当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 糖尿病学杂志 > 2006年 > 第6期 > 正文
编号:11257098
The Role of Phosphoinositide 3-Kinase/Akt Signaling in Low-Dose MercuryeCInduced Mouse Pancreatic -Cell Dysfunction In Vitro and In Vivo
     1 Institute of Toxicology, National Taiwan University, Taipei, Taiwan

    2 Department of Laboratory Medicine, National Taiwan University, Taipei, Taiwan

    3 Department of Orthopaedics, College of Medicine, National Taiwan University, Taipei, Taiwan

    4 Department of Occupational Safety and Health, College of Health Care and Management, Chung San Medical University, Taichung, Taiwan

    5 Department of Traumatology, National Taiwan University, Taipei, Taiwan

    6 Department of Surgery, National Taiwan University, Taipei, Taiwan

    7 Department of Emergency Medicine, National Taiwan University Hospital, Taipei, Taiwan

    DCF, dichlorofluorescein; DCFH-DA, DCF diacetate; ELISA, enzyme-linked immunosorbent assay; KRB, Krebs Ringer buffer; MeHg, methylmercury; NAC, N-acetyl-L-cysteine; PI3K, phosphoinositide 3-kinase; ROS, reactive oxygen species

    ABSTRACT

    The relationship between oxidation stress and phosphoinositide 3-kinase (PI3K) signaling in pancreatic -cell dysfunction remains unclear. Mercury is a well-known toxic metal that induces oxidative stress. Submicromolar-concentration HgCl2 or methylmercury triggered reactive oxygen species (ROS) production and decreased insulin secretion in -celleCderived HIT-T15 cells and isolated mouse islets. Mercury increased PI3K activity and its downstream effector Akt phosphorylation. Antioxidant N-acetyl-L-cysteine (NAC) prevented mercury-induced insulin secretion inhibition and Akt phosphorylation but not increased PI3K activity. Inhibition of PI3K/Akt activity with PI3K inhibitor or by expressing the dominant-negative p85 or Akt prevented mercury-induced insulin secretion inhibition but not ROS production. These results indicate that both PI3K and ROS independently regulated Akt signalingeCrelated, mercury-induced insulin secretion inhibition. We next observed that 2- or 4-week oral exposure to low-dose mercury to mice significantly caused the decrease in plasma insulin and displayed the elevation of blood glucose and plasma lipid peroxidation and glucose intolerance. Akt phosphorylation was shown in islets isolated from mercury-exposed mice. NAC effectively antagonized mercury-induced responses. Mercury-induced in vivo effects and increased blood mercury were reversed after mercury exposure was terminated. These results demonstrate that low-dose mercuryeCinduced oxidative stress and PI3K activation cause Akt signalingeCrelated pancreatic -cell dysfunction.

    Oxidative stress has been implicated in a wide variety of disease processes, including diabetes (1). Pancreatic -cells are vulnerable to oxidative stress (2,3). It has been shown that chronic exposure to elevated free fatty acids or glucose levels increases apoptosis in rat pancreatic islets and that these cytotoxic effects could be mediated by oxidative stress (4). On the other hand, the phosphoinositide 3-kinase (PI3K) pathway is implicated in human diseases, including diabetes and cancer (5). A PI3K-dependent signaling pathway has been demonstrated to exist in -cells, and it might function to restrain glucose-induced insulin secretion from -cells (6,7). Moreover, various studies have shown that reactive oxygen species (ROS) could regulate the activation of Akt, a downstream effector of PI3K, in cultured cells such as fibroblasts (8), mesangial cells (9), or vascular smooth muscle cells (10). It has also been reported that activation of PI3K/Akt signaling during the cellular response to oxidant injury was important for survival (11). However, the relationship between oxidation stress and PI3K/Akt signaling in pancreatic -cell dysfunction is still unclear.

    Various studies (12eC15) have indicated that heavy metals acted as catalysts in the oxidative reactions of biological macromolecules; therefore, the toxicities associated with these metals might be due to oxidative tissue damage. Mercury is a well-known toxic metal that induces oxidative stress (16eC19). Among humans, the major source of exposure to methylmercury (MeHg) is the consumption of fish and sea mammals (20). The industrial release of MeHg into Minamata Bay and the Agano River in Japan resulted in the accumulation of the toxicant in fish and, subsequently, in two large epidemics related to fish consumption (20). Elemental and inorganic mercury are found in scientific instruments, electrical equipment, dental amalgams, disinfectants, production of caustic soda, disk batteries, and the atmosphere (21). These mercury sources are now usually related to accidental and occupational exposures (20,22). Recently, increased oxidative DNA damage and serum redox status have been detected in individuals exposed to mercury (23). Moreover, previous in vitro studies have shown that HgCl2 altered intracellular Ca2+ homeostasis in pancreatic -cells isolated from mouse islets (24) and decreased insulin secretion from secretion granules isolated from toadfish islets (25). It has also been shown that an increased incidence of diabetes existed in patients with documented Minamata disease (MeHg poisoning) in Japan (26). Nevertheless, the precise action and mechanism of mercury-induced oxidative stress on the pancreatic -cell function and blood glucose regulation remain unclear.

    Taken together, in the current study, we try to explore the role of PI3K/Akt signaling in low-dose mercuryeCinduced mouse pancreatic -cell dysfunction. To this aim, we sought to investigate the in vitro effects of low concentrations of inorganic mercury (HgCl2) and organic mercury (MeHg) (0.2eC1 e蘭ol/l) on ROS generation, investigate PI3K/Akt activation and insulin secretion in the isolated mouse islets and pancreatic -cell line HIT-T15, and test whether low-dose exposure to HgCl2 (50 and 500 e蘥/kg) or MeHg (20 e蘥/kg), which approaches the human-exposed mercury levels in blood, would generate lipid peroxidation, activate PI3K signaling, and alter the regulation of insulin secretion and blood glucose in mice. Moreover, the potential protective effects of N-acetyl-L-cysteine (NAC; ROS scavenger) on the pancreatic -cell functions in vitro and in vivo were investigated. These observations may provide evidence that oxidative stress causes pancreatic -cell dysfunction through a PI3K/Akt-related pathway and that mercury is a possible environmental risk factor to diabetes.

    RESEARCH DESIGN AND METHODS

    We purchased 18eC25 g male ICR mice from the Animal Center of the College of Medicine, National Taiwan University (Taipei, Taiwan). The Animal Research Committee of College of Medicine, National Taiwan University, conducted the study in accordance with the guidelines for the care and use of laboratory animals. Mice were housed in a room at a constant temperature of 22 ± 2°C with 12-h light-dark cycles. Mice were randomly assigned to pretreatment groups, weighed, and administered HgCl2, MeHg, or vehicle (oral application by gavage). These groups were given 0, 50, and 500 e蘥 · kgeC1 · dayeC1 HgCl2 or 20 e蘥 · kgeC1 · dayeC1 MeHg (7 days/week) in the presence or absence of NAC (150 mg/kg) for 2eC4 weeks. Each group contained 16 mice. All mice were observed daily for clinical signs. Following collection of blood samples, the animals were exsanguinated, while still under anesthesia, and necropsied. A necropsy examination was performed on all animals from each group. All pathological findings were recorded. A histopathological examination was performed on organs from groups treated with 0 and 500 e蘥/kg HgCl2 and 20 e蘥/kg MeHg.

    Plasma lipid peroxidation assay.

    Blood samples were collected from the peripheral vessels of mice under ketamine anesthesia (85 mg/kg i.p.). Whole blood was centrifuged at 3,000g for 10 min, and plasma was obtained and assayed immediately using the lipid peroxidation assay kit (Calbiochem). An absorbance of 586 nm was measured using an enzyme-linked immunosorbent assay (ELISA) microplate reader.

    Blood glucose measurement and glucose tolerance test.

    Blood samples were collected after an overnight fast. Blood glucose levels were determined using the SureStep blood glucose meter (Lifescan, Milpitas, CA). For oral glucose tolerance testing, D-glucose (1 g/kg) was administered by stomach tube after an overnight fast. Blood samples were collected before and 30, 60, 90, 120, and 180 min after delivery of the glucose load.

    Pancreatic islet isolation.

    Islets of Langerhans were isolated by collagenase digestion from the mouse pancreas as previously described (27,28). After separation on a Ficoll gradient, the islets were further purified by handpicking to eliminate any remaining exocrine tissue. Whole islets were maintained in culture medium consisting of RPMI-1640 medium supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin/amphotericin B at 37°C in an atmosphere of 95% air/5% CO2 before experimentation.

    HIT-T15 cell culture.

    Pancreatic -celleCderived HIT-T15 cells were cultured in a humidified chamber with a 95% air/5% CO2 mixture at 37°C and maintained in RPMI-1640 medium supplemented with 10% fetal bovine serum and containing 11.1 mmol/l glucose as described (29). In some experiments, transfection in HIT-T15 cells was performed using the Lipofectin reagent (BD Clontech) according to the recommendations of the manufacturer with 2 e蘥 total DNA (or vector) per sample. The efficiency of transfection (80%) was determined by using an equal amount of a plasmid encoding the green fluorescent protein under the cytomegalovirus promoter. Plasmids containing the DN-p85 (-p85) or DN-Akt (Akt K179A) (30,31) were kindly provided by Dr. R.H. Chen and Dr. M.L. Kuo (Institutes of Molecular Medicine and Toxicology, National Taiwan University, Taiwan).

    Insulin secretion studies.

    HIT-T15 insulin secretion experiments were performed in Krebs Ringer buffer (KRB), and islet secretion studies were performed in islet culture media for a 1-h experimental incubation with a 95% air/5% CO2 mixture at 37°C, as previously described (32). All experimental agents were mixed together in experimental solutions (KRB or media) and then added to the cells at the start of the experimental incubation. To measure the amount of insulin secreted, aliquots of samples were collected from the plasma or experimental solutions at indicated time points and subjected to insulin antiserum immunoassay according to the manufacturer’s instruction (Mercodia, Uppsala, Sweden).

    Cell viability.

    Cells were washed with fresh media from dishes and cultured in 96-well plates (2 x 105/well), then stimulated with HgCl2 and MeHg. After 24 h, the medium was replaced with fresh medium containing 30 e蘬 (2 mg/ml) 3-(4,5-dimethyl thiazol-2-yl-)-2,5-diphenyl tetrazolium bromide. After incubation for 4 h, the medium was removed and 1 ml dimethyl sulfoxide added to dissolve blue formazan crystal. An ELISA reader was used for fluorescence detection; absorption was 570 nm.

    Detection of intracellular ROS.

    Intracellular ROS generation was monitored by flow cytometry using peroxide-sensitive fluorescent probe (2',7'-dichlorofluorescin diacetate [DCFH-DA; Molecular Probes]). In brief, HIT-T15 cells or islets were coincubated with 20 e蘭ol/l DCFH-DA for 15 min at 37°C. In the presence of a proper oxidant, DCFH was oxidized into the highly fluorescent 2',7'-dichlorofluorescein (DCF). After incubation with the dye, cells and islets were resuspended in ice-cold PBS and placed on ice in darkness for flow cytometry analysis. Using trypsin, the dispersed islets and intracellular peroxide levels were measured with the FACScan flow cytometry device (Becton Dickinson, Franklin Lakes, NJ). More than 100 islets from at least three separate islet isolations were studied for each group. The results were expressed as the mean fluorescence intensity. In some experiments, after 15 min of incubation of DCFH-DA, the islets were washed twice and images captured by a Leica DMIL inverted microscope equipped with a charged-coupled device camera.

    Analysis of cellular ATP levels.

    Cellular ATP concentration was determined by the ATP bioluminescent assay kit (Sigma), which measures the intracellular ATP level using the ATP-dependent luciferin-luciferase reaction.

    Western blotting.

    Fifty micrograms of protein of each cell lysate were subjected to electrophoresis on 10% SDS-polyacrylamide gels. The samples were then electroblotted on polyvinylidene difluoride membranes. After blocking, blots were incubated with anti-Akt and antieCphospho-Akt (New England BioLabs) antibodies in PBS and 0.1% Tween 20 for 1 h followed by two washes (15 min each) in PBS and 0.1% Tween 20. The membranes were then incubated with horseradish peroxidaseeCconjugated secondary antibodies for 30 min. Enhanced chemiluminescence reagents (Amersham) were used to depict the protein bands on membranes.

    PI3K activity assay.

    PI3K activity was assayed as described previously (33). Cell extracts were incubated with 2 e蘥 anti-p85 antibody overnight at 4°C. The immunocomplex was precipitated with protein A sepharose for 1 h at 4°C and washed three times with lysis buffer, twice with LiCl buffer, and twice with TNE buffer (10 mmol/l Tris, 100 mmol/l NaCl, and 1 mmol/l EDTA), and then the immunoprecipitated enzymes were subjected to a kinase assay by competitive ELISA, according to the manufacturer’s instructions (Echelon Biosciences, Salt Lake City, UT).

    Mercury contents.

    The levels of mercury in blood were determined by cold vapor atomic absorption spectrophotometer combined with the flow-injection analysis system (34), and analysis was performed with a PerkinElmer 5100PC AAS equipped with a PerkinElmer FIAS-200 flow-injection analysis system and AA WinLab software. The detection limit for mercury was 0.1 ppb (e蘥/l).

    Statistics.

    The values in the text are given as means ± SE. The significance of difference was evaluated by the paired Student’s t test. When more than one group was compared with one control, significance was evaluated according to one-way ANOVA. Statistical difference was indicated by P < 0.05.

    RESULTS

    Effects of low-concentration mercury on ROS formation and insulin secretion and the involvement of PI3K/Akt in HIT-T15 cells.

    To understand whether mercury-induced oxidative stress could affect the function of pancreatic -cells, we investigated the in vitro effects of low-concentration HgCl2 and MeHg (0.2eC1 e蘭ol/l) on the -celleCderived HIT-T15 cells. Exposure of cells to mercury for 24 h did not affect cell viability (data not shown). Measurement of DCF fluorescence as an indicator of ROS formation, after exposure of HIT-T15 cells to 0.5 and 1 e蘭ol/l mercury for 1 h, significantly increased ROS levels, which were inhibited by the antioxidant NAC (1 mmol/l) (Fig. 1A). Furthermore, measurement of insulin secretion from HIT-T15 cells after exposure to mercury under various concentrations of glucose (5.5eC30 mmol/l) showed that HgCl2 or MeHg (0.5 and 1 e蘭ol/l, respectively) significantly inhibited insulin secretion (Fig. 1B). Cells treated with NAC effectively prevented the inhibition of insulin secretion in HIT-T15 cells exposed to mercury (Fig. 2A). The PI3K inhibitor LY294002 (5 e蘭ol/l) also effectively prevented the mercury-induced inhibition of insulin secretion in HIT-T15 cells (Fig. 2B). In a 4-h incubation, 5 e蘭ol/l LY294002 alone did not affect the insulin secretion; however, it was capable of increasing the insulin secretion in HIT-T15 cells at concentration of 10 e蘭ol/l (131.6 ± 1.2% of control, n = 5).

    To further evaluate the involvement of PI3K/Akt signaling in the responses triggered by mercury-induced oxidative stress, the PI3K and Akt activities in HIT-T15 cells were examined by determining the PI3K activity and phosphorylated status of Akt. Immunoprecipitates with the anti-p85 antibody revealed a substantial increase in PI3K activity 5 min after the exposure of mercury (1 e蘭ol/l) (Fig. 3A, a). The significant activation of Akt was also shown in mercury-treated cells (Fig. 3A, b). LY294002 markedly reduced the PI3K activity and phosphorylation of Akt induced by mercury in HIT-T15 cells (data not shown). NAC effectively inhibited the mercury-induced Akt phosphorylation (Fig. 3A, b) but not the increased PI3K activity (Fig. 3A, a).

    The next aim of the investigation was to ascertain whether inhibition of PI3K/Akt activities might affect the mercury-induced inhibition of insulin secretion in HIT-T15 cells. To address this issue, cells were transfected with a dominant-negative form of the regulatory subunit (p85) of PI3K (DN-p85) or a dominant-negative form of Akt (DN-Akt) (35). As shown in Fig. 3B, a, DN-p85 or DN-Akt transfection markedly decreased the phosphorylation of Akt. The levels of Akt protein expression in DN-AkteCtransfected cells were increased compared with control. Blocking the PI3K and Akt activities with DN-p85 and DN-Akt greatly prevented the 1 e蘭ol/l mercuryeCtriggered insulin secretion inhibition (Fig. 3B, b). The control pcDNA3 transfection did not affect the mercury-induced responses. However, blocking the PI3K activity with DN-p85 did not affect ROS generation by mercury (Fig. 3B, c). Therefore, these results imply that both PI3K and ROS independently regulated Akt signalingeCrelated mercury-induced insulin secretion inhibition.

    Effects of low-concentration mercury on ROS formation and insulin secretion and the involvement of PI3K/Akt in isolated mouse islets.

    Exposure of isolated mouse islets to 1 e蘭ol/l mercury for 1 h increased DCF-sensitive ROS formation, which was prevented by 1 mmol/l NAC (Fig. 4). The role of PI3K/Akt signaling was further investigated. A significant phosphorylation of Akt in isolated mouse islets was shown 1 h after the treatment of mercury (1 e蘭ol/l) (Fig. 4B, a). NAC and LY294002 (5 e蘭ol/l) markedly reversed the increase of Akt phosphorylation by mercury (Fig. 4B, a). Furthermore, mercury (1 e蘭ol/l) was capable of inhibiting the insulin secretion in mouse islets, which could also be reversed by NAC and LY294002 (Fig. 4B, b).

    Effects of low-dose mercury on blood glucose, plasma insulin secretion, plasma lipid peroxidation, and islet Akt phosphorylation in mice.

    The next aim of the investigation was to ascertain whether mercury-induced oxidative stress might affect the function of pancreatic -cells in vivo and the involvement of PI3K/Akt signaling. A 2- or 4-week exposure to low-dose HgCl2 (50 or 500 e蘥 · kgeC1 · dayeC1) or MeHg (20 e蘥 · kgeC1 · dayeC1) (oral application by gavage) to mice caused a significant decrease in plasma insulin levels (Fig. 5A). Moreover, a significant phosphorylation of Akt was shown in islets isolated from mice exposed to mercury for 2 weeks (Fig. 5B). Mercury-exposed mice also showed an elevation in blood glucose (Fig. 6A) and glucose intolerance (Fig. 6B). Moreover, the result of plasma lipid peroxidation assay after exposure to HgCl2 or MeHg to mice showed that plasma malondialdehyde levels were significantly increased at 2eC4 weeks (Fig. 7). These mercury-triggered responses in mice could also be reversed by the treatment of NAC (150 mg/kg) (Figs. 5eC7).

    Blood mercury and insulin secretion and blood glucose in mice after HgCl2 or MeHg exposure was terminated.

    Mice exposed to mercury over a 2- to 4-week period had elevated blood mercury levels ranging from 4.5 to 60 e蘥/l (Fig. 8A). There was a similar blood mercury range for treatment with 500 e蘥/kg HgCl2 and 20 e蘥/kg MeHg. The elevated blood mercury and decreased plasma insulin and elevated blood glucose levels were gradually reversed after mercury withdrawal (Fig. 8A, blood mercury; Fig. 8B, plasma insulin; blood glucose [mg/dl]: HgCl2 4 weeks 144.2 ± 8.4, withdrawal 2 weeks 112.4 ± 5.8, withdrawal 4 weeks 89.7 ± 6.7, n = 8; MeHg 4 weeks 122.1 ± 7.7, withdrawal 2 weeks 93.6 ± 5.4, withdrawal 4 weeks 82.9 ± 4.2, n = 8; age-matched controls 81.6 ± 4.9, n = 8).

    There were no clinical signs and grossly detectable lesions (including brain, liver, kidney, lung, heart, adrenal, gonad, and intestine) related to the administration of mercury for 2eC4 weeks in mice, and there was no effect on whole-body weight (data not shown).

    DISCUSSION

    Oxidative stress has been demonstrated to be produced under diabetic conditions and was likely involved in the progression of pancreatic -cell dysfunction found in diabetes (3,36). It has been demonstrated that mitochondrial ROS suppressed glucose-induced insulin secretion from -cells (2). Despite several studies showing that mercury could induce oxidative stress with subsequent oxidative damage in several organs or systems (16eC19,37), the precise action and mechanism of mercury-induced oxidative stress on the pancreatic -cell function is unknown. To investigate the effects of mercury-induced oxidative stress on the function of pancreatic -cells, we first investigated the in vitro effects of HgCl2 and MeHg on the -celleCderived HIT-T15 cells and isolated mouse islets. Our results showed that submicromolar-concentration exposure to mercury was capable of triggering ROS production and decreasing insulin secretion in HIT-T15 cells and isolated islets, which could be prevented completely by treatment of cells with antioxidant NAC, indicating that ROS was involved in mercury-induced -cell insulin secretion inhibition. On the other hand, changes in glucose levels to insulin secretion are related to intracellular ATP levels. Following glucose entry into pancreatic -cells, products of glucose metabolism enter the mitochondrial respiratory chain, which uses them to generate ATP. The increase in ATP inhibits ATP-sensitive potassium channels, which in turn stimulate insulin secretion (38). Here we found that increasing doses of mercury did not change the intracellular ATP levels in HIT-T15 cells after exposure to 0.2eC1 e蘭ol/l HgCl2 or MeHg at 1eC4 h (data not shown), and hence, it may not be a primary toxic mechanism in dysfunction of pancreatic -cells induced by low-concentration mercury. Next, we investigated the in vivo effects of mercury-induced oxidative stress on the insulin secretion and blood glucose regulation in mice. The results showed that a 2- or 4-week oral exposure to low-dose mercury caused an increase in plasma lipid peroxidation levels, a decrease in plasma insulin levels, and an elevation in blood glucose and glucose intolerance. NAC could prevent these mercury-induced responses. These findings indicate that oxidative stress is involved in the toxic mechanism in mercury-induced hypoinsulinemia and hyperglycemia. In addition, it has been found that endogenous nitric oxide (NO) production could be stimulated by glucose and implicated this glucose-stimulated NO production in physiological glucose-stimulated insulin secretion (32). Whether dysfunction or deregulation of the NO system is involved in mercury-induced -cell damage may need to be clarified in the future.

    Some studies (6,7) have suggested that as insulin resistance developed in peripheral tissues, a potential result of impaired PI3K activation in -cells promoted a linked increase in insulin secretion to maintain glucose homeostasis. Eto et al. (39) have also indicated that PI3K suppressed glucose-stimulated insulin secretion by affecting postcytosolic Ca2+ elevation signals. It has been shown that redox stress regulated cell proliferation and apoptosis of human hepatoma through PI3K/Akt pathway (40). Angiotensin IIeCinduced Akt phosphorylation has been demonstrated to be mediated by ROS, which contributed to angiotensin IIeCinduced hypertrophy in vascular smooth muscle cells (10). However, the role of PI3K/Akt signaling pathway in the oxidative stresseCinduced pancreatic -cell dysfunction is still unclear. The present work showed that low-dose mercury could enhance the phosphorylation of Akt, which was reversed by NAC. Moreover, the inhibition of PI3K/Akt activity with the PI3K inhibitor LY294002, or by expressing the dominant-negative p85 or Akt, effectively attenuated the inhibition of insulin secretion induced by mercury. LY294002 was capable of increasing the insulin secretion in HIT-T15 cells at a concentration of 10 e蘭ol/l. However, unexpectedly, we found that NAC could not inhibit mercury-increased PI3K activity, and the inhibition of PI3K activity by expressing the dominant-negative p85 also could not suppress mercury-induced ROS production in -cells. Therefore, these results implicate that both PI3K and ROS independently regulate Akt signalingeCrelated, mercury-induced insulin secretion inhibition and suggest that low-dose mercuryeCinduced oxidative stress and PI3K activation cause pancreatic -cell dysfunction through an Akt signaling pathway.

    A recent study of mercury exposure in an urban pediatric population has shown that mean urinary mercury was 1.08 ± 1.82 e蘥/l (range 0.2eC11.7) and found that 5% of subjects had unsuspected elevated urinary mercury levels (>5 e蘥/l) (41). The Food and Drug Administration advises women to limit the consumption of fish with up to 0.5 ppm (e蘥/ml) mercury to 14 oz/week or with up to 1.0 ppm mercury to 7 oz/week (42). Recently, the Wisconsin Department of Natural Resources lowered its mercury in fish action level by a factor of 10x to 0.05 ppm based on subtle alterations in neurodevelopment in children born to mothers who eat large amounts of fish (43). A study (44) of blood mercury levels in workers exposed to the preparation of mercury cadmium telluride layers has shown that the mercury value was estimated at 1.60 ± 0.20 e蘥 Hg/l in control and at 10.72 ± 1.34 e蘥 Hg/l in phase I and 8.08 ± 1.15 e蘥 Hg/l in phase II; an individual who met with a mercury accident showed 226 e蘥 Hg/l of blood, which decreased to 25 e蘥 Hg/l after 3 months. It has been reported that sensitive nonhuman mammals showed significant adverse effects of mercury when dietary levels were 1,100 e蘥/kg body wt (45). The study of Nakagawa has shown that the total mercury concentrations in the hair of ordinary diseased people, including diabetes, were from 2.08 to 36.5 ppm; those values were considerably higher than that of healthy people of the same age-groups (46). In the present study, we used low doses of inorganic mercury (50 and 500 e蘥/kg) and organic methylmercury (20 e蘥/kg) to expose mice. The blood mercury content ranges are 3.9eC5.8 and 38.4eC72.2 e蘥/l in 50 and 500 e蘥 · kgeC1 · dayeC1 HgCl2eCtreated mice, respectively, or 40.2eC64.6 e蘥/l in 20 e蘥 · kgeC1 · dayeC1 MeHgeCtreated mice, which were exposed over a 2- to 4-week period (0.83eC1.62 e蘥/l in age-matched controls). Moreover, no evidence of mercury-induced target organ toxicity was seen in either the clinical pathology parameters or histomorphologic evaluations. Thus, the increased blood mercury levels in the present study approach the human-exposed levels.

    An increased incidence of diabetes has been found to exist in patients with documented Minamata disease (blood mercury >200 e蘥/l [20]) in Japan (26). Takeuchi and Eto (47) reported that disturbance of pancreatic islet cells were found in autopsy cases of Minamata disease. Nevertheless, a health-surveillance study in the population living in a methylmercury-polluted area 30 years after the outbreak of Minamata disease by Futatsuka et al. (48) showed that the prevalence of liver disease, renal disease, and diabetes was not higher in this methylmercury-polluted area compared with other areas in Japan, contrary to what was expected based on standard mortality ratios and pathological findings. However, the results of Sakamoto et al. (49) and Harada et al. (50) showed that no significant difference in the mercury concentrations in the erythrocytes and scalp hair was observed between the cases with Minamata disease and the controls about several decades after the outbreak of Minamata disease. Around 1960, marine products were heavily polluted by methylmercury in the Minamata area. When the cause of Minamata disease had already proved to be methylmercury-contaminated marine products, the residents on the local coast were hesitant to eat those marine products, so the patient’s mercury levels were gradually reduced (50). These findings may explain why there are no positive correlations between those diseases and methylmercury exposure 30 years after the outbreak of Minamata disease, as indicated by the study of Futatsuka et al. (48). These findings also imply that some diseases induced by methylmercury exposure may be reversible if the exposure was terminated. Our present results support this possibility; we found that the elevated blood mercury, decreased plasma insulin, and elevated blood glucose levels in mercury-treated mice were gradually reversed after mercury exposure was terminated. Thus, the present study indicates that low-dose mercury is capable of altering blood insulin and glucose homeostasis, although individuals may tend to ignore this risk of long-term exposure to low-dose mercury.

    In conclusion, as indicated in Fig. 8C, our results strongly suggest an essential and specific role for the Akt signaling in low-dose mercuryeCinduced, oxidative stresseCcaused pancreatic -cell dysfunction. Moreover, low-dose mercuryeCinduced oxidative stress causes pancreatic islet Akt activation and induces hypoinsulinemia and hyperglycemia in mice in the absence of overt mercury toxicity. These observations also give further evidence to confirm the possibility that mercury is an environmental risk factor for diabetes.

    ACKNOWLEDGMENTS

    This study was supported by grants from the National Science Council of Taiwan (NSC93-2314-B-002-178); the Department of Health, Executive Yuan of Taiwan (DOH95-TD-B-111-TM003); and the National Taiwan University Hospital (93-A018).

    FOOTNOTES

    Y.W.C., C.F.H., K.S.T., and R.S.Y. contributed equally to this work.

    The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

    REFERENCES

    Finkel T, Holbrook NJ: Oxidants, oxidative stress and the biology of ageing. Nature 408:239eC247, 2000

    Sakai K, Matsumoto K, Nishikawa T, Suefuji M, Nakamaru K, Hirashima Y, Kawashima J, Shirotani T, Ichinose K, Brownlee M, Araki E: Mitochondrial reactive oxygen species reduce insulin secretion by pancreatic beta-cells. Biochem Biophys Res Commun 300:216eC222, 2003

    Kajimoto Y, Kaneto H: Role of oxidative stress in pancreatic beta-cell dysfunction. Ann N Y Acad Sci 1011:168eC176, 2004

    Piro S, Anello M, Di Pietro C, Lizzio MN, Patane G, Rabuazzo AM, Vigneri R, Purrello M, Purrello F: Chronic exposure to free fatty acids or high glucose induces apoptosis in rat pancreatic islets: possible role of oxidative stress. Metabolism 51:1340eC1347, 2002

    Cantley LC: The phosphoinositide 3-kinase pathway (Review). Science 296:1655eC1657, 2002

    Zawalich WS, Zawalich KC: A link between insulin resistance and hyperinsulinemia: inhibitors of phosphatidylinositol 3-kinase augment glucose-induced insulin secretion from islets of lean, but not obese, rats. Endocrinology 141:3287eC3295, 2000

    Zawalich WS, Tesz GJ, Zawalich KC: Inhibitors of phosphatidylinositol 3-kinase amplify insulin release from islets of lean but not obese mice. J Endocrinol 174:247eC258, 2002

    Esposito F, Chirico G, Montesano Gesualdi N, Posadas I, Ammendola R, Russo T, Cirino G, Cimino F: Protein kinase B activation by reactive oxygen species is independent of tyrosine kinase receptor phosphorylation and requires SRC activity. J Biol Chem 278:20828eC20834, 2003

    Gorin Y, Ricono JM, Kim NH, Bhandari B, Choudhury GG, Abboud HE: Nox4 mediates angiotensin II-induced activation of Akt/protein kinase B in mesangial cells. Am J Physiol Renal Physiol 285:F219eCF229, 2003

    Ushio-Fukai M, Alexander RW, Akers M, Yin Q, Fujio Y, Walsh K, Griendling KK: Reactive oxygen species mediate the activation of Akt/protein kinase B by angiotensin II in vascular smooth muscle cells. J Biol Chem 274:22699eC22704, 1999

    Martindale JL, Holbrook NJ: Cellular response to oxidative stress: signaling for suicide and survival. J Cell Physiol 192:1eC15, 2002

    Stohs SJ, Bagchi D: Oxidative mechanisms in the toxicity of metal ions. Free Radic Biol Med 18:321eC336, 1995

    Sarafian TA: Methylmercury-induced generation of free radicals: biological implications. Met Ions Biol Syst 36:415eC444, 1999

    Ercal N, Gurer-Orhan H, Aykin-Burns N: Toxic metals and oxidative stress part I: mechanisms involved in metal-induced oxidative damage. Curr Top Med Chem 1:529eC539, 2001

    Valko M, Morris H, Cronin MT: Metals, toxicity and oxidative stress. Curr Med Chem 12:1161eC1208, 2005

    Mahboob M, Shireen KF, Atkinson A, Khan AT: Lipid peroxidation and antioxidant enzyme activity in different organs of mice exposed to low level of mercury. J Environ Sci Health B 36:687eC697, 2001

    Huang YL, Cheng SL, Lin TH: Lipid peroxidation in rats administrated with mercuric chloride. Biol Trace Elem Res 52:193eC206, 1996

    Miller DM, Woods JS: Urinary porphyrins as biological indicators of oxidative stress in the kidney: interaction of mercury and cephaloridine. Biochem Pharmacol 46:2235eC2241, 1993

    Reus IS, Bando I, Andres D, Cascales M: Relationship between expression of HSP70 and metallothionein and oxidative stress during mercury chloride induced acute liver injury in rats. J Biochem Mol Toxicol 17:161eC168, 2003

    Clarkson TW, Magos L, Myers GJ: The toxicology of mercuryeCcurrent exposures and clinical manifestations. N Engl J Med 349:1731eC1737, 2003

    Aschner M, Walker SJ: The neuropathogenesis of mercury toxicity. Mol Psychiatry 7 (Suppl. 2):S40eCS41, 2002

    Beck C, Krafchik B, Traubici J, Jacobson S: Mercury intoxication: it still exists. Pediatr Dermatol 21:254eC259, 2004

    Chen C, Qu L, Li B, Xing L, Jia G, Wang T, Gao Y, Zhang P, Li M, Chen W, Chai Z: Increased oxidative DNA damage, as assessed by urinary 8-hydroxy-2'-deoxyguanosine concentrations, and serum redox status in persons exposed to mercury. Clin Chem 51:759eC767, 2005

    Liu SH, Lin-Shiau SY: Mercuric chloride alters the membrane potential and intracellular calcium level in mouse pancreatic islet cells. J Toxicol Environ Health A 65:317eC326, 2002

    Bloom GD, Hellman B, Idahl LA, Lernmark A, Sehlin J, Taljedal IB: Effects of organic mercurials on mammalian pancreatic -cells: insulin release, glucose transport, glucose oxidation, membrane permeability and ultrastructure. Biochem J 129:241eC254, 1972

    Uchino M, Tanaka Y, Ando Y, Yonehara T, Hara A, Mishima I, Okajima T, Ando M: Neurologic features of chronic minamata disease (organic mercury poisoning) and incidence of complications with aging. J Environ Sci Health B 30:699eC715, 1995

    Lacy PE, Kostianovsky M: Method for the isolation of intact islets of Langerhans from the rat pancreas. Diabetes 16:35eC39, 1967

    Lernmark : The preparation of, and studies on, free cell suspensions from mouse pancreatic islets. Diabetologia 10:431eC438, 1974

    Zhang HJ, Walseth TF, Robertson RP: Insulin secretion and cAMP metabolism in HIT cells: reciprocal and serial passage-dependent relationships. Diabetes 38:44eC48, 1989

    Lin MT, Lee RC, Yang PC, Ho FM, Kuo ML: Cyclooxygenase-2 inducing Mcl-1-dependent survival mechanism in human lung adenocarcinoma cl1.0 cells: involvement of phosphatidylinositol 3-kinase/Akt pathway. J Biol Chem 276:48997eC49002, 2001

    Kuo ML, Chuang SE, Lin MT, Yang SY: The involvement of PI3-K/Akt-dependent up-regulation of Mcl-1 in the prevention of apoptosis of Hep3B cells by interleukin-6. Oncogene 20:677eC685, 2001

    Smukler SR, Tang L, Wheeler MB, Salapatek AM: Exogenous nitric oxide and endogenous glucose-stimulated beta-cell nitric oxide augment insulin release. Diabetes 51:3450eC3460, 2002

    Flaxenburg JA, Melter M, Lapchak PH, Briscoe DM, Pal S: The CD40-induced signaling pathway in endothelial cells resulting in the overexpression of vascular endothelial growth factor involves ras and phosphatidylinositol 3-kinase. J Immunol 172:7503eC7509, 2004

    Yen CC, Liu SH, Chen WK, Lin RH, Lin-Shiau SY: Tissue distribution of different mercurial compounds analyzed by the improved FI-CVAAS. J Anal Toxicol 26:286eC295, 2002

    Sheu ML, Ho FM, Chao KF, Kuo ML, Liu SH: Activation of phosphoinositide 3-kinase in response to high glucose leads to regulation of reactive oxygen species-related nuclear factor-kappaB activation and cyclooxygenase-2 expression in mesangial cells. Mol Pharmacol 66:187eC196, 2004

    Kaneto H, Kajimoto Y, Miyagawa J, Matsuoka T, Fujitani Y, Umayahara Y, Hanafusa T, Matsuzawa Y, Yamasaki Y, Hori M: Beneficial effects of antioxidants for diabetes: possible protection of pancreatic -cells against glucose toxicity. Diabetes 48:2398eC2406, 1999

    Kim SH, Sharma RP: Mercury-induced apoptosis and necrosis in murine macrophages: role of calcium-induced reactive oxygen species and p38 mitogen-activated protein kinase signaling. Toxicol Appl Pharmacol 196:47eC57, 2004

    Langin D: Diabetes, insulin secretion, and the pancreatic beta-cell mitochondrion (Review). N Engl J Med 345:1772eC1774, 2001

    Eto K, Yamashita T, Tsubamoto Y, Terauchi Y, Hirose K, Kubota N, Yamashita S, Taka J, Satoh S, Sekihara H, Tobe K, Iino M, Noda M, Kimura S, Kadowaki T: Phosphatidylinositol 3-kinase suppresses glucose-stimulated insulin secretion by affecting post-cytosolic [Ca2+] elevation signals. Diabetes 51:87eC97, 2002

    Dong-Yun S, Yu-Ru D, Shan-Lin L, Ya-Dong Z, Lian W: Redox stress regulates cell proliferation and apoptosis of human hepatoma through Akt protein phosphorylation. FEBS Lett 542:60eC64, 2003

    Ozuah PO, Lesser MS, Woods JS, Choi H, Markowitz M: Mercury exposure in an urban pediatric population. Ambul Pediatr 3:24eC26, 2003

    Agency for Toxic Substances and Disease Registry: Mercury (Update). Atlanta, GA, U.S. Public Health Service, 1999

    Wisconsin Department of Natural Resources: Choose Wisely: A Health Guide for Eating Fish in Wisconsin. Madison, WI, Wisconsin Department of Natural Resources, 2002

    Gupta M, Bansal JK, Khanna CM: Blood mercury in workers exposed to the preparation of mercury cadmium telluride layers on cadmium telluride base. Ind Health 34:421eC425, 1996

    Eisler R: Mercury hazards from gold mining to humans, plants, and animals. Rev Environ Contam Toxicol 181:139eC198, 2004

    Nakagawa R: Concentration of mercury in hair of diseased people in Japan. Chemosphere 30:135eC140, 1995

    Takeuchi T, Eto K: Pathology and pathogenesis of Minamata disease. In Minamata Disease-Methyl Mercury Poisoning in Minamata and Niigata, Japan. Tsubaki T, Kirukayama, Eds. Tokyo, Kodansya, 1997, p.103eC141

    Futatsuka M, Kitano T, Shono M, Fukuda Y, Ushijima K, Inaoka T, Nagano M, Wakamiya J, Miyamoto K: Health surveillance in the population living in a methyl mercury-polluted area over a long period. Environ Res 83:83eC92, 2000

    Sakamoto M, Nakano A, Kinjo Y, Higashi H, Futatsuka M: Present mercury levels in red blood cells of nearby inhabitants about 30 years after the outbreak of Minamata disease. Ecotoxicol Environ Saf 22:58eC66, 1991

    Harada M, Nakanishi J, Konuma S, Ohno K, Kimura T, Yamaguchi H, Tsuruta K, Kizaki T, Ookawara T, Ohno H: The present mercury contents of scalp hair and clinical symptoms in inhabitants of the Minamata area. Environ Res 77:160eC164, 1998(Ya Wen Chen, Chun Fa Huan)