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4期 > 正文
编号:11257045
Genistein Acutely Stimulates Insulin Secretion in Pancreatic -Cells Through a cAMP-Dependent Protein Kinase Pathway
     1 Department of Human Nutrition, Foods, and Exercise, College of Agriculture and Life Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia

    2 Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Virginia, Charlottesville, Virginia

    GLP-1, glucagon-like peptide-1; GSIS, glucose-stimulated insulin secretion; IBMX, isobutylmethylxanthine; KRBB, Krebs-Ringer bicarbonate buffer; PDE, phosphodiesterase; PKA, protein kinase A; PKAC and PKAC, PKA catalytic subunits and ; PTK, protein tyrosine kinase; RIA, radioimmunoassay; siRNA, small interfering RNA

    ABSTRACT

    Although genistein, a soy isoflavone, has beneficial effects on various tissues, it is unclear whether it plays a role in physiological insulin secretion. Here, we present evidence that genistein increases rapid glucose-stimulated insulin secretion (GSIS) in both insulin-secreting cell lines (INS-1 and MIN6) and mouse pancreatic islets. Genistein elicited a significant effect at a concentration as low as 10 nmol/l with a maximal effect at 5 e蘭ol/l. The effect of genistein on GSIS was not dependent on estrogen receptor and also not related to an inhibition of protein tyrosine kinase (PTK). Consistent with its effect on GSIS, genistein increases intracellular cAMP and activates protein kinase A (PKA) in both cell lines and the islets by a mechanism that does not involve estrogen receptor or PTK. The induced cAMP by genistein, at physiological concentrations, may result primarily from enhanced adenylate cyclase activity. Pharmacological or molecular intervention of PKA activation indicated that the insulinotropic effect of genistein is primarily mediated through PKA. These findings demonstrated that genistein directly acts on pancreatic -cells, leading to activation of the cAMP/PKA signaling cascade to exert an insulinotropic effect, thereby providing a novel role of soy isoflavones in the regulation of insulin secretion.

    Soy isoflavones have received widespread attention over the past few years because of their potential for preventing some highly prevalent chronic diseases. Genistein, the primary soy-derived isoflavone, has various biological actions, including a weak estrogenic effect by binding to estrogen receptors (1) and inhibiting protein tyrosine kinases (PTKs) (2). Studies on whether genistein has an effect on diabetes are very limited. Recent studies performed in animals and humans have shown that ingestion of soy protein associated with isoflavones moderates hyperglycemia (3,4), suggesting a beneficial role for soy in diabetes. However, it is not clear whether the beneficial effect of soy protein is due to genistein or other components. Data from recent animal studies suggest an antidiabetic effect of genistein presumably by a hypolipidemic effect (5). However, recent reports demonstrated that soy isoflavone administration lowered plasma glucose, whereas triglyceride levels were unaffected in obese and diabetic animals (6) and postmenopausal women (7). Therefore, although these data suggest that genistein may have a protective role in diabetes, the mechanism underlying these beneficial effects is still largely unknown.

    Few data exist on whether genistein has a direct effect on pancreatic -cells. Several earlier studies demonstrated that genistein stimulates insulin secretion from a clonal pancreatic -cell line (8) and cultured islets (9,10), whereas other studies have found an inhibitory effect on insulin secretion (11,12). These discrepant data may be the result of variations in the experimental conditions and model used. Nevertheless, the doses used in most of these studies (>20 e蘭ol/l) are well above those concentrations physiologically achievable through dietary means (<5 e蘭ol/l). Therefore, it is still unclear whether genistein, at physiological doses, can act on pancreatic -cells to modulate insulin secretion. In the present study, we examined whether genistein can directly regulate pancreatic -cell function through stimulation of insulin secretion. We focused on the acute effects of genistein on insulin secretion, and the cellular signaling related to this effect.

    RESEARCH DESIGN AND METHODS

    Reagents and materials.

    Culture media and supplements were from Gibco (Gaithersburg, MD); an insulin radioimmunoassay (RIA) kit was obtained from Crystal Chem (Downer Grove, IL); nitrocellulose membranes were from Schleicher & Schuell (Keene, NH); chemiluminescence detection system, stripping buffer, protein assay, and PTK assay kits were purchased from Pierce (Rockville, IL); small interfering RNA (siRNA) of protein kinase A (PKA) catalytic subunits (PKAC and PKAC) was from Dharmacon Research (Lafayette, CO); antibodies to PKAC (sc-903), PKAC (sc-904), and PKA regulatory subunit II were from Santa Cruz Biotechnology (Santa Cruz, CA); cAMP enzyme immunoassay kits were obtained from Cayman Chemical (Madison, WI); Chariot protein transfection reagent was bought from Active Motif (Carlsbad, CA); siRNA transfection reagent was from Targeting System (Santee, CA); ICI 182,780 was purchased from Tocris Cookson (Balwin, MO); and other reagents and chemicals were from Sigma (St. Louis, MO).

    Cell culture and islet isolation.

    INS-1 (a generous gift from Dr. Joseph Dillon, The University of Iowa) and MIN6 cells (provided by the Islet Core facility at the University of Virginia) were cultured in RPMI-1640 medium containing 5.5 mmol/l glucose and supplemented with 10% heat-inactivated fetal bovine serum, 1 mmol/l sodium pyruvate, 10 mmol/l HEPES, 2 mmol/l L-glutamine, 50 e蘭ol/l -mercaptoethanol, 100 units/ml penicillin, and 100 e蘥/ml streptomycin. The medium was changed every other day until the cells became confluent. Mouse islets were isolated from female C57BL/6J mice as described previously (13) and maintained in complete RPMI-1640. Before the experiment, INS-1 and MIN6 cells were preincubated in Krebs-Ringer bicarbonate buffer (KRBB; 129 mmol/l NaCl, 4.8 mmol/l KCl, 1.2 mmol/l MgSO4, 1.2 mmol/l KH2PO4, 2.5 mmol/l CaCl2, 5 mmol/l NaHCO3, 0.1% BSA, and 10 mmol/l HEPES, pH 7.4) containing 1.0 mmol/l glucose at 37°C for 2 h. Mouse islets were preincubated in KRBB containing 2.8 mmol/l glucose for the same duration.

    Insulin secretion.

    INS-1 and MIN6 cells were incubated in KRBB containing various concentrations of glucose with or without stimulating reagents. In some experiments, cells were preincubated with antagonists for 30 min before addition of genistein. Mouse islets (25 islets/tube) were incubated in different glucose concentrations with 1 e蘭ol/l genistein or various concentrations of genistein with 8 mmol/l glucose in 37°C water bath with gentle shaking for 30 min. Insulin secreted in experimental samples was measured by a RIA kit. Our preliminary experiments show that exposure of the cells to genistein for 30 min had no effect on insulin or protein content. All insulin secretion data in the present study were therefore normalized to cellular protein as determined by a protein assay kit.

    PTK activity assay.

    INS-1 cells were exposed to genistein (10 nmol/leC100 e蘭ol/l) or vehicle in the presence of 5.6 mmol/l glucose for 30 min in KRBB at 37°C. PTK activity in cell extracts was measured as previously described (14). The relative fluorescence data were normalized to corresponding protein levels.

    Intracellular cAMP assay.

    INS-1 cells were incubated in KRBB containing different glucose concentrations with 1 e蘭ol/l genistein or various concentrations of genistein with 5.6 mmol/l glucose at 37°C for 20 min. Mouse islets (25 islets/tube) were stimulated with genistein (1 and 5 e蘭ol/l) in the presence of 8 mmol/l glucose in a 37°C water bath with gentle shaking for 20 min. In some experiments, INS-1 cells were preincubated with 0.25 mmol/l isobutylmethylxanthine (IBMX), an inhibitor of phosphodiesterases (PDEs), before addition of 2.5 e蘭ol/l genistein or vehicle. Intracellular cAMP levels were determined by an enzyme immunoassay kit as previously described (15). Data were normalized to the protein concentration in samples.

    Adenylate cyclase assay.

    The plasma membranes from INS-1 cells and the mouse islets were isolated by differential centrifugation (15). The islets (50 islets/tube) were stimulated with genistein (1 and 5 e蘭ol/l) or vehicle in the presence of 8 mmol/l glucose at 37°C for 10 min before the membrane preparations. The adenylate cyclase activity assays were as described previously (15).

    PKA-specific kinase assay.

    -Cells or the islets treated with different agents or vehicle were collected in PBS supplemented with protease (1:500) and phosphatase inhibitor (1:200) cocktails. Protein was extracted and harvested by sonication and centrifugation. The enzymatic activity of PKA in lysates was assessed by measuring phosphorylation of kemptide, a highly specific peptide substrate for PKA, as described previously (15).

    Transfection for siRNA construct of PKAC and -C.

    Exponentially growing MIN6 cells were transfected with 25 pmol mouse-specific siRNA-PKAC plus 25 pmol PKAC or 50 pmol scrambled control by using a siRNA transfection reagent according to the manufacturer’s protocol. After transfection, cells were incubated with complete medium containing 10% fetal bovine serum for 48 h and then with KRBB containing 1 mmol/l glucose for 2 h before stimulation with 2.5 e蘭ol/l genistein or vehicle for 30 min. Supernatants were collected for insulin assay, and cells were either harvested by scraping in PBS for PKA activity assay or collected in lysis buffer for immunoblot analysis.

    Antibody transfection.

    Polyclonal antibodies against PKAC plus -C or a preimmune IgG were transfected into mouse islets using a Chariot reagent according to the manufacturer’s protocol. Following 5 h for the islets in culture medium and 2 h in KRBB containing 2.8 mmol/l glucose after transfection, the islets were treated with genistein for 30 min, and the supernatants were assayed for glucose-stimulated insulin secretion (GSIS). Efficacy for neutralization of the target proteins PKAC and -C by the antibodies was confirmed by PKA activity assay.

    Immunoblot analysis.

    Cell extracts were first equalized to the same protein levels and were then subjected to immunoblot analysis as previously described (14,15). Membranes were probed, stripped, and re-probed sequentially with antibodies against PKAC, PKAC, and PKA regulatory subunit II. The protein bands were digitally imaged for densitometric quantitation with a software program (ImageJ; National Institutes of Health).

    Statistical analysis.

    Data were analyzed with one-way ANOVA using the General Linear Model procedure of SAS and expressed as means ± SE. Treatment differences were subjected to a Duncan’s multiple comparison test or paired t test when designated. Differences were considered significant at P < 0.05.

    RESULTS

    Effects of genistein on GSIS.

    We first examined whether genistein has an effect on GSIS in INS-1 cells. Cells were stimulated with various concentrations of glucose with 2.5 e蘭ol/l genistein or vehicle for 30 min. The basal level of insulin secretion was 380 ± 29 pg · mgeC1 protein · mineC1. As shown in Fig. 1A, genistein alone had no effect on insulin secretion but significantly augmented GSIS. Dose-response studies demonstrated that exposure of the cells to genistein at a concentration as low as 10 nmol/l potentiated GSIS, although a maximal increase was observed at 5 e蘭ol/l genistein (Fig. 1B).

    Estrogen receptoreCindependent effect of genistein on GSIS.

    Because genistein has weak estrogenic effects in some tissues by binding to estrogen receptors (1), we examined whether the genistein effect was mediated through the estrogen receptors. Consistently, genistein increased GSIS by >50%. The estrogen receptor antagonist ICI 182,780 caused no change in genistein-potentiated GSIS (Fig. 2). In addition, exposure of INS-1 cells to 17-estradiol (1 and 10 nmol/l) for 30 min failed to significantly affect GSIS (Fig. 2). The activity of ICI 182,780 used in this study was validated through our recent study (15). These results suggest that the effect of genistein on insulin secretion is independent of estrogen signaling mechanisms.

    Effect of genistein on GSIS is independent of PTK.

    Because genistein is often used as a PTK inhibitor in studies of PTK-mediated cellular events and because PTK may be involved in regulation of insulin secretion (12), we therefore evaluated whether genistein enhances GSIS by inhibition of PTK in INS-1 cells. We first compared the effect of genistein with that of daidzein, an analog of genistein that is inactive for PTK inhibition, on GSIS. As expected, exposure of cells to genistein for 30 min augmented GSIS. Daidzein was slightly less potent than genistein but increased insulin secretion in a similar way (Fig. 3A). We then directly measured PTK activity in cells treated with genistein. Genistein, at the highest concentration used in the present study (10 e蘭ol/l), did not inhibit the basal PTK activity (Fig. 3B). Significant inhibition of PTK activity in this study was observed only at 100 e蘭ol/l genistein, consistent with previous findings that PTK inhibition requires higher concentrations of genistein (16).

    Genistein stimulates accumulation of intracellular cAMP.

    Previous studies established that the cAMP signaling pathway plays an important role in insulin secretion (17). We therefore determined whether genistein can induce intracellular cAMP, thereby elevating GSIS. Genistein, in the absence of IBMX, significantly elevated cAMP concentrations both in low and high glucoseeCcultured INS-1 cells (Fig. 4A). Dose-response studies showed that genistein as low as 10 nmol/l elevated intracellular cAMP level, with a maximal increase at 5 e蘭ol/l genistein, which was also significantly higher than those induced by 0.1 e蘭ol/l genistein (Fig. 4B).

    Genistein stimulates adenylate cyclase activity.

    To determine whether genistein elevates cAMP through stimulation of cAMP production and/or inhibition of cAMP hydrolysis, we preincubated the cells with 0.25 mmol/l IBMX before addition of 2.5 e蘭ol/l genistein. As shown in Fig. 5A, genistein or IBMX alone increased cAMP levels from 6.2 ± 0.5 to 9.5 ± 1.1 and 10.1 ± 0.7 pmol/mg, respectively. In the presence of IBMX, genistein-stimulated cAMP production was further increased by 85%, suggesting that genistein may at least partially elevate cAMP by activation of adenylate cyclase activity. Genistein (2.5 e蘭ol/l) had no additive effect on adenylate cyclase agonist forskolin-induced cAMP production, whereas IBMX potentiated forskolin-induced cAMP in INS-1 cells (data not shown), confirming a role for adenylate cyclase in genistein-induced cAMP accumulation in -cells. The level of genistein-induced cAMP was about one-third of that achieved by exposure to forskolin (9.5 ± 1.1 vs.27.7 ± 3.4 pmol/mg protein) but only 10% lower than that induced by 10 nmol/l glucagon-like peptide-1 (GLP-1) (7eC36) amide (10.7 ± 1.4 pmol/mg protein), a potent incretin that activates adenylate cyclase and increases the cAMP concentration in -cells. We further determined whether genistein activates adenylate cyclase in isolated plasma membranes of INS-1 cells. As shown in Fig. 5B, genistein significantly increased adenylate cyclase activity in plasma membranes (15.8 ± 0.9 pmol · mgeC1 protein · mineC1) compared with basal values (9.7 ± 1.0 pmol · mgeC1 protein · mineC1), a change that was comparable with that induced by GLP-1 but was about one-third that achieved by exposure to forskolin (33.1 ± 1.4 pmol · mgeC1 protein · mineC1).

    Genistein activates cAMP-dependent PKA activity that is independent of the estrogen receptors and PTK.

    Because cAMP is a direct activator of PKA, we next investigated whether the elevation of cAMP by genistein is sufficient to activate PKA in INS-1 cells. As shown in Fig. 6A, genistein potently stimulated PKA activity. The PKA activity increased in response to increasing concentrations of genistein, with a maximal response observed at 5 e蘭ol/l genistein. To further characterize the cAMP/PKA pathway, INS-1 cells were preincubated with SQ 22536 (10 e蘭ol/l), an adenylate cyclase inhibitor. As shown in Fig. 6B, inhibiting the generation of cAMP completely blocked the genistein-induced PKA activity. As expected, SQ 22536 also completely ablated forskolin-augmented PKA activity (data not shown). These data clearly demonstrated that genistein is a potent activator of the cAMP/PKA cascade. Consistently, inhibition of estrogen receptors with ICI 182,780 had no effect on genistein-stimulated PKA (Fig. 6C). In addition, daidzein (5 e蘭ol/l) was potent in stimulation of PKA activity, whereas 17-estradiol (10 nmol/l) had no obvious effect (Fig. 6C), suggesting that activation of PKA by genistein is PTK independent and estrogen receptor independent.

    Genistein-enhanced insulin secretion is mediated by PKA.

    Next, we examined whether PKA regulates the genistein-induced insulin secretion. INS-1 cells were preincubated with a specific PKA inhibitor H89 (10 e蘭ol/l) or SQ 22536 (10 e蘭ol/l) for 30 min, followed by stimulation with genistein (2.5 e蘭ol/l) for 30 min. As shown in Fig. 7A, SQ 22536 blocked the genistein-induced GSIS by 88% and H89 resulted in 66% inhibition. To further examine the specificity and role of the PKA and also to ensure that the genistein effect was not a species-specific phenomenon, we used siRNA to ablate the catalytic subunits of PKA in mouse MIN6 cells. As shown in Fig. 7B, transfection of MIN6 cells with siRNA of PKAC and -C knocked down the PKAC and -C protein expression by 73 ± 3 and 79 ± 6%, respectively, whereas scrambled construct had no effect. As a result, this transfection ablated genistein-induced PKA activity by 64%. Accordingly, ablation of PKA reduced 56% of genistein-augmented GSIS, whereas the scrambled control did not suppress the insulinotropic effect of genistein (Fig. 7C). These results further support an important role of the cAMP/PKA pathway in genistein-induced insulin secretion.

    Genistein activates the adenylate cyclase/cAMP/PKA cascade and enhances GSIS in mouse pancreatic islets.

    Finally, we tested whether genistein has a similar effect on the cAMP signaling and insulin secretion in pancreatic islets. As shown in Fig. 8A, genistein greatly potentiated GSIS in mouse islets, a similar response to that observed in INS-1 cells, suggesting that in vitro findings may have physiological relevance. In parallel to its effect on insulin secretion, genistein also significantly enhanced adenylate cyclase activity (Fig. 8B), elevated cAMP (Fig. 8C), and activated PKA (Fig. 8D) in the islets. To confirm the role of PKA in the regulation of genistein effect in the islets, we either preincubated the islets with H89 or delivered PKAC plus -C antibodies into the islets with a recently developed protein delivery reagent that has been successfully used in previous studies (18). Preincubation with the PKA inhibitor or transfection of the cells with PKA antibodies significantly attenuated genistein-induced GSIS (Fig. 8E) and PKA activity (Fig. 8F), whereas preimmune IgG had no effects.

    DISCUSSION

    The soy isoflavone genistein has various biological functions. Recent studies suggest a potential antidiabetic role for isoflavones in animals and humans (3,4). However, it is unknown whether genistein, at physiological doses, directly acts on pancreatic -cells to modulate insulin secretion. In the present study, we found that genistein, at physiologically achievable concentrations for individuals consuming soy products, potentiated GSIS both in insulin-secreting cell lines and mouse pancreatic islets. This effect of genistein was not mediated through the estrogen receptor mechanism and also was independent of PTK or NO signaling pathway. However, genistein stimulates intracellular cAMP accumulation, which subsequently activates PKA that at least partially mediates the genistein-augmented GSIS.

    Genistein at a concentration as low as 10 nmol/l significantly augmented GSIS in INS-1 cells, although the maximal effect was achieved at 5 e蘭ol/l genistein. The augmentation of GSIS by genistein in the INS-1 cells was maximal at 5.6 mmol/l glucose, with a 50% increase over control, and then gradually declined with the increase of glucose concentration. These results agreed with previous studies demonstrating that glucose-dependent insulinotropic polypeptide (19) and GLP-1 (20) maximally enhance insulin secretion in INS-1 cells at this glucose concentration. The reported serum concentrations of genistein are 0.16eC0.89 e蘭ol/l in Japanese men (21,22). However, the serum genistein level in humans consuming three meals per day containing soy milk can reach 4.6 e蘭ol/l (23). Given the fact that the plasma glucose concentration usually changes between 5 and 8 mmol/l under physiological conditions (24), our results are therefore physiologically relevant. The results observed in INS-1 cells were confirmed with mouse MIN6 -cells and isolated mouse islets, suggesting a noneCspecies-specific effect of genistein and further suggesting that physiologically relevant concentrations of genistein may augment GSIS in vivo and thereby have antidiabetic implications by directly acting on -cells.

    Genistein has weak estrogenic effects by binding to estrogen receptors (1). However, unlike genistein, 17-estradiol did not significantly alter insulin secretion. Our result is in agreement with previous findings that 17-estradiol had no effect on basal and stimulated insulin secretion in INS-1 cells overexpressing estrogen receptors (25). In addition, blocking estrogen receptors with ICI 182,780 did not inhibit the cellular response to genistein. It is unlikely that the inability of this agent to block the effect of genistein on GSIS is due to a lack of efficacy, because we previously reported that, at the same concentration used, ICI 182,780 completely abolished the 17-estradioleCinduced endothelial NO synthase activity (14). These results suggest that the observed insulinotropic effect of genistein is not related to its estrogenic effect.

    Genistein is an inhibitor of PTK (2) and is often used to study PTK-mediated signaling events. However, we provide evidence that genistein-induced GSIS is not related to PTK inhibition (8eC10) because genistein at the concentrations used in the present study (10 nmol/leC10 e蘭ol/l) failed to inhibit PTK activity. As reported (26), genistein only inhibits PTK at a much higher concentration (100 e蘭ol/l). In addition, daidzein, an analog of genistein that does not inhibit PTK, also significantly increased insulin secretion and stimulated PKA activity, although these effects are less potent than those of genistein as previously observed in other tissues (27), further supporting a PTK-independent effect of genistein. Additionally, these results suggest that genistein should be cautiously used in studies of PTK signal transduction, because increased activation of PKA by genistein may contribute to the observed effects.

    cAMP is a central signaling molecule in a variety of cellular systems. It is well established that an elevation of cAMP level and consequent activation of PKA in -cells plays an important role in incretin-stimulated insulin secretion (28,29). We found in the present study that genistein stimulated cAMP accumulation over the same concentration range as its effect on insulin secretion. We further demonstrated that in a pattern similar to the stimulated cAMP accumulation, genistein potently activated PKA activity, an effect that was blocked by inhibition of adenylate cyclase, suggesting that the elevated cAMP by genistein is essential to stimulate PKA activity, an effect that is also independent of estrogen receptors or PTK inhibition. Our results demonstrated that activation of PKA by genistein in response to cAMP plays an important role in mediating genistein-induced insulin secretion because ablating the production of cAMP or inhibition of PKA attenuated the genistein effect on insulin secretion by 88 and 66%, respectively. The efficacy of inhibitors in our assay was confirmed by demonstrating their ability to block the genistein-stimulated PKA activity (data not shown). This result was confirmed by our observation that transfecting MIN6 cells with both PKAC and PKAC siRNA or mouse islets with their polyclonal antibodies diminished insulin secretion in response to genistein, whereas the scrambled control or preimmune IgG had no effect, further emphasizing the important role of PKA in mediating genistein effect.

    PKA is a heterotetramer composed of a regulatory subunit (R) dimer and a catalytic subunit (C) dimer. It was shown that rat -cell line expresses both PKAC and -C (29), which was confirmed in the preliminary study. Although it is known that elevation of cAMP causes translocation of PKAC to the plasma membranes in -cells (30), where it may interact with L-type Ca2+ channel (31) and thereby may participate in the insulin exocytotic process, it is unclear whether PKAC primarily regulates rapid insulin secretion, thereby mediating the genistein effect. In addition, our finding that although both SQ 22536 and H89 maximally inhibited PKA, SQ 22536 inhibited genistein-induced insulin secretion to a greater extent than H89, suggesting that a PKA-independent mechanism may exist to contribute to the genistein action, although PKA apparently plays a major role. A PKA-independent pathway involving a cAMP-regulated guanine nucleotide exchange factor (Epac) has recently been suggested to play a role in cAMP stimulation of insulin exocytosis (32,33). Whether genistein also activates Epac remains to be determined.

    AC- and cAMP-specific PDEs are the primary enzymes responsible for regulation of cAMP. Recent studies by others suggested that genistein, at pharmacological concentrations, may control the degradation of cAMP by inhibiting PDEs in anterior pituitary cells (34) and airway epithelial cells (35). However, our results suggest that genistein, at physiological concentrations, may elevate cAMP primarily via activation of adenylate cyclase. First, genistein (2.5 e蘭ol/l) effect on cAMP was potentiated in the presence of potent inhibitor of PDEs. Second, unlike IBMX, genistein (2.5 e蘭ol/l) had no additive effect on adenylate cyclase agonist forskolin-induced cAMP accumulation. Third, genistein induced adenylate cyclase activity in plasma membranes of INS-1 cells and the islets. Previous studies have shown that various adenylate cyclase isoforms are expressed in -cells that are differentially regulated by Ca2+, G-proteins, and protein kinases (36eC38). A recent study reported that genistein may enhance cAMP accumulation by modifying -adrenoceptors in rat brain (39). However, how genistein activates adenylate cyclase in -cells remains to be clarified.

    Besides playing an important role in insulin secretion, it has been recently shown that cAMP has an array of beneficial effects on -cells, including protection of cells from proinflammatory cytokine- and lipid-induced damage and apoptosis (40,41), stimulation of -cell survival and proliferation (31,42), and direct regulation of insulin gene expression (43). In addition, activation of the cAMP/PKA pathway in -cells increases the expression of insulin receptor substrate-2 and activates pancreatic duodenal homeobox-1 protein, a transcriptional factor with essential functions for pancreas development and islet formation (44,45). Given the present results and the previous evidence showing a wide range of roles for cAMP in maintaining -cell function, it is tempting to speculate that, besides regulation of insulin secretion, genistein may have various beneficial effects on -cells by targeting the cellular cAMP/PKA pathway.

    ACKNOWLEDGMENTS

    D.L. has received grants from Virginia Polytechnic Institute and State University and Jeffress Memorial Trust Foundation.

    The mouse islets were generously supplied by the Islet and Cell Core Facility at the University of Virginia. We are grateful to Dr. Joseph Dillon (Department of Internal Medicine, The University of Iowa) for helpful discussions.

    FOOTNOTES

    Z.Y. is currently affiliated with Merck, Rahway, New Jersey.

    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

    Kuiper GG, Carlsson B, Grandien K, Enmark E, Haggblad J, Nilsson S, Gustafsson JA: Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors alpha and beta. Endocrinology 138:863eC870, 1997

    Akiyama T, Ishida J, Nakagawa S, Ogawara H, Watanabe S, Itoh N, Shibuya M, Fukami Y: Genistein, a specific inhibitor of tyrosine-specific protein kinases. J Biol Chem 262:5592eC5595, 1987

    Lavigne C, Marette A, Jacques H: Cod and soy proteins compared with casein improve glucose tolerance and insulin sensitivity in rats. Am J Physiol Endocrinol Metab 278:E491eCE500, 2000

    Jayagopal V, Albertazzi P, Kilpatrick ES, Howarth EM, Jennings PE, Hepburn DA, Atkin SL: Beneficial effects of soy phytoestrogen intake in postmenopausal women with type 2 diabetes. Diabetes Care 25:1709eC1714, 2002

    Mezei O, Banz WJ, Steger RW, Peluso MR, Winters TA, Shay N: Soy isoflavones exert antidiabetic and hypolipidemic effects through the PPAR pathways in obese Zucker rats and murine RAW 264.7 cells. J Nutr 133:1238eC1243, 2003

    Ali AA, Velasquez MT, Hansen CT, Mohamed AI, Bhathena SJ: Modulation of carbohydrate metabolism and peptide hormones by soybean isoflavones and probiotics in obesity and diabetes. J Nutr Biochem 16:693eC9, 2005

    Cheng SY, Shaw NS, Tsai KS, Chen CY: The hypoglycemic effects of soy isoflavones on postmenopausal women. J Womens Health (Larchmt ) 13:1080eC1086, 2004

    Ohno T, Kato N, Ishii C, Shimizu M, Ito Y, Tomono S, Kawazu S: Genistein augments cyclic adenosine 3'5'-monophosphate(cAMP) accumulation and insulin release in MIN6 cells. Endocr Res 19:273eC285, 1993

    Sorenson RL, Brelje TC, Roth C: Effect of tyrosine kinase inhibitors on islets of Langerhans: evidence for tyrosine kinases in the regulation of insulin secretion. Endocrinology 134:1975eC1978, 1994

    Jonas JC, Plant TD, Gilon P, Detimary P, Nenquin M, Henquin JC: Multiple effects and stimulation of insulin secretion by the tyrosine kinase inhibitor genistein in normal mouse islets. Br J Pharmacol 114:872eC880, 1995

    Jones PM, Persaud SJ: Tyrosine kinase inhibitors inhibit glucose-stimulated insulin secretion. Biochem Soc Trans 22:209S, 1994

    Persaud SJ, Harris TE, Burns CJ, Jones PM: Tyrosine kinases play a permissive role in glucose-induced insulin secretion from adult rat islets. J Mol Endocrinol 22:19eC28, 1999

    Yang Z, Chen M, Ellett JD, Fialkow LB, Carter JD, Nadler JL: The novel anti-inflammatory agent lisofylline prevents autoimmune diabetic recurrence after islet transplantation. Transplantation 77:55eC60, 2004

    Liu D, Homan LL, Dillon JS: Genistein acutely stimulates nitric oxide synthesis in vascular endothelial cells by a cyclic adenosine 5'-monophosphate-dependent mechanism. Endocrinology 145:5532eC5539, 2004

    Liu D, Jiang H, Grange RW: Genistein activates the 3',5'-cyclic adenosine monophosphate signaling pathway in vascular endothelial cells and protects endothelial barrier function. Endocrinology 146:1312eC1320, 2005

    Makela S, Savolainen H, Aavik E, Myllarniemi M, Strauss L, Taskinen E, Gustafsson JA, Hayry P: Differentiation between vasculoprotective and uterotrophic effects of ligands with different binding affinities to estrogen receptors alpha and beta. Proc Natl Acad Sci U S A 96:7077eC7082, 1999

    Boo YC, Sorescu G, Boyd N, Shiojima I, Walsh K, Du J, Jo H: Shear stress stimulates phosphorylation of endothelial nitric-oxide synthase at Ser1179 by Akt-independent mechanisms: role of protein kinase A. J Biol Chem 277:3388eC3396, 2002

    Aoshiba K, Yokohori N, Nagai A: Alveolar wall apoptosis causes lung destruction and emphysematous changes. Am J Respir Cell Mol Biol 28:555eC562, 2003

    Ehses JA, Casilla VR, Doty T, Pospisilik JA, Winter KD, Demuth HU, Pederson RA, McIntosh CH: Glucose-dependent insulinotropic polypeptide promotes beta-(INS-1) cell survival via cyclic adenosine monophosphate-mediated caspase-3 inhibition and regulation of p38 mitogen-activated protein kinase. Endocrinology 144:4433eC4445, 2003

    Wang Q, Li L, Xu E, Wong V, Rhodes C, Brubaker PL: Glucagon-like peptide-1 regulates proliferation and apoptosis via activation of protein kinase B in pancreatic INS-1 beta cells. Diabetologia 47:478eC487, 2004

    Adlercreutz H, Markkanen H, Watanabe S: Plasma concentrations of phyto-oestrogens in Japanese men. Lancet 342:1209eC1210, 1993

    Morton MS, Arisaka O, Miyake N, Morgan LD, Evans BA: Phytoestrogen concentrations in serum from Japanese men and women over forty years of age. J Nutr 132:3168eC3171, 2002

    Xu X, Harris KS, Wang HJ, Murphy PA, Hendrich S: Bioavailability of soybean isoflavones depends upon gut microflora in women. J Nutr 125:2307eC2315, 1995

    Yamada S, Komatsu M, Sato Y, Yamauchi K, Kojima I, Aizawa T, Hashizume K: Time-dependent stimulation of insulin exocytosis by 3',5'-cyclic adenosine monophosphate in the rat islet beta-cell. Endocrinology 143:4203eC4209, 2002

    Horn PA, Mohlig M, Osterhoff M, Wolter S, Hofmann J, Stocking C, Ostertag W, Wahl M, Schatz H, Pfeiffer A: Effect of estradiol on insulin secreting INS-1 cells overexpressing estrogen receptors. Eur J Endocrinol 142:84eC91, 2000

    Peterson G, Barnes S: Genistein inhibits both estrogen and growth factor-stimulated proliferation of human breast cancer cells. Cell Growth Differ 7:1345eC1351, 1996

    Vissac-Sabatier C, Bignon YJ, Bernard-Gallon DJ: Effects of the phytoestrogens genistein and daidzein on BRCA2 tumor suppressor gene expression in breast cell lines. Nutr Cancer 45:247eC255, 2003

    Holz GG, Leech CA, Habener JF: Activation of a cAMP-regulated Ca(2+)-signaling pathway in pancreatic beta-cells by the insulinotropic hormone glucagon-like peptide-1. J Biol Chem 270:17749eC17757, 1995

    Yang S, Fransson U, Fagerhus L, Holst LS, Hohmeier HE, Renstrom E, Mulder H: Enhanced cAMP protein kinase A signaling determines improved insulin secretion in a clonal insulin-producing beta-cell line (INS-1 832/13). Mol Endocrinol 18:2312eC2320, 2004

    Gao Z, Young RA, Trucco MM, Greene SR, Hewlett EL, Matschinsky FM, Wolf BA: Protein kinase A translocation and insulin secretion in pancreatic beta-cells: studies with adenylate cyclase toxin from Bordetella pertussis. Biochem J 368:397eC404, 2002

    Gomez E, Pritchard C, Herbert TP: cAMP-dependent protein kinase and Ca2+ influx through L-type voltage-gated calcium channels mediate Raf-independent activation of extracellular regulated kinase in response to glucagon-like peptide-1 in pancreatic beta-cells. J Biol Chem 277:48146eC48151, 2002

    Kang G, Joseph JW, Chepurny OG, Monaco M, Wheeler MB, Bos JL, Schwede F, Genieser HG, Holz GG: Epac-selective cAMP analog 8-pCPT-2'-O-Me-cAMP as a stimulus for Ca2+-induced Ca2+ release and exocytosis in pancreatic beta-cells. J Biol Chem 278:8279eC8285, 2003

    Dyachok O, Gylfe E: Ca(2+)-induced Ca(2+) release via inositol 1,4,5-trisphosphate receptors is amplified by protein kinase A and triggers exocytosis in pancreatic beta-cells. J Biol Chem 279:45455eC45461, 2004

    Ogiwara T, Chik CL, Ho AK: Tyrosine kinase inhibitors enhance GHRH-stimulated cAMP accumulation and GH release in rat anterior pituitary cells. J Endocrinol 152:193eC199, 1997

    Burvall KM, Palmberg L, Larsson K: The tyrosine kinase inhibitor genistein increases basal cAMP and potentiates forskolin-induced cAMP accumulation in A549 human airway epithelial cells. Mol Cell Biochem 240:131eC133, 2002

    Thams P, Capito K, Hedeskov CJ: Differential effects of Ca2+-calmodulin on adenylate cyclase activity cyclase activity in mouse and rat pancreatic islets. Biochem J 206:97eC102, 1982

    Tian Y, Laychock SG: Protein kinase C and calcium regulation of adenylyl cyclase in isolated rat pancreatic islets. Diabetes 50:2505eC2513, 2001

    Portela-Gomes GM, Abdel-Halim SM: Overexpression of Gs proteins and adenylyl cyclase in normal and diabetic islets. Pancreas 25:176eC181, 2002

    Quesada A, Etgen AM: Tyrosine kinase effects on adrenoceptor-stimulated cyclic AMP accumulation in preoptic area and hypothalamus of female rats: modulation by estradiol. Brain Res 861:117eC125, 2000

    Li L, El-Kholy W, Rhodes CJ, Brubaker PL: Glucagon-like peptide-1 protects beta cells from cytokine-induced apoptosis and necrosis: role of protein kinase B. Diabetologia 48:1339eC1349, 2005

    Kwon G, Pappan KL, Marshall CA, Schaffer JE, McDaniel ML: cAMP dose-dependently prevents palmitate-induced apoptosis by both protein kinase A- and cAMP-guanine nucleotide exchange factor-dependent pathways in beta-cells. J Biol Chem 279:8938eC8945, 2004

    Jhala US, Canettieri G, Screaton RA, Kulkarni RN, Krajewski S, Reed J, Walker J, Lin X, White M, Montminy M: cAMP promotes pancreatic beta-cell survival via CREB-mediated induction of IRS2. Genes Dev 17:1575eC1580, 2003

    Philippe J, Missotten M: Functional characterization of a cAMP-responsive element of the rat insulin I gene. J Biol Chem 265:1465eC1469, 1990

    Hennige AM, Burks DJ, Ozcan U, Kulkarni RN, Ye J, Park S, Schubert M, Fisher TL, Dow MA, Leshan R, Zakaria M, Mossa-Basha M, White MF: Upregulation of insulin receptor substrate-2 in pancreatic beta cells prevents diabetes. J Clin Invest 112:1521eC1532, 2003

    Wang X, Zhou J, Doyle ME, Egan JM: Glucagon-like peptide-1 causes pancreatic duodenal homeobox-1 protein translocation from the cytoplasm to the nucleus of pancreatic beta-cells by a cyclic adenosine monophosphate/protein kinase A-dependent mechanism. Endocrinology 142:1820eC1827, 2001(Dongmin Liu, Wei Zhen, Za)