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3期 > 正文
编号:11256989
Novel Mechanism for Plasma GlucoseeCLowering Action of Metformin in Streptozotocin-Induced Diabetic Rats
     1 Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan City, Taiwan, R.O.C

    2 Department of Pharmacy, Tajen University, Yen-Pou, Ping Tung Shien, Taiwan, R.O.C

    3 Department of Internal Medicine, Pao Chien Hospital, Ping Tung City, Taiwan, R.O.C

    4 Department of Family Medicine, College of Medicine, National Cheng Kung University, Tainan City, Taiwan, R.O.C

    AMPK, 5'AMP-activated protein kinase; BER, -endorphineClike immunoreactivity; PKC, protein kinase C; STZ, streptozotocin

    ABSTRACT

    To better understand the insulin-independent plasma glucoseeClowering action of metformin, we used streptozotocin (STZ)-induced diabetic rats to investigate the possible mechanisms. Oral intake of metformin decreased the plasma glucose of STZ-induced diabetic rats with a parallel increase of plasma -endorphineClike immunoreactivity (BER). Mediation of opioid e?receptors in the action of metformin was identified by the blockade of receptors with antagonist in STZ-induced diabetic rats and the failure of action in opioid e?receptor knockout diabetic mice. Release of BER from adrenal glands by metformin was characterized, using bilateral adrenalectomy and the release of BER from isolated adrenal medulla of STZ-induced diabetic rats. Repeated treatment with metformin in STZ-induced diabetic rats increased the mRNA and protein levels of GLUT-4 in soleus muscle that was blocked by naloxonazine. Reduction of the mRNA or protein levels of hepatic PEPCK was also impeded in the same group of STZ-induced diabetic rats. In conclusion, our results provide novel mechanisms for the plasma glucoseeClowering action of metformin, via an increase of -endorphin secretion from adrenal glands to stimulate opioid e?receptor linkage, leading to an increase of GLUT-4 gene expression and an attenuation of hepatic PEPCK gene expression in STZ-induced diabetic rats.

    Diabetes and its complications constitute a major health problem in modern societies. Both type 1 and type 2 diabetes comprise abnormalities of insulin action, including deficiency and insulin resistance (1).

    Metformin is a widely prescribed antihyperglycemic agent for type 2 diabetes. Because it lowers blood glucose without increasing insulin secretion, metformin has been considered an insulin sensitizer (2). In fact, metformin showed beneficial effects in type 2 diabetes, including weight reduction, improved lipid profiles, and enhanced endothelial function (3). Thus, metformin is introduced for use in insulin-resistant states even before the development of hyperglycemia (4).

    The mechanisms of metformin action have remained obscure, despite multiple pathways of action being proposed, including a decrease of hepatic glucose production, an increase of peripheral glucose disposal, and a reduction of intestinal glucose absorption (1). It has been documented that metformin activated 5'AMP-activated protein kinase (AMPK) in hepatocytes, thereby reducing activity of acetyl-CoA carboxylase and lowering expression of a lipogenic transcription factor as well as inhibiting hepatic gluconeogenesis (5,6). Thus, AMPK seems a major signal for the action of metformin to suppress lipogenesis and induce fatty acid oxidation (5,6). In fact, plasma glucoseeClowering action is not entirely dependent on insulin (7). It has been mentioned that exercise causes an increase of glucose uptake in the skeletal muscle of diabetic and nondiabetic subjects through the translocation of GLUT-4 to cell membranes (8). This translocation of GLUT-4 is mediated through insulin-independent phosphorylation and activation of AMPK (9,10). However, direct evidence for the linkage of these two signals is lacking.

    Additionally, it has been indicated that exogenous -endorphin induces an increase of circulating insulin in humans with or without diabetes (11,12). The effect of opioids on glucose homeostasis may in fact be produced by other mechanisms in addition to insulin. Intravenous injection of synthetic -endorphin lowered plasma glucose in streptozotocin (STZ)-induced diabetic rats, as observed in our previous study (13). Chemical agents such as loperamide or tramadol increased glucose utilization via activation of opioid e?receptors to lower plasma glucose in STZ-induced diabetic rats (14,15). In obese Zucker rats, mediation of -endorphin in exercise-induced improvement of insulin resistance has also been observed (16). These findings support a beneficial effect of opioid e?receptor activation on plasma glucose regulation. Thus, the aim of this study is to clarify whether -endorphin is involved in the plasma glucoseeClowering action of metformin. The deficient functions of pancreas -cells in STZ-induced diabetic rats has been documented (17), and rats with STZ-induced diabetes were thus used in the current study as an animal model of type 1eClike diabetes.

    RESEARCH DESIGN AND METHODS

    Male Wistar rats weighing 200eC250 g were obtained from the Animal Center of the National Cheng Kung University Medical College. Male BDF1 mice (as the wild-type controls) and opioid e?receptor knockout BDF1 mice (18), aged 8eC10 weeks, all of which had been bred in the same animal center, were obtained from Dr. H.H. Loh (University of Minnesota Medical School, Minneapolis, MN). STZ-induced diabetic rats were prepared by intravenously injecting STZ (60 mg/kg) into the male Wistar rats at age 8eC10 weeks. Mice with or without opioid e?receptors also received an intraperitoneal injection of STZ at 50 mg/kg to induce diabetes (19). At 2 weeks after the injection of STZ, animals were considered to be diabetic if they had plasma glucose levels 20 mmol/l and other diabetic features, including polyuria, polydipsia, and hyperphagia. Also, plasma insulin in these STZ-induced diabetic rats was reduced to 1.23 ± 0.6 pmol/l (n = 8), a level markedly lower than that of normal rats (154.8 ± 5.2 pmol/l, n = 8). All animal procedures were performed according to the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health, as well as the guidelines of the Animal Welfare Act.

    Effect of metformin on plasma glucose or -endorphin level in STZ-induced diabetic rats.

    Metformin was dissolved in distilled water containing 0.9% (wt/vol) sodium chloride for oral administration into fasted rats at desired doses. It has been documented that rats that have received sodium pentobarbital show no changes of plasma glucose and glucagon (20). Thus, under anesthesia with sodium pentobarbital (30 mg/kg i.p.), blood samples (0.1 ml) were collected from the tail vein for the measurement of plasma glucose or -endorphineClike immunoreactivity (BER). The time course of the effect of metformin on plasma glucose in STZ-induced diabetic rats was preliminarily determined; the plasma glucoseeClowering effect of metformin at an oral dosage of 100 mg/kg reached a plateau within 60 min and was maintained for 120 min. Thus, the effects of metformin on plasma glucose and plasma BER were determined, using blood samples collected 60 min after oral administration. As controls, we used animals that had received a similar administration of saline used to dissolve metformin at the same volume. The opioid receptor antagonists known to antagonize e?subtypes, including naloxonazine and naloxone, were injected into the tail vein of animals 30 min before oral treatment with metformin.

    Effect of metformin on plasma glucose or BER level in opioid e?receptor knockout diabetic mice.

    Fasting STZ-induced diabetic mice with or without opioid e?receptors were given an oral administration of metformin at 100 mg/kg, the dose showing maximal effect in STZ-induced diabetic rats. After 60 min, blood samples (0.1 ml) were collected from the lower eyelid of mice under anesthesia with pentobarbital (30 mg/kg i.p.) for determination of plasma glucose and BER.

    Isolation and incubation of adrenal medulla.

    Adrenal glands were removed from the killed STZ-induced diabetic rats, and medullas were immediately dissected after removal of the cortex as previously described (21). The tissues were cut into 1-mm thick slices and transferred to a glass tube fitted with a mesh of nylon at the bottom to permit free interchange with the medium. The tissues were incubated with metformin at indicated concentrations with continuous shaking at 40 cycles/min under 37°C for 30 min. Incubation was terminated by placing the tubes on ice. The medium from each incubated sample was collected and frozen at eC70°C until the -endorphin assay was performed.

    Adrenalectomized rats.

    Bilateral adrenalectomy was performed in Wistar rats, using the dorsal approach as described previously (22). The adrenalectomized rats were also fed standard rat chow and 0.9% sodium chloride in their drinking water ad libitum. Sham-operated rats were fed standard rat chow and water ad libitum. Animals were allowed to recover for 2 weeks after the operation. The animals appeared alert and in good health. Then, diabetes was induced by an injection of STZ as described above.

    Laboratory determinations.

    The concentration of plasma glucose was measured by the glucose oxidase method, using an analyzer (Quik-Lab; Ames, Miles, Elkhart, IN). An enzyme-linked immunosorbent assay for the determination of BER in plasma or medium incubating adrenal medulla was carried out, using a commercially available kit (Peninsula Laboratories, San Carlos, CA).

    Determination of gene expression.

    STZ-induced diabetic rats were orally administered vehicle or metformin (100 mg/kg) every 8 h, three times daily. Naloxonazine was intravenously injected into another group of STZ-induced diabetic rats 30 min before receiving an oral administration of metformin in the same manner. In preliminary experiments, metformin was found to modify the mRNA and protein levels for GLUT-4 and hepatic PEPCK in STZ-induced diabetic rats after a 3-day treatment. Therefore, animals were killed after 3 days of treatment. Normal rats receiving a similar treatment of vehicle were used as controls. Liver and soleus muscle were immediately removed, frozen in liquid nitrogen, and stored at eC70°C for the determination of gene transcripts. Changes in hepatic PEPCK mRNA were determined by RT-PCR. PEPCK-specific primers were 5'-AGTTGAATGTGTGGGTGATGACA-3' and 5'-AAAACCGTTTTCTGGGTTGATG-3' for forward and reverse primers, respectively (23). Prophobilinogene deaminaseeCspecific primers were 5'-GGAGCCATGTCTGGTAACGGCA-3' and 5'-GGTACCCACGCGAATCACTCTCA-3' for forward and reverse primers, respectively (24). Quantification of the mRNA level was examined, using the ratio of PEPCK to prophobilinogene deaminase. The mRNA of GLUT-4 in soleus muscle was investigated by Northern blot analysis (15). The effects of testing agents on protein levels of PEPCK and GLUT-4 were evaluated by Western blot analysis (15). AntieCrat GLUT-4 antibody was purchased from Genzyme Diagnostics (Cambridge, MA). The antibody specific to PEPCK was a gift from Dr. D.K. Granner (Vanderbilt University School of Medicine, Nashville, TN). The blots were incubated with a goat polyclonal actin antibody (Santa Cruz Biotechnology, Santa Cruz, CA) or a mouse monoclonal -tubulin antibody (Zymed Laboratories, San Francisco, CA) as an internal control. Blood samples were also collected from the tail vein of these rats before death to assay the plasma levels of glucose and BER as described above. Also, the daily amount of food and water intake as well as changes of body weight in metformin-treated STZ-induced diabetic rats were measured to compare with vehicle-administered controls.

    Statistical analysis.

    Plasma glucoseeClowering activity was noted in the animals that had received metformin. Results of plasma glucoseeClowering activity were calculated as the percentage reduction of the initial value according to the formula (Gi eC Gt)/Gi x 100%, where Gi was the initial plasma glucose and Gt was the plasma glucose concentration after treatment with metformin.

    Data are expressed as the means ± SE for the number (n) of animals in the group, as indicated in tables and figures. Repeated-measures ANOVA was used to analyze the changes in plasma glucose and other parameters. Dunnett range post hoc comparisons were used to determine the source of significant differences where appropriate. P < 0.05 was considered statistically significant.

    RESULTS

    Effects of metformin on plasma glucose and BER levels in STZ-induced diabetic rats.

    At 60 min after treatment, a dose-dependent decrease of plasma glucose was observed in STZ-induced diabetic rats receiving an oral administration of metformin (Fig. 1A). The plasma glucoseeClowering activity produced by metformin at 100 mg/kg was 21.4 ± 1.8% in STZ-induced diabetic rats. Increasing the dosage of metformin to 115 mg/kg yielded a plasma glucoseeClowering activity of 21.9 ± 2.3% that was not more effective. Plasma glucose decreased from 5.3 ± 0.9 mmol/l to 4.6 ± 0.8, 4.4 ± 0.5, and 3.9 ± 0.7 mmol/l at 60 min later in normal rats receiving oral administration of metformin at 50, 75, and 100 mg/kg, respectively. The plasma glucoseeClowering activity of metformin in normal rats was as effective as that produced in STZ-induced diabetic rats. The effect of metformin at 100 mg/kg was investigated in subsequent experiments.

    Plasma BER was raised 30 min later in STZ-induced diabetic rats by oral administration of metformin. The action of metformin was maximal at 60 min, which was used as the optimal time in subsequent experiments. A dose-dependent elevation of plasma BER level was observed in the same group of STZ-induced diabetic rats receiving metformin (Fig. 1B).

    Effect of bilateral adrenalectomy on the action of metformin in STZ-induced diabetic rats.

    The basal plasma glucose or BER in STZ-induced diabetic rats was not modified by adrenalectomy, compared with the sham-operated group (Table 1). However, both the decrease of plasma glucose and the increase of plasma BER by metformin (100 mg/kg) disappeared in STZ-induced diabetic rats with bilateral adrenalectomy, whereas these effects persisted in sham-operated STZ-induced diabetic rats receiving the same treatment (Table 1).

    Effect of metformin on the secretion of BER from isolated adrenal medulla of STZ-induced diabetic rats.

    The amount of BER in the medium was increased by metformin in a concentration-dependent manner (Fig. 2). Metformin at 1 e蘭ol/l increased BER in the medium and reached a plateau, and no further effect was produced by metformin at higher concentrations (Fig. 2).

    Effects of opioid e?receptor antagonists on the action of metformin in STZ-induced diabetic rats.

    Table 2 shows the dose-dependent action of naloxone and naloxonazine to inhibit the plasma glucoseeClowering activity of metformin in STZ-induced diabetic rats. In the presence of 1 mg/kg naloxone, the plasma glucose level in STZ-induced diabetic rats treated with 100 mg/kg metformin was not statistically different from the basal level. Also, naloxonazine (1 mg/kg) prevented the ability of metformin (100 mg/kg) to lower plasma glucose in STZ-induced diabetic rats. However, both naloxone and naloxonazine at the highest dose did not affect the basal plasma glucose level of STZ-induced diabetic rats (Table 2).

    Change of metformin-induced plasma glucoseeClowering activity in opioid e?receptor knockout diabetic mice.

    Plasma glucose in opioid e?receptor knockout diabetic mice was not modified by oral administration with metformin (100 mg/kg), whereas plasma BER was elevated (Fig. 3). Similar administration of metformin (100 mg/kg) in diabetic mice with opioid e?receptors also increased plasma BER, but this action was in parallel with the plasma glucoseeClowering action (Fig. 3). The plasma glucose-lowering activity of metformin in these wild-type diabetic mice was 21.7 ± 2.1%, similar to the activity produced in STZ-induced diabetic rats.

    General characteristics of STZ-induced diabetic rats repeatedly receiving oral administration of metformin.

    Plasma glucose levels of STZ-induced diabetic rats were reduced after repeated oral administration of metformin (100 mg/kg) for 3 days (Table 3), showing a plasma glucoseeClowering activity of 23.9 ± 2.3%. Elevation of plasma BER occurred in the same group of STZ-induced diabetic rats, and it was significantly (P < 0.01) higher than in the vehicle-administered group (Table 3). The 3-day treatment with metformin (100 mg/kg) did not influence the feeding behavior and/or body weight of STZ-induced diabetic rats (Table 3).

    In the presence of naloxonazine (1 mg/kg), plasma BER levels in the STZ-induced diabetic rats treated with metformin (100 mg/kg) were still markedly raised (Table 3). However, the reduction of plasma glucose by metformin (100 mg/kg) in STZ-induced diabetic rats was abolished by naloxonazine (Table 3). The body weight of STZ-induced diabetic rats was not influenced by naloxonazine (Table 3).

    Effect of metformin on the mRNA and protein levels of GLUT-4 in soleus muscle of STZ-induced diabetic rats.

    The mRNA level of GLUT-4 in soleus muscle isolated from vehicle-administered STZ-induced diabetic rats was 47% of that from vehicle-administered normal rats (Fig. 4A). Repeated oral treatment of STZ-induced diabetic rats with metformin (100 mg/kg) for 3 days resulted in an elevation of GLUT-4 mRNA level in the soleus muscle to a level 76% of that in vehicle-administered normal rats. Also, the effect of metformin (100 mg/kg) on GLUT-4 mRNA levels in STZ-induced diabetic rats was reversed by naloxonazine (1 mg/kg) to the same level as in the vehicle-administered STZ-induced diabetic rats (Fig. 4A).

    The protein level of GLUT-4 in soleus muscle of vehicle-administered STZ-induced diabetic rats was significantly reduced to 45% of that in the vehicle-administered normal rats (Fig. 4B). Repeated oral treatment with metformin (100 mg/kg) elevated the protein level of GLUT-4 in soleus muscle of STZ-induced diabetic rats to a level 80% of that in vehicle-administered normal rats. Naloxonazine (1 mg/kg) reversed this action of metformin (Fig. 4B). However, naloxonazine alone did not affect basal GLUT-4 gene expression. Quantification of the mRNA and protein levels of GLUT-4 induced by these treatments is shown in Table 4.

    Effect of metformin on the mRNA and protein levels of hepatic PEPCK in STZ-induced diabetic rats.

    Fig. 4C shows the level of mRNA encoding PEPCK that was elevated nearly 3.4-fold in untreated STZ-induced diabetic rats compared with normal rats. This increase of mRNA level in the liver of STZ-induced diabetic rats was reduced to 33% of vehicle-administered STZ-induced diabetic rats by repeated oral treatment with metformin (100 mg/kg) for 3 days.

    The protein levels of PEPCK in the liver of vehicle-administered STZ-induced diabetic rats were about threefold of the vehicle-administered normal rats. Repeated oral treatment of STZ-induced diabetic rats with metformin (100 mg/kg) for 3 days resulted in a marked reduction of the protein level of PEPCK to near the level in vehicle-administered normal rats; which was reversed on blockade of opioid e?receptors by naloxonazine (1 mg/kg) (Fig. 4D). However, the basal levels of mRNA and protein in hepatic PEPCK were not influenced by naloxonazine alone. Quantification of all data from these treatments is also shown in Table 4.

    DISCUSSION

    Metformin has oral bioavailability, with peak plasma concentration reached after 2eC3 h (25). We found that oral treatment with metformin for 1 h can dose-dependently lower plasma glucose in a manner parallel with an increase of plasma BER in STZ-induced diabetic rats. In fact, plasma insulin levels in STZ-induced diabetic rats was only 1/120 of that in normal rats. Thus, mediation of endogenous insulin is negligible in this STZ-induced diabetic rat model. The effective dose of metformin in STZ-induced diabetic rats was similar to that used in type 2 diabetic rats (26). Also, the plasma glucoseeClowering activity of metformin in STZ-induced diabetic rats was the same as that in normal rats. These results indicated that metformin has an ability to lower plasma glucose without the help of endogenous insulin; elevation of -endorphin seems related to this action of metformin.

    Although -endorphin is released with adrenocorticotrophic hormone from the pituitary gland (27), adrenal glands are also a source of -endorphin (28,29). We have demonstrated that secretion of opioids from adrenal glands is associated with a decrease of plasma glucose in STZ-induced diabetic rats (19), which is consistent with the view that pituitary glandeCindependent release of endogenous opioids is operative in other organs (28,29). In an attempt to make certain that adrenal glands are the main source of metformin-induced release of -endorphin, we used bilateral adrenalectomy. The plasma glucoseeClowering action of metformin was eliminated by bilateral adrenalectomy in STZ-induced diabetic rats. Also, no increase of plasma -endorphin was obtained in adrenalectomized diabetic rats receiving metformin at the effective doses. Thus, adrenal glands seem responsible for the secretion of -endorphin by metformin. Moreover, metformin enhanced -endorphin secretion from the isolated adrenal medullas of STZ-induced diabetic rats in a concentration-dependent manner. Taken together, one can conclude that a release of -endorphin from adrenal glands by metformin is related to metformin’s plasma glucoseeClowering action in STZ-induced diabetic rats.

    The actions of -endorphins are mediated in part by opioid e?receptors, which are believed to be expressed in specialized neurons for pain transmission at both spinal and supraspinal sites (30). Recently, we observed that -endorphin enhanced the uptake of radioactive glucose into the isolated soleus muscle of STZ-induced diabetic rats and stimulated glycogen synthesis in the hepatocytes isolated from STZ-induced diabetic rats (13). Both actions of -endorphin were naloxone and naloxonazine sensitive (13). Thus, opioid e?receptors are also located in peripheral tissues that can be activated to lower plasma glucose by improving glucose utilization (13,15). The action of metformin in STZ-induced diabetic rats was inhibited by blockade of opioid e?receptors, using naloxone or naloxonazine. In fact, two subtypes of the e?receptors (e? and e?) have been postulated, although the e? subtype is naloxonazine sensitive (30). This suggests that the plasma glucoseeClowering action of metformin in STZ-induced diabetic rats may be mediated by peripheral opioid e?-receptor activation, which is worthwhile to be investigated in advance. However, these antagonists may have nonspecific effects in addition to the blockade of opioid e?receptors. Therefore, for further investigation, we used opioid e?receptor knockout mice receiving STZ. The plasma glucoseeClowering action of metformin was eliminated in opioid e?receptor knockout diabetic mice, although the BER-elevating action of metformin was still observed. This result supports the essential role of opioid e?receptors in plasma glucoseeClowering action of metformin during the absence of insulin. The role of cerebral opioid receptors in the regulation of food intake has been well-established (31). Although the evidence indicating the cerebral action of metformin is not documented, chronic treatment with metformin produced an anorectic effect to lower body weight in genetically obese Zucker rats (32). In fact, metformin did not influence the food intake of STZ-induced diabetic rats. This is probably because the -endorphin release induced by metformin mainly exerts its effect on opioid e?receptors located on peripheral tissues. However, this view shall be clarified in the future.

    In diabetes, elevation of blood glucose is a consequence of increased hepatic glucose output in concert with reduced peripheral glucose utilization (33). PEPCK is one of the key enzymes in hepatic gluconeogenesis (33). Insulin deficiency is clearly associated with a change in hepatic metabolism (33). Moreover, a reduction in insulin-mediated glucose uptake caused by decreasing gene expression of GLUT-4 has been reported in skeletal muscle, a major site for glucose disposal, in diabetic rats (34). Recent studies strongly support the reduction in hepatic gluconeogenesis as the primary route through which metformin exerts its glucose-lowering action (5,6). In addition, metformin has been shown to facilitate the trafficking of GLUT-4 to membrane of skeletal muscle (8eC10). Because long-term exposure is required for the activation of mRNA level, the gene expression associated with glucose regulation was examined in STZ-induced diabetic rats that had received a 3-day repeated treatment with metformin. Consistent with previous studies (5,6), the plasma glucoseeClowering activity of metformin was associated with an attenuation of the raised hepatic PEPCK gene expression in STZ-induced diabetic rats. Meanwhile, an increase in the gene expression of GLUT-4 may contribute to plasma glucose regulation in metformin-treated STZ-induced diabetic rats. Both actions of metformin were reversed by blockade of opioid e?receptors, using naloxonazine. This is consistent with the previous view that -endorphin, via activation of opioid e?receptors, is a positive regulator in glucose utilization and a negative modulator in hepatic gluconeogenesis in the insulin-deficient state (15). However, the effect of -endorphin on the brain to affect these genes cannot be excluded, and this needs more studies in the future. Also, the effect of metformin on the regulation of hepatic PEPCK gene expression in STZ-induced diabetic rats seems more effective than that in the GLUT-4 gene expression. This finding suggests that metformin exerts its antihyperglycemic effect mainly through a reduction of hepatic gluconeogenesis; this view is consistent with previous reports (5,6).

    It has been indicated that phospholipase C and protein kinase C (PKC) play a key role in opioid signals (13). Also, PKC is involved in a rate-limiting step in GLUT-4 mRNA expression (35). Therefore, the phospholipase CeCPKC pathway is related to the signals of opioid e?receptors in the regulation of GLUT-4 gene expression, although the detailed action mechanism needs further investigation. In fact, the suppression of PEPCK gene expression in STZ-induced diabetic rats by metformin was blocked by opioid e?receptor antagonist, indicating the mediation of opioid e?receptors. The gene expression of PEPCK in liver is regulated by a number of hormones (36), and signals for opioid e?receptors to regulate the hepatic PEPCK gene expression need to be clarified in the near future. Nevertheless, we demonstrated that metformin possesses an ability to enhance the secretion of -endorphin from adrenal glands of STZ-induced diabetic rats. Then, the released -endorphin can activate peripheral opioid e?receptors to modify gene expressions of GLUT-4 and PEPCK for lowering the plasma glucose level in the insulin-deficient state.

    It has been documented that splanchnic nerve stimulation increases the release of opioids from adrenal glands (37,38). Activation of 1-adrenoceptors on adrenal medulla in the regulation of -endorphin secretion has also been mentioned (19). In fact, it has been proposed that metformin releases norepinephrine by an indirect sympathomimetic-like action (39). Also, metformin inducing the release of norepinephrine from postganglionic sympathetic nerve endings (40) has been demonstrated. The role of norepinephrine in the mechanism(s) for metformin to enhance -endorphin secretion from adrenal grand needs further evidence.

    In conclusion, our results suggest that metformin exerts its antihyperglycemic effect primarily through enhancement of -endorphin secretion from adrenal glands to stimulate opioid e?receptors located on peripheral tissue, thereby leading to the amelioration of GLUT-4 gene expression and an attenuation of raised hepatic PEPCK gene expression in rats with insulin-deficient diabetes. This finding provides a new insight on the mechanisms of metformin action.

    ACKNOWLEDGMENTS

    The current study was supported in part by a grant from the National Science Council (NSC93-2320-B006-010) of China.

    We are grateful to Dr. H.H. Loh (Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN) for the kind supply of opioid e?receptor knockout mice and the wild-type mice. We also thank Dr. C. Makepeace (Department of Cell Biology and Physiology, School of Medicine, Washington University, St. Louis, MO) and Dr. S.S. Liu (Department of Microbiology and Immunology, National Cheng Kung University, Tainan City, Taiwan, China) for kindly providing us the plasmid containing cDNAs. Moreover, the kind donation of antibodies specific to PEPCK from Dr. D.K. Granner (Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN) is respectfully acknowledged.

    FOOTNOTES

    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

    El-Atat F, McFarlane SI, Sowers JR: Diabetes, hypertension, and cardiovascular derangements: pathophysiology and management. Curr Hypertens Rep 6:215eC223, 2004

    Hundal RS, Inzucchi SE: Metformin: new understandings, new uses. Drugs 63:1879eC1894, 2003

    Wulffele MG, Kooy A, de Zeeuw D, Stehouwer CD, Gansevoort RT: The effect of metformin on blood pressure, plasma cholesterol and triglycerides in type 2 diabetes mellitus: a systematic review. J Intern Med 256:1eC14, 2004

    Davies MJ, Tringham JR, Troughton J, Khunti KK: Prevention of type 2 diabetes mellitus: a review of the evidence and its application in a UK setting. Diabet Med 21:403eC414, 2004

    Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, Wu M, Ventre J, Doebber T, Fujii N, Musi N, Hirshman MF, Goodyear LJ, Moller DE: Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest 108:1167eC1174, 2001

    Lochhead PA, Salt IP, Walker KS, Hardie DG, Sutherland C: 5-aminoimidazole-4-carboxamide riboside mimics the effects of insulin on the expression of the 2 key gluconeogenic genes PEPCK and glucose-6-phosphatase. Diabetes 49:896eC903, 2000

    Santeusanio F, Di Loreto C, Lucidi P, Murdolo G, De Cicco A, Parlanti N, Piccioni F, De Feo P: Diabetes and exercise. J Endocrinol Invest 26:937eC940, 2003

    Kennedy JW, Hirshman MF, Gervino EV, Ocel JV, Forse RA, Hoenig SJ, Aronson D, Goodyear LJ, Horton ES: Acute exercise induces GLUT4 translocation in skeletal muscle of normal human subjects and subjects with type 2 diabetes. Diabetes 48:1192eC1197, 1999

    Kurth-Kraczek EJ, Hirshman MF, Goodyear LJ, Winder WW: 5' AMP-activated protein kinase activation causes GLUT4 translocation in skeletal muscle. Diabetes 48:1667eC1671, 1999

    Hayashi T, Hirshman MF, Kurth EJ, Winder WW, Goodyear LJ: Evidence for 5' AMP-activated protein kinase mediation of the effect of muscle contraction on glucose transport. Diabetes 47:1369eC1373, 1998

    Locatelli A, Spotti D, Caviezel F: The regulation of insulin and glucagon secretion by opiates: a study with naloxone in healthy humans. Acta Diabetol 22:25eC31, 1985

    Feldman M, Kiser RS, Unger RH, Li CH: Beta-endorphin and the endocrine pancreas: studies in healthy and diabetic human beings. N Engl J Med 308:349eC353, 1983

    Cheng JT, Liu IM, Tzeng TF, Tsai CC, Lai TY: Plasma glucose lowering effect of -endorphin in streptozotocin-induced diabetic rats. Horm Meta Res 34:570eC576, 2002

    Liu IM, Chi TC, Chen YC, Lu FH, Cheng JT: Activation of opioid e?receptor by loperamide to lower plasma glucose in streptozotocin-induced diabetic rats. Neurosci Lett 265:183eC186, 1999

    Cheng JT, Liu IM, Chi TC, Tzeng TF, Lu FH, Chang CJ: Plasma glucose-lowering effect of tramadol in streptozotocin-induced diabetic rats. Diabetes 50:2815eC2821, 2001

    Su CF, Chang YY, Pai HH, Liu IM, Lo CY, Cheng JT: Mediation of beta-endorphin in exercise-induced improvement in insulin resistance in obese Zucker rats. Diabetes Metab Res Rev 21:175eC182, 2005

    Wohaieb SA, Godin DV: Alterations in free radical tissue-defense mechanisms in streptozotocin-induced diabetes in rat: effect of insulin treatment. Diabetes 36:1014eC1018, 1987

    Loh HH, Liu HC, Cavalli A, Yang W, Chen YF, Wei LN: Opioid e?receptor knockout in mice: effects on ligand-induced analgesia and morphine lethality. Mol Brain Res 54:321eC326, 1998

    Liu IM, Chi TC, Shiao GC, Lin MT, Cheng JT: Loss of plasma glucose lowering response to cold stress in opioid mu-receptor knock-out diabetic mice. Neurosci Lett 307:81eC84, 2001

    Johansen O, Vaaler S, Jorde R, Reikeras O: Increased plasma glucose levels after Hypnorm anaesthesia, but not after Pentobarbital anaesthesia in rats. Lab Anim 28:244eC248, 1994

    Cheng JT, Liu IM, Kuo DH, Lin MT: Stimulatory effect of phenylephrine on the secretion of -endorphin from rat adrenal medulla in vitro. Auton Neurosci 93:31eC35, 2001

    Suzuki H, Zweifach BW, Forrest MJ, Schmid-Schnbein GW: Modification of leukocyte adhesion in spontaneously hypertensive rats by adrenal corticosteroids. J Leukoc Biol 57:20eC26, 1995

    Lamont BJ, Andrikopoulos S, Funkat A, Favaloro J, Ye JM, Kraegen EW, Howlett KF, Zajac JD, Proietto J: Peripheral insulin resistance develops in transgenic rats overexpressing phosphoenolpyruvate carboxykinase in the kidney. Diabetologia 46:1338eC1347, 2003

    Cassinat B, Zassadowski F, Balitrand N, Barbey C, Rain JD, Fenaux P, Degos L, Vidaud M, Chomienne C: Quantitation of minimal residual disease in acute promyelocytic leukemia patients with t(15;17) translocation using real-time RT-PCR. Leukemia 14:324eC328, 2000

    Scheen AJ: Clinical pharmacokinetics of metformin. Clin Pharmacokinet 30:359eC371, 1996

    Rouru J, Koulu M, Peltonen J, Santti E, Hanninen V, Pesonen U, Huupponen R: Effects of metformin treatment on glucose transporter proteins in subcellular fractions of skeletal muscle in (fa/fa) Zucker rats. Br J Pharmacol 115:1182eC1187, 1995

    Vargo T, Rossier J, Minick S, Ling N, Rivier C, Vale W, Bloom F: -Endorphin and adrenal corticotropin are secreted concomitantly by the pituitary gland. Science 197:1367eC1369, 1977

    Arefolov VA, Dmitriev AD, Tennov AV, Val’dman AV: Detection of the pro-opiomelanocortin peptide fragments—beta-endorphin and ACTH—in the adrenals of rats and mice by immunohistochemistry. Biull Eksp Biol Med 101:445eC447, 1986

    Mitsuma T, Nogimori T, Sun DH, Chaya M: Thyrotropin-releasing hormone reduces the plasma levels of beta-endorphin-like immunoreactivity in rats. Exp Clin Endocrinol 89:55eC60, 1987

    Gourlay GK: Advances in opioid pharmacology. Support Care Cancer 13:153eC159, 2005

    Pupovac J, Anderson GH: Dietary peptides induce satiety via cholecystokinin-A and peripheral opioid receptors in rats. J Nutr 132:2775eC2780, 2002

    Rouru J, Pesonen U, Koulu M, Huupponen R, Santti E, Virtanen K, Rouvari T, Jhanwar-Uniyal M: Anorectic effect of metformin in obese Zucker rats: lack of evidence for the involvement of neuropeptide Y. Eur J Pharmacol 273:99eC106, 1995

    Consoli A, Nurjhan N, Capani F, Gerich J: Predominant role of gluconeogenesis in increased hepatic glucose production in NIDDM. Diabetes 38:550eC557, 1989

    Berger J, Biswas C, Vicario PP, Strout HV, Saperstein R, Pilch PF: Decreased expression of the insulin-responsive glucose transporter in diabetes and fasting. Nature 340:70eC72, 1989

    Pessin JE, Bell GI: Mammalian facilitative glucose transporter family: structure and molecular regulation. Annu Rev Physiol 54:911eC930, 1992

    Hanson RW, Reshef L: Regulation of phosphoenolpyruvate carboxykinase (GTP) gene expression. Annu Rev Biochem 66:581eC611, 1997

    Hanbauer I, Kelly GD, Saiani L, Yang HY: [Met5]-enkephalin-like peptides of the adrenal medulla: release by nerve stimulation and functional implications. Peptides 3:469eC473, 1982

    Hexum TD, Russett LR: Stimulation of cholinergic receptor mediated secretion from the bovine adrenal medulla by neuropeptide Y. Neuropeptides 13:35eC41, 1989

    Lee JM, Peuler JD: A possible indirect sympathomimetic action of metformin in the arterial vessel wall of spontaneously hypertensive rats. Life Sci 69:1085eC1092, 2001

    Peuler JD: Opposing adrenergic actions of intravenous metformin on arterial pressure in female spontaneously hypertensive rats. Cardiovasc Res 43:237eC247, 1999(Juei-Tang Cheng, Ching-Ch)