Вы находитесь на странице: 1из 9

Beneficial Effects of Antioxidants in Diabetes

Possible Protection of Pancreatic b-Cells Against Glucose


Toxicity
Hideaki Kaneto, Yoshitaka Kajimoto, Jun-ichiro Miyagawa, Taka-aki Matsuoka, Yoshio Fujitani,
Yutaka Umayahara, Toshiaki Hanafusa, Yuji Matsuzawa, Yoshimitsu Yamasaki, and Masatsugu Hori

Oxidative stress is produced under diabetic conditions for treating diabetes and provides further support for
and possibly causes various forms of tissue damage in the implication of oxidative stress in b-cell dysfunc-
patients with diabetes. The aim of this study was to tion in diabetes. Diabetes 48:23982406, 1999
examine the involvement of oxidative stress in the pro-
gression of pancreatic b-cell dysfunction in type 2 dia-
betes and to evaluate the potential usefulness of
antioxidants in the treatment of type 2 diabetes. We
used diabetic C57BL/KsJ-db/db mice, in whom antioxi-
dant treatment (N-acetyl-L-cysteine [NAC], vitamins C
plus E, or both) was started at 6 weeks of age; its
effects were evaluated at 10 and 16 weeks of age.
According to an intraperitoneal glucose tolerance test,
the treatment with NAC retained glucose-stimulated
I n general, the development of type 2 diabetes is asso-
ciated with pancreatic b-cell dysfunction occurring
together with insulin resistance. Normal b-cells can
compensate for insulin resistance by increasing
insulin secretion, but insufficient compensation leads to the
onset of glucose intolerance. Once hyperglycemia becomes
apparent, b-cell function progressively deteriorates: glu-
insulin secretion and moderately decreased blood glu-
cose levels. Vitamins C and E were not effective when cose-induced insulin secretion becomes further impaired
used alone but slightly effective when used in combi- and degranulation of b-cells becomes evident, often accom-
nation with NAC. No effect on insulin secretion was panied by a decrease in the number of b-cells (14). The signi-
observed when the same set of antioxidants was given ficance of hyperglycemia as a direct cause of these phe-
to nondiabetic control mice. Histologic analyses of the nomena, i.e., b-cell glucose toxicity, has been demonstrated
pancreases revealed that the b-cell mass was signifi- by various studies in vivo (5) and in vitro (610). Chronic
cantly larger in the diabetic mice treated with the hyperglycemia may impair b-cell function at the level of
antioxidants than in the untreated mice. As a possible insulin synthesis as well as insulin secretion; when b-cell
cause, the antioxidant treatment suppressed apoptosis
derived cell lines are exposed to a high glucose concentra-
in b-cells without changing the rate of b-cell prolifera-
tion, supporting the hypothesis that in chronic hyper- tion for a long period of time, insulin gene transcription and
glycemia, apoptosis induced by oxidative stress causes insulin content are dramatically reduced (610). These
reduction of b-cell mass. The antioxidant treatment changes are often accompanied by a decrease in expres-
also preserved the amounts of insulin content and sion of pancreatic and duodenal homeobox factor-1 (PDX-1)
insulin mRNA, making the extent of insulin degranula- (8,10). PDX-1 (also known as islet duodenum homeobox-1
tion less evident. Furthermore, expression of pancreatic [IDX-1], somatostatin transactivating factor-1 [STF-1], and
and duodenal homeobox factor-1 (PDX-1), a b-cell insulin promoter factor-1 [IPF1]) (1115), is a b-cellspe-
specific transcription factor, was more clearly visible in cific transcription factor that plays a major role in main-
the nuclei of islet cells after the antioxidant treatment. taining normal b-cell function, probably by regulating mul-
In conclusion, our observations indicate that anti-
tiple genes expressed in b-cells (14,1618).
oxidant treatment can exert beneficial effects in dia-
betes, with preservation of in vivo b-cell function. This Under diabetic conditions, reactive oxygen species (ROS)
finding suggests a potential usefulness of antioxidants are produced mainly through the glycation reaction (19,20),
which occurs in various tissues (21) and may play a role in the
From the Department of Internal Medicine and Therapeutics (H.K., Y.K., development of complications in diabetes (22). Although the
T.M., Y.F., Y.U., Y.Y., M.H.) and the Department of Internal Medicine and induction of the glycation reaction in diabetes was originally
Molecular Science (J.M., T.H., Y.M.), Osaka University Graduate School of found in neural cells and the lens crystalline, which are also
Medicine, Suita, Japan.
Address correspondence and reprint requests to Dr. Y. Kajimoto, known targets of diabetic complications, another target was
Department of Internal Medicine and Therapeutics, A8, Osaka University recently shown to be the b-cell (2325). Indeed, advanced gly-
Graduate School of Medicine, 2-2 Yamadaoka, Suita City, Osaka 565-0871, cosylation end products (AGEs) were shown to be
Japan. E-mail: kajimoto@medone.med.osaka-u.ac.jp.
Received for publication 12 May 1999 and accepted in revised form
detectable in b-cells kept under high glucose concentrations
16 August 1999. (24), and the level of 8-hydroxy-29-deoxyguanosine (8-OHdG),
ABC, avidin-biotin complex; AEC, 3-amino-9-ethylcarbozol; BrdU, 5-bromo- a marker for oxidative stress, is increased in b-cells of diabetic
2-deoxyuridine; BSA, bovine serum albumin; DAB, 3,39-diaminobenzidine Goto-Kakizaki (GK) rats (25). Also, the expression of antiox-
tetrahydrochloride; NAC, N-acetyl- L-cysteine; PDX-1, pancreatic and duo-
denal homeobox gene-1; ROS, reactive oxygen species; TUNEL, TdT- idant enzymes, such as superoxide dismutase, catalase, and
mediated dUTP-biotin nick end labeling. glutathione peroxidase, is known to be very low in islet cells
2398 DIABETES, VOL. 48, DECEMBER 1999
H. KANETO AND ASSOCIATES

compared with other tissues and cells (26). Therefore, once


b-cells face oxidative stress, they may be rather sensitive to
it, suggesting that glycation and subsequent oxidative stress
may in part mediate the toxic effect of hyperglycemia. As
direct support for this, we recently showed that glycation-
mediated ROS production reduces insulin gene transcrip-
tion (27) and also causes apoptosis of b-cells (23).
Although animals have their own antioxidant defense sys-
tems, the defense can be externally strengthened. This might
be especially true for the pancreas, since it has a relatively
weak intrinsic defense system against oxidative stress (26).
Antioxidants include N-acetyl-L-cysteine (NAC), which scav-
enges hydrogen peroxide, and vitamins C and E, which are
well-known dietary antioxidants. Vitamin E is lipophilic and
inhibits lipid peroxidation, scavenging lipid peroxyl radicals
to yield lipid hydroperoxides and the tocopheroxyl radical
(28). Vitamin C, a water-soluble vitamin, functions coopera-
tively with vitamin E by regenerating tocopherol from the
tocopheroxyl radical. To investigate the implication of
oxidative stress in b-cell glucose toxicity in vivo and also to
find a potential tool for protecting the b-cell function from
glucose toxicity, we examined the possible effects of antiox-
idant treatment (NAC, vitamins C plus E, or both) on the
preservation of b-cell function in diabetic C57BL/KsJ-db/db
mice. In this mouse, a well-known obese model for type 2 dia-
betes, hyperglycemia is induced because of increasing
insulin resistance and the subsequent insufficiency of the
b-cell compensation (29,30). Our results show that the
antioxidant treatment is beneficial for treating diabetes and
can provide protection to b-cells against glucose toxicity in
C57BL/KsJ-db/db mice.

RESEARCH DESIGN AND METHODS


Experimental protocol. We used C57BL/KsJ-db/db and nondiabetic C57BL/KsJ-
misty/misty female mice purchased from Clea Japan (Tokyo). The mice were
allowed free access to food and water in a specific pathogenfree environment.
At 6 weeks of age, C57BL/KsJ-db/db mice were divided into four groups. Mice in
the control group (group 1, n = 40) were kept on regular diet without antioxidants,
and mice in the other three groups were given high-antioxidant diets: group 2
(n = 20), 2.5% NAC; group 3 (n = 20), 0.5% vitamin C plus 0.5% vitamin E; group 4
(n = 40), both 2.5% NAC and 0.5% vitamin C plus 0.5% vitamin E. At 10 and 16 weeks
of age, certain numbers of mice in each group (see figure legends) were used for
physiologic or histologic analyses. The animal studies were conducted in accor-
dance with Principles of Laboratory Animal Care (National Institutes of Health FIG. 1. Body weights (A) and nonfasting plasma glucose levels (B) in
publication no. 8523). C57BL/KsJ-db/db mice (10 and 16 weeks of age) maintained on a diet
Glucose tolerance test. After an overnight fast, mice were injected intraperi- with and without antioxidants. , group 1; , group 2; , group 3;
toneally with glucose (1.0 g/kg body weight). Blood samples were taken at vari- , group 4. Data are means SE (n = 10). *P < 0.05 vs. group 1.
ous time points (0120 min). Blood glucose concentrations were measured by the
glucose oxidase method using a glucose analyzer (MS-GR101; Terumo, Tokyo),
and serum insulin concentrations were determined using a Lebis radioim- sections were then incubated with ABC reagent for 30 min, and positive reactions
munoassay kit (Shibayagi, Gunma, Japan) with mouse insulin as the standard. were visualized by incubation with the peroxidase substrate solution containing
Insulin tolerance test. After an overnight fast, mice were injected intraperi- 3,39-diaminobenzidine tetrahydrochloride (DAB) (Zymed Laboratories, San Fran-
toneally with 2 U/kg human regular insulin. Blood samples were taken at various cisco, CA). Nuclei were counterstained with methyl green.
time points (090 min), and blood glucose concentrations were measured as PDX-1 staining followed a similar procedure, except the mounted sections were
described above. incubated with Target Retrieval Solution (Dako) at 90C for 5 min before ABC was
Preparation of pancreas sections and immunohistochemical analyses. performed and antiPDX-1 antiserum (16) diluted 1:1,000 in PBS containing
The mice were anesthetized using pentobarbital sodium. A midline abdominal inci- 1% BSA was used.
sion was made, and pancreases were removed from the mice and fixed overnight Northern blot analyses. Total RNA was isolated from mouse pancreases, and
in a solution of 4% paraformaldehyde. Fixed tissues were processed routinely for Northern blot analyses were performed. Ten micrograms of total RNA was size-
paraffin embedding, and ~5-m sections were prepared and mounted on slides. fractionated and transferred onto a Hybond-N+ membrane. After hybridization with
Before each incubation with antibodies, the mounted sections were rinsed three a [32P]-labeled mouse insulin cDNA probe at 42C in the presence of 50% for-
times with phosphate-buffered saline (PBS). mamide, the membrane was washed twice with 13 SSPE (180 mmol/l NaCl,
For detection of insulin, the avidin-biotin complex (ABC) method was per- 10 mmol/l sodium phosphate, 1 mmol/l EDTA, pH 7.4) and 0.1% SDS at 50C for
formed using Vectastain ABC Kit (Vector Laboratories, Burlingame, CA). The 15 min. Kodak XAR film was exposed with an intensifying screen at 80C.
mounted sections were incubated for 30 min with guinea pig polyclonal anti-insulin Measurement of insulin content. Four mice in each group (10 and 16 weeks
antibody (Dako, Glostrup, Denmark) diluted 1:3,000 in PBS containing 1% bovine of age) were used to measure the insulin content. After a 6-h fast, the whole pan-
serum albumin (BSA). They were then incubated for 30 min with biotinylated anti- creas was excised, and insulin content in the pancreatic tissue was determined using
guinea pig IgG (Vector Laboratories), diluted 1:200, as the secondary antibody. The a Lebis radioimmunoassay kit (Shibayagi) with mouse insulin as the standard. The

DIABETES, VOL. 48, DECEMBER 1999 2399


ANTIOXIDANT TREATMENT PROTECTS b-CELLS

data were normalized with respect to protein concentration in the extract, which group 4, NAC and vitamins C plus E) at 10 or 16 weeks of
was measured using a protein assay (BioRad Laboratories, Richmond, CA). age (Fig. 1A). As shown in Fig. 1B, the nonfasting blood glu-
Detection of apoptosis. Apoptotic cells were detected by the TdT-mediated
dUTP-biotin nick end labeling (TUNEL) method using an in situ apoptosis detec-
cose levels in mice in groups 2 and 4 were lower than those
tion kit (Takara Biochemicals, Kyoto, Japan). Sections were treated with pro- in mice in group 1 at both 10 and 16 weeks of age. No signi-
teinase K (20 g/ml) for 15 min at room temperature and incubated with TdT ficant difference was observed between groups 1 and 3
enzyme for 60 min at 37C. After washing in PBS, the sections were further incu- (Fig. 1B). Intraperitoneal glucose tolerance tests performed
bated with antifluorescein isothiocyanate HRP conjugate for 30 min at 37C and
at 10 and 16 weeks of age revealed that glucose tolerance in
visualized with DAB. Using the same sections, insulin staining was also per-
formed as described above except 3-amino-9-ethylcarbozol (AEC) (Dako) was mice in groups 2 and 4 was ameliorated (Fig. 2A and B). Also,
used for visualization and nuclei were counterstained with hematoxylin. insulin secretion during the glucose tolerance test was
Evaluation of cell proliferation. 5-Bromo-2-deoxyuridine (BrdU) (Sigma, St. improved in mice in groups 2 and 4 compared with those in
Louis, MO), freshly dissolved in saline, was injected intraperitoneally into group 1 (Fig. 2C and D), indicating that NAC is beneficial for
C57BL/KsJ-db/db mice at a dose of 100 mg/kg 6 h before excision of the pancreas.
Sections were incubated for 60 min at 37C with a mouse monoclonal anti-BrdU
preservation of glucose tolerance. In contrast, the effect of
antibody diluted 1:10. After washing in PBS, sections were incubated for 30 min antioxidative vitamins is not evident when used alone: there
at 37C with sheep anti-mouse IgG antibody (diluted 1:10) conjugated with alka- was no difference in blood glucose or insulin levels
line phosphate (Cell Proliferation Kit; Amersham Japan, Tokyo) followed by between groups 1 and 3 at either 10 or 16 weeks of age
visualization with DAB. To detect insulin-producing cells, the same sections were
(Fig. 2AD). When used in combination with NAC, how-
stained for insulin as described above using AEC for visualization, and nuclei were
counterstained with hematoxylin. ever, the antioxidative vitamins exerted some beneficial
Morphometry. The total area of the pancreas was measured using a television effect: there were significant differences in blood glucose
monitor connected to a light microscope and microvideoscope system with a tablet levels (at 30 and 60 min after the glucose injection) and
measure unit (Krypton-40; Flovel, Tokyo). b-Cell numbers were calculated by insulin levels (at 30 and 120 min) between groups 2 and 4 at
counting the nuclei surrounded by cytoplasm positive for insulin staining. The mea-
surement and calculation were done with a total of 12 sections of the pancreas
16 weeks of age (Fig. 2B and D). In contrast to the positive
from 4 mice (3 sections per mouse) in each group. The total number of islets effects of the antioxidants in diabetic C57BL/KsJ-db/db
included in the 12 sections from each group was approximately 500700. mice, the same set of antioxidants (NAC and vitamins C plus
Statistical analyses. Data are means SE. Statistical comparisons of means E) did not alter the glucose tolerance in nondiabetic
among individual groups used analysis of variance (ANOVA) followed by post-hoc
C57BL/KsJ-misty/misty mice (data not shown).
testing with Fishers least significant difference test.
To investigate the possible effects of antioxidant adminis-
tration on insulin sensitivity/resistance, an intraperitoneal
RESULTS insulin tolerance test was performed. As shown in Fig. 3,
Glucose tolerance. No differences in food intake or body reduction of blood glucose levels in response to the injected
weight were observed among the four groups (group 1, insulin (2 U/kg) was similar in the four groups at both 10 and
untreated; group 2, NAC alone; group 3, vitamins C plus E; 16 weeks of age.

FIG. 2. Glucose tolerance in C57BL/KsJ-db/db


mice treated with antioxidants. Intraperitoneal
glucose tolerance tests were performed in
C57BL/KsJ-db/db mice in each group at 10
(A and C) and 16 (B and D) weeks of age. After
an overnight fast, glucose was injected intra-
peritoneally at a dose of 1 g/kg, and blood glu-
cose (A and B) and insulin (C and D) levels
were measured. s, Group 1 (untreated);
d, group 2 (diet with NAC supplementation);
h, group 3 (diet with vitamin C plus E supple-
mentation); j, group 4 (diet with NAC and vit-
amin C plus E supplementation). Data are
means SE (A, n = 10; B, n = 12; C and D, n = 6
each). *P < 0.05 vs. group 1; **P < 0.01 vs. group
1; #P < 0.05 vs. group 2.

2400 DIABETES, VOL. 48, DECEMBER 1999


H. KANETO AND ASSOCIATES

FIG. 3. Insulin resistance in C57BL/KsJ-db/db mice


treated with antioxidants. Intraperitoneal insulin tol-
erance tests were performed in C57BL/KsJ-db/db mice
(10 and 16 weeks of age) given diet with and without
antioxidants. After an overnight fast, insulin was
injected at a dose of 2 U/kg. s, Group 1; d, group 2; h,
group 3; j, group 4. Data are means SE (n = 8).

Islet morphology. To elucidate how glucose tolerance is pre- in group 2 showed immunostaining results similar to group 4,
served in antioxidant-treated mice, insulin immunostaining while mice in group 3 were similar to group 1 (data not
was performed with the pancreases from C57BL/KsJ-db/db shown), suggesting that the antioxidative vitamins did not
mice in each group. The results revealed that islets of the exert significant effects on islet morphology.
16-week-old mice in group 4 (Fig. 4B) were larger and more To further elucidate the potency of antioxidants in pre-
numerous than those in group 1 mice (Fig. 4A and B). Mice serving b-cell function in diabetes, we tried to examine the

FIG. 4. Effects of antioxidants on b-cell


mass. A representative result is shown
for the insulin immunostaining per-
formed with pancreatic tissue sections
derived from C57BL/KsJ-db/db mice (16
weeks of age) in group 1 (A) and group 4
(B). In group 1 mice, islets were irregu-
lar in shape and islet cells showed
marked insulin degranulation. In con-
trast, islets in group 4 mice were plump
and revealed intense insulin immuno-
staining. Bar, 200 m. C: b-Cell mass was
quantitatively evaluated by counting
b-cell numbers per square millimeter of
the pancreatic section. The counting was
performed in the mice (10 and 16 weeks
of age) in group 1 (j) and group 4 (h).
Data are means SE (n = 4). *P < 0.05.

DIABETES, VOL. 48, DECEMBER 1999 2401


ANTIOXIDANT TREATMENT PROTECTS b-CELLS

FIG. 5. Insulin content in C57BL/KsJ-db/db mice treated with antiox-


idants. Insulin content in pancreases was measured at 10 and
16 weeks of age in C57BL/KsJ-db/db mice in group 1 (j) and group 4
(h). Data are means SE (n = 4). *P < 0.05; **P < 0.01.

mechanism underlying the preservation of insulin secretion


and islet morphology. For this purpose, we focused on the
mice in group 4, in which we assumed the maximal potency
of antioxidants might be revealed.
b-Cell mass. Using pancreas sections from mice in groups
1 and 4, we quantitatively evaluated the b-cell mass. For
each group, sections containing ~500700 islets were pre-
pared, and the b-cell number per square millimeter was cal-
culated. As shown in Fig. 4C, the number of b-cells in the
10-week-old mice in group 4 was higher than that of the
mice in group 1, and this difference became more evident
at 16 weeks of age.
Insulin synthesis. The results of insulin staining also
revealed that insulin degranulation was less evident in the
islets of mice in group 4 (Fig. 4A and B). Data for the insulin
content (Fig. 5), which showed a significant difference FIG. 6. Insulin mRNA levels in C57BL/KsJ-db/db mice treated with
between the mice in groups 1 and 4, supported this observa- antioxidants. A representative result is shown for Northern blot
tion. Although the decrease in insulin content in the whole analyses performed using mouse insulin cDNA as the probe. Total
RNA was isolated from pancreases of C57BL/KsJ-db/db mice in group
pancreas was in part due to the decrease in b-cell mass 1 and group 4 at 10 (A) and 16 (B) weeks of age. C: Relative insulin
(Fig. 4C), the insulin content per cell, estimated by normal- mRNA levels were evaluated by densitometry, with those of 10-week-
izing the data (Fig. 5) with respect to b-cell number (Fig. 4C), old mice in group 1 arbitrarily set at 1. Data are means SE (n = 4).
was also higher in the mice in group 4 (data not shown). *P < 0.05; **P < 0.01.
Consistent with this observation, the results of Northern blot
analyses revealed that the insulin mRNA level in the mice in
group 4 is significantly higher than that in the mice in group 1 PDX-1 expression was markedly retained, with clear
(Fig. 6). Again, the preservation in the insulin mRNA in part immunostaining in the nuclei (Fig. 7).
depends on the preservation in b-cell mass (Fig. 4C), but the b-Cell apoptosis. To examine the background for the
insulin mRNA per cell, normalized with respect to b-cell num- preservation of b-cell mass in the antioxidant-treated mice,
ber, was also higher in the mice in group 4 (data not shown). b-cell apoptosis was evaluated. Using double-immunostain-
Immunostaining for PDX-1. To understand the back- ing for insulin and apoptotic cells, ~500700 islets were
ground for the preservation in insulin biosynthesis, we examined for each group of mice. In the islets of mice in
examined the possible effects of the antioxidants on the group 1, 4.8 TUNEL-positive b-cells per 100 islets were rec-
expression of a transcription factor, PDX-1 (1118). ognized. On the other hand, only a few TUNEL-positive b-cells
Immunostaining for PDX-1 was performed in the pancreases (0.6 per 100 islets) could be identified in the islets of the
of C57BL/KsJ-db/db mice in groups 1 and 4. Consistent with mice in group 4 (Fig. 8).
a previous observation obtained with b-cells after partial b-Cell proliferation rate. To further understand the back-
pancreatectomy (5), we found that PDX-1 expression was ground for the preservation of b-cell mass in antioxidant-
reduced in the mice in group 1. In group 4 mice, however, treated mice, we examined the possible effects of the antiox-
2402 DIABETES, VOL. 48, DECEMBER 1999
H. KANETO AND ASSOCIATES

FIG. 7. Immunostaining for PDX-1 in C57BL/KsJ-db/db mice treated with antioxidants. Pancreases were immunostained for PDX-1 in mice (16
weeks of age) in group 1 (A) and group 4 (B). In group 1 mice, weak immunostaining for PDX-1 was observed in islet cells. This contrasts with
the clear immunostaining in nuclei of the islet cells in the mice in group 4. Bar, 50 m. Similar results were obtained with 12 pairs of sections
from 4 mice each of groups 1 and 4.

idants on the cell proliferation rate of the b-cells. For this pur- of pancreatic b-cell function in diabetic C57BL/KsJ-db/db
pose, we measured the percentage of S-phase cells by mon- mice. Since the same set of antioxidants did not alter the glu-
itoring BrdU incorporation (31). In mice from groups 1 and 4, cose tolerance in nondiabetic C57BL/KsJ-misty/misty mice,
~500700 islets were examined, and the ratio of the BrdU-pos- the antioxidant treatment probably exerts its effect in asso-
itive b-cells was calculated. As shown in Fig. 9A and B, some ciation with the presence of hyperglycemia; i.e., by protect-
BrdU-positive cells were detectable in the islets of both ing b-cells from the toxic effects of ROS produced under
group 1 and group 4. Double-immunostaining for BrdU and hyperglycemic conditions. Indeed, the antioxidant treatment
insulin revealed that most of the BrdU-positive cells in the increased the b-cell mass (Fig. 4) and preserved insulin con-
islets were insulin-containing b-cells. There was no difference tent (Fig. 5) and mRNA amount (Fig. 6). Since decreases in
in the ratio of BrdU-positive b-cells between groups 1 and 4 b-cell mass and insulin biosynthesis are considered to be
at either 10 or 16 weeks of age (Fig. 9C). associated with the development of diabetes in various ani-
mal models with type 2 diabetes (32,33), it is likely that those
DISCUSSION effects of the antioxidants contributed to partial preservation
In this study, we have shown that antioxidant treatment of the glucose tolerance in the antioxidant-treated mice
using NAC can improve glycemic control with preservation (Figs. 1B and 2).

FIG. 8. Effects of antioxidants on b-cell apoptosis. A representative result for double-immunostaining for insulin (red) and apoptotic cells
(brown) (visualized by TUNEL method) performed on pancreases of C57BL/KsJ-db/db mice (16 weeks of age) in group 1 (A) and group 4 (B).
The arrow indicates the b-cell identified as being apoptotic. Bar, 50 m. In each group, 12 pairs of sections from 4 mice each of groups 1 and 4
were examined, and similar results were obtained.

DIABETES, VOL. 48, DECEMBER 1999 2403


ANTIOXIDANT TREATMENT PROTECTS b-CELLS

FIG. 9. Effects of antioxidants on b-cell proliferation. A representative result for double-immunostaining for insulin (red) and BrdU (dark brown)
performed in pancreases of C57BL/KsJ-db/db mice (16 weeks of age) in group 1 (A) and group 4 (B). Several BrdU-positive b-cells per islet
(arrows) were recognized in both groups. Bar, 50 m. In each group, 12 pairs of sections from 4 mice each of groups 1 and 4 were examined,
and similar results were obtained. C: Percentage of BrdU-positive cells in b-cells calculated in mice (10 and 16 weeks of age) in group 1 (j)
or group 4 (h). Data are means SE (n = 4).

There was no difference in insulin sensitivity between reduced glucose toxicity and contributed in part to the pre-
groups 1, 2, 3, and 4 in the insulin tolerance test (Fig. 3), sug- vention of a decrease of b-cell mass and insulin content.
gesting that improvement of glycemic control was mainly Once the vicious circle of the glucose toxicity is prevented by
initiated by a beneficial effect of antioxidant on b-cells. How- somehow achieving good glycemic control, the circle then
ever, we cannot totally deny the possibility that the antioxi- starts moving in the opposite direction: the toxic effects of
dant treatment could have exerted an influence on target tis- high glucose will be reduced, resulting in further improvement
sues other than the b-cells such as muscle and fat; it was of glycemic control. Nonetheless, we assume that the antiox-
recently reported that oxidative stress impaired insulin- idants used in this study probably kept providing protection
induced GLUT4 translocation in 3T3-L1 adipocytes (34). of b-cells from the toxic effects of glucose. While the average
Thus, it would be safe to conclude that antioxidant treat- nonfasting glucose levels in mice treated with antioxidants
ment has beneficial effects on preservation of b-cell function (NAC and vitamins C plus E) were >20 mmol/l at 10 and
in diabetes, although the effects may not be exerted totally 16 weeks of age (Fig. 1B), Robertson and colleagues (610)
through its direct action on b-cells. Also, regardless of the have shown that chronic exposure of the b-cellderived HIT-
influence on insulin sensitivity, because the antioxidant treat- T15 or bTC6 cells to a glucose level of 11.1 mmol/l caused
ment indeed reduced blood glucose levels, it must have dramatic reductions in insulin gene transcription and insulin
2404 DIABETES, VOL. 48, DECEMBER 1999
H. KANETO AND ASSOCIATES

content. Also, according to a result of fluorescence-activated these observations suggest that the amount of PDX-1 expres-
cell sorting (FACS) analysis, there was a significant increase sion is important for maintaining normal b-cell function, it is
in ROS amount in the HIT-T15 cells kept under 20 mmol/l glu- possible that the suppression of PDX-1 function by ROS in dia-
cose compared with cells kept under 5 mmol/l glucose (H.K., betes is implicated in further deterioration of b-cell function
Y.K., unpublished observations). Thus, we assume that even and thus mediates glucose toxicity.
after the moderate decrease of blood glucose levels was Although both NAC and vitamins C and E have antioxida-
achieved in the antioxidant-treated mice, the blood glucose tive activity, only NAC ameliorates glucose tolerance when
levels in those mice remained high enough to cause glucose used alone (Figs. 1B and 2). The main function of vitamins C
toxicity, and that the antioxidants kept protecting the b-cells and E is to suppress lipid peroxidation, which occurs in the
against it by neutralizing the toxic effects of oxidative stress plasma membrane and damages membrane structure and
provoked due to hyperglycemia. permeability. Therefore, the antioxidative vitamins C and E
As possible mechanisms underlying the preservation of may not have been able to exert an effect on intracellular
the b-cell mass, we found that apoptosis was suppressed in events, such as those involved in apoptosis and gene tran-
the mice in group 4 (Fig. 8). b-Cell apoptosis has been sug- scription. In other words, inhibition of lipid peroxidation by
gested as being involved in physiologic and pathologic antioxidative vitamins (C and E) was not enough to improve
decrease of b-cell mass (33,35,36). According to a report by b-cell function. On the other hand, NAC scavenges hydrogen
Donath et al. (35), hyperglycemia induces b-cell apoptosis in peroxide, which is produced in the cytoplasm and probably
Psammomys obesus islets. Also, Pick et al. (33) showed that the nuclei of cells as a direct consequence of the glycation
apoptosis plays a role in the failure of b-cell mass compen- reaction (19,20). Once produced, hydrogen peroxide has a rel-
sation in Zucker diabetic fatty rats, providing support for the atively long lifespan and can transfer anywhere by penetrat-
possible involvement of apoptosis in b-cell glucose toxicity. ing nuclear and plasma membranes. Indeed, hydrogen per-
Although not very many TUNEL-positive cells were identified oxide is known to mediate the glycation-dependent degra-
even in the untreated mice (group 1), it should be noted that dation of several proteins and is widely involved in the
the frequency of apoptosis tends to be underestimated, since damage of various tissues in diabetes (20,44). Therefore, it
apoptotic cells are usually eliminated rather quickly. While seems reasonable that NAC, which is known to decrease the
glycation-mediated ROS production induces apoptosis in intracellular hydrogen peroxide level in b-cells (23), was
b-cells in vitro (17), our present results support the hypoth- effective for preventing b-cell damage. Although the antiox-
esis that ROS, provoked by hyperglycemia in vivo, play a idative vitamins did not reveal significant effects when used
significant role in decreasing the b-cell mass in type 2 diabetes alone, they exerted some beneficial effects when used in
through induction of apoptosis. Failure to detect a differ- combination with NAC (Fig. 2B and D). While many of the
ence in rate of b-cell proliferation between the groups with molecules constituting the machinery for the glucose-respon-
and without antioxidant treatment also supports this possi- sive insulin secretion are membrane proteins, it seems pos-
bility (Fig. 9). sible that vitamins C and E improved the outcome of NAC
The homeodomain containing transcription factor PDX-1 is treatment through their effects on such membrane proteins.
involved in pancreas development and, possibly, also in In conclusion, our present results show for the first time
regeneration after birth (18,3740). It is expressed in mature that antioxidants can exert beneficial effects on pancreatic
b-cells and functions as an important transcription factor b-cell function in diabetes. Thus a sufficient supply of antiox-
common to multiple genes essential for the b-cell function idants may prevent or delay b-cell dysfunction in diabetes by
(1418). When the b-cellderived cell line HIT-T15 was kept providing protection against glucose toxicity.
under a high glucose concentration for a long period of time,
a reduction of PDX-1 activity was identified in association ACKNOWLEDGMENTS
with a reduction of insulin gene transcription (8,10). Also, This work was supported in part by grants from Suzuken
Zangen et al. (5) have shown that the expression level of Memorial Foundation (to Y.K.) and a grant-in-aid for Scien-
PDX-1 is reduced in islets from partially pancreatectomized tific Research from the Ministry of Education of Japan (to
rats, which were exposed to chronic hyperglycemia in vivo. Y.K. and Y.Y.).
Although the limitations of mouse islet cell preparation pre- We thank Noriko Fujita and Katsumi Yamamori for the
vented reliable quantification concerning PDX-1 expression excellent technical assistance. We also thank Dr. M. Alan
levels and DNA-binding activity, we observed clear immuno- Permutt of the University of Washington School of Medicine
staining in nuclei of the islet cells in group 4, which contrasts and Dr. John M. Chirgwin of the University of Texas Health
with weak immunostaining for PDX-1 in islet cells in group 1 Science Center for kindly providing the mouse insulin II
(Fig. 7). These results provide further support for the vulner- cDNA plasmid.
ability of PDX-1 to chronic high glucose conditions and also
suggest the involvement of oxidative stress in hyperglycemia- REFERENCES
dependent reduction of PDX-1 activity, agreeing with the 1. Porte D Jr: Banting lecture 1990: b-cells in type II diabetes mellitus. Diabetes
40:166180, 1991
observation that PDX-1 activity in HIT cells was reduced by 2. DeFronzo RA, Bonadonna RC, Ferrannini E: Pathogenesis of NIDDM: a bal-
induction of oxidative stress in vitro (27). Clinically, it was anced overview. Diabetes Care 15:318368, 1992
shown that heterozygous mutations of PDX-1 cause diabetes, 3. Yki-Jarvinen H: Glucose toxicity. Endocrine Rev 13:415431, 1992
typically maturity-onset diabetes of the young type 4 4. Vinik A, Pittenger G, Rafaeloff R, Rosenberg L, Duguid W: Determinants of pan-
(MODY 4) (41,42). Also, a phenotype of the heterozygous creatic islet cell mass: a balance between neogenesis and senescence/apo-
ptosis. Diabetes Rev 4:235263, 1996
PDX-1 knockout mouse, which displays a decreased b-/a-cell 5. Zangen DH, Bonner-Weir S, Lee CH, Latimer JB, Miller CP, Habener JF, Weir
ratio and subsequent impaired glucose tolerance, indicates that GC: Reduced insulin, GLUT2, and IDX-1 in b-cells after partial pancreatectomy.
the PDX-1 gene has a dose-dependent effect (43). Whereas Diabetes 46:258264, 1997

DIABETES, VOL. 48, DECEMBER 1999 2405


ANTIOXIDANT TREATMENT PROTECTS b-CELLS

6. Robertson RP, Zhang H-J, Pyzdrowski KL, Walseth TF: Preservation of insulin release and biosynthesis: evidence that the formation of advanced glycosy-
mRNA levels and insulin secretion in HIT cells by avoidance of chronic expo- lation end products inhibits B cell function. Endocrinology 138:273280, 1997
sure to high glucose concentrations. J Clin Invest 90:320325, 1992 25. Ihara Y, Toyokuni S, Uchida K, Odaka H, Tanaka T, Ikeda H, Hiai H, Seino Y,
7. Olson LK, Redmon JB, Towle HC, Robertson RP: Chronic exposure of HIT cells Yamada Y: Hyperglycemia causes oxidative stress in pancreatic b-cells of GK
to high glucose concentrations paradoxically decreases insulin gene tran- rats, a model of type 2 diabetes. Diabetes 48:927932, 1999
scription and alters binding of insulin gene regulatory protein. J Clin Invest 26. Tiedge M, Lortz S, Drinkgern J, Lenzen S: Relation between antioxidant
92:514519, 1993 enzyme gene expression and antioxidative defense status of insulin-produc-
8. Sharma A, Olson LK, Robertson RP, Stein R: The reduction of insulin gene tran- ing cells. Diabetes 46:17331742, 1997
scription in HIT-T15b cells chronically exposed to high glucose concentration 27. Matsuoka T, Kajimoto Y, Watada H, Kaneto H, Kishimoto M, Umayahara Y, Fuji-
is associated with loss of RIPE3b1 and STF-1 transcription factor expression. tani Y, Kamada T, Kawamori R, Yamasaki Y: Glycation-dependent, reactive oxy-
Mol Endocrinol 9:11271134, 1995 gen species-mediated suppression of the insulin gene promoter activity in HIT
9. Poitout V, Olson LK, Robertson RP: Chronic exposure of bTC-6 cells to sup- cells. J Clin Invest 99:144150, 1997
raphysiologic concentrations of glucose decreases binding of the RIPE3b1 28. Stahl W, Sies H: Antioxidant defense: vitamins E and C and carotenoids. Dia -
insulin gene transcription activator. J Clin Invest 97:10411046, 1996 betes 46 (Suppl. 2):1418, 1997
10. Moran A, Zhang H-J, Olson LK, Harmon JS, Poitout V, Robertson RP: Differ- 29. Hummel KP, Dick MM, Coleman DL: Diabetes, a new mutation in the mouse.
entiation of glucose toxicity from beta cell exhaustion during the evolution Science 153:11271128, 1966
of defective insulin gene expression in the pancreatic islet cell line, HIT-T15. 30. Coleman DL, Hummel KP: Studies with the mutation, diabetes, in the mouse.
J Clin Invest 99:534539, 1997 Diabetologia 3:238248, 1966
11. Ohlsson H, Karlsson K, Edlund T: IPF1, a homeodomain-containing-transac- 31. deFrazio A, Leary JA, Hedley DW, Tattersall NHN: Immunohistochemical
tivator of the insulin gene. EMBO J 12:42514259, 1993 detection of proliferating cells in vivo. J Histochem Cytochem 35:571577, 1987
12. Leonard J, Peers B, Johnson T, Ferreri K, Lee S, Montminy MR: Characteri- 32. Tokuyama Y, Sturis J, DePaoli AM, Takeda J, Stoffel M, Tang J, Sun X,
zation of somatostatin transactivating factor-1, a novel homeobox factor that Polonsky KS, Bell GI: Evolution of b-cell dysfunction in the male Zucker dia-
stimulates somatostatin expression in pancreatic islet cells. Mol Endocrinol betic fatty rat. Diabetes 44:14471457, 1995
7:12751283, 1993 33. Pick A, Clark J, Kubstrup C, Levisetti M, Pugh W, Bonner-Weir S, Polonsky KS:
13. Miller CP, McGehee RE, Habener JF: IDX-1: a new homeodomain transcrip- Role of apoptosis in failure of b-cell mass compensation for insulin resistance
tion factor expressed in rat pancreatic islets and duodenum that transactivates and b-cell defects in the male Zucker diabetic fatty rat. Diabetes 47:358364,
the somatostatin gene. EMBO J 13:11451156, 1994 1998
14. Stoffers DA, Thomas MK, Habener JF: Homeodomain protein IDX-1: a mas- 34. Rudich A, Tirosh A, Potashnik R, Hemi R, Kanety H, Bashan N: Prolonged
ter regulator of pancreas development and insulin gene expression. Trends oxidative stress impairs insulin-induced GLUT4 translocation in 3T3-L1
Endocrinol Metab 8:145151, 1997 adipocytes. Diabetes 47:15621569, 1998
15. Weir GC, Sharma A, Zangen DH, Bonner-Weir S: Transcription factor abnor- 35. Donath MY, Gross DJ, Cerasi E, Kaiser N: Hyperglycemia-induced b-cell apo-
malities as a cause of beta cell dysfunction in diabetes: a hypothesis. Acta Dia - ptosis in pancreatic islets of Psammomys obesus during development of dia-
betol 34:177184, 1997 betes. Diabetes 48:783744, 1999
16. Watada H, Kajimoto Y, Umayahara Y, Matsuoka T, Kaneto H, Fujitani Y, 36. Scaglia L, Smith FE, Bonner-Weir S: Apoptosis contributes to the involution
Kamada Y, Kawamori R, Yamasaki Y: The human glucokinase gene b-cell-type of b cell mass in the post partum rat pancreas. Endocrinology 136:54615468,
promoter: an essential role of insulin promoter factor 1 (IPF1)/PDX-1 in its acti- 1995
vation in HIT-T15 cells. Diabetes 45:14781488, 1996 37. Jonsson J, Carlsson L, Edlund T, Edlund H: Insulin-promoter-factor 1 is
17. Waeber G, Thompson N, Nicod P, Bonny C: Transcriptional activation of the required for pancreas development in mice. Nature 37:606609, 1994
GLUT2 gene by the IPF-1/STF-1/IDX-1 homeobox factor. Mol Endocrinol 38. Offield MF, Jetton TL, Labosky PA, Ray M, Stein RW, Magnuson MA, Hogan
10:13271334, 1996 BLM, Wright CVE: PDX-1 is required for pancreas outgrowth and differenti-
18. Kaneto H, Miyagawa J, Kajimoto Y, Yamamoto K, Watada H, Umayahara Y, ation of the rostral duodenum. Development 122:983995, 1996
Hanafusa T, Matsuzawa Y, Yamasaki Y, Higashiyama S, Taniguchi N: Expres- 39. Waguri M, Yamamoto K, Miyagawa J, Tochino Y, Yamamori K, Kajimoto Y,
sion of heparin-binding epidermal growth factor-like growth factor during pan- Nakajima H, Watada H, Yoshiuchi I, Itoh N, Imagawa A, Namba M, Kuwajima
creas development: a potential role of PDX-1 in transcriptional activation. M, Yamasaki Y, Hanafusa T, Matsuzawa Y: Demonstration of two different
J Biol Chem 272:2913729143, 1997 processes of b-cell regeneration in a new diabetic mouse model induced by
19. Sakurai T, Tsuchiya S: Superoxide production from nonenzymatically glyca- a selective perfusion of alloxan. Diabetes 46:12811290, 1997
tion protein. FEBS Lett 236:406410, 1988 40. Sharma A, Zangen DH, Reitz P, Taneja M, Lissauer ME, Miller CP, Weir GC,
20. Hunt JV, Smith CC, Wolff SP: Autoxidative glycosylation and possible involve- Habener JF, Bonner-Weir S: The homeodomain protein IDX-1 increases after
ment of peroxides and free radicals in LDL modification by glucose. Diabetes an early burst of proliferation during pancreatic regeneration. Diabetes
39:14201424, 1991 48:507513, 1999
21. Myint T, Hoshi S, Ookawara T, Miyazawa N, Suzuki K, Taniguchi N: Immuno- 41. Stoffers DA, Zinkin NT, Stanojevic V, Clarke WL, Habener JF: Pancreatic age-
logical detection of glycated proteins in normal and streptozotocin-induced nesis attributable to a single nucleotide deletion in the human IPF1 gene
diabetic rats using anti hexitol-lysine IgG. Biochem Biophys Acta 1272:7379, coding sequence. Nat Genet 15:106110, 1997
1995 42. Stoffers DA, Ferrer J, Clarke WL, Habener JF: Early-onset type-II diabetes mel-
22. Baynes JW: Role of oxidative stress in development of complications in dia- litus (MODY4) linked to IPF1. Nat Genet 17:138139, 1997
betes. Diabetes 40:405412, 1991 43. Dutta S, Bonner-Weir S, Montminy M, Wright C: Regulatory factor linked to
23. Kaneto H, Fujii J, Myint T, Miyazawa N, Islam KN, Kawasaki Y, Suzuki K, Naka- late-onset diabetes? Nature 392:560, 1998
mura M, Tatsumi H, Yamasaki Y, Taniguchi N: Reducing sugars trigger oxida- 44. Sagara M, Satoh J, Wada R, Yagihashi S, Takahashi K, Fukuzawa M, Muto G,
tive modification and apoptosis in pancreatic b-cells by provoking oxidative Toyota T: Inhibition with N-acetylcysteine of development of peripheral neu-
stress through the glycation reaction. Biochem J 320:855863, 1996 ropathy in streptozotocin-induced diabetic rats. Diabetologia 39:263269,
24. Tajiri Y, Moller C, Grill V: Long term effects of aminoguanidine on insulin 1996

2406 DIABETES, VOL. 48, DECEMBER 1999

Вам также может понравиться