Skip to main content
Erschienen in: Diabetologia 2/2013

Open Access 01.02.2013 | Article

Pharmacological reduction of NEFA restores the efficacy of incretin-based therapies through GLP-1 receptor signalling in the beta cell in mouse models of diabetes

verfasst von: Z. F. Kang, Y. Deng, Y. Zhou, R. R. Fan, J. C. N. Chan, D. R. Laybutt, J. Luzuriaga, G. Xu

Erschienen in: Diabetologia | Ausgabe 2/2013

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Aims/hypothesis

Type 2 diabetes mellitus is associated with reduced incretin effects. Although previous studies have shown that hyperglycaemia contributes to impaired incretin responses in beta cells, it is largely unknown how hyperlipidaemia, another feature of type 2 diabetes, contributes to impaired glucagon-like peptide 1 (GLP-1) response. Here, we investigated the effects of NEFA on incretin receptor signalling and examined the glucose-lowering efficacy of incretin-based drugs in combination with the lipid-lowering agent bezafibrate.

Methods

We used db/db mice to examine the in vivo efficacy of the treatment. Beta cell lines and mouse islets were used to examine GLP-1 and glucose-dependent insulinotropic peptide receptor signalling.

Results

Palmitate treatment decreased Glp1r expression in rodent insulinoma cell lines and isolated islets. This was associated with impairment of the following: GLP-1-stimulated cAMP production, phosphorylation of cAMP-responsive elements binding protein (CREB) and insulin secretion. In insulinoma cell lines, the expression of exogenous Glp1r restored cAMP production and the phosphorylation of CREB. Treatment with bezafibrate in combination with des-fluoro-sitagliptin or exendin-4 led to more robust glycaemic control, associated with improved islet morphology and beta cell mass in db/db mice.

Conclusions/interpretation

Elevated NEFA contributes to impaired responsiveness to GLP-1, partially through downregulation of GLP-1 receptor signalling. Improvements in lipid control in mouse models of obesity and diabetes increase the efficacy of incretin-based therapy.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1007/​s00125-012-2776-x) contains peer-reviewed but unedited supplementary material, which is available to authorised users.
Abkürzungen
Ad
Adenoviral vector
Ad-GFP
Adenoviral vector with green fluorescent protein (GFP)
CREB
cAMP-responsive elements binding protein
d-GIP
[d-Ala2]GIP(1-30)
DPP-4
Dipeptidyl peptidase-4
GFP
Green fluorescent protein
GIP
Glucose-dependent insulinotropic polypeptide
GIPR
GIP receptor
GLP-1
Glucagon-like peptide 1
GLP1R
GLP-1 receptor
p-CREB
CREB phosphorylation
PPAR
Peroxisome proliferator-activated receptor
SREBP1c
Sterol regulatory element binding protein 1c

Introduction

The gastrointestinal hormones, glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), cause glucose-dependent insulin secretion from pancreatic beta cells within minutes of nutrient ingestion [1, 2]. One characteristic of type 2 diabetes mellitus is impaired incretin effect [3, 4], although the secretion of GIP and GLP-1 is not always decreased [46]. This indicates that the reduced incretin effect is due to defects in incretin receptor signalling pathways, rather than to the concentration of incretin hormones. The insulinotropic activity of GIP is largely impaired in patients with type 2 diabetes [4, 7]. In contrast, the insulinotropic effects of GLP-1 are partially preserved, which is important for its therapeutic potential, but insulin responses are substantially reduced, especially when studies are done at comparable glucose levels [7, 8]. Moreover, a growing body of evidence has shown that the glucose-lowering effects of GLP-1 are mediated by various mechanisms, including stimulation of glucose-dependent insulin secretion in pancreatic beta cells [1, 2], promotion of pancreatic beta cell proliferation and inhibition of beta cell apoptosis [911], inhibition of pancreatic alpha cell glucagon release [12, 13] and regulation of appetite and the central nervous system [2, 14]. These attributes of GLP-1 provide a strong basis for novel pharmacotherapies in type 2 diabetes. Currently, synthetic versions of GLP-1 mimetics (e.g. exenatide and liraglutide) and dipeptidyl peptidase-4 (DPP-4) inhibitors (e.g. sitagliptin and vildagliptin), which reduce GLP-1 and GIP degradation by DPP-4, have been approved for the treatment of type 2 diabetes [2, 15].
Type 2 diabetes develops as a result of impaired beta cell function and is closely associated with increased plasma NEFA, which are thought to be an important link between obesity and type 2 diabetes [1618]. NEFA can result in a state of insulin resistance [19], induce pancreatic beta cell dysfunction and cause beta cell death [18, 20]. Although acute exposure to elevated plasma NEFA enhances glucose- and non-glucose-stimulated insulin secretion in vitro and in vivo [18, 21], long-term exposure to NEFA impairs glucose-stimulated insulin secretion [22]. Recently, it was reported that while incretin secretion is similar between obese and non-obese type 2 diabetic patients [23], obesity impairs the incretin effect independently of glucose tolerance [24]. It has also been reported that loss of the incretin effects was more extensive in obese than in lean type 2 diabetic patients [25]. More recently, Bando et al reported that obesity may attenuate the HbA1c-lowering effect of sitagliptin in Japanese type 2 diabetic patients [26]. This suggests that lipids may be involved in the regulation of incretin responsiveness in pancreatic beta cells. However, little is known about the influence of NEFA on incretin receptor signalling. Our previous study showed that hyperglycaemia downregulates GLP-1 receptor (GLP1R), which potentially contributes to the impaired incretin response in beta cells [27]. Furthermore, the normalisation of blood glucose concentrations improves the insulin response to GLP-1 and GIP in patients with type 2 diabetes [28]. In the present study, therefore, we used in vitro and in vivo approaches to investigate the role of NEFA in the impairment of incretin responses.

Methods

Chemicals and reagents
For details, see electronic supplementary material (ESM), Chemicals and reagents.
Cell culture and treatment
INS-1E cell line (passages 65 to 75) was a kind gift from P. Maechler (Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland). INS-1E and MIN6 cells were grown as described previously [11, 29]. Cells were cultured in 6- or 12-well plates for 24 to 48 h before treatment with palmitate or 1.05% (wt/vol) BSA (in RPMI 1640 or DMEM with 1% FBS). GLP-1, GIP, exendin-4 and [d-Ala2]GIP(1-30) (d-GIP) were dissolved in PBS and stored at −80°C. To achieve expression of exogenous Glp1r, cells were infected with an adenoviral vector (Ad), Ad-GLP1R, for 6 h prior to treatment with palmitate or 1.05% BSA.
Islet isolation and culture, construction of Ad-GLP1R, RT-PCR, western blotting and measurement of cAMP production, and insulin secretion
Pancreatic islets were isolated from 8- to 9-week-old male C57BL/6J mice as previously described (ESM Methods, Isolation and cell culture). The gateway-compatible adenoviral expression system was used to generate the recombinant adenoviruses (ESM Methods, Construction of Ad-GLPR1). Real-time PCR (ESM Methods, RNA extraction and quantitative RT-PCR) and western blotting (ESM Methods, Analysis of phosphorylation of CREB) were performed with standard procedures. Intracellular cAMP content was determined using a kit (Cyclic AMP EIA; Cayman, Ann Arbor, MI, USA) (ESM Methods, Measurement of cAMP production) and protein levels were assayed by BCA for the correction. Insulin secretion was measured in INS1-E cells or mouse islets after exposure to 3 or 16.7 mmol/l glucose (ESM Methods, Measurement of insulin secretion).
Animals and experimental protocols
All animal procedures were performed in accordance with the Guidelines for Care and Use of Laboratory Animals and approved by The Animal Subjects Committee of The Chinese University of Hong Kong. Male db/db and db/+ (control) mice (aged 7 to 8 weeks) were obtained from The Chinese University of Hong Kong and housed in specific pathogen-free conditions with a 12 h light–dark cycle and free access to water and food. Experiments were performed after 1 week of acclimatisation. For drug treatments, des-fluoro-sitagliptin (200 mg/kg) and bezafibrate (100 mg/kg) were dissolved in 0.5% (wt/vol) CMC and given by gavage; exendin-4 (10 nmol/kg) and d-GIP (24 nmol/kg) were dissolved in PBS and given by intraperitoneal injection. Mice were treated daily (16:00 to 18:00 hours) by gavage or intraperitoneal injection for the indicated time. Fed random blood glucose was monitored weekly at 09:00 to 10:00 hours. For measurement of the acute glucose-lowering actions of exendin-4 and d-GIP, db/db mice were treated with vehicle or bezafibrate for 2 weeks and then injected intraperitoneally with saline, exendin-4 or d-GIP. Glucose levels were determined at 0, 30, 60 and 240 min after injection.
OGTT, insulin tolerance test and serum lipid profile measurement
For the OGTT, mice were fasted overnight (~17 h). Glucose levels were determined using a glucometer (Johnson & Johnson, Milpitas, CA, USA) at 0, 30, 60 and 120 min after oral administration of 0.3 g/kg glucose. For the insulin tolerance test (ITT), done after 6 h of fasting, mice were intraperitoneally injected with 2 IU/kg human insulin (Novo Nordisk, Bagsvaerd, Denmark). Glucose levels were measured at 0, 30, 60 and 120 min after the injection. Triacylglycerol, NEFA and total cholesterol concentrations were measured using related kits (Wako Lab Assays, Richmond, VA, USA). HDL-cholesterol was determined by enzymatic assays using an automated analyser (Olympus, Tokyo, Japan).
Histological analysis
Pancreases were quickly dissected from mice and fixed in 4% (wt/vol.) paraformaldehyde, after which paraffin-embedded 4-μm sections were immunostained overnight at 4°C with guinea pig anti-insulin (Dako, Glostrup, Denmark) and mouse anti-glucagon (1:200; Accurate Chemical & Scientific, Westbury, NY, USA), or with mouse anti-BrdU (BD Biosciences, Franklin Lakes, NJ, USA) antibodies. Following this, staining with cy2-goat anti-guinea pig or cy3-donkey anti-mouse (1:400; Jackson, West Grove, PA, USA) was done at room temperature for 2 h. The sample slides were washed three times with 0.1% PBS Tween (vol./vol., PBST) and stained with DAPI (Invitrogen, Grand Island, NY, USA) before microscopic analysis. The insulin-positive area vs total pancreas or total islet area was evaluated using Image J (NIH, Bethesda, Maryland, USA) [10].
Statistical analysis
Animal data are expressed as means ± SEM. Differences between the groups were examined for statistical significance using one-way or two-way ANOVA, followed by Dunnett’s post tests or t tests (as appropriate). For in vitro experiments, quantitative RT-PCR data are expressed as means ± SEM; other data are presented as means ± SD. Statistical significance was determined by Student’s t test. A value of p < 0.05 was considered to be statistically significant.

Results

Reduced expression of Glp1r and reduction of GLP-1-stimulated insulin secretion in palmitate-treated beta cells and mouse islets
In rat INS-1E cells, palmitate treatment significantly reduced Glp1r mRNA expression and levels of GLP1R in a dose-dependent manner (Fig. 1a, ESM Fig. 1a, c), while no effect on Gipr mRNA expression was observed (Fig. 1a). Similar results were found in mouse MIN6 cells (Fig. 1b, ESM Fig. 1b, d). Consistent with these results, exposure to palmitate also led to decreases in Glp1r mRNA expression in isolated islets (Fig. 1c). We next examined transcription factor 7-like 2 (TCF7L2), which has been reported to regulate the expression of Glp1r and Gipr in beta cells [30]. However, Tcf7l2 mRNA expression was not changed by palmitate treatment in INS-1E or MIN6 cells (Fig. 1a, b). Similarly, Irs2 mRNA was not affected by palmitate (Fig. 1a, b). Upregulation of uncoupling protein-2 (UCP2) and sterol regulatory element binding protein 1c (SREBP1c) is known to play a crucial role in NEFA-induced beta cell dysfunction [31]. As expected, the mRNA expression of Ucp2 and Srebp1c (also known as Srebf1) was significantly increased by palmitate in INS-1E cells (Fig. 1a). However, in MIN6 cells, Ucp2 was upregulated while Srebp1c was unchanged by palmitate (Fig. 1b).
In islets isolated from db/db mice, we also found that Glp1r and Gipr mRNA were significantly reduced compared with expression in control mice (Fig. 1d, e). Interestingly, db/db mice that were treated with the lipid-lowering agent bezafibrate for 2 weeks displayed partial restoration of Glp1r and Gipr mRNA to levels not different from control mice (Fig. 1d, e), with lowered triacylglycerol and NEFA, but no obvious changes in glucose levels [32].
To further assess the functional consequences of the reductions in Glp1r and Gipr expression, we assayed insulin secretion in INS-1E cells and isolated islets. In INS-1E control cells, both GLP1R agonists (GLP-1, exendin-4) and the GIP receptor (GIPR) agonists (GIP, d-GIP) markedly increased insulin secretion in the presence of 16.7 mmol/l glucose (Fig. 1f). However, after pre-incubation with palmitate, this response was significantly attenuated (Fig. 1f). Similar results were found in isolated mouse islets, with palmitate treatment decreasing the fold induction of GLP-1-stimulated glucose-dependent insulin secretion (Fig. 1g).
Palmitate impairs GLP-1-stimulated cAMP production and phosphorylation of cAMP-responsive elements binding protein in rodent insulinoma cell lines
GLP-1 and GIP exert their effects by binding to their respective receptors, GLP1R and GIPR, leading to activation of adenylate cyclase and elevation of intracellular cAMP. In INS-1E cells, palmitate treatment decreased GLP-1- and GIP-stimulated cAMP production (Fig. 2a), while in MIN6 cells, GLP-1- but not GIP-stimulated cAMP production was reduced by palmitate (Fig. 2b). We then examined cAMP-responsive elements binding protein (CREB) phosphorylation (p-CREB), as CREB is an important transcription factor in glucose homeostasis and beta cell survival [33]. In INS-1E cells, palmitate treatment significantly reduced GLP-1-stimulated p-CREB in a time- and dose-dependent manner (Fig. 2c, e). In contrast, GIP-stimulated p-CREB was unchanged throughout the time course of activation (Fig. 2c, f). Similarly, in MIN6 cells, palmitate treatment decreased GLP-1- but not GIP-stimulated p-CREB (Fig. 2d).
Expression of exogenous Glp1r restores GLP-1-stimulated cAMP production and p-CREB in palmitate-treated rodent insulinoma cell lines
As GLP1R signalling is crucial for glucose homeostasis, we examined whether the expression of exogenous Glp1r could reverse the impairment of GLP1R signalling by palmitate. We constructed and expressed adenoviral vectors expressing mouse Glp1r (Ad-GLP1R) in INS-1E and MIN6 cells. Our RT-PCR results show that mouse Glp1r was expressed in rat INS-1E cells after infection with Ad-GLP1R (ESM Fig. 2). Functionally, INS-1E cells infected with Ad-GLP1R also showed increases in basal levels of GLP-1-stimulated cAMP production compared with cells infected with a control Ad with green fluorescent protein (GFP) (Ad-GFP) (Fig. 3a). This result indicated enhanced GLP1R signalling. Palmitate treatment resulted in reduced GLP-1-stimulated cAMP production in INS-1E cells infected with Ad-GFP or Ad-GLP1R (Fig. 3a). However, Ad-GLP1R-mediated expression of exogenous Glp1r still restored GLP-1-stimulated cAMP production to the normal level in the presence of palmitate (Fig. 3a). Expression of exogenous Glp1r also restored GLP-1-stimulated p-CREB to the normal level in the presence of palmitate (Fig. 3c). Similarly, in MIN6 cells, infection with Ad-GLP1R protected against the palmitate-mediated reductions of GLP-1-stimulated cAMP production and p-CREB (Fig. 3b, d). To summarise, the above data suggest that the impairment of GLP-1-stimulated cAMP production and p-CREB in rodent insulinoma cell lines by palmitate can be at least partially restored by the expression of exogenous Glp1r.
Lipid lowering enhances the efficacy of the DPP-4 inhibitor, des-fluoro-sitagliptin, in db/db mice
Since our findings implicate increased fatty acids in the impairment of GLP1R signalling through downregulation of GLP1R, we next investigated whether an improvement in the lipid profile would enhance responsiveness to incretin hormones in mouse models of diabetes. We first used the DPP-4 inhibitor, des-fluoro-sitagliptin, combined with the lipid-lowering drug, bezafibrate, in db/db mice. Administration of des-fluoro-sitagliptin or bezafibrate alone had no effect on blood glucose levels during the 8 week treatment period compared with the vehicle-treated control group (Fig. 4a, b). In contrast, treatment with des-fluoro-sitagliptin and bezafibrate together significantly decreased blood glucose levels from 3 weeks onwards until the end of the experiment (Fig. 4a, b). We next examined the effects of these treatments on glucose tolerance by performing OGTTs. Glucose tolerance in db/db mice was not affected by 6 weeks of treatment with des-fluoro-sitagliptin or bezafibrate alone (Fig. 4c, d). However, treatment with des-fluoro-sitagliptin and bezafibrate combined significantly improved glucose excursions during the OGTT compared with the vehicle control group (Fig. 4c, d). This was not due to changes in insulin sensitivity, as there was no difference in the blood glucose response between the treatment groups during the ITT (Fig. 4e, f). The lipid-lowering effects of bezafibrate were confirmed by measuring circulating triacylglycerol and NEFA concentrations. As expected, serum triacylglycerol and NEFA were significantly decreased after bezafibrate and combined des-fluoro-sitagliptin and bezafibrate treatments (ESM Fig. 3). In addition, HDL and total cholesterol levels were elevated in the bezafibrate and combined des-fluoro-sitagliptin and bezafibrate groups (ESM Fig. 3).
Pancreatic beta cells are important targets of incretin-based drugs. We analysed the islet morphology and beta cell mass of db/db mice after the various drug treatments. As shown in Fig. 5a, the normal islet architecture of control mice comprised a large core of insulin-positive beta cells ringed by a mantle of glucagon-positive alpha cells. In contrast, islets of vehicle-treated db/db mice exhibited an abnormal architecture, with reduced beta cells and increased alpha cells that were scattered throughout the islet core (Fig. 5a). A similar abnormal islet morphology was observed in db/db mice that were treated with des-fluoro-sitagliptin or bezafibrate alone (Fig. 5a). Only mice treated with a combination of des-fluoro-sitagliptin and bezafibrate displayed an improved islet architecture approaching that of normal islets, as evidenced by increased beta cell mass (measured as beta cell area per pancreas area) (Fig. 5b), an increased proportion of beta cells per islet (Fig. 5c) and the appearance of normal beta cell distribution within the islets (Fig. 5a).
Lipid lowering enhances the efficacy of an agonist to GLP1R (exendin-4) but not to GIPR (d-GIP) in db/db mice
DPP-4 inhibition prevents the degradation of GLP-1 and GIP, resulting in increased levels of biologically active GLP-1 and GIP in vivo. To investigate the effects of lipid lowering on the activity of each incretin separately, we tested the efficacy of the GLP1R agonist, exendin-4, or the GIPR agonist, d-GIP, either alone or in combination with bezafibrate, in db/db mice. First, db/db mice were treated with vehicle or bezafibrate for 2 weeks. As expected, these treatments significantly reduced plasma triacylglycerol and NEFA levels (data not show) without changes in blood glucose levels (Fig. 6c, d). We then assessed the glucose-lowering effect of acute GLP1R or GIPR activation after an intraperitoneal injection of a single dose of exendin-4 (1 nmol/kg) or d-GIP (24 nmol/kg). Acute treatment of db/db mice with exendin-4 significantly lowered blood glucose to a similar level to that in vehicle- and bezafibrate-treated groups compared with PBS injection (Fig. 6a). In contrast, the administration of d-GIP did not affect the hyperglycaemic status in either vehicle- or bezafibrate-treated db/db mice (Fig. 6b). Following the acute glucose-lowering experiments, the mice were treated with exendin-4 or d-GIP for an additional 3 weeks as described in the Methods. Under these chronic experimental conditions, neither exendin-4 nor d-GIP administration alone altered blood glucose levels (Fig. 6c, d). In mice treated with exendin-4 and bezafibrate combined, fed blood glucose levels were significantly reduced after 1 week and for the remainder of the treatment period (Fig. 6c). In contrast, treatment with d-GIP and bezafibrate combined did not affect blood glucose levels (Fig. 6d). In response to the OGTT only the group treated with exendin-4 and bezafibrate combined exhibited significantly improved glucose tolerance (Fig. 6e). In contrast, d-GIP slightly worsened glucose excursions during the OGTT and combined treatment with d-GIP and bezafibrate did not improve glucose tolerance (Fig. 6f). Chronic treatment of db/db mice with bezafibrate, exendin-4 or exendin-4 and bezafibrate combined did not alter insulin sensitivity, as indicated by the unchanged blood glucose levels compared with the vehicle control group during the ITT (ESM Fig. 4a). On the other hand, in mice treated with d-GIP or d-GIP and bezafibrate combined, the AUC for blood glucose levels during the ITT was slightly, but significantly elevated, indicating a modest impairment of insulin action (ESM Fig. 4b). As expected, exendin-4 or d-GIP treatments had no effects on serum triacylglycerol, NEFA and HDL-cholesterol levels, while bezafibrate alone or in combination with exendin-4 or d-GIP lowered serum triacylglycerol and NEFA levels, and increased serum total- and HDL-cholesterol levels (ESM Fig. 5).
Finally, we analysed the islet morphology of db/db mice after the various treatments using immunohistochemistry. As seen in Fig. 7a, islets from bezafibrate-, exendin-4- or d-GIP-treated db/db mice displayed a disordered islet morphology, which was similar to that in vehicle-treated db/db mice. Similarly, db/db mice treated with a combination of d-GIP and bezafibrate did not show any improvements in islet architecture compared with the vehicle group (Fig. 7a). However, mice treated with a combination of exendin-4 and bezafibrate displayed normalised islet architecture, with an increased proportion of beta cells, as well as normal distribution of alpha cells (Fig. 7a). Furthermore, only the exendin-4 plus bezafibrate group exhibited significantly increased beta cell mass and ratios of beta cell area to islet area compared with the vehicle-treated group (Fig. 7b, c). BrdU staining also showed that only the exendin-4 plus bezafibrate group significantly increased the number of BrdU-positive beta cells in islets compared with the vehicle-treated group (Fig. 7d, e). These data indicate that lipid lowering selectively enhanced the efficacy of GLP1R but not GIPR agonists to improve islet morphology and glucose homeostasis, possibly through increases in pancreatic beta cell proliferation in db/db mice.

Discussion

Impaired incretin effects are found in type 2 diabetes [3, 4]. Our study was designed to investigate the role of hyperlipidaemia in the impairment of the incretin response in vitro and in vivo. In the in vitro models, we found that exposure to palmitate was sufficient for impairment of GLP1R, but not GIPR signalling to occur. This was evidenced by a reduced ability of GLP-1 to stimulate cAMP production, p-CREB and insulin secretion. The specificity of these defects was demonstrated by the partial restoration of signalling after Ad-GLP1R-mediated expression of exogenous Glp1r. In the in vivo models, we found that hyperlipidaemia was necessary for the downregulation of incretin receptor expression in islets of a mouse model of diabetes. Furthermore, in the db/db mouse model of diabetes, normalisation of the lipid profile by bezafibrate dramatically improved the efficacy of incretin-based therapies, including the DPP-4 inhibitor, des-fluoro-sitagliptin, and the GLP1R agonist, exendin-4. These findings, together with the work of others [34], indicate crucial roles of fatty acids and GLP1R in maintaining incretin signalling, beta cell function and glucose homeostasis.
We and others have reported that GLP1R and GIPR levels were decreased in islets from mouse and rat models of diabetes, and from type 2 diabetic patients [27, 30, 35]. In the present study, we found that in vitro palmitate treatment resulted in reduced GLP1R levels. In islets isolated from db/db mice, we also observed a significant reduction of Glp1r expression. Furthermore, treatment of db/db mice with bezafibrate for 2 weeks, which significantly improved the serum lipid profile, partially restored Glp1r expression in islets, even though the hyperglycaemic status remained. These findings imply that, apart from hyperglycaemia, hyperlipidaemia is required for downregulation of Glp1r expression in diabetes. GIPR was less sensitive to regulation by palmitate. This difference in the regulation of incretin receptors by fatty acids is reminiscent of the effects of hyperglycaemia; thus conscious rats receiving glucose infusions and isolated rat islets exposed to high glucose exhibited decreases in Glp1r but not Gipr expression [27]. Although Gipr expression was unaltered in palmitate-treated INS-1E cells, GIP-stimulated cAMP production and insulin secretion were significantly decreased. This discrepancy in findings for GIP is possibly due to NEFA-induced global beta cell dysfunction via other pathways involving endoplasmic reticulum stress, oxidative stress and Ca2+ homeostasis [29, 31]. For example, SREBP1c, which was reported to mediate palmitate-induced impairment of insulin secretion in islets [36], was increased in INS-1E, but not in MIN6 cells after palmitate treatment. The results probably reflect the complexity of the effects of hyperlipidaemia on beta cell function, with impaired incretin receptor signalling contributing to beta cell glucolipotoxicity in concert with other pathways involving endoplasmic reticulum and oxidative stress [29, 31].
Although the glucose-lowering efficacy of incretin agonists and DPP-4 inhibitors has been shown in animal models [3739], it is worth noting that chronic treatment of db/db mice with incretin agonists or DPP-4 inhibitors alone only delays the onset of diabetes at the early stages of disease progression [40]. Exendin-4 treatment does not prevent the ongoing deterioration of glucose intolerance in severely diabetic db/db mice [41]. Likewise, clinical evidence shows that the efficacy of incretin-based drugs for the treatment of type 2 diabetes is variable, and may be affected by various factors such as age [42], stage and severity of diabetes, differences in responsiveness to GLP-1 in diverse ethnic groups, genetic variance of GIPR and GLP1R [43, 44], as well as hyperglycaemia [27]. The current study demonstrates for the first time that hyperlipidaemia should be included as a contributing factor to the reduced efficacy of incretin-based drugs in mouse models of diabetes.
Hyperlipidaemia is closely associated with type 2 diabetes, and glucose-lowering drugs such as thiazolidinediones and metformin improve glucose and lipid metabolism [45]. Our in vitro data show that elevated NEFA is sufficient to cause impaired GLP1R signalling, prompting us to test the relationship between hyperlipidaemia and the efficacy of incretin-based therapy in animal models of diabetes. The lipid-lowering agent bezafibrate significantly improved the serum lipid profile, without affecting blood glucose levels in db/db mice. Strikingly, after lipid lowering, the DPP-4 inhibitor des-fluoro-sitagliptin and the GLP1R agonist exendin-4 both had a more robust effect on glycaemic control than co-treatment with vehicle or treatment with each agent alone. The effects were not due to increases in insulin sensitivity. Rather the improved glucose tolerance was associated with restoration of normal islet morphology and increased beta cell mass. This effect was only apparent with prolonged incretin activation, since the lowering of fatty acids did not enhance glucose disappearance after acute treatment of db/db mice with exendin-4. These results suggest that the improved glucose homeostasis induced by chronic administration of exendin-4 and bezafibrate to db/db mice is effected via long-term improvements in beta cell mass and function, which may be due to restored expression of Glp1r and thus GLP1R signalling after lipid lowering. However, although Gipr expression was also partially restored by bezafibrate treatment, the GIPR agonist d-GIP did not improve glucose metabolism. Recent reports have demonstrated that GLP1R signalling exerts more robust control of beta cell survival and regeneration than does GIPR signalling in mice [46]. It has also been reported that GLP-1-stimulated p-CREB plays important roles in the regulation of beta cell survival through GLP1R activation [33]. In this study, we only observed reduced GLP-1-stimulated p-CREB during in vitro palmitate treatment. Moreover, only combined treatment with exendin-4 plus bezafibrate improved islet morphology and increased beta cell mass, associated with increased beta cell proliferation in db/db mice. On the other hand, it has been reported that GIP was associated with impaired insulin sensitivity [38, 47], and this may partially explain the differences in efficacy of the incretin receptor agonists.
The peroxisome proliferator-activated receptor (PPAR)-α agonist WY14643 has been reported to increase Gipr expression [48]; at the same time, PPAR-α activation is associated with improved beta cell survival and function through reduction of lipid accumulation by increased fatty acid oxidation in beta cells and human islets [49, 50]. It has also been reported that metformin increases Glp1r expression in INS-1 cells via a PPAR-α-dependent pathway [51]. Our results demonstrate that palmitate downregulates GLP1R in insulinoma cell lines and bezafibrate can improve the efficacy of des-fluoro-sitagliptin and exendin-4 in mouse models of diabetes. Intriguingly, the related nuclear receptor PPAR-γ has been reported to regulate Gipr expression by binding to the PPAR response elements within the rat Gipr promoter [52]. The question of whether PPAR-α activation regulates incretin receptors in beta cells through direct binding or indirect improvement in lipotoxicity requires further investigation.
In summary, our results show that hyperlipidaemia contributes to impaired beta cell responsiveness to GLP-1, partially through downregulation of GLP1R. Combined treatment with incretin-based drugs (des-fluoro-sitagliptin or exendin-4) and lipid-lowering drugs (bezafibrate) results in synergistic improvements of glucose metabolism and islet morphology and function. These findings reinforce the importance of lipid management in type 2 diabetes and provide important information for the design of new incretin-based therapies for the treatment of type 2 diabetes mellitus.

Funding

This work was supported by the Hong Kong Government Research Grant Committee (478110), the National Natural Science Foundation of China (81170722), the National Health and Medical Research Council of Australia (1030715) and a grant from Merck Sharp & Dohme (C2709; Whitehouse Station, NJ, USA). Z.F. Kang was supported by a studentship from Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, People’s Republic of China.

Duality of interest

The authors declare that there is no duality of interest associated with this manuscript.

Contribution statement

ZK, RF, YZ, JL and DRL performed experiments, analysed the data and contributed to the revision of the article. YD and JC contributed to the acquisition of data and revision of the article. ZK, YD and DRL analysed the data and wrote the manuscript. GX conceived and designed the experiments, performed experiments and wrote the manuscript. All authors approved the final version of the article.

Open Access

This article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution Noncommercial License which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Anhänge

Electronic Supplementary Materials

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Baggio LL, Drucker DJ (2007) Biology of incretins: GLP-1 and GIP. Gastroenterology 132:2131–2157PubMedCrossRef Baggio LL, Drucker DJ (2007) Biology of incretins: GLP-1 and GIP. Gastroenterology 132:2131–2157PubMedCrossRef
2.
Zurück zum Zitat Holst JJ, Vilsboll T, Deacon CF (2009) The incretin system and its role in type 2 diabetes mellitus. Mol Cell Endocrinol 297:127–136PubMedCrossRef Holst JJ, Vilsboll T, Deacon CF (2009) The incretin system and its role in type 2 diabetes mellitus. Mol Cell Endocrinol 297:127–136PubMedCrossRef
3.
Zurück zum Zitat Nauck M, Stockmann F, Ebert R, Creutzfeldt W (1986) Reduced incretin effect in type 2 (non-insulin-dependent) diabetes. Diabetologia 29:46–52PubMedCrossRef Nauck M, Stockmann F, Ebert R, Creutzfeldt W (1986) Reduced incretin effect in type 2 (non-insulin-dependent) diabetes. Diabetologia 29:46–52PubMedCrossRef
4.
Zurück zum Zitat Meier JJ, Nauck MA (2010) Is the diminished incretin effect in type 2 diabetes just an epi-phenomenon of impaired beta-cell function? Diabetes 59:1117–1125PubMedCrossRef Meier JJ, Nauck MA (2010) Is the diminished incretin effect in type 2 diabetes just an epi-phenomenon of impaired beta-cell function? Diabetes 59:1117–1125PubMedCrossRef
5.
Zurück zum Zitat Nauck MA, Vardarli I, Deacon CF, Holst JJ, Meier JJ (2011) Secretion of glucagon-like peptide-1 (GLP-1) in type 2 diabetes: what is up, what is down? Diabetologia 54:10–18PubMedCrossRef Nauck MA, Vardarli I, Deacon CF, Holst JJ, Meier JJ (2011) Secretion of glucagon-like peptide-1 (GLP-1) in type 2 diabetes: what is up, what is down? Diabetologia 54:10–18PubMedCrossRef
6.
Zurück zum Zitat Lee S, Yabe D, Nohtomi K et al (2010) Intact glucagon-like peptide-1 levels are not decreased in Japanese patients with type 2 diabetes. Endocr J 57:119–126PubMedCrossRef Lee S, Yabe D, Nohtomi K et al (2010) Intact glucagon-like peptide-1 levels are not decreased in Japanese patients with type 2 diabetes. Endocr J 57:119–126PubMedCrossRef
7.
Zurück zum Zitat Nauck MA, Heimesaat MM, Orskov C, Holst JJ, Ebert R, Creutzfeldt W (1993) Preserved incretin activity of glucagon-like peptide 1 [7-36 amide] but not of synthetic human gastric inhibitory polypeptide in patients with type-2 diabetes mellitus. J Clin Invest 91:301–307PubMedCrossRef Nauck MA, Heimesaat MM, Orskov C, Holst JJ, Ebert R, Creutzfeldt W (1993) Preserved incretin activity of glucagon-like peptide 1 [7-36 amide] but not of synthetic human gastric inhibitory polypeptide in patients with type-2 diabetes mellitus. J Clin Invest 91:301–307PubMedCrossRef
8.
Zurück zum Zitat Kjems LL, Holst JJ, Volund A, Madsbad S (2003) The influence of GLP-1 on glucose-stimulated insulin secretion: effects on beta-cell sensitivity in type 2 and nondiabetic subjects. Diabetes 52:380–386PubMedCrossRef Kjems LL, Holst JJ, Volund A, Madsbad S (2003) The influence of GLP-1 on glucose-stimulated insulin secretion: effects on beta-cell sensitivity in type 2 and nondiabetic subjects. Diabetes 52:380–386PubMedCrossRef
9.
Zurück zum Zitat Li Y, Hansotia T, Yusta B, Ris F, Halban PA, Drucker DJ (2003) Glucagon-like peptide-1 receptor signaling modulates beta cell apoptosis. J Biol Chem 278:471–478PubMedCrossRef Li Y, Hansotia T, Yusta B, Ris F, Halban PA, Drucker DJ (2003) Glucagon-like peptide-1 receptor signaling modulates beta cell apoptosis. J Biol Chem 278:471–478PubMedCrossRef
10.
Zurück zum Zitat Xu G, Stoffers DA, Habener JF, Bonner-Weir S (1999) Exendin-4 stimulates both beta-cell replication and neogenesis, resulting in increased beta-cell mass and improved glucose tolerance in diabetic rats. Diabetes 48:2270–2276PubMedCrossRef Xu G, Stoffers DA, Habener JF, Bonner-Weir S (1999) Exendin-4 stimulates both beta-cell replication and neogenesis, resulting in increased beta-cell mass and improved glucose tolerance in diabetic rats. Diabetes 48:2270–2276PubMedCrossRef
11.
Zurück zum Zitat Fan R, Li X, Gu X, Chan JC, Xu G (2010) Exendin-4 protects pancreatic beta cells from human islet amyloid polypeptide-induced cell damage: potential involvement of AKT and mitochondria biogenesis. Diabetes Obes Metab 12:815–824PubMedCrossRef Fan R, Li X, Gu X, Chan JC, Xu G (2010) Exendin-4 protects pancreatic beta cells from human islet amyloid polypeptide-induced cell damage: potential involvement of AKT and mitochondria biogenesis. Diabetes Obes Metab 12:815–824PubMedCrossRef
12.
Zurück zum Zitat Nauck MA, Heimesaat MM, Behle K et al (2002) Effects of glucagon-like peptide 1 on counterregulatory hormone responses, cognitive functions, and insulin secretion during hyperinsulinemic, stepped hypoglycemic clamp experiments in healthy volunteers. J Clin Endocrinol Metab 87:1239–1246PubMedCrossRef Nauck MA, Heimesaat MM, Behle K et al (2002) Effects of glucagon-like peptide 1 on counterregulatory hormone responses, cognitive functions, and insulin secretion during hyperinsulinemic, stepped hypoglycemic clamp experiments in healthy volunteers. J Clin Endocrinol Metab 87:1239–1246PubMedCrossRef
13.
Zurück zum Zitat Hare KJ, Vilsboll T, Asmar M, Deacon CF, Knop FK, Holst JJ (2010) The glucagonostatic and insulinotropic effects of glucagon-like peptide 1 contribute equally to its glucose-lowering action. Diabetes 59:1765–1770PubMedCrossRef Hare KJ, Vilsboll T, Asmar M, Deacon CF, Knop FK, Holst JJ (2010) The glucagonostatic and insulinotropic effects of glucagon-like peptide 1 contribute equally to its glucose-lowering action. Diabetes 59:1765–1770PubMedCrossRef
14.
Zurück zum Zitat Turton MD, O'Shea D, Gunn I et al (1996) A role for glucagon-like peptide-1 in the central regulation of feeding. Nature 379:69–72PubMedCrossRef Turton MD, O'Shea D, Gunn I et al (1996) A role for glucagon-like peptide-1 in the central regulation of feeding. Nature 379:69–72PubMedCrossRef
15.
Zurück zum Zitat Lovshin JA, Drucker DJ (2009) Incretin-based therapies for type 2 diabetes mellitus. Nat Rev Endocrinol 5:262–269PubMedCrossRef Lovshin JA, Drucker DJ (2009) Incretin-based therapies for type 2 diabetes mellitus. Nat Rev Endocrinol 5:262–269PubMedCrossRef
16.
Zurück zum Zitat Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, Butler PC (2003) Beta-cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes. Diabetes 52:102–110PubMedCrossRef Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, Butler PC (2003) Beta-cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes. Diabetes 52:102–110PubMedCrossRef
17.
Zurück zum Zitat Paolisso G, Tataranni PA, Foley JE, Bogardus C, Howard BV, Ravussin E (1995) A high concentration of fasting plasma non-esterified fatty acids is a risk factor for the development of NIDDM. Diabetologia 38:1213–1217PubMedCrossRef Paolisso G, Tataranni PA, Foley JE, Bogardus C, Howard BV, Ravussin E (1995) A high concentration of fasting plasma non-esterified fatty acids is a risk factor for the development of NIDDM. Diabetologia 38:1213–1217PubMedCrossRef
18.
Zurück zum Zitat McGarry JD, Dobbins RL (1999) Fatty acids, lipotoxicity and insulin secretion. Diabetologia 42:128–138PubMedCrossRef McGarry JD, Dobbins RL (1999) Fatty acids, lipotoxicity and insulin secretion. Diabetologia 42:128–138PubMedCrossRef
19.
Zurück zum Zitat Randle PJ, Garland PB, Hales CN, Newsholme EA (1963) The glucose fatty-acid cycle. Its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus. Lancet 1:785–789PubMedCrossRef Randle PJ, Garland PB, Hales CN, Newsholme EA (1963) The glucose fatty-acid cycle. Its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus. Lancet 1:785–789PubMedCrossRef
20.
Zurück zum Zitat Lee Y, Hirose H, Ohneda M, Johnson JH, McGarry JD, Unger RH (1994) Beta-cell lipotoxicity in the pathogenesis of non-insulin-dependent diabetes mellitus of obese rats: impairment in adipocyte-beta-cell relationships. Proc Natl Acad Sci U S A 91:10878–10882PubMedCrossRef Lee Y, Hirose H, Ohneda M, Johnson JH, McGarry JD, Unger RH (1994) Beta-cell lipotoxicity in the pathogenesis of non-insulin-dependent diabetes mellitus of obese rats: impairment in adipocyte-beta-cell relationships. Proc Natl Acad Sci U S A 91:10878–10882PubMedCrossRef
21.
Zurück zum Zitat Crespin SR, Greenough WB 3rd, Steinberg D (1973) Stimulation of insulin secretion by long-chain free fatty acids. A direct pancreatic effect. J Clin Invest 52:1979–1984PubMedCrossRef Crespin SR, Greenough WB 3rd, Steinberg D (1973) Stimulation of insulin secretion by long-chain free fatty acids. A direct pancreatic effect. J Clin Invest 52:1979–1984PubMedCrossRef
22.
Zurück zum Zitat Zhou YP, Grill VE (1994) Long-term exposure of rat pancreatic islets to fatty acids inhibits glucose-induced insulin secretion and biosynthesis through a glucose fatty acid cycle. J Clin Invest 93:870–876PubMedCrossRef Zhou YP, Grill VE (1994) Long-term exposure of rat pancreatic islets to fatty acids inhibits glucose-induced insulin secretion and biosynthesis through a glucose fatty acid cycle. J Clin Invest 93:870–876PubMedCrossRef
23.
Zurück zum Zitat Kozawa J, Okita K, Imagawa A et al (2010) Similar incretin secretion in obese and non-obese Japanese subjects with type 2 diabetes. Biochem Biophys Res Commun 393:410–413PubMedCrossRef Kozawa J, Okita K, Imagawa A et al (2010) Similar incretin secretion in obese and non-obese Japanese subjects with type 2 diabetes. Biochem Biophys Res Commun 393:410–413PubMedCrossRef
24.
Zurück zum Zitat Muscelli E, Mari A, Casolaro A et al (2008) Separate impact of obesity and glucose tolerance on the incretin effect in normal subjects and type 2 diabetic patients. Diabetes 57:1340–1348PubMedCrossRef Muscelli E, Mari A, Casolaro A et al (2008) Separate impact of obesity and glucose tolerance on the incretin effect in normal subjects and type 2 diabetic patients. Diabetes 57:1340–1348PubMedCrossRef
25.
Zurück zum Zitat Knop FK, Aaboe K, Vilsbøll T et al (2012) Impaired incretin effect and fasting hyperglucagonaemia characterizing type 2 diabetic subjects are early signs of dysmetabolism in obesity. Diabetes Obes Metab 14:500–510PubMedCrossRef Knop FK, Aaboe K, Vilsbøll T et al (2012) Impaired incretin effect and fasting hyperglucagonaemia characterizing type 2 diabetic subjects are early signs of dysmetabolism in obesity. Diabetes Obes Metab 14:500–510PubMedCrossRef
26.
Zurück zum Zitat Bando Y, Aoki H, Hisada K, Toya A, Tanaka D (2012) Obesity may attenuate the HbA1c-lowering effect of sitagliptin in Japanese type 2 diabetic patients. J Diabetes Investig 3:170–174CrossRef Bando Y, Aoki H, Hisada K, Toya A, Tanaka D (2012) Obesity may attenuate the HbA1c-lowering effect of sitagliptin in Japanese type 2 diabetic patients. J Diabetes Investig 3:170–174CrossRef
27.
Zurück zum Zitat Xu G, Kaneto H, Laybutt DR et al (2007) Downregulation of GLP-1 and GIP receptor expression by hyperglycemia: possible contribution to impaired incretin effects in diabetes. Diabetes 56:1551–1558PubMedCrossRef Xu G, Kaneto H, Laybutt DR et al (2007) Downregulation of GLP-1 and GIP receptor expression by hyperglycemia: possible contribution to impaired incretin effects in diabetes. Diabetes 56:1551–1558PubMedCrossRef
28.
Zurück zum Zitat Hojberg PV, Vilsboll T, Rabol R et al (2009) Four weeks of near-normalisation of blood glucose improves the insulin response to glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide in patients with type 2 diabetes. Diabetologia 52:199–207PubMedCrossRef Hojberg PV, Vilsboll T, Rabol R et al (2009) Four weeks of near-normalisation of blood glucose improves the insulin response to glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide in patients with type 2 diabetes. Diabetologia 52:199–207PubMedCrossRef
29.
Zurück zum Zitat Laybutt DR, Preston AM, Akerfeldt MC et al (2007) Endoplasmic reticulum stress contributes to beta cell apoptosis in type 2 diabetes. Diabetologia 50:752–763PubMedCrossRef Laybutt DR, Preston AM, Akerfeldt MC et al (2007) Endoplasmic reticulum stress contributes to beta cell apoptosis in type 2 diabetes. Diabetologia 50:752–763PubMedCrossRef
30.
Zurück zum Zitat Shu L, Matveyenko AV, Kerr-Conte J, Cho JH, McIntosh CH, Maedler K (2009) Decreased TCF7L2 protein levels in type 2 diabetes mellitus correlate with downregulation of GIP- and GLP-1 receptors and impaired beta-cell function. Hum Mol Genet 18:2388–2399PubMedCrossRef Shu L, Matveyenko AV, Kerr-Conte J, Cho JH, McIntosh CH, Maedler K (2009) Decreased TCF7L2 protein levels in type 2 diabetes mellitus correlate with downregulation of GIP- and GLP-1 receptors and impaired beta-cell function. Hum Mol Genet 18:2388–2399PubMedCrossRef
31.
Zurück zum Zitat Poitout V, Robertson RP (2008) Glucolipotoxicity: fuel excess and beta-cell dysfunction. Endocr Rev 29:351–366PubMedCrossRef Poitout V, Robertson RP (2008) Glucolipotoxicity: fuel excess and beta-cell dysfunction. Endocr Rev 29:351–366PubMedCrossRef
32.
Zurück zum Zitat Kjorholt C, Akerfeldt MC, Biden TJ, Laybutt DR (2005) Chronic hyperglycemia, independent of plasma lipid levels, is sufficient for the loss of beta-cell differentiation and secretory function in the db/db mouse model of diabetes. Diabetes 54:2755–2763PubMedCrossRef Kjorholt C, Akerfeldt MC, Biden TJ, Laybutt DR (2005) Chronic hyperglycemia, independent of plasma lipid levels, is sufficient for the loss of beta-cell differentiation and secretory function in the db/db mouse model of diabetes. Diabetes 54:2755–2763PubMedCrossRef
33.
Zurück zum Zitat Jhala US, Canettieri G, Screaton RA et al (2003) cAMP promotes pancreatic beta-cell survival via CREB-mediated induction of IRS2. Genes Dev 17:1575–1580PubMedCrossRef Jhala US, Canettieri G, Screaton RA et al (2003) cAMP promotes pancreatic beta-cell survival via CREB-mediated induction of IRS2. Genes Dev 17:1575–1580PubMedCrossRef
34.
Zurück zum Zitat Lamont BJ, Li Y, Kwan E, Brown TJ, Gaisano H, Drucker DJ (2012) Pancreatic GLP-1 receptor activation is sufficient for incretin control of glucose metabolism in mice. J Clin Invest 122:388–402PubMedCrossRef Lamont BJ, Li Y, Kwan E, Brown TJ, Gaisano H, Drucker DJ (2012) Pancreatic GLP-1 receptor activation is sufficient for incretin control of glucose metabolism in mice. J Clin Invest 122:388–402PubMedCrossRef
35.
Zurück zum Zitat Piteau S, Olver A, Kim SJ et al (2007) Reversal of islet GIP receptor down-regulation and resistance to GIP by reducing hyperglycemia in the Zucker rat. Biochem Biophys Res Commun 362:1007–1012PubMedCrossRef Piteau S, Olver A, Kim SJ et al (2007) Reversal of islet GIP receptor down-regulation and resistance to GIP by reducing hyperglycemia in the Zucker rat. Biochem Biophys Res Commun 362:1007–1012PubMedCrossRef
36.
Zurück zum Zitat Kato T, Shimano H, Yamamoto T et al (2008) Palmitate impairs and eicosapentaenoate restores insulin secretion through regulation of SREBP-1c in pancreatic islets. Diabetes 57:2382–2392PubMedCrossRef Kato T, Shimano H, Yamamoto T et al (2008) Palmitate impairs and eicosapentaenoate restores insulin secretion through regulation of SREBP-1c in pancreatic islets. Diabetes 57:2382–2392PubMedCrossRef
37.
Zurück zum Zitat Young AA, Gedulin BR, Bhavsar S et al (1999) Glucose-lowering and insulin-sensitizing actions of exendin-4: studies in obese diabetic (ob/ob, db/db) mice, diabetic fatty Zucker rats, and diabetic rhesus monkeys (Macaca mulatta). Diabetes 48:1026–1034PubMedCrossRef Young AA, Gedulin BR, Bhavsar S et al (1999) Glucose-lowering and insulin-sensitizing actions of exendin-4: studies in obese diabetic (ob/ob, db/db) mice, diabetic fatty Zucker rats, and diabetic rhesus monkeys (Macaca mulatta). Diabetes 48:1026–1034PubMedCrossRef
38.
Zurück zum Zitat Lamont BJ, Drucker DJ (2008) Differential antidiabetic efficacy of incretin agonists versus DPP-4 inhibition in high fat fed mice. Diabetes 57:190–198PubMedCrossRef Lamont BJ, Drucker DJ (2008) Differential antidiabetic efficacy of incretin agonists versus DPP-4 inhibition in high fat fed mice. Diabetes 57:190–198PubMedCrossRef
39.
Zurück zum Zitat Mu J, Woods J, Zhou YP et al (2006) Chronic inhibition of dipeptidyl peptidase-4 with a sitagliptin analog preserves pancreatic beta-cell mass and function in a rodent model of type 2 diabetes. Diabetes 55:1695–1704PubMedCrossRef Mu J, Woods J, Zhou YP et al (2006) Chronic inhibition of dipeptidyl peptidase-4 with a sitagliptin analog preserves pancreatic beta-cell mass and function in a rodent model of type 2 diabetes. Diabetes 55:1695–1704PubMedCrossRef
40.
Zurück zum Zitat Wang Q, Brubaker PL (2002) Glucagon-like peptide-1 treatment delays the onset of diabetes in 8 week-old db/db mice. Diabetologia 45:1263–1273PubMedCrossRef Wang Q, Brubaker PL (2002) Glucagon-like peptide-1 treatment delays the onset of diabetes in 8 week-old db/db mice. Diabetologia 45:1263–1273PubMedCrossRef
41.
Zurück zum Zitat Rolin B, Larsen MO, Gotfredsen CF et al (2002) The long-acting GLP-1 derivative NN2211 ameliorates glycemia and increases beta-cell mass in diabetic mice. Am J Physiol Endocrinol Metab 283:E745–E752PubMed Rolin B, Larsen MO, Gotfredsen CF et al (2002) The long-acting GLP-1 derivative NN2211 ameliorates glycemia and increases beta-cell mass in diabetic mice. Am J Physiol Endocrinol Metab 283:E745–E752PubMed
42.
Zurück zum Zitat Fan R, Kang ZF, He L, Chan JC, Xu G (2011) Exendin-4 improves blood glucose control in both young and aging normal non-diabetic mice, possible contribution of beta cell independent effects. PLoS One 6:e20443PubMedCrossRef Fan R, Kang ZF, He L, Chan JC, Xu G (2011) Exendin-4 improves blood glucose control in both young and aging normal non-diabetic mice, possible contribution of beta cell independent effects. PLoS One 6:e20443PubMedCrossRef
43.
Zurück zum Zitat Sathananthan A, Man CD, Micheletto F et al (2010) Common genetic variation in GLP1R and insulin secretion in response to exogenous GLP-1 in nondiabetic subjects: a pilot study. Diabetes Care 33:2074–2076PubMedCrossRef Sathananthan A, Man CD, Micheletto F et al (2010) Common genetic variation in GLP1R and insulin secretion in response to exogenous GLP-1 in nondiabetic subjects: a pilot study. Diabetes Care 33:2074–2076PubMedCrossRef
44.
Zurück zum Zitat Saxena R, Hivert MF, Langenberg C et al (2010) Genetic variation in GIPR influences the glucose and insulin responses to an oral glucose challenge. Nat Genet 42:142–148PubMedCrossRef Saxena R, Hivert MF, Langenberg C et al (2010) Genetic variation in GIPR influences the glucose and insulin responses to an oral glucose challenge. Nat Genet 42:142–148PubMedCrossRef
45.
Zurück zum Zitat Bolen S, Feldman L, Vassy J et al (2007) Systematic review: comparative effectiveness and safety of oral medications for type 2 diabetes mellitus. Ann Intern Med 147:386–399PubMed Bolen S, Feldman L, Vassy J et al (2007) Systematic review: comparative effectiveness and safety of oral medications for type 2 diabetes mellitus. Ann Intern Med 147:386–399PubMed
46.
Zurück zum Zitat Maida A, Hansotia T, Longuet C, Seino Y, Drucker DJ (2009) Differential importance of glucose-dependent insulinotropic polypeptide vs glucagon-like peptide 1 receptor signaling for beta cell survival in mice. Gastroenterology 137:2146–2157PubMedCrossRef Maida A, Hansotia T, Longuet C, Seino Y, Drucker DJ (2009) Differential importance of glucose-dependent insulinotropic polypeptide vs glucagon-like peptide 1 receptor signaling for beta cell survival in mice. Gastroenterology 137:2146–2157PubMedCrossRef
47.
Zurück zum Zitat Nie Y, Ma R, Chan JC, Xu H, Xu G (2012) Glucose-dependent insulinotropic peptide impairs insulin signaling via inducing adipocyte inflammation in glucose-dependent insulinotropic peptide receptor over-expressing adipocytes. FASEB J 26:2383–2393PubMedCrossRef Nie Y, Ma R, Chan JC, Xu H, Xu G (2012) Glucose-dependent insulinotropic peptide impairs insulin signaling via inducing adipocyte inflammation in glucose-dependent insulinotropic peptide receptor over-expressing adipocytes. FASEB J 26:2383–2393PubMedCrossRef
48.
Zurück zum Zitat Lynn FC, Thompson SA, Pospisilik JA et al (2003) A novel pathway for regulation of glucose-dependent insulinotropic polypeptide (GIP) receptor expression in beta cells. FASEB J 17:91–93PubMed Lynn FC, Thompson SA, Pospisilik JA et al (2003) A novel pathway for regulation of glucose-dependent insulinotropic polypeptide (GIP) receptor expression in beta cells. FASEB J 17:91–93PubMed
49.
Zurück zum Zitat Ravnskjaer K, Boergesen M, Rubi B et al (2005) Peroxisome proliferator-activated receptor alpha (PPARalpha) potentiates, whereas PPARgamma attenuates, glucose-stimulated insulin secretion in pancreatic beta-cells. Endocrinology 146:3266–3276PubMedCrossRef Ravnskjaer K, Boergesen M, Rubi B et al (2005) Peroxisome proliferator-activated receptor alpha (PPARalpha) potentiates, whereas PPARgamma attenuates, glucose-stimulated insulin secretion in pancreatic beta-cells. Endocrinology 146:3266–3276PubMedCrossRef
50.
Zurück zum Zitat Lalloyer F, Vandewalle B, Percevault F et al (2006) Peroxisome proliferator-activated receptor alpha improves pancreatic adaptation to insulin resistance in obese mice and reduces lipotoxicity in human islets. Diabetes 55:1605–1613PubMedCrossRef Lalloyer F, Vandewalle B, Percevault F et al (2006) Peroxisome proliferator-activated receptor alpha improves pancreatic adaptation to insulin resistance in obese mice and reduces lipotoxicity in human islets. Diabetes 55:1605–1613PubMedCrossRef
51.
Zurück zum Zitat Maida A, Lamont BJ, Cao X, Drucker DJ (2011) Metformin regulates the incretin receptor axis via a pathway dependent on peroxisome proliferator-activated receptor-alpha in mice. Diabetologia 54:339–349PubMedCrossRef Maida A, Lamont BJ, Cao X, Drucker DJ (2011) Metformin regulates the incretin receptor axis via a pathway dependent on peroxisome proliferator-activated receptor-alpha in mice. Diabetologia 54:339–349PubMedCrossRef
52.
Zurück zum Zitat Gupta D, Peshavaria M, Monga N, Jetton TL, Leahy JL (2010) Physiologic and pharmacologic modulation of glucose-dependent insulinotropic polypeptide (GIP) receptor expression in beta-cells by peroxisome proliferator-activated receptor (PPAR)-gamma signaling: possible mechanism for the GIP resistance in type 2 diabetes. Diabetes 59:1445–1450PubMedCrossRef Gupta D, Peshavaria M, Monga N, Jetton TL, Leahy JL (2010) Physiologic and pharmacologic modulation of glucose-dependent insulinotropic polypeptide (GIP) receptor expression in beta-cells by peroxisome proliferator-activated receptor (PPAR)-gamma signaling: possible mechanism for the GIP resistance in type 2 diabetes. Diabetes 59:1445–1450PubMedCrossRef
Metadaten
Titel
Pharmacological reduction of NEFA restores the efficacy of incretin-based therapies through GLP-1 receptor signalling in the beta cell in mouse models of diabetes
verfasst von
Z. F. Kang
Y. Deng
Y. Zhou
R. R. Fan
J. C. N. Chan
D. R. Laybutt
J. Luzuriaga
G. Xu
Publikationsdatum
01.02.2013
Verlag
Springer-Verlag
Erschienen in
Diabetologia / Ausgabe 2/2013
Print ISSN: 0012-186X
Elektronische ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-012-2776-x

Weitere Artikel der Ausgabe 2/2013

Diabetologia 2/2013 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

CAR-M-Zellen: Warten auf das große Fressen

22.05.2024 Onkologische Immuntherapie Nachrichten

Auch myeloide Immunzellen lassen sich mit chimären Antigenrezeptoren gegen Tumoren ausstatten. Solche CAR-Fresszell-Therapien werden jetzt für solide Tumoren entwickelt. Künftig soll dieser Prozess nicht mehr ex vivo, sondern per mRNA im Körper der Betroffenen erfolgen.

Chronische Verstopfung: „Versuchen Sie es mit grünen Kiwis!“

22.05.2024 Obstipation Nachrichten

Bei chronischer Verstopfung wirken Kiwis offenbar besser als Flohsamenschalen. Das zeigen die Daten aus einer randomisierten Studie, die der Gastroenterologe Oliver Pech beim Praxis-Update vorstellte.

So häufig greift rheumatoide Arthritis auf Organe über

21.05.2024 Rheumatoide Arthritis Nachrichten

Im Verlauf von rheumatoider Arthritis entwickeln viele Patienten extraartikuläre Manifestationen. Schwedische Forscher haben sich mit der Inzidenz und den Risikofaktoren befasst.

Blutdrucksenkung könnte Uterusmyome verhindern

Frauen mit unbehandelter oder neu auftretender Hypertonie haben ein deutlich erhöhtes Risiko für Uterusmyome. Eine Therapie mit Antihypertensiva geht hingegen mit einer verringerten Inzidenz der gutartigen Tumoren einher.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.