidation. These data demonstrate that hemin can improve lipid metabolism and insulin sensitivity in both cultured hepatocytes and mice fed a high-fat diet, and show the potential beneficial effects of hemin from food on lipid and glucose metabolism.
... Heme oxygenase-1 also contributes to metabolic control in both in vitro and in vivo models [20]. High glucose exposure leads to induction of heme oxygenase-1 gene expression and enzyme activity in islets in accordance with elevation in intracellular peroxide concentration [6,21,22], while chronic hyperglycemia results in a decrease in heme oxygenase activity in rat models [23,24]. Also, dissimilar expression of heme oxygenase-1 according to diabetes stage has been found, which indicates an increase in heme oxygenase-1 level in the early phases and a subsequent decrease in the late stages [11,12]. ...
Article
Full-text available
Aim Serum bilirubin level was negatively associated with the prevalence of metabolic syndrome (MetS) in previous cross-sectional studies. However, bilirubin variance preceding the development of MetS has yet to be investigated. We aimed to determine the effect of change in bilirubin concentration on the risk of incident MetS in healthy Korean adults. Methods We conducted a retrospective longitudinal study of subjects who had undergone at least four yearly health check-ups between 2006 and 2012. Of 24,185 total individuals who received annual check-ups, 11,613 non-MetS participants with a baseline bilirubin level not exceeding 34.2 μmol/l were enrolled. We evaluated the association between percent change in bilirubin and risk of incident MetS. Results During 55,407 person-years of follow-up, 2,439 cases of incident MetS developed (21.0%). Baseline serum bilirubin level clearly showed no association with the development of MetS in men but an independent significant inverse association in women which attenuated (hence may be mediated) by elevated homeostatic model assessment index 2 for insulin resistance (HOMA2-IR). However, increased risk for incident MetS was observed in higher percent change in bilirubin quartiles, with hazard ratios of 2.415 (95% CI 2.094–2.785) in men and 2.156 (95% CI 1.738–2.675) in women in the fourth quartile, compared to the lowest quartile, after adjusting for age, smoking status, medication history, alanine aminotransferase, uric acid, estimated glomerular filtration rate, fasting glucose, baseline diabetes mellitus prevalence, systolic blood pressure, waist circumference, and body mass index. The hazard ratios per one standard deviation increase in percent change in bilirubin as a continuous variable were 1.277 (95% CI 1.229–1.326) in men and 1.366 (95% CI 1.288–1.447) in women. Conclusions Increases in serum bilirubin concentration were positively associated with a higher risk of incident MetS. Serum bilirubin increment might be a sensitive marker for the development of MetS.
Article
Ferroptosis, the iron‐dependent, lipid peroxide‐mediated cell death, has garnered attention due to its critical involvement in crucial physiological and pathological cellular processes. Indeed, several studies have attributed its role in developing a range of disorders, including diabetes. As accumulating evidence further the understanding of ferroptotic mechanisms, the impact this specialized mode of cell death has on diabetic pathogenesis is still unclear. Several in vivo and in vitro studies have highlighted the association of ferroptosis with beta‐cell death and insulin resistance, supported by observations of marked alterations in ferroptotic markers in experimental diabetes models. The constant improvement in understanding ferroptosis in diabetes has demonstrated it as a potential therapeutic target in diabetic management. In this regard, ferroptosis inhibitors promise to rescue pancreatic beta‐cell function and alleviate diabetes and its complications. This review article elucidates the key ferroptotic pathways that mediate beta‐cell death in diabetes, and its complications. In particular, we share our insight into the cross talk between ferroptosis and other hallmark pathogenic mediators such as oxidative and endoplasmic reticulum stress regulators relevant to diabetes progression. Further, we extensively summarize the recent developments on the role of ferroptosis inhibitors and their therapeutic action in alleviating diabetes and its complications.
Article
Carbon monoxide (CO), a member of the multifunctional gasotransmitters family produced by heme oxygenases (i.e., HO-1 and HO-2), has received significant attention because of its involvement in carbohydrate metabolism. Experimental evidence indicates that both HO-2- and HO-1-derived CO stimulate insulin secretion, but the latter mainly acts as a compensatory response in pre-diabetes conditions. CO protects pancreatic β-cell against cytokine- and hypoxia-induced apoptosis and promotes β-cell regeneration. CO cross-talks with nitric oxide (NO) and hydrogen sulfide (H2S), other important gasotransmitters in carbohydrate metabolism, in regulating β-cell function and insulin secretion. These data speak in favor of the potential therapeutic application of CO in type 2 diabetes mellitus (T2DM) and preventing the progression of pre-diabetes to diabetes. Either CO (as both gaseous form and CO-releasing molecule) or pharmacological formulations made of natural HO inducers (i.e., bioactive components originating from plant-based foods) are potential candidates for developing CO-based therapeutics in T2DM. Future studies are needed to assess the safety/efficacy and potential therapeutic applications of CO in T2DM.
Article
Full-text available
Scope: Aspalathin, the main polyphenolic phytochemical of rooibos (Aspalathus linearis), has been attributed with health promoting properties, including a glucose lowering effect that could prove interesting for application as nutraceutical or therapeutic in (pre-) diabetics. Preservation of β cell mass in the pancreas is considered a key issue for diabetes prevention or treatment, therefore we aimed to investigate whether aspalathin also has β cell cytoprotective potential. Methods and results: Rat pancreatic islets and the β cell line INS1E were studied in vitro after exposure to various cytotoxic agents, namely streptozotocin, hydrogen peroxide or chronic high glucose. The effect of aspalathin on cell survival and apoptosis was studied. Expression of relevant cytoprotective genes was analyzed by qRT-PCR and proteins by Western blot. Aspalathin was found to protect β cells against cytotoxicity and apoptosis. This was associated with increased translocation of nuclear factor erythroid 2-related factor 2 (NRF2) and expression of its antioxidant target genes heme oxygenase 1 (Hmox1), NAD(P)H quinone dehydrogenase 1 (Nqo-1) and superoxide dismutase 1 (Sod1). Conclusion: It is proposed that aspalathin protects β cells against glucotoxicity and oxidative stress by increasing the expression of NRF2-regulated antioxidant enzymes. This indicates that aspalathin is an interesting β cell cytoprotectant. This article is protected by copyright. All rights reserved.
Article
Full-text available
In islet transplantation, in addition to immunologic and ischemic factors, the diabetic/hyperglycemic state of the recipient has been proposed, although not yet validated, as a possible cause of islet toxicity, contributing to islet loss during the engraftment period. Using a miniature swine model of islet transplantation, we have now assessed the effect of a persistent state of hyperglycemia on islet engraftment and subsequent function. An islet–kidney (IK) model previously described by our laboratory was utilized. Three experimental donor animals underwent total pancreatectomy and autologous islet transplantation underneath the renal capsule to prepare an IK at a load of ≤1,000 islet equivalents (IE)/kg donor weight, leading to a chronic diabetic state during the engraftment period (fasting blood glucose >250 mg/dL). Three control donor animals underwent partial pancreatectomy (sufficient to maintain normoglycemia during islet engraftment period) and IK preparation. As in vivo functional readout for islet engraftment, the IKs were transplanted across an immunologic minor or class I mismatch barrier into diabetic, nephrectomized recipients at an islet load of ∼4,500 IE/kg recipient weight. A 12-d course of cyclosporine was administered for tolerance induction. All experimental donors became diabetic and showed signs of end organ injury, while control donors maintained normoglycemia. All recipients of IK from both experimental and control donors achieved glycemic control over long-term follow-up, with reversal of diabetic nephropathy and with similar glucose tolerance tests. In this preclinical, large animal model, neither islet engraftment nor subsequent long-term islet function after transplantation appear to be affected by the diabetic state.
Article
Full-text available
The aim of this study was to investigate the protective effects of a chokeberry anthocyanin extract (CAE) on pancreatic β-cells (βTC3) exposed to hydrogen peroxide- (H2O2-) and high glucose- (HG-) induced oxidative stress conditions. In order to quantify individual anthocyanins high performance liquid chromatography (HPLC) coupled to photodiode array (PDA) was used. The identification of the fragment ion pattern of anthocyanins was carried out by electrospray ionization mass spectrometry (LC-ESI-MS). The results showed that physiologically achievable concentrations of CAE (1, 5, and 10 μM) protect βTC3 against H2O2- and HG-induced cytotoxicity. Antioxidant enzymes such as superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx) were increased in pancreatic β-cells pretreated with CAE compared to cells exposed to the prooxidant agents. GSH levels initially reduced after exposure to H2O2 and HG were restored by pretreatment with CAE. Insulin secretion in βTC3 cells was enhanced by CAE pretreatment. CAE restored the insulin pool and diminished the intracellular reactive oxygen species level in glucose-induced stress condition in βTC3 cells. These results demonstrate that anthocyanins from CAE were biologically active, showing a secretagogue potential and an antioxidative protection of enzymatic systems, conferring protection against H2O2 and glucose toxicity in βTC3 cells.
Article
Full-text available
Treatment of pancreatic islets with interleukin 1 (IL-1) results in a time-dependent inhibition of glucose-stimulated insulin secretion which has recently been demonstrated to be dependent on the metabolism of L-arginine to nitric oxide. In this report IL-1 beta is shown to induce the accumulation of cyclic GMP (cGMP) in a time-dependent fashion that mimics the time-dependent inhibition of insulin secretion by IL-1 beta. The accumulation of cGMP is dependent on nitric oxide synthase activity, since NG-monomethyl-L-arginine (a competitive inhibitor of nitric oxide synthase) prevents IL-1 beta-induced cGMP accumulation. cGMP formation and nitrite production induced by IL-1 beta pretreatment of islets are also blocked by the protein synthesis inhibitor, cycloheximide. The formation of cGMP does not appear to mediate the inhibitory effects of IL-1 beta on insulin secretion since a concentration of cycloheximide (1 microM) that blocks IL-1 beta-induced inhibition of glucose-stimulated insulin secretion and nitric oxide formation does not prevent cGMP accumulation, thus dissociating the two events. By using e.p.r. spectroscopy, IL-1 beta is shown to induce the formation of a g = 2.04 iron-nitrosyl feature in islets which is prevented by cycloheximide, demonstrating the requirement of protein synthesis for IL-1 beta-induced nitric oxide formation. Iron-nitrosyl complex-formation by islets confirms that IL-1 beta induces the generation of nitric oxide by islets, and provides evidence indicating that nitric oxide mediates destruction of iron-sulphur clusters of iron-containing enzymes. Consistent with the destruction of iron-sulphur centres is the finding that pretreatment of islets with IL-1 beta results in an approx. 60% inhibition of mitochondrial oxidation of D-glucose to CO2. Inhibition of islet glucose oxidation appears to be mediated by nitric oxide since both NMMA and cycloheximide prevent IL-1 beta-induced inhibition of glucose oxidation. These results show that IL-1 beta-induced nitric oxide formation parallels the ability of IL-1 beta to inhibit glucose-stimulated insulin secretion by islets, and that protein synthesis is required for IL-1 beta-induced nitric oxide formation. These results also suggest that nitric oxide mediates IL-1 beta-induced inhibitory effects on the pancreatic beta-cell by functioning as an effector molecule responsible for the destruction of iron-sulphur centres of iron-containing proteins, resulting in an impairment of mitochondrial function.
Article
Full-text available
Glucose toxicity of the pancreatic beta cell is considered to play a secondary role in the pathogenesis of type II diabetes mellitus. To gain insights into possible mechanisms of action of glucose toxicity, we designed studies to assess whether the loss of insulin secretion associated with serial passages of HIT-T15 cells might be caused by chronic exposure to high glucose levels since these cells are routinely cultured in media containing supramaximal stimulatory concentrations of glucose. We found that late passages of HIT cells serially cultured in media containing 11.1 mM glucose lost insulin responsivity and had greatly diminished levels of insulin content and insulin mRNA. In marked contrast, late passages of HIT cells cultured serially in media containing 0.8 mM glucose retained insulin mRNA, insulin content, and insulin responsivity to glucose in static incubations and during perifusion with glucose. No insulin gene mutation or alteration of levels of GLUT-2 were found in late passages of HIT cells cultured with media containing 11.1 mM glucose. These data uniquely indicate that loss of beta cell function in HIT cells passed serially under high glucose conditions is caused by loss of insulin mRNA, insulin content, and insulin secretion and is preventable by culturing HIT cells under low glucose conditions. This strongly suggests potential genetic mechanisms of action for glucose toxicity of beta cells.
Article
In order to further characterize the actions of recombinant interleukin-1 beta (rIL-1 beta) on the function of insulin-producing cells, the effects of different concentrations of the cytokine were studied on islets obtained from four different mouse strains (NMRI, NOD, C57BL/6, and C57BL/Ks). For this purpose the islets were exposed to rIL-1 beta (25, 50, or 100 U/ml) for a 48-h period in medium RPMI 1640 containing 10% calf serum and 11.1 mM glucose. In all groups and at the various rIL-1 beta concentrations tested, there was a similar 30-50% inhibition in glucose-induced insulin release, a 70-80% decrease in islet insulin content, and no significant differences in islet DNA content or insulin accumulation in the culture medium. To clarify the mechanisms underlying the decreased islet insulin content, rates of (pro)insulin biosynthesis and insulin messenger RNA (mRNA) contents were determined. Exposure of NMRI and C57BL/6 islets to 50 U/ml rIL-1 beta reduced the (pro)insulin biosynthesis by 40-50% and the insulin mRNA contents by 80-90%. The cytokine also induced an increased cellular content of the heat shock protein hsp70, as measured by western blot analysis, and a decrease in DNA biosynthesis, as measured by [methyl-3H]thymidine incorporation. However, exposure to rIL-1 beta did not decrease islet total protein biosynthesis, glucose oxidation, ATP content, ATP/ADP ratio, cAMP content, or polyamine contents. In conclusion, these data suggest that exposure of mouse islets to rIL-1 beta reduces DNA synthesis, insulin mRNA levels, and the biosynthesis of (pro)insulin, without equally impairing other cellular functions. The mechanisms behind these reductions seem to be different from those observed in rat islets, where a rIL-1 beta-induced impairment of substrate metabolism at the mitochondrial level seems to be related to the decrease of several cellular functions.
Article
The aim of this study was to characterize the dynamics and functional relevance of interleukin-1β (IL-1β)-induced nitric oxide production in isolated pancreatic islets. Thus, islets were isolated from adult rats, precultured for 3-5 days in medium RPMI-1640 plus 10% fetal calf serum, and then exposed to IL-1β for different time periods, after which islet nitrite production and aconitase activty were determined. IL-1β (5 ng/ml) did not increase islet nitrite production during the first hour of incubation. Moreover, the nitric oxide synthase inhibitor NG-monomethyl-L-arginine (Meth-arg; 5 mM) failed to prevent the initial (90 min) IL-1β-induced increase in islet insulin release. After 4, 7, and 24 h, however, nitrite production was increased by 50%, 93%, and 139%, respectively. Islet aconitase activity and glucose oxidation rates were decreased by 70% after incubation for 24 h with IL-1β. Both Meth-arg and Nα-p-tosyl-L-lysine chloromethyl ketone (0.1 mM), a protease inhibitor, could completely counteract the IL-1β-induced increases in nitrite production and inhibition of aconitase activity and glucose oxidation rates. In a separate series of experiments, islets were incubated for 60 min with or without IL-1β and the RNA synthesis inhibitor actinomycin-D (5 μg/ml) and subsequently incubated for another 9 h without any additions. The presence of actinomycin-D during the 1-h IL-1β incubation period prevented the IL-1β-induced rise in nitrite production and the IL-1β-induced inhibition of aconitase activity and insulin release. It is concluded that IL-1β-induced nitric oxide production is a late event which requires gene transcription and does not mediate the initial stimulatory effects of IL-1β on β-cell function. However, the gradually augmented rate of nitric oxide production may inhibit the enzyme aconitase, leading to a suppressed mitochondrial activity and a defective insulin release in response to nutrient secretagogues.
Article
BetaTC6-F7 cells like normal Beta cells were found to be highly sensitive to hydrogen peroxide and to possess very low levels of catalase. Therefore we tested whether overexpression of catalase could enhance resistance to hydrogen peroxide. Enzyme activity was increased forty fold by transient transfection of a catalase transgene. To assess protection from hydrogen peroxide a cotransfection method using a human growth hormone reporter gene was developed. Human growth hormone secretion was shown to be a suitable marker for insulin secretion since both hormones demonstrated virtually identical glucose dose response curves. Catalase transfection was found to provide significant protection against hydrogen peroxide indicating that low catalase may contribute to the sensitivity of cells to hydrogen peroxide.
Article
New insulin-secreting cell lines (INS-1 and INS-2) were established from cells isolated from an x-ray-induced rat transplantable insulinoma. The continuous growth of these cells was found to be dependent on the reducing agent 2-mercaptoethanol. Removal of this thiol compound caused a 15-fold drop in total cellular glutathione levels. These cells proliferated slowly (population doubling time about 100 h) and, in general, showed morphological characteristics typical of native beta-cells. Most cells stained positive for insulin and did not react with antibodies against the other islet hormones. The content of immunoreactive insulin was about 8 micrograms/10(6) cells, corresponding to 20% of the native beta-cell content. These cells synthesized both proinsulin I and II and displayed conversion rates of the two precursor hormones similar to those observed in rat islets. However, glucose failed to stimulate the rate of proinsulin biosynthesis. In static incubations, glucose stimulated insulin secretion from floating cell clusters or from attached cells. Under perifusion conditions, 10 mM but not 1 mM glucose enhanced secretion 2.2-fold. In the presence of forskolin and 3-isobutyl-1-methylxanthine, increase of glucose concentration from 2.8-20 mM caused a 4-fold enhancement of the rate of secretion. Glucose also depolarized INS-1 cells and raised the concentration of cytosolic Ca2+. This suggests that glucose is still capable of eliciting part of the ionic events at the plasma membrane, which leads to insulin secretion. The structural and functional characteristics of INS-1 cells remained unchanged over a period of 2 yr (about 80 passages). Although INS-2 cells have not been fully characterized, their insulin content was similar to that of INS-1 cells and they also remain partially sensitive to glucose as a secretagogue. INS-1 cells retain beta-cell surface antigens, as revealed by reactivity with the antigangloside monoclonal antibodies R2D6 and A2B5. These findings indicate that INS-1 cells have remained stable and retain a high degree of differentiation which should make them a suitable model for studying various aspects of beta-cell function.
Article
Interleukin 1 beta, potentiated by tumour necrosis factor alpha, is cytotoxic to pancreatic Beta cells in vitro. We have hypothesized that interleukin 1 beta induces oxygen free radicals in Beta cells. Since cytotoxicity induced by free radicals and by heat may activate the same cellular repair mechanism (the heat shock response), the aim of this study was to investigate the pattern of protein synthesis in isolated islets after exposure to interleukin 1 beta (150 pg/ml, 24 h), tumour necrosis factor alpha (50 ng/ml, 24 h) heat shock (43 degrees C, 30 min) and H2O2 (0.1 mmol/l, 20 min). By polyacrylamide gel electrophoresis, autoradiography, Western-blot analysis and partial peptide mapping of 35S-methionine labelled islets, interleukin 1 beta was found to induce a 73 kilodalton protein belonging to the heat shock protein family heat shock protein 70, a heat shock protein 90, and haem oxygenase. A minor induction of heat shock protein 73 and haem oxygenase was seen after H2O2. Interleukin 1 beta did not induce heat shock proteins in rat thyroid cells, rat mesangial cells or in human monocytes. Tumour necrosis factor alpha did not induce selective protein synthesis. Pre-exposure of islets to heat, tumour necrosis factor alpha, or H2O2 did not prevent the impairment of glucose-stimulated insulin release seen after 24 h of interleukin 1 beta exposure. The data are compatible with free radical induction by interleukin 1 beta. However, the heat shock response is not specific for oxidative injury, and previous studies have shown discrepant effects as to a protective effect of free radical scavengers against interleukin 1 beta-mediated beta-cytotoxicity.(ABSTRACT TRUNCATED AT 250 WORDS)
Article
Recently it has been demonstrated that heat shock protein 70 (hsp70) is induced in pancreatic islet cells during prolonged exposure to interleukin 1 beta (IL-1 beta). It is unclear whether this represents a cellular defense against the noxious action of IL-1 beta or whether hsp70 is involved in the suppressive action of the cytokine. To assess the role for hsp70 in isolated islets exposed to IL-1 beta, hsp70 was purified and introduced into cells of isolated rat pancreatic islets via the liposome technique. Delivery of hsp70 was efficient according to immunoblot analysis, but delivered hsp70 disappeared within 16 h. Hsp70-containing liposomes did not affect protein synthesis, insulin secretion, or islet insulin mRNA content. However, when hsp70 liposome-incubated islets were further exposed to IL-1 beta (25 U/ml) for 16 h, these islets released more insulin in response to glucose stimulation and contained more insulin mRNA than islets incubated with control liposomes and subsequently exposed to the cytokine. No protective effect of liposomes containing bovine serum albumin or ovalbumin were observed. We conclude that hsp70 may protect against IL-1 beta-induced impairment of pancreatic beta-cell function.
Article
Bilirubin is generally considered to be a diagnostically useful, sometimes toxic, metabolic waste product--and nothing more. Many studies, however, summarized in this article, have shown that bilirubin is an effective lipid-soluble antioxidant in vitro, even at physiologic concentrations, vying with even vitamin E in its ability to intercept and inhibit free radical chain reactions that generate hazardous lipid peroxides. These studies suggest that bilirubin may have a biochemical function in vivo and belong to a group of low-molecular weight antioxidants that together provide protection from cellular damage by endogenous-organic free radicals. Dovetailing with the notion that bilirubin may have a protective function is the recent discovery that heme oxygenase, one of the enzymes responsible for bilirubin formation, is a heat-shock protein, one of a group of proteins that are thought to protect organisms from oxidative and other forms of biochemical stress. Thus, the biochemical path from red to green to yellow may defend as well as degrade, and modest levels of the end product may possibly be physiologically beneficial.
Article
Inhibition of glucose-induced insulin secretion by interleukin-1 beta (IL-1 beta), or IL-1 beta plus tumour necrosis factor-alpha (TNF-alpha), was less marked when rat islets of Langerhans were cultured for 12 h with these cytokines in L-arginine-free medium as opposed to medium containing L-arginine (1 mM). Inhibition of secretion by IL-1 beta was further alleviated when islets were maintained in L-arginine-free medium supplemented with N-omega-nitro-L-arginine methyl ester (NAME), while synergism between IL-1 beta plus TNF-alpha was completely abolished. Tissue culture medium nitrite levels were raised in islets treated with IL-1 beta or TNF-alpha (48 h). Cytokine-stimulated nitrite production was not observed in islets cultured with NAME (1 mM). In conclusion, an L-arginine-dependent nitric oxide generating mechanism is involved in the inhibition of insulin secretion by IL-1 beta, and accounts for the phenomenon of synergism between IL-1 beta and TNF-alpha.