Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

The Anthocyanin Delphinidin 3-Rutinoside Stimulates Glucagon-Like Peptide-1 Secretion in Murine GLUTag Cell Line via the Ca2+/Calmodulin-Dependent Kinase II Pathway

Abstract

Glucagon-like peptide-1 (GLP-1) is an incretin hormone secreted from enteroendocrine L-cells. Although several nutrients induce GLP-1 secretion, there is little evidence to suggest that non-nutritive compounds directly increase GLP-1 secretion. Here, we hypothesized that anthocyanins induce GLP-1 secretion and thereby significantly contribute to the prevention and treatment of diabetes. Delphinidin 3-rutinoside (D3R) was shown to increase GLP-1 secretion in GLUTag L cells. The results suggested that three hydroxyl or two methoxyl moieties on the aromatic ring are essential for the stimulation of GLP-1 secretion. Notably, the rutinose moiety was shown to be a potent enhancer of GLP-1 secretion, but only in conjunction with three hydroxyl moieties on the aromatic ring (D3R). Receptor antagonist studies revealed that D3R-stimulates GLP-1 secretion involving inositol 1,4,5-trisphosphate receptor-mediated intracellular Ca2+ mobilization. Treatment of GLUTag cells with a Ca2+/calmodulin-dependent kinaseII (CaMKII) inhibitor (KN-93) abolished D3R-stimulated GLP-1 secretion. In addition, treatment of GLUTag cells with D3R resulted in activation of CaMKII. Pre-treatment of cells with a G protein-coupled receptor (GPR) 40/120 antagonist (GW1100) also significantly decreased D3R-stimulated GLP-1 secretion. These observations suggest that D3R stimulates GLP-1 secretion in GLUTag cells, and that stimulation of GLP-1 secretion by D3R is mediated via Ca2+-CaMKII pathway, which may possibly be mediated by GPR40/120. These findings provide a possible molecular mechanism of GLP-1 secretion in intestinal L-cells mediated by foods or drugs and demonstrate a novel biological function of anthocyanins in regards to GLP-1 secretion.

Introduction

Glucagon-like peptide-1 (GLP-1) secreted from enteroendocrine L-cells is one type of incretin and stimulates glucose-dependent insulin secretion and proliferation of pancreatic β-cells [13]. Due to its established role in the metabolic response, particularly glucose homeostasis, GLP-1 is an important factor in the treatment and prevention of type 2 diabetes. Several therapeutic approaches to enhance GLP-1 action are being studied and include the use of GLP-1 analogs, which improve glycemic control in type 2 diabetes patients [4, 5]. However, GLP-1 analogs are not suitable for oral administration and must be hypodermically injected. As circulating GLP-1 is rapidly inactivated by the enzyme dipeptidyl peptidase IV (DPP-4) through cleavage of the N-terminal region of intact GLP-1 [6, 7], DPP-4 inhibitors are promising therapeutic agents for extending the half-life of endogenously secreted GLP-1. To date, several DPP-4 antagonists have been identified that ameliorate hyperglycemia in type 2 diabetes patients [8, 9].

These approaches are effective for controlling blood glucose levels in type 2 diabetic patients. However, an alternative therapeutic approach is to increase endogenous GLP-1 secretion through modulation of the secretory mechanisms in intestinal L cells using pharmaceutical agents or dietary factors. This novel therapeutic strategy may help treat diabetes and decrease the required doses of other diabetic medicines. A number of nutrients and small molecules are reported to increase GLP-1 secretion in vitro and in vivo and include certain fatty acids [1012], as well as glutamine and arginine, which are well-characterized GLP-1 secretagogues [1315]. Protein hydrolysates have also been reported to induce enhanced GLP-1 secretion [1619].

We previously demonstrated that curcumin, a yellow pigment isolated from turmeric, markedly increases GLP-1 secretion in the murine GLUTag cell line [20]. Despite the abundant evidence that several nutrients and drug candidates stimulate GLP-1 secretion, there is little evidence that non-nutritive food compounds, and not the nutrients themselves, are able to directly enhance GLP-1 secretion.

Anthocyanins are flavonoid phytopigments [21] that are found naturally in plants in the form of glycosides and are widely available in fruits and vegetables commonly consumed by humans. Recent research suggests that the consumption of anthocyanin-rich foods is associated with various health benefits [2124]. Our research group demonstrated that anthocyanin-rich extract (bilberry and black soybean) reduces blood glucose levels and improves insulin sensitivity in type 2 diabetic mice [25, 26]. A recent epidemiological study showed that a higher consumption of anthocyanins and anthocyanin-rich fruit is associated with a lower risk of type 2 diabetes [24]. The molecular mechanism underlying this effect can be explained by the activation of AMP-activated protein kinase [25, 26]. However, at least six principal types of anthocyanidins have been identified to date (Fig 1), and many anthocyanins can be derived from these six types through structural modifications, such as the addition of substituent groups on the B ring, conjugation of various types and numbers of sugars, and the presence or absence of an acyl group. For this reason, anthocyanins may have various anti-diabetic effects via mechanisms other than AMP-activated protein kinase activation, such as the modification of GLP-1 activity. We hypothesized that anthocyanins facilitate GLP-1 secretion and thereby contribute to the prevention and treatment of diabetes.

In the present study, we demonstrated that delphinidin 3-rutinoside (D3R) significantly increased GLP-1 secretion in GLUTag cells, and clarified the structure-activity relationship using anthocyanin derivatives. Moreover, this increase was found to involve the inositol 1,4,5-trisphosphate receptor (IP3R)-mediated intracellular Ca2+ mobilization-Ca2+/calmodulin-dependent kinase II (CaMKII) pathway.

Materials and Methods

Chemicals

The purity of all administered chemicals was over 98%. Commercially available purified anthocyanins (pelargonidin, Pel; pelargonidin 3-glucoside, P3G; cyanidin, Cy; cyanidin 3-glucoside, C3G; cyanidin 3-rutinoside, C3R; delphinidin, Del; delphinidin 3-glucoside, D3G; D3R; peonidin, Peo; peonidin 3-glucoside, Peo3G; peonidin 3-rutinoside, Peo3R; Petunidin, Pet; petunidin 3-glucoside, Pet3G; malvidin, Mal; malvidin 3-glucoside, Mal3G; and malvidin 3,5-diglucoside, Mal3,5dG) were obtained from Tokiwa Phytochemical (Chiba, Japan) and Extrasynthèse (Genay, France). Malvidin 3-rutinoside (Mal3R, > 96% purity) was isolated and purified from tuber dry powder of black ginger (Kaempferia parviflora L.) using preparative ODS-HPLC. The structure of isolated Mal3R was verified by MS and NMR. The chemical structures of these anthocyanins are shown in Fig 1. Forskolin (Fos), ionomycin, 3-isobutyl-1-methylxanthine (IBMX), sodium dantrolene, verapamil chloride, H-89, KN-93, and Gö6983 were obtained from Wako Pure Chemical Industries (Osaka, Japan). 2-Aminoethyl diphenylborinate (2-APB) and GW1100 were purchased from Cayman Chemical (Ann Arbor, MI). NF449 was obtained from Abcam (Cambridge, MA). Rabbit polyclonal phospho-CaMKII (Thr286) antibody (#3361), rabbit monoclonal CaMKII antibody (#4436), and rabbit polyclonal β-actin antibody (#4967) were purchased from Cell Signaling Technology (Beverly, MA). BAPTA-AM and PD98059 were obtained from Merck (Darmstadt, Germany).

Cell culture and GLP-1 secretion

The murine GLUTag L cell line [27] (a gift from Dr. D. J. Drucker, University of Toronto, Toronto, Canada) was routinely cultured in Dulbecco’s modified Eagle’s medium containing 25 mM glucose supplemented with 10% fetal bovine serum at 37°C in a humidified atmosphere containing 5% CO2. For secretion experiments, once the cells reached 80% confluence after seeded on 24-well plates at a density of 2 x 105 cells/well, the medium was replaced with glucose free Krebs-Ringer bicarbonate buffer (120 mM NaCl, 5 mM KCl, 2 mM CaCl2, 1 mM MgCl2, and 22 mM NaHCO3) supplemented with 0.5% (w/v) fatty acid-free BSA to starve the cells for 1 h. The cells were then incubated with various test compounds in glucose free Krebs-Ringer bicarbonate buffer containing 0.5% fatty acid-free BSA for 2 h [20]. After treatment, the medium was collected and centrifuged at 800 × g for 5 min at 4°C to remove any floating cells. Secreted GLP-1 was assayed using an ELISA specific for GLP-1(7–36 amide) and GLP-1(7–37) (GLP-1 ELISA-kit, Millipore, St. Charles, MS) according to the manufacturer’s instructions.

GLUTag cell treatment and western blotting

The medium of cultured GLUTag cells was replaced with serum-free Dulbecco’s modified Eagle’s medium containing 1% BSA for 3 h. After incubation, the cells were treated with vehicle (0.1% DMSO) or D3R for the indicated time periods and conditions and then lysed [28]. Supernatant aliquots were treated with Laemmli sample buffer for 5 min at 100°C [29], and the samples (20 μg protein) were separated by SDS-PAGE. After electrophoresis, proteins were transblotted onto nitrocellulose membranes and probed with various primary antibodies for 16 h at 4°C. The proteins were then reacted with horseradish peroxidase-conjugated anti-rabbit antibody, and immunoreactivity was visualized using Pierce Western Blotting Substrate (Thermo Fisher Scientific, Yokohama, Japan). The relative density of the stained proteins was evaluated using Multi Gauge Ver 3.0 Densitograph Software (Fuji Film, Tokyo, Japan).

Measurement of intracellular cAMP levels

Cells were extracted by treatment with 0.1 M HCl to avoid degradation of cAMP, and the cAMP levels of the extract were measured using a cyclic AMP EIA kit (Cayman Chemical, Ann Arbor, MI) according to the manufacturer’s instructions.

Statistical analysis

All data are expressed as the means ± SEM. Differences in the GLP-1 secretion or cytosolic cAMP concentrations of GLUTag cells were compared by the Tukey-Kramer test. In other cases, the data were analyzed by two-way ANOVA. If the interaction effect of two components (D3R and inhibitor) was significant, the Tukey-Kramer test was performed to compare the differences between the groups. For all statistical tests, values without a common letter (a, b, c, d and e) on the bar graphs are significantly different at P values < 0.05.

Results

D3R significantly stimulates GLP-1 secretion from GLUTag cells

We first screened various types of anthocyanins for their ability to stimulate GLP-1 secretion in GLUTag cells. The screening revealed that three anthocyanins, D3R, which contains three B-ring hydroxyl moieties and rutinose, Del (the aglycone of D3R), and Mal, which has two methoxyl moieties on the B-ring, resulted in a significant increase in the secretion of GLP-1 by GLUTag cells (Fig 2A). Notably, however, the GLP-1 secretion induced by D3R was markedly higher compared with that induced by Del and Mal treatment. In addition, D3R stimulated GLP-1 secretion in a concentration-dependent manner (Fig 2B). Treatment of GLUTag cells with 10 μM D3R was sufficient to significantly stimulate GLP-1 secretion. However, we administered a final concentration of 100 μM of D3R to obtain clearer measurements of the inhibitory effects in the cell signaling pathway experiments (Fig 2B). Interestingly, glucosyl anthocyanins (P3G, C3G, D3G, Peo3G, Pet3G, Mal3G, and Mal3,5dG) did not significantly increase GLP-1 secretion (data not shown). Additionally, the other examined aglycones (Pel, Cy, Peo, and Pet) and rutinosyl anthocyanins (C3R, Peo3R, and Mal3R) had no detectable effect on GLP-1 secretion. In addition, we confirmed that rutinose itself did not stimulate GLP-1 secretion (Fig 2A), and that the treatment of GLUTag cells with up to 100 μM of each tested anthocyanin for 2 h had no cytotoxic effects, as shown by a cell viability >98% based on analysis using an automatic live cell counter system (Countess, Life Technologies, Tokyo, Japan) (data not shown). Based on the results of this initial screening, D3R was selected for further studies of the GLP-1 secretion mechanism.

thumbnail
Fig 2. GLP-1 secretion in the medium of GLUTag cells treated with various anthocyanins.

All examined (A) anthocyanins (100 μM) and sugars (rutinose, glucose, and rhamnose; 100 μM), or (B) varying concentrations of D3R were administered for 2 h. The GLP-1 concentration in the medium was then determined by ELISA. Secreted GLP-1 levels are expressed as the fold change of the control levels (= 1.0). Values are expressed as the means ± SEM, n = 3–9. Values without a common letter (a, b, c, d, and e) are significantly different at P < 0.05 (Tukey-Kramer test).

https://doi.org/10.1371/journal.pone.0126157.g002

D3R-induced GLP-1 secretion is regulated by endogenous Ca2+ mobilization

GLP-1 secretion occurs through exocytosis and is regulated by various signaling pathways [3033]. In particular, increases in cytosolic Ca2+ levels via the opening of Ca2+ channels or mobilization of intracellular Ca2+ stores leads to GLP-1 secretion [34]. To investigate whether D3R affected intracellular Ca2+ levels, GLUTag cells were first treated with various Ca2+ fluorescent indicators (Fura2-AM: excitation at 340/380 nm and emission at 510 nm, Fluo3-AM: excitation at 508 nm and emission at 527 nm, Fluo4-AM: excitation at 495 nm and emission at 518 nm, and CaSiR-1 AM: excitation at 650 nm and emission at 664 nm), and the intracellular fluorescence was monitored during exposure to D3R. However, the modulation of intracellular Ca2+ levels in response to D3R administration could not be detected, because the inherent fluorescence of D3R interfered with the detection of the fluorescent Ca2+ indicators (data not shown). Therefore, to indirectly elucidate the intracellular Ca2+ response of GLUTag cells to D3R, we used BAPTA-AM (intracellular Ca2+ chelator) and examined the effect of intracellular Ca2+ chelation on D3R-stimulated GLP-1 secretion. Pre-treatment of GLUTag cells with a BAPTA-AM completely blocked D3R-induced GLP-1 secretion (Fig 3A).

thumbnail
Fig 3. Effect of Ca2+ signaling pathway inhibitor on D3R-stimulated GLP-1 secretion in GLUTag cells.

GLUTag cells were pre-treated with vehicle (0.1% DMSO) or (A) endogenous Ca2+ chelator (BAPTA-AM, 10 μM), (B) L-type Ca2+ channel blocker (verapamil, 20 μM), (C, D) endogenous Ca2+ channel blocker (dantrolene, 25 μM; 2-APB, 50 μM) for 15 min, followed by treatment with vehicle or D3R (100 μM) for 2 h without washing out. GLP-1 levels in the medium were measured by ELISA. Secreted GLP-1 levels are expressed as the fold change of the control levels (= 1.0). Values are expressed as the means ± SEM, n = 3. Values without a common letter (a, b, and c) are significantly different at P < 0.05 (Tukey-Kramer test followed by two-way ANOVA).

https://doi.org/10.1371/journal.pone.0126157.g003

To examine the GLP-1 secretion-related Ca2+ response triggered by D3R in more detail, the effect of various Ca2+ signaling inhibitors on D3R-stimulated GLP-1 secretion was investigated. In response to increasing intracellular glucose concentration, the ATP/ADP ratio increases and KATP channels close. This change triggers the opening of Ca2+ channels and stimulates GLP-1 exocytosis. Therefore, we examined the effect of the L-type Ca2+ channel blocker verapamil on D3R-stimulated GLP-1 secretion. Pre-treatment of GLUTag cells with verapamil did not affect GLP-1 secretion induced by D3R (Fig 3B).

The release of Ca2+ from intracellular Ca2+ stores is enhanced following activation of the ryanodine receptor (RyR) and IP3R. Here, GLUTag cells pre-treated with the RyR antagonist dantrolene did not canceled increased GLP-1 secretion after exposure to D3R (Fig 3C). In contrast, the pre-treatment of cells with the IP3 receptor antagonist 2-APB to inhibit intracellular Ca2+ mobilization via the IP3 receptor significantly reduced D3R-stimulated GLP-1 secretion (Fig 3D).

Activation of CaMKII is involved in D3R-induced GLP-1 secretion

Ca2+ is a well-known regulator of exocytosis [35]. One of the main factors involved in Ca2+-mediated signaling is Ca2+/calmodulin-dependent protein kinase II (CaMKII), which is regulated via the binding of Ca2+-calmodulin, resulting in autophosphorylation of stimulatory and inhibitory sites [36]. Previous studies have shown that activation of CaMKII is involved in drug candidate-stimulated secretion of insulin from β-cells [37, 38]. As we recently demonstrated that CaMKII activation is involved in GLP-1 secretion in GLUTag cells treated with curcumin [20], we speculated that D3R-stimulated GLP-1 secretion proceeds by a similar mechanism. Therefore, we examined the effect of a CaMKII inhibitor on GLP-1 secretion induced by D3R. Pre-treatment of GLUTag cells with the CaMKII inhibitor KN-93 completely blocked D3R-induced GLP-1 secretion (Fig 4A). Based on this result, the effect of D3R on the phosphorylation of CaMKII was next examined. In GLUTag cells treated with D3R, phosphorylation of CaMKII protein was significantly induced in a time-dependent manner (Fig 4B). CaMKII phosphorylation was first detected at 5 min and continued to increase, reaching peak levels by 60 min (Fig 4B). The D3R-induced phosphorylation of CaMKII protein proceeded in a dose-dependent manner (Fig 4C). The observed phosphorylation pattern is consistent with the GLP-1 secretion profile of D3R-treated cells.

thumbnail
Fig 4. Effect of D3R on CaMKII in GLUTag cells.

(A) GLUTag cells were pre-treated with vehicle (0.1% DMSO) or CaMKII inhibitor (KN-93, 10 μM) for 15 min, followed by treatment with vehicle or D3R (100 μM) for 2 h without washing out. GLP-1 levels in the medium were measured by ELISA. Secreted GLP-1 levels are expressed as the fold change of the control levels (= 1.0). Values are expressed as the means ± SEM, n = 3. Values without a common letter (a, b, and c) are significantly different at P < 0.05 (Tukey-Kramer test followed by two-way ANOVA). (B, C) Immunoblot analysis of the effect of D3R treatment duration (B) and dose (C) on phosphorylated CaMKII, total CaMKII, and β-actin protein. Cells were treated with 100 μM D3R for the indicated durations (B) or with concentrations of D3R ranging from 10 to 100 μM for 60 min (C). Protein intensity was expressed relative to the control (= 1.0) after normalization using the protein intensity of total CaMKII. Values are expressed as the means ± SEM, n = 3. Values without a common letter are significantly different at P < 0.05 (Tukey-Kramer test).

https://doi.org/10.1371/journal.pone.0126157.g004

GPR40/120 is involved in D3R-stimulated GLP-1 secretion, but not other possible signaling pathways

It is known that GLP-1 secretion is stimulated via the activation of G protein-coupled receptors (GPRs) in intestinal L cells [39]. For example, GPR40, GPR119, GPR120, and TGR5 mediate the secretion of GLP-1 enteroendocrine cells [4043]. The coupling of GPR40/120 to the Gαq subunit leads to the IP3-mediated release of intracellular Ca2+ [42, 44], whereas Gαs-coupled GPR119 and TGR5 stimulate GLP-1 secretion via the cAMP/protein kinase A (PKA)-dependent pathway [43, 45].

In addition, recent studies have shown that GLP-1 secretagogues are stimulated by the protein kinase C (PKC) or mitogen-activated or extracellular signal-regulated protein kinase (MEK)-extracellular signal-regulated kinase (ERK) pathways [17, 4648]. Here, to identify the mechanism underlying D3R-induced GLP-1 secretion, we investigated the possible involvement of these pathways.

The effect of the GPR40/120 antagonist GW1100 on D3R-stimulated GLP-1 secretion was examined (Fig 5A). The pre-treatment of GLUTag cells with GW1100 significantly decreased GLP-1 secretion in D3R-treated cells compared to vehicle. In contrast, D3R-induced GLP-1 secretion was not affected by the pre-treatment of cells with NF449, a Gαs subunit-selective antagonist that serves as an indicator of GPR119 and TGR5 inhibition (Fig 5B). The treatment of GLUTag cells with D3R did not significantly affect intracellular cAMP levels (Fig 5C), and D3R-stimulated GLP-1 secretion was also not affected by the pre-treatment of cells with a PKA inhibitor (H-89; Fig 5D). In addition, pre-treatment of GLUTag cells with a PKC inhibitor (Gö6983) or a MEK inhibitor (PD98059) did not affect GLP-1 secretion induced by D3R (S1 Fig).

thumbnail
Fig 5. Effect of D3R on GPR signaling pathway in GLUTag cells.

(A, B, D) GLUTag cells were pre-treated with vehicle (0.1% DMSO) or (A) GPR40/120 antagonist (GW1100, 10 μM), (B) Gαs subunit antagonist (NF449, 10 μM), (D) PKA inhibitor (H-89, 10 μM) for 15 min, followed by treatment with vehicle or D3R (100 μM) for 2 h without washing out. GLP-1 levels in the medium were measured by ELISA. Secreted GLP-1 levels are expressed as the fold change of the control levels (= 1.0). (C) Cytosolic cAMP concentrations in GLUTag cells treated with vehicle (0.1% DMSO), positive control (Fos, 10 μM + IBMX, 10 μM; F/I), or D3R (50 or 100 μM) after 15 min. Values are expressed as the means ± SEM, n = 3. Values without a common letter (a, b, c, and d) are significantly different at P < 0.05 (A, B, D, Tukey-Kramer test followed by two-way ANOVA; C, Tukey-Kramer test).

https://doi.org/10.1371/journal.pone.0126157.g005

Discussion

In the present study, we hypothesized that anthocyanins have multiple biological functions and aid in GLP-1 secretion, and that anthocyanins have an anti-diabetic effect through the stimulation of GLP-1 secretion. Our findings demonstrate that anthocyanins, particularly D3R, have a significant effect on GLP-1 secretion and therefore have a unique pharmacological function in the regulation of glucose homeostasis.

The treatment of GLUTag cells with various anthocyanins significantly stimulated GLP-1 secretion. The results suggested that modification of the aromatic ring with at least three hydroxyl or two methoxyl moieties is essential for stimulating GLP-1 secretion. Notably, the rutinose moiety was shown to be a potent enhancer of GLP-1 secretion, but only in conjunction with three hydroxyl moieties on the aromatic ring (D3R), as rutinose alone did not stimulate GLP-1 secretion. Interestingly, Mal3R, which contains two methoxyl groups on the aromatic ring and a rutinose moiety, did not significantly stimulate GLP-1 secretion. To our knowledge, this is the first study to demonstrate that certain anthocyanins enhance GLP-1 secretion.

In general, anthocyanins exhibit low bioavailability [49]. In this study, although 10 μM of D3R significantly stimulate GLP-1 secretion (Fig 2B), the higher levels of concentrations that were tested are not achievable in blood in vivo. However, it is not necessary for such high concentrations of D3R to be absorbed into the body and directly distributed to target cells, as these concentrations are achievable in the gut lumen, where they can exert a significant impact on enteroendocrine L cells.

To explain how D3R induces GLP-1 secretion in GLUTag cells, two molecular mechanisms are possible: 1) elevation of cytosolic Ca2+ via endogenous Ca2+ store mobilization and/or extracellular influx from L-type Ca2+ channels results in activation of CaMKII, which may be involved in GLP-1 secretion, as well as curcumin-induced GLP-1 secretion [20]; and 2) stimulation of the cAMP/PKA, PKCζ, or ERK pathways [17, 43, 4548]. In the present study, we demonstrated that D3R-stimulated GLP-1 secretion is completely abolished by the pre-administration of an intracellular Ca2+ chelator (BAPTA-AM). Furthermore, D3R-induced GLP-1 secretion was significantly inhibited by an IP3 receptor antagonist (2-APB), but not by a RyR antagonist (dantrolene) or L-type Ca2+ channel blocker (verapamil). Taken together, these results indicate that D3R stimulates intracellular Ca2+ mobilization via IP3R.

One important target of Ca2+-mediated signaling molecules is CaMKII, which is regulated via binding to Ca2+-calmodulin, resulting in autophosphorylation of stimulatory and inhibitory sites [36]. Interestingly, evidence suggests that the activation of CaMKII in response to elevated cytosolic Ca2+ levels stimulates insulin secretion in β-cells [37, 38], possibly indicating that GLP-1 secretion is induced by CaMKII activation via cytosolic Ca2+ elevation. Supporting this speculation, we recently showed that curcumin-induced GLP-1 secretion is regulated by the Ca2+-CaMKII pathway [20]. As shown in Fig 4A, the pre-treatment of GLUTag cells with the CaMKII inhibitor KN-93 abolished D3R-induced GLP-1 secretion. In addition, D3R induced the phosphorylation of CaMKII in GLUTag cells in both a time- and concentration-dependent manner. Together, these results indicate that the Ca2+-CaMKII pathway regulates D3R-induced GLP-1 secretion in GLUTag cells. However, the exact exocytosis mechanism of GLP-1 from L cell remains unclear. A recent study indicated that vesicle-associated membrane protein 2 (VAMP2), which is involved in vesicular transport and membrane fusion, has an essential role in GLP-1 exocytosis [30]. Interestingly, binding of CaMKII to syntaxin1a, which interacts with VAMP2, is an important process in the regulation of exocytosis [50], suggesting that the D3R-mediated stimulation of CaMKII modulates VAMP2-mediated GLP-1 exocytosis.

The present study raises another question regarding the primary molecular target of D3R. GPR agonists have been shown to stimulate GLP-1 secretion in intestinal L cells [4043]. Here, the pre-treatment of GLUTag cells with a GPR40/120 antagonist, but not a GPR119 or TGR5-related Gαs subunit antagonist, resulted in a significant decrease of D3R-stimulated GLP-1 secretion, suggesting that GPR40/120-intracellular Ca2+ mobilization via IP3R is involved in this process. Although it is also possible that D3R-stimulated GLP-1 secretion is regulated by the GPR119- or TGR5-related cAMP/PKA, PKCζ, or MEK-ERK pathways, our results do not support this conclusion.

Conclusions

We demonstrated that the anthocyanin D3R significantly stimulates GLP-1 secretion in GLUTag cells through increased Ca2+-CaMKII pathway activation, which may be mediated by GPR40/120 (Fig 6). However, the secretion of GLP-1 is independent of the cAMP/PKA, PKC, and MEK-ERK pathways. These findings provide a possible molecular mechanism of GLP-1 secretion in intestinal L-cells mediated by foods or drugs and demonstrate a novel biological function of anthocyanins in regards to GLP-1 secretion.

thumbnail
Fig 6. Proposed mechanism for stimulation of GLP-1 secretion by D3R in intestinal L-cells.

D3R activates GPR, e.g. GPR40/120, on the L-cell surface. Activation induces IP3R-mediated release of intracellular Ca2+ from the endoplasmic reticulum. The elevation of cytosolic Ca2+ stimulates phosphorylation of CaMKII, and CaMKII activation leads to an increase in GLP-1 secretion from intestinal L-cells.

https://doi.org/10.1371/journal.pone.0126157.g006

Supporting Information

S1 Fig. Effect of PKC and MEK inhibitoron D3R-stimulated GLP-1 secretion in GLUTag cells.

GLUTag cells were pre-treated with vehicle (0.1% DMSO) or (A) PKC inhibitor (Gö6983, 1 μM), (B) MEK inhibitor (PD98059, 50 μM) for 15 min, followed by treatment with vehicle or D3R (100 μM) for 2 h without washing out. GLP-1 levels in the medium were measured by ELISA. Secreted GLP-1 levels are expressed as the fold change of the control levels (= 1.0). Values are expressed as the means ± SEM, n = 3. Values without a common letter (a and b) are significantly different at P < 0.05 (Tukey-Kramer test followed by two-way ANOVA).

https://doi.org/10.1371/journal.pone.0126157.s001

(TIF)

Acknowledgments

We thank Dr. Toshiro Matsui (Kyusyu University) for confirming structure of Mal3R using MS and NMR and Taeko Kawai (Chubu University) for her technical assistance.

Author Contributions

Conceived and designed the experiments: T. Tsuda. Performed the experiments: MK T. Tsubasa. Analyzed the data: T. Tsuda MK T. Tsubasa. Contributed reagents/materials/analysis tools: NT. Wrote the paper: T. Tsuda MK NT.

References

  1. 1. Mojsov S, Weir GC, Habener JF (1987) Insulinotropin: glucagon-like peptide I (7–37) co-encoded in the glucagon gene is a potent stimulator of insulin release in the perfused rat pancreas. J Clin Invest 79: 616–619. pmid:3543057
  2. 2. Kreymann B, Williams G, Ghatei MA, Bloom SR (1987) Glucagon-like peptide-1 7–36: a physiological incretin in man. Lancet 2: 1300–1304. pmid:2890903
  3. 3. Buteau J, Foisy S, Joly E, Prentki M (2003) Glucagon-like peptide 1 induces pancreatic beta-cell proliferation via transactivation of the epidermal growth factor receptor. Diabetes 52: 124–132. pmid:12502502
  4. 4. Zander M, Madsbad S, Madsen JL, Holst JJ (2002) Effect of 6-week course of glucagon-like peptide 1 on glycaemic control, insulin sensitivity, and beta-cell function in type 2 diabetes: a parallel-group study. Lancet 359: 824–830. pmid:11897280
  5. 5. Vilsbøll T, Brock B, Perrild H, Levin K, Lervang HH, Kølendorf K, et al. (2008) Liraglutide, a once-daily human GLP-1 analogue, improves pancreatic B-cell function and arginine-stimulated insulin secretion during hyperglycaemia in patients with Type 2 diabetes mellitus. Diabet Med 25: 152–156. pmid:18201212
  6. 6. Kieffer TJ, McIntosh CH, Pederson RA (1995) Degradation of glucose-dependent insulinotropic polypeptide and truncated glucagon-like peptide 1 in vitro and in vivo by dipeptidyl peptidase IV. Endocrinology 136: 3585–3596. pmid:7628397
  7. 7. Deacon CF, Nauck MA, Toft-Nielsen M, Pridal L, Willms B, Holst JJ (1995) Both subcutaneously and intravenously administered glucagon-like peptide I are rapidly degraded from the NH2-terminus in type II diabetic patients and in healthy subjects. Diabetes 44: 1126–1131. pmid:7657039
  8. 8. Herman GA, Bergman A, Stevens C, Kotey P, Yi B, Zhao P, et al. (2006) Effect of single oral doses of sitagliptin, a dipeptidyl peptidase-4 inhibitor, on incretin and plasma glucose levels after an oral glucose tolerance test in patients with type 2 diabetes. J Clin Endocrinol Metab 91: 4612–4619. pmid:16912128
  9. 9. Rosenstock J, Sankoh S, List JF (2008) Glucose-lowering activity of the dipeptidyl peptidase-4 inhibitor saxagliptin in drug-naive patients with type 2 diabetes. Diabetes Obes Metab 10: 376–386. pmid:18355324
  10. 10. Rocca AS, Brubaker PL (1995) Stereospecific effects of fatty acids on proglucagon-derived peptide secretion in fetal rat intestinal cultures. Endocrinology 136: 5593–5599. pmid:7588313
  11. 11. Rocca AS, LaGreca J, Kalitsky J, Brubaker PL (2001) Monounsaturated fatty acids improve glycemic tolerance through increased secretion of glucagon-like peptide-1. Endocrinology 142: 1148–1155. pmid:11181530
  12. 12. Thomsen C, Rasmussen O, Lousen T, Holst JJ, Fenselau S, Schrezenmeir J, et al. (1999) Differential effects of saturated and monounsaturated fatty acids on postprandial lipemia and incretin responses in healthy subjects. Am J Clin Nutr 69: 1135–1143. pmid:10357731
  13. 13. Reimann F, Williams L, da Silva Xavier G, Rutter GA, Gribble FM (2004) Glutamine potently stimulates glucagon-like peptide-1 secretion from GLUTag cells. Diabetologia 47: 1592–1601. pmid:15365617
  14. 14. Tolhurst G, Zheng Y, Parker HE, Habib AM, Reimann F, Gribble FM (2011) Glutamine triggers and potentiates glucagon-like peptide-1 secretion by raising cytosolic Ca2+ and cAMP. Endocrinology 152: 405–413. pmid:21209017
  15. 15. Clemmensen C, Smajilovic S, Smith EP, Woods SC, Bräuner-Osborne H, Seeley RJ, et al. (2013) Oral L-arginine stimulates GLP-1 secretion to improve glucose tolerance in male mice. Endocrinology 154: 3978–3983. pmid:23959939
  16. 16. Cordier-Bussat M, Bernard C, Levenez F, Klages N, Laser-Ritz B, Philippe J, et al. (1998) Peptones stimulate both the secretion of the incretin hormone glucagon-like peptide-1 and the transcription of the proglucagon gene. Diabetes 47: 1038–1045. pmid:9648826
  17. 17. Reimer RA (2006) Meat hydrolysate and essential amino acid-induced glucagon-like peptide-1 secretion, in the human NCI-H716 enteroendocrine cell line, is regulated by extracellular signal-regulated kinase1/2 and p38 mitogen-activated protein kinases. J Endocrinol 191: 159–170. pmid:17065399
  18. 18. Hira T, Mochida T, Miyashita K, Hara H (2009) GLP-1 secretion is enhanced directly in the ileum but indirectly in the duodenum by a newly identified potent stimulator, zein hydrolysate, in rats. Am J Physiol Gastrointest Liver Physiol 297: G663–G671. pmid:19661152
  19. 19. Higuchi N, Hira T, Yamada N, Hara H (0213) Oral administration of corn zein hydrolysate stimulates GLP-1 and GIP secretion and improves glucose tolerance in male normal rats and Goto-Kakizaki rats. Endocrinology 15: 3089–3098.
  20. 20. Takikawa M, Kurimoto Y, Tsuda T (2013) Curcumin stimulates glucagon-like peptide-1 secretion in GLUTag cells via Ca2+/calmodulin-dependent kinase II activation. Biochem Biophys Res Commun 435: 165–170. pmid:23660191
  21. 21. Tsuda T (2012) Dietary anthocyanin-rich plants: biochemical basis and recent progress in health benefits studies. Mol Nutr Food Res 56: 159–170. pmid:22102523
  22. 22. Tsuda T (2012) Anthocyanins as Functional Food Factors—Chemistry, Nutrition and Health Promotion-. Food Sci Technol Res 18: 315–324.
  23. 23. Alvarez-Suarez JM, Giampieri F, Tulipani S, Casoli T, Di Stefano G, González-Paramás AM, et al. (2014) One-month strawberry-rich anthocyanin supplementation ameliorates cardiovascular risk, oxidative stress markers and platelet activation in humans. J Nutr Biochem 25: 289–294. pmid:24406274
  24. 24. Wedick NM, Pan A, Cassidy A, Rimm EB, Sampson L, Rosner B, et al. (2012) Dietary flavonoid intakes and risk of type 2 diabetes in US men and women. Am J Clin Nutr 95: 925–933. pmid:22357723
  25. 25. Takikawa M, Inoue S, Horio F, Tsuda T (2010) Dietary anthocyanin-rich bilberry extract ameliorates hyperglycemia and insulin sensitivity via activation of AMP-activated protein kinase in diabetic mice. J Nutr 140: 527–533. pmid:20089785
  26. 26. Kurimoto Y, Shibayama Y, Inoue S, Soga M, Takikawa M, Ito C, et al. (2013) Black soybean seed coat extract ameliorates hyperglycemia and insulin sensitivity via the activation of AMP-activated protein kinase in diabetic mice. J Agric Food Chem 61: 5558–5564. pmid:23683106
  27. 27. Drucker DJ, Jin T, Asa SL, Young TA, Brubaker PL (1994) Activation of proglucagon gene transcription by protein kinase-A in a novel mouse enteroendocrine cell line. Mol Endocrinol 8: 1646–1655. pmid:7535893
  28. 28. Mahadev K, Wu X, Zilbering A, Zhu L, Lawrence JT, Goldstein BJ (2001) Hydrogen peroxide generated during cellular insulin stimulation is integral to activation of the distal insulin signaling cascade in 3T3-L1 adipocytes. J Biol Chem 276: 48662–48669. pmid:11598110
  29. 29. Laemmli UK (1970) Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature 227: 680–685. pmid:5432063
  30. 30. Li SK, Zhu D, Gaisano HY, Brubaker PL (2014) Role of vesicle-associated membrane protein 2 in exocytosis of glucagon-like peptide-1 from the murine intestinal L cell. Diabetologia 57: 809–818. pmid:24356748
  31. 31. Reimann F, Habib AM, Tolhurst G, Parker HE, Rogers GJ, Gribble FM (2008) Glucose sensing in L cells: a primary cell study. Cell Metab 8: 532–539. pmid:19041768
  32. 32. Symonds EL, Peiris M, Page AJ, Chia B, Dogra H, Masding A, et al. (2015) Mechanisms of activation of mouse and human enteroendocrine cells by nutrients. Gut 64: 618–626. pmid:25015642
  33. 33. Lim GE, Xu M, Sun J, Jin T, Brubaker PL (2009) The rho guanosine 5'-triphosphatase, cell division cycle 42, is required for insulin-induced actin remodeling and glucagon-like peptide-1 secretion in the intestinal endocrine L cell. Endocrinology 150: 5249–5261. pmid:19819966
  34. 34. Reimann F, Ward PS, Gribble FM (2006) Signaling Mechanisms Underlying the Release of Glucagon-Like Peptide 1. Diabetes 55: S78–S85.
  35. 35. Rorsman P, Braun M, Zhang Q (2012) Regulation of calcium in pancreatic α- and β-cells in health and disease. Cell Calcium 51: 300–308. pmid:22177710
  36. 36. Hudmon A, Schulman H (2002) Neuronal Ca2+/calmodulin-dependent protein kinaseII: the role of structure and autoregulation in cellular function. Annu Rev Biochem 71: 473–510. pmid:12045104
  37. 37. Choi SE, Shin HC, Kim HE, Lee SJ, Jang HJ, et al. (2007) Involvement of Ca2+, CaMK II and PKA in EGb 761-induced insulin secretion in INS-1 cells. J Ethnopharmacol 110: 49–55. pmid:17046186
  38. 38. Lee SJ, Kim HE, Choi SE, Shin HC, Kwag WJ, Lee KW, et al. (2009) Involvement of Ca2+/calmodulin kinase II (CaMK II) in genistein-induced potentiation of leucine/glutamine-stimulated insulin secretion. Mol Cells 28: 167–174. pmid:19756396
  39. 39. Janssen S, Depoortere I (2013) Nutrient sensing in the gut: new roads to therapeutics? Trends Endocrinol Metab 24: 92–100. pmid:23266105
  40. 40. Edfalk S, Steneberg P, Edlund H (2008) Gpr40 is expressed in enteroendocrine cells and mediates free fatty acid stimulation of incretin secretion. Diabetes 57: 2280–2287. pmid:18519800
  41. 41. Lan H, Lin HV, Wang CF, Wright MJ, Xu S, Kang L, et al. (2012) Agonists at GPR119 mediate secretion of GLP-1 from mouse enteroendocrine cells through glucose-independent pathways. Br J Pharmacol 165: 2799–2807. pmid:22029751
  42. 42. Hirasawa A, Tsumaya K, Awaji T, Katsuma S, Adachi T, Yamada M, et al. (2005) Free fatty acids regulate gut incretin glucagon-like peptide-1 secretion through GPR120. Nat Med 11: 90–94. pmid:15619630
  43. 43. Parker HE, Wallis K, le Roux CW, Wong KY, Reimann F, Gribble FM (2012) Molecular mechanisms underlying bile acid-stimulated glucagon-like peptide-1 secretion. Br J Pharmacol 165: 414–423. pmid:21718300
  44. 44. Itoh Y, Kawamata Y, Harada M, Kobayashi M, Fujii R, Fukusumi S, et al. (2003) Free fatty acids regulate insulin secretion from pancreatic beta cells through GPR40. Nature 422: 173–176. pmid:12629551
  45. 45. Lauffer LM, Iakoubov R, Brubaker PL (2009) GPR119 is essential for oleoylethanolamide-induced glucagon-like peptide-1 secretion from the intestinal enteroendocrine L-cell. Diabetes 58: 1058–1066. pmid:19208912
  46. 46. Lim GE, Huang GJ, Flora N, LeRoith D, Rhodes CJ, Brubaker PL (2009) Insulin regulates glucagon-like peptide-1 secretion from the enteroendocrine L cell. Endocrinology 150: 580–591. pmid:18818290
  47. 47. Sangle GV, Lauffer LM, Grieco A, Trivedi S, Iakoubov R, Brubaker PL (2012) Novel biological action of the dipeptidylpeptidase-IV inhibitor, sitagliptin, as a glucagon-like peptide-1 secretagogue. Endocrinology 153: 564–573. pmid:22186413
  48. 48. Iakoubov R, Izzo A, Yeung A, Whiteside CI, Brubaker PL (2007) Protein kinase Czeta is required for oleic acid-induced secretion of glucagon-like peptide-1 by intestinal endocrine L cells. Endocrinology148: 1089–1098. pmid:17110421
  49. 49. Manach C, Williamson G, Morand C, Scalbert A, Rémésy C (2005) Bioavailability and bioefficacy of polyphenols in humans. I. Review of 97 bioavailability studies. Am J Clin Nutr 81: 230S–242S. pmid:15640486
  50. 50. Ohyama A, Hosaka K, Komiya Y, Akagawa K, Yamauchi E, Taniguchi H, et al. (2002) Regulation of exocytosis through Ca2+/ATP-dependent binding of autophosphorylated Ca2+/calmodulin-activated protein kinase II to syntaxin 1A. J Neurosci 22: 3342–3351. pmid:11978810