Skip to main content
Erschienen in: Cardiovascular Diabetology 1/2012

Open Access 01.12.2012 | Original investigation

Telmisartan ameliorates insulin sensitivity by activating the AMPK/SIRT1 pathway in skeletal muscle of obese db/db mice

verfasst von: Asuka Shiota, Michio Shimabukuro, Daiju Fukuda, Takeshi Soeki, Hiromi Sato, Etsuko Uematsu, Yoichiro Hirata, Hirotsugu Kurobe, Norikazu Maeda, Hiroshi Sakaue, Hiroaki Masuzaki, Iichiro Shimomura, Masataka Sata

Erschienen in: Cardiovascular Diabetology | Ausgabe 1/2012

Abstract

Background

Telmisartan is a well-established angiotensin II type 1 receptor blocker that improves insulin sensitivity in animal models of obesity and insulin resistance, as well as in humans. Telmisartan has been reported to function as a partial agonist of the peroxisome proliferator-activated receptor (PPAR) γ, which is also targeted by the nicotinamide adenine dinucleotide (NAD)-dependent deacetylase (SIRT1). Here, we investigated the pathways through which telmisartan acts on skeletal muscle, in vitro as well as in vivo.

Methods

Nine-week-old male db/db mice were fed a 60% high-fat diet, with orally administrated either vehicle (carboxymethyl-cellulose, CMC), 5 mg/kg telmisartan, or 5 mg/kg telmisartan and 1 mg/kg GW9662, a selective irreversible antagonist of PPARγ, for 5 weeks. Effects of telmisartan on Sirt1 mRNA, AMPK phosphorylation, and NAD+/NADH ratio were determined in C2C12 cultured myocytes.

Results and discussion

Telmisartan treatment improved insulin sensitivity in obese db/db mice fed a high-fat diet and led to reduction in the size of hypertrophic pancreatic islets in these mice. Moreover, in vitro treatment with telmisartan led to increased expression of Sirt1 mRNA in C2C12 skeletal muscle cells; the increase in Sirt1 mRNA in telmisartan-treated C2C12 myoblasts occurred concomitantly with an increase in AMPK phosphorylation, an increase in NAD+/NADH ratio, and increases in the mRNA levels of PGC1α, FATP1, ACO, and GLUT4.

Conclusions

Our results indicate that telmisartan acts through a PPARγ-independent pathway, but at least partially exerts its effects by acting directly on skeletal muscle AMPK/SIRT1 pathways.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1475-2840-11-139) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

AS, MS, MS: design and conducting the study, data collection, analysis, and manuscript writing; HA, MH, YH, HK, YN, HM: contribution to design and discussion; HA and HA: contribution to cell distribution study; NM and IS: contribution to C2C12 study. We sincerely thank Mss. ST, JK and HG for her assistance. All authors read and approved the final manuscript.
Abkürzungen
ANOVA
Analysis of variance
AT1
Angiotensin II type 1 receptor
DMEM
Dulbecco’s modified Eagle’s medium
DMSO
Dimethyl sulfoxide
G6PDH
Glucose-6-phosphate dehydrogenase
ITT
Insulin tolerance test
NAD
Nicotinamide adenine dinucleotide
PPAR
Peroxisome proliferator-activated receptor
RT-PCR
Real-time-polymerase chain reaction
Sirt1
Nicotinamide adenine dinucleotide (NAD)-dependent deacetylase.

Introduction

The nicotinamide adenine dinucleotide (NAD)-dependent deacetylase (SIRT1) has been reported to be involved in protection against metabolic disorders as well as in enhancing life span [1]. Evidence suggests that SIRT1 also plays a fundamental role in the metabolism and differentiation of skeletal muscle cells [2], and transgenic or knockout Sirt1 mouse models have implicated the protein in protection of skeletal muscle against oxidative stress [3, 4]. It has been speculated that SIRT1 plays an important role in the regulation of transcriptional networks in various critical metabolic processes [1]. The SIRT1 signal seems to be mediated by an increase in fatty acid oxidation by activation of AMP-activated protein kinase (AMPK) in skeletal muscle [5]. The AMPK activators metformin [6] and A-769662 [7], as well as resveratrol [8], a polyphenolic SIRT1 activator, ameliorate insulin resistance in animals given a high-fat diet.
Telmisartan is a well-established angiotensin II type 1 receptor (AT1) blocker that improves insulin sensitivity in rodents that have received high-fat–containing diets [912], as well as in diabetic [12] and nondiabetic patients [13]. Telmisartan has been reported to function as a partial agonist of the peroxisome proliferator-activated receptor (PPAR) γ, a member of the ligand-activated nuclear receptor superfamily that is expressed at high levels in adipose tissue [14]. PPARγ regulates genes that modulate lipid utilization and storage, lipoprotein metabolism, adipocyte differentiation, and insulin action [14]. Thus, it is possible that the antidiabetic effects of telmisartan depend largely on PPARγ-dependent mechanisms [15]. PPARγ and the PPARγ coactivator (PGC)-1α, which play key roles in various metabolic disorders, are also known to be targeted by SIRT1 [16, 17], but the effects of telmisartan on SIRT1- and PPARγ-signaling remain unclear.
In the present study, we investigated whether telmisartan acts on skeletal muscle through an AMPK/SIRT1 pathway in skeletal muscle.

Materials and methods

Animals and experimental protocol

Study protocols were approved by the Committee on Animal Research, the University of Tokushima, and have been done on the basis of ethical principles and guidelines for experiments on animals (http://​freedownload.​is/​pdf/​ethical-principles-and-guidelines-for-experiments-on-animals-22328952.​html). Nine-week-old male db/db mice (Charles River Laboratories Japan Inc. Tokyo, Japan) were fed a high-fat diet (HFD-60, Oriental Yeast Co., Ltd., Tokyo) ad libitum for 5 weeks. HFD-60 contains energy content 62% fat (lard 33, milk-casein 26, corn-starch 16 g/100 g·diet; fatty acid 16:0 24.4, C18:0 13.8, C:18:1 41.8, 18:2(n-6) 12.0 g/100 g of total fatty acids), 18% protein and 20% carbohydrate, compatible with the AIN-93 G recommendations [18]. db/db mice were separated into 3 groups and were orally administrated either vehicle (carboxymethyl-cellulose, CMC), 5 mg/kg telmisartan, or 5 mg/kg telmisartan and 1 mg/kg GW9662 (Sigma, St. Louis, MO), a selective irreversible antagonist of PPARγ, once a day, for 5 weeks. Age-matched male wild type +/+ mice (Charles River Laboratories Japan) fed a normal diet, containing energy content 10% fat, 14% protein and 76% carbohydrate (AIN-93 M, Oriental Yeast Co. Ltd.) were used as a control group.
We defined the dose usage of telmisartan and GW9662 as follows. (1) telmisartan (5 mg/kg/day). For hypertensive patients, telmisartan is orally administered at the dose of 20‐160 mg per day, which is equivalent to ≈ 0.3-3 mg/kg/day. A pharmacokinetic study showed that steady state Cmax of telmisartan was 28.3 ng/ml at 20 mg and 592 ng/ml at 120 mg (≈ 0.06-1.15 μmol/L) in normotensive elderly subjects [19]. We had preliminary confirmed telmisartan show a PPAR-γ agonistic effect at doses of 0.5-10 μM in HEK293 cells (data not shown). Combined, we selected the dose of 5 mg/kg/day to obtain a PPAR-γ agonistic effect at a clinical dose. (2) GW9662 (1 mg/kg/day). We determined the dose of GW9662 following Goyal et al [20]. They showed that the protective effects of telmisartan (10 mg/kg/day) on myocardial infarction model in experimental diabetes could be modulated by down-regulated PPAR-γ expression by 1 mg/kg/day of GW9662 [20].
Body weight, food intake, systolic blood pressure and blood glucose were measured weekly. Blood pressure was measured using a tail-cuff system (Softron, Co., Tokyo) and glucose was determined by a glucose oxidase test. Mice were housed in a light- and temperature-controlled room, with a 12-hour light/dark cycle.
At the end of the experimental period, an insulin tolerance test (ITT) was performed. Mice were given an intraperitoneal injection of 0.75 U/kg insulin, after which blood glucose concentrations were measured at 0, 15, 30, 60, and 120 min. Moreover, at the end of the experimental period, serum, and subcutaneous and visceral fat, were collected and stored at -80°C until assayed. Insulin was determined by a mouse insulin ELISA kit (AKRIN-011 T, Shibayagi, Gunma, Japan).
Distribution of adipocyte size was determined as previously described [21]. Briefly, adipocytes isolated from visceral and subcutaneous adipose tissue were fixed with 2% osmium tetroxide and passed through a 250-μm nylon filter to remove the fibrous elements, after which the cells were washed extensively with isotonic saline. 10000 cells were analyzed using the Coulter Multisizer III (Beckman Coulter, High Wycombe, England). Paraffin-embedded serial sections of pancreata (5-μm thickness) were stained for h ematoxylin and e osin (HE) and indirect insulin immunostaining [22].

Cell culture

C2C12 myoblasts were cultured in Dulbecco’s modified Eagle’s medium (DMEM; Sigma) with 4.5 g/L glucose supplemented with 10% fetal bovine serum (FBS; Sigma), 100 U/mL penicillin, and 100 μg/mL streptomycin as previously described [23]. One day after C2C12 myoblasts reached confluence (day 0), cells were induced to differentiate into myocytes by replacing the medium with a low serum differentiation medium (2% horse serum; Gibco®, Life Technologies Japan, Ltd., Tokyo) for 2 days. After 48 h, the induction medium was removed and cells were maintained in DMEM with 10% FBS and, thereafter, the medium was changed every 2 days until day 5. Telmisartan was dissolved in dimethyl sulfoxide (DMSO) and added to culture media within 0.1% of volume (telmisartan final concentration: 0 - 1 μM). Telmisartan treatment was performed on day 5.

NAD+/NADH measurements

NAD+ and NADH nucleotides were measured with an enzymatic NADH recycling assay, using the NAD+/NADH Quantification kit (BioVision, Mountain View, CA) following the manufacturer’s recommended protocol [24]. C2C12 myotubes were collected in 200 μl of NADH/NAD+ extraction buffer by subjecting the cells to 2 cycles of freeze/thaw. The supernatants were divided into 2 sets: to detect NADH, 1 set of supernatants was heated to 60°C for 30 min to decompose NADH, while those from the other set was used to measure the total NADH plus NAD+-content, by performing the cycling assay in the absence of thermal decomposition. Then, the NAD+/NADH ratio was calculated.

Semi-quantitative RT-PCR analysis

Total RNA was isolated from C2C12 myotubes using RNAiso plus (Takara Bio Inc., Shiga, Japan). Complementary DNA synthesis was performed, using a QuantiTect® Reverse Transcription kit (SA Biosciences, Boston, MA), from 1 μg of the extracted total RNA. Then, we performed real-time RT-PCR with gene-specific primers and SYBR green dye using an Applied Biosystems 7500 Real-Time PCR System (Life Technologies Japan Ltd, Tokyo). The forward (fwd) and reverse (rev) primer sequences were as follows: Sirt1 (fwd: 5-TGCAGACGTGGTAATGTCCAAAC-3; rev: 5-ACATCTTGGCAGTATTTGTGGTGAA-3), GLUT4 (fwd: 5-TTTCCAGCAGATCGGCTCTGACGA-3; rev: 5-TAGCCAAACTGAAGGGAGCCAAGC-3), ACO (fwd: 5-GGTTGTCATCGCTTTGGTGCCTGT-3; rev: 5-TAACTCTGGATTGAAGGTGGCGGCG-3), FATP-1 (fwd: 5-CGGTGTGGTGGCTGCTCTTCTCAA-3; rev: 5-CGCTGCCATCTCCCCGCCATAAAT-3), GAPDH (fwd: 5-CAAGGTCATCCATGACAACTTTG-3; rev: 5-GGCCATCCACAGTCTTCTGG-3). The reaction mixture containing reverse-transcribed cDNAs was preheated for 10 min at 95°C to activate the Taq polymerase. Fifty cycles of PCR, each consisting of a 10-s denaturation step at 95°C, a 15-s annealing step at 60°C, and a 15-s extension step at 72°C, was then performed. Throughout real-time PCR analysis, product identities were confirmed by melting curve analysis. The quantification of each gene was determined relative to a standard curve generated from a serially diluted sample. The ratios of the amounts of target mRNA to the amount of the internal standard (GAPDH) mRNA was determined as an arbitrary unit.

Western blotting

Whole cell lysates for western blotting were harvested in RIPA buffer (Wako Pure Chemical Industries, Ltd., Tokyo) containing protease inhibitors (Takara Bio Inc., Shiga) and phosphatase inhibitor (Nacalai Tesque, Kyoto, Japan). Protein concentrations in the supernatants were determined using the Bio-Rad protein assay (Bio-Rad Laboratories, Inc., Tokyo). Twenty micrograms of protein were separated on a 5–20% gradient SDS-PAGE gel (ATTO Corporation, Tokyo) and transferred to polyvinylidene difluoride membranes (Millipore Corp., Billerica, MA). Membranes were blocked for 1 h at 20-25°C with 2% bovine serum albumin (BSA) or 5% Skim milk tris buffer saline Tween 20 (TBST) buffer. The membranes were incubated with appropriate dilutions of the primary antibodies (Toyobo Co., Ltd., Tokyo), viz., anti-Adiponectin (PA1-054 [1:2500 dilution]; Thermo Fisher Scientific Inc., Rockford, Illinois), anti-Phospho-AMPKα (Thr172) (2531 [1:5000 dilution]; Cell Signaling Technology, Inc., Beverly, MA), anti-AMPKα (2532 [1:5000 dilution]; Cell Signaling Technology, Inc.), and anti-GAPDH (2118 [1:5000 dilution]; Cell Signaling Technology, Inc.), overnight at 4°C. After washing, the blots were then incubated with HRP-conjugated anti-rabbit secondary antibody (7074 [dilution 1:5000], Cell Signaling Technology, Inc.) for 1 h in a room with a temperature maintained at 22 ± 2°C. Signals were detected using the Amersham™ ECLPrime enhanced chemiluminescence system (GE Healthcare Japan, Tokyo) and LAS-3000 mini imaging system (FujiFilm, Tokyo).

Statistical analysis

All data are expressed as the means ± standard error (SE). Statistical analysis was analyzed with Student’s t-test for parametric comparison between 2 groups, or analysis of variance (ANOVA) with post-hoc testing by Fisher’s protected least significant difference (PLSD) test for multiple comparisons. Values of p < 0.05 were considered statistically significant.

Results

In vivo study

During the 5 weeks of the study period, db/db mice fed a high-fat diet gained weight, irrespective of whether they were treated with CMC, telmisartan, or telmisartan + GW9662 (Figures 1A and B). Systolic blood pressure remained unchanged in all groups during the study period (Figure 1C). Overnight fasting glucose levels were higher in the db/db mice than in the wild-type mice, but were not different among the 3 treatment db/db groups (Figure 1D). Plasma levels of adiponectin were not significantly different among groups [wild-type 5.22 ± 0.49 μg/mL (n = 6), CMC 9.14 ± 2.88 (n = 5) telmisartan 9.09 ± 1.07 (n = 4), and telmisartan + GW9662 6.67 ± 0.49 (n = 4)].
The ITT at the end of the 5-week study period demonstrated that the 120-min glucose levels were significantly decreased in telmisartan-treated and telmisartan + GW9662-treated mice as compared to CMC-treated mice (Figure 1E). The baseline insulin levels, measured before the ITT, had been comparable among the 3 db/db groups (Figure 1F).

Effects of telmisartan on Sirt1, PGC1α, and GLUT4 mRNA expression

In skeletal muscle, expression of Sirt1 mRNA was increased in telmisartan-treated mice compared to CMC-treated mice, but the expression was similar in telmisartan- and telmisartan + GW9662-treated mice (Figure 2). Expression of PGC1α and GLUT4 mRNA was not different between CMC-treated and telmisartan-treated mice.
In visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT), expression of Sirt1, PGC1α, and GLUT4 mRNA were profoundly decreased in CMC-treated db/db mice as compared to wild type. The expression was comparable between telmisartan-treated and telmisartan + GW9662-treated mice.

Effects of telmisartan on the size of adipocytes and pancreatic islets in db/db mice fed a high-fat diet

The mean size of adipocyte in VAT and SAT was ≈ 2 times larger in db/db mice as compared to wild type (mean size of VAT 88 μm and SAT 94 μm). There were no significant differences in the size distribution (Figure 2) and the mean size among CMC-, telmisartan-, and telmisartan + GW9662-treated mice: 171 μm, 179 μm, and 182 μm in VAT and 181 μm, 180 μm, and 178 μm in SAT. However, although pancreatic islet size was increased in CMC-treated compared to wild-type mice, it was significantly decreased in telmisartan- and telmisartan + GW9662-treated mice (Figure 3).

Effect of telmisartan on AMPK-SIRT1 pathway

Since telmisartan increased Sirt1 mRNA levels in the skeletal muscle of db/db mice, we investigated whether telmisartan administration could directly affect SIRT1 signals in C2C12 myoblasts. Treatment with telmisartan increased Sirt1 mRNA in C2C12 cells (Figure 4A). Treatment of C2C12 cells with telmisartan resulted in activation of AMPK, by means of phosphorylation, to a level comparable to that induced by 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR), an activator of AMPK (Figure 4B). Treatment of C2C12 cells with telmisartan increased NAD+ and decreased NADH, resulting in an increase in the NAD+/NADH ratio, to a level comparable to that induced by AICAR (Figure 4C).

Effect of telmisartan on mRNA expression of AMPK/SIRT1 pathway-related molecules in C2C12 myocytes

Treatment with telmisartan increased the mRNA levels of PGC1, FATP1, ACO and GLUT4, but not of GLUT1, in C2C12 myocytes (Figure 5).

Discussion

Here, we showed that telmisartan treatment improved insulin sensitivity in obese db/db mice fed a high-fat diet and led to reduction in the size of hypertrophic pancreatic islets in these mice. Moreover, we demonstrated that in vitro treatment with telmisartan led to increased expression of Sirt1 mRNA in skeletal muscle cells; the increase in Sirt1 mRNA in telmisartan-treated C2C12 myoblasts occurred concomitantly with an increase in AMPK phosphorylation, an increase in NAD+/NADH ratio, and increases in the mRNA levels of PGC1, FATP1, ACO, and GLUT4.
It has previously been shown that levels of Sirt1 mRNA and SIRT1 activity are decreased in an animal obesity model [25, 26] and in obese humans [27]. To investigate the potential consequences of decreased SIRT1, we utilized db/db mice, a genetic model of obesity and insulin resistance resulting from a leptin receptor mutation [28]. On a high-fat diet, this db/db strain demonstrates severe insulin resistance and hyperglycemia [29]. In the current study, all mice fed a high-fat diet gained body weigh comparably (Figures 1A and B) and casual glucose levels did not differ among the 3 treatment groups (Figure 1D), but ITT-determined glucose levels were significantly decreased in telmisartan-treated mice. This indicates that administration of telmisartan ameliorates insulin resistance in the obesity and insulin resistance model. Because co-administration of GW9662, a selective PPARγ inhibitor, could not reverse this in vivo effect of telmisartan, the effects of telmisartan appear to act via a PPARγ-independent pathway. It has been suggested that some of angiotensin II receptor blockers (ARBs) have beneficial effects on lipid metabolism [30] and insulin sensitivity possibly through PPAR-γ activation [913]. We confirmed that 0.5-10 μM of telmisartan, but not up to 10 μM of olmesartan nor valsartan, have a PPAR-γ agonistic effect in HEK293 cells (data not shown). It should be clarified how different ARBs show mechanism(s) beyond blockade of renin-angiotensin system (RAS) in future studies.
Since it has been reported that AMPK activity and SIRT1 expression are decreased in the white adipose tissue (WAT) of db/db and HFD mice [26], we evaluated mRNA levels of Sirt1, PGC1α, and GLUT4 in the VAT and SAT of the various db/db mouse treatment groups. Consistent with a previous study [26], vehicle-treated db/db mice showed a decreased expression of Sirt1, PGC1α, and GLUT4 mRNAs in the VAT and SAT. Moreover, treatment with telmisartan did not change these expression levels, suggesting that telmisartan improved insulin resistance via a non-adipose tissue metabolic network in this db/db model. This hypothesis is supported by the finding that the adipocyte size distribution was not changed by telmisartan treatment. However, telmisartan administration did upregulate expression of Sirt1 mRNA and levels of phospho-Thr172-AMPK in skeletal muscle.
Metabolic sensors such as AMPK and SIRT1 are the gatekeepers of the activity of the master regulator, the mitochondrion, and are crucial to the regulatory network for metabolic homeostasis [1, 31, 32]. Mice models mildly overexpressing SIRT1 demonstrated ameliorated glucose tolerance when insulin resistance and/or diabetes were induced [33, 34]. Similarly, treatment with different SIRT1 agonists prevents weight gain and insulin resistance when mice were challenged with high-fat diets [8, 35]. Similarly, pharmacological activation of AMPK by metformin [6] and A-769662 [7] ameliorated insulin resistance in animals fed a high-fat diet. These results indicate a role for AMPK/SIRT1 in the control of metabolic homeostasis. It is believed that metformin treatment is the successful approach to reduce insulin resistance mainly through activation of liver AMPK [6], whereas DPP-4 inhibitors exhibit the best effects after oral glucose loading, emphasizing the incretin effect after oral stimulation [36]. However, metformin and DPP-4 inhibitors may involve different mechanisms in the treatment of mice fed high-fat diet [37]. Bourron et al. suggested a possible role of metformin on AMPK dependent lipolysis in adipocytes that may lead to lower plasma levels of fatty acids and to alleviating insulin resistance [38]. Sitagliptin also decreased adipocyte size efficiently and, even though the precise mechanisms remain to be elucidated, DPP-4 inhibition produced extra-pancreatic effects, such as enhanced postprandial lipid mobilization and oxidation [36]. Altogether, upregulation of Sirt1 mRNA and activation of AMPK in the skeletal muscle are associated with the improved insulin sensitivity observed upon telmisartan treatment.
PGC1α and GLUT4 mRNA expression also tended to be increased in skeletal muscle cells. AMPK and SIRT1 have both been described to directly affect PGC-1α activity through phosphorylation and deacetylation, respectively [32]. Thus, we next evaluated the direct effect of telmisartan on PGC-1α activity in muscle using C2C12 mouse myoblast cells. We found that in vitro treatment with telmisartan increased Sirt1 mRNA concomitantly with the increase in AMPK phosphorylation, an increase in the NAD+/NADH ratio, and the mRNA levels of PGC1, FATP1, ACO, and GLUT4 in C2C12 cells. To our knowledge, the current study, for the first time, showed that treatment with telmisartan enhanced Sirt1 mRNA and protein in the skeletal muscle.
SIRT1, an NAD+-dependent regulator of energy metabolism, has been reported to be activated through AMPK-mediated induction of nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme for NAD+ biosynthesis [1, 8]. We thus examined the direct effects of telmisartan on AMPK. Telmisartan resulted in a significant increase in AMPKα phosphorylation at Thr172, to a level comparable to that induced by AICAR, an activator of AMPK.
Taken altogether, we propose a possible mechanism by which telmisartan improves insulin sensitivity in skeletal muscle in Figure 6D. Impaired adipocytokine signaling, caused by suppression of expression of leptin and adipoR2 receptors, could lead to decreased activation of AMPK. This, in turn, may result in decreased NAMPT transcription, and consequently decreased SIRT1 activity and expression. Our results are also consistent with the notion that telmisartan, besides blocking AT1, acts to promote adipocyte differentiation and improve insulin sensitivity.
As previously reported, pancreatic islets in db/db mice on a high-fat diet were profoundly hypertrophic, presumably as compensation for severe insulin resistance. Saitoh et al. reported that telmisartan decreased the accumulation of palmitate-induced reactive oxygen species in MIN6 cells by 25%, and by 55% in mouse islet cells, suggesting that RAS blockade may be a possible mechanism for protecting β-cell survival and preserving insulin secretion capacity [39]. Yuan et al. reported that telmisartan and perindopril, a RAS inhibitor, comparably improved islet morphology, and significantly reduced IL-1β, HIF-1α, and CHOP mRNA in Wistar rats fed a high-fat diet for 16 weeks [40]. They suggested that blockade of RAS may protect the islet function of rats on a long-term high-fat diet, via down-regulation of islet inflammation, oxidative stress, endoplasmic reticulum stress, and apoptosis [41]. Our study showed that such hypertrophic pancreatic islets were improved in telmisartan-treated, and in telmisartan + GW9662-treated mice. The distribution in pancreatic β cell mass may also be affected by telmisartan, but we could not determine variation in the β cell mass distribution by our method. Although we could not determine the mechanism by which telmisartan affects islet morphology and function, future studies should determine the precise mechanism of the antimetabolic effects of this drug.
Our study had some limitations. First, the C2C12 model has limitations in comparison to freshly prepared frozen skeletal muscle cells [23]: C2C12 cells were originally obtained by Yaffe and Saxel through serial passage of myoblasts cultured from the thigh muscle of C3H mice after a crush injury. C2C12 cells are a useful tool for studying the differentiation of myoblasts and osteoblasts, expressing various proteins, and exploring mechanistic pathways. Nevertheless, the C2C12 cell line originated from a single clone, and thus has clone-specific traits, and fails to recapitulate primary cells. Second, the use of a single concentration of GW9662 may not be specific for verifying inhibition of PPARγ activity. We used a single dose of 1 mg/kg of GW9662 for the in vivo studies, and a single dose of 5 μM of GW9662 for the in vitro studies. We determined the effect of telmisartan on the activity of a PPARγ2 promoter luciferase gene reporter construct, indicating that 0.5–10.1 mM of telmisartan increased the activity of the PPARγ2 gene promoter by 1.8- to 2-fold, to a ≃30% level comparable to that achieved by pioglitazone, a known PPARγ agonist (in submission). The effects of telmisartan through PPARγ-dependent and -independent pathways need to be confirmed in further studies. Third, nonspecific effects of the drugs used in this study cannot be eliminated. GW9662 has been shown to inhibit COX-2 activity in addition to having an inhibitory effect on PPARγ, the active derivative of AICAR modulates other AMP-sensitive enzymes, and the selectivity of compound C for inhibition of AMPK remains uncertain. The findings presented in this study should be confirmed by using dominant-negative vectors or siRNA knockdown of AMPK and SIRT1 in the future. Fourth, our study could not determine whether telmisartan regulates AMPK/SIRT1 pathways in a tissue- or model-specific manner. Caton et al. reported that AMPK activity, NAMPT expression and SIRT1 expression were decreased in the WAT of db/db and HFD mice [26]. In addition, they showed that metformin increased the AMPK activity in the WAT of db/db mice and in adipocytes treated with metformin, with increased NAMPT and SIRT1 levels. In our study, telmisartan showed significant changes in protein and mRNA levels in skeletal muscle, but not in WAT. Although we cannot explain this discrepancy between the 2 studies, Chen et al. reported that SIRT1 expression is differentially regulated, in a tissue-specific manner upon calorie restriction, which may reflect tissue-specific roles of SIRT1 [41]. Tissue-specific effects of telmisartan need to be elucidated in future studies. Finally, the estimation of cell size has a limitation: in the fat pad, central adipocytes are usually bigger than peripheral adipocytes, and islets are roughly spheres and sections could cut spheres at its poles and distort the results.
In conclusion, telmisartan treatment improved glucose intolerance in db/db mice fed a high-fat diet, which was not inhibited by GW9662 co-administration. Telmisartan increased Sirt1 mRNA concomitantly with activation of AMPK via phosphorylation, an increase in NAD+/NADH ratio, and increase in mRNA levels of PGC1α, FATP1, ACO, and GLUT4 in C2C12 cell lines, suggesting that these improvements were at least partially due to the direct actions of telmisartan on AMPK/SIRT1 pathways. Precise understanding of this molecular mechanism will require further investigations.

Acknowledgements

This study was supported by grants from the Ministry of Education, Culture, Sports, Science, and Technology, and the Ministry of Health, Labor, and Welfare (MHLW) and Joint Research Association for Japanese Diabetes. We thank Nippon Boehringer Ingelheim Co., Ltd., Japan, for kindly providing telmisartan.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

AS, MS, MS: design and conducting the study, data collection, analysis, and manuscript writing; HA, MH, YH, HK, YN, HM: contribution to design and discussion; HA and HA: contribution to cell distribution study; NM and IS: contribution to C2C12 study. We sincerely thank Mss. ST, JK and HG for her assistance. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Houtkooper RH, Pirinen E, Auwerx J: Sirtuins as regulators of metabolism and healthspan. Nat Rev Mol Cell Biol. 2012, 13: 225-238.CrossRefPubMed Houtkooper RH, Pirinen E, Auwerx J: Sirtuins as regulators of metabolism and healthspan. Nat Rev Mol Cell Biol. 2012, 13: 225-238.CrossRefPubMed
2.
Zurück zum Zitat Fulco M, Schiltz RL, Iezzi S: Sir2 regulates skeletal muscle differentiation as a potential sensor of the redox state. Mol Cell. 2003, 12: 51-62. 10.1016/S1097-2765(03)00226-0.CrossRefPubMed Fulco M, Schiltz RL, Iezzi S: Sir2 regulates skeletal muscle differentiation as a potential sensor of the redox state. Mol Cell. 2003, 12: 51-62. 10.1016/S1097-2765(03)00226-0.CrossRefPubMed
3.
Zurück zum Zitat Vinciguerra M, Fulco M, Ladurner A, Sartorelli V, Rosenthal N: SirT1 in muscle physiology and disease: lessons from mouse models. Dis Model Mech. 2010, 3: 298-303. 10.1242/dmm.004655.PubMedCentralCrossRefPubMed Vinciguerra M, Fulco M, Ladurner A, Sartorelli V, Rosenthal N: SirT1 in muscle physiology and disease: lessons from mouse models. Dis Model Mech. 2010, 3: 298-303. 10.1242/dmm.004655.PubMedCentralCrossRefPubMed
4.
Zurück zum Zitat Tonkin J, Villarroya F, Puri PL, Vinciguerra M: SIRT1 signaling as potential modulator of skeletal muscle diseases. Curr Opin Pharmacol. 2012, Epub ahead of print Tonkin J, Villarroya F, Puri PL, Vinciguerra M: SIRT1 signaling as potential modulator of skeletal muscle diseases. Curr Opin Pharmacol. 2012, Epub ahead of print
5.
Zurück zum Zitat Zhang BB, Zhou G, Li C: AMPK: an emerging drug target for diabetes and the metabolic syndrome. Cell Metab. 2009, 9: 407-416. 10.1016/j.cmet.2009.03.012.CrossRefPubMed Zhang BB, Zhou G, Li C: AMPK: an emerging drug target for diabetes and the metabolic syndrome. Cell Metab. 2009, 9: 407-416. 10.1016/j.cmet.2009.03.012.CrossRefPubMed
6.
Zurück zum Zitat Ouyang J, Parakhia RA, Ochs RS: Metformin activates AMP kinase through inhibition of AMP deaminase. J Biol Chem. 2011, 286: 1-11. 10.1074/jbc.M110.121806.PubMedCentralCrossRefPubMed Ouyang J, Parakhia RA, Ochs RS: Metformin activates AMP kinase through inhibition of AMP deaminase. J Biol Chem. 2011, 286: 1-11. 10.1074/jbc.M110.121806.PubMedCentralCrossRefPubMed
7.
Zurück zum Zitat Cool B, Zinker B, Chiou W: Identification and characterization of a small molecule AMPK activator that treats key components of type 2 diabetes and the metabolic syndrome. Cell Metab. 2006, 3: 403-416. 10.1016/j.cmet.2006.05.005.CrossRefPubMed Cool B, Zinker B, Chiou W: Identification and characterization of a small molecule AMPK activator that treats key components of type 2 diabetes and the metabolic syndrome. Cell Metab. 2006, 3: 403-416. 10.1016/j.cmet.2006.05.005.CrossRefPubMed
8.
Zurück zum Zitat Lagouge M, Argmann C, Gerhart-Hines Z: Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1alpha. Cell. 2006, 127: 1109-1122. 10.1016/j.cell.2006.11.013.CrossRefPubMed Lagouge M, Argmann C, Gerhart-Hines Z: Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1alpha. Cell. 2006, 127: 1109-1122. 10.1016/j.cell.2006.11.013.CrossRefPubMed
9.
Zurück zum Zitat Benson SC, Pershadsingh HA, Ho CI: Identification of telmisartan as a unique angiotensin II receptor antagonist with selective PPARgamma-modulating activity. Hypertension. 2004, 43: 993-1002. 10.1161/01.HYP.0000123072.34629.57.CrossRefPubMed Benson SC, Pershadsingh HA, Ho CI: Identification of telmisartan as a unique angiotensin II receptor antagonist with selective PPARgamma-modulating activity. Hypertension. 2004, 43: 993-1002. 10.1161/01.HYP.0000123072.34629.57.CrossRefPubMed
10.
Zurück zum Zitat Araki K, Masaki T, Katsuragi I, Tanaka K, Kakuma T, Yoshimatsu H: Telmisartan prevents obesity and increases the expression of uncoupling protein 1 in diet-induced obese mice. Hypertension. 2006, 48: 51-57. 10.1161/01.HYP.0000225402.69580.1d.CrossRefPubMed Araki K, Masaki T, Katsuragi I, Tanaka K, Kakuma T, Yoshimatsu H: Telmisartan prevents obesity and increases the expression of uncoupling protein 1 in diet-induced obese mice. Hypertension. 2006, 48: 51-57. 10.1161/01.HYP.0000225402.69580.1d.CrossRefPubMed
11.
Zurück zum Zitat Sugimoto K, Qi NR, Kazdova L, Pravenec M, Ogihara T, Kurtz TW: Telmisartan but not valsartan increases caloric expenditure and protects against weight gain and hepatic steatosis. Hypertension. 2006, 47: 1003-1009. 10.1161/01.HYP.0000215181.60228.f7.CrossRefPubMed Sugimoto K, Qi NR, Kazdova L, Pravenec M, Ogihara T, Kurtz TW: Telmisartan but not valsartan increases caloric expenditure and protects against weight gain and hepatic steatosis. Hypertension. 2006, 47: 1003-1009. 10.1161/01.HYP.0000215181.60228.f7.CrossRefPubMed
12.
Zurück zum Zitat Pershadsingh HA, Kurtz TW: Insulin-sensitizing effects of telmisartan: implications for treating insulin-resistant hypertension and cardiovascular disease. Diabetes Care. 2004, 27: 1015.CrossRefPubMed Pershadsingh HA, Kurtz TW: Insulin-sensitizing effects of telmisartan: implications for treating insulin-resistant hypertension and cardiovascular disease. Diabetes Care. 2004, 27: 1015.CrossRefPubMed
13.
Zurück zum Zitat Shimabukuro M, Tanaka H, Shimabukuro T: Effects of telmisartan on fat distribution in subjects with the metabolic syndrome. J Hypertens. 2007, 25: 841-848. 10.1097/HJH.0b013e3280287a83.CrossRefPubMed Shimabukuro M, Tanaka H, Shimabukuro T: Effects of telmisartan on fat distribution in subjects with the metabolic syndrome. J Hypertens. 2007, 25: 841-848. 10.1097/HJH.0b013e3280287a83.CrossRefPubMed
14.
Zurück zum Zitat Evans RM, Barish GD, Wang YX: PPARs and the complex journey to obesity. Nat Med. 2004, 10: 355-361. 10.1038/nm1025.CrossRefPubMed Evans RM, Barish GD, Wang YX: PPARs and the complex journey to obesity. Nat Med. 2004, 10: 355-361. 10.1038/nm1025.CrossRefPubMed
15.
Zurück zum Zitat Kurtz TW: Beyond the classic angiotensin-receptor-blocker profile. Nat Clin Pract Cardiovasc Med. 2008, 5: S19-26.CrossRefPubMed Kurtz TW: Beyond the classic angiotensin-receptor-blocker profile. Nat Clin Pract Cardiovasc Med. 2008, 5: S19-26.CrossRefPubMed
16.
Zurück zum Zitat Chaudhary N, Pfluger PT: Metabolic benefits from Sirt1 and Sirt1 activators. Curr Opin Clin Nutr Metab Care. 2009, 12: 431-437. 10.1097/MCO.0b013e32832cdaae.CrossRefPubMed Chaudhary N, Pfluger PT: Metabolic benefits from Sirt1 and Sirt1 activators. Curr Opin Clin Nutr Metab Care. 2009, 12: 431-437. 10.1097/MCO.0b013e32832cdaae.CrossRefPubMed
17.
Zurück zum Zitat Sugden MC, Caton PW, Holness MJ: PPAR control: it’s SIRTainly as easy as PGC. J Endocrinol. 2010, 204: 93-104. 10.1677/JOE-09-0359.CrossRefPubMed Sugden MC, Caton PW, Holness MJ: PPAR control: it’s SIRTainly as easy as PGC. J Endocrinol. 2010, 204: 93-104. 10.1677/JOE-09-0359.CrossRefPubMed
18.
Zurück zum Zitat Reeves PG, Nielsen FH, Fahey GC: AIN-93 purified diets for laboratory rodents: final report of the American Institute of Nutrition ad hoc writing committee on the reformulation of the AIN-76A rodent diet. J Nutr. 1993, 123: 1939-1951.PubMed Reeves PG, Nielsen FH, Fahey GC: AIN-93 purified diets for laboratory rodents: final report of the American Institute of Nutrition ad hoc writing committee on the reformulation of the AIN-76A rodent diet. J Nutr. 1993, 123: 1939-1951.PubMed
19.
Zurück zum Zitat Stangier J, Su CA, Roth W: Pharmacokinetics of orally and intravenously administered telmisartan in healthy young and elderly volunteers and in hypertensive patients. J Int Med Res. 2000, 28: 149-67.CrossRefPubMed Stangier J, Su CA, Roth W: Pharmacokinetics of orally and intravenously administered telmisartan in healthy young and elderly volunteers and in hypertensive patients. J Int Med Res. 2000, 28: 149-67.CrossRefPubMed
20.
Zurück zum Zitat Goyal S, Arora S, Bhatt TK, Das P, Sharma A, Kumari S, Arya DS: Modulation of PPAR-gamma by telmisartan protects the heart against myocardial infarction in experimental diabetes. Chem Biol Interact. 2010, 185: 271-80. 10.1016/j.cbi.2010.03.030.CrossRefPubMed Goyal S, Arora S, Bhatt TK, Das P, Sharma A, Kumari S, Arya DS: Modulation of PPAR-gamma by telmisartan protects the heart against myocardial infarction in experimental diabetes. Chem Biol Interact. 2010, 185: 271-80. 10.1016/j.cbi.2010.03.030.CrossRefPubMed
21.
Zurück zum Zitat Sakai T, Sakaue H, Nakamura T, Okada M, Matsuki Y, Watanabe E, Hiramatsu R, Nakayama K, Nakayama KI, Kasuga M: Skp2 controls adipocyte proliferation during the development of obesity. J Biol Chem. 2007, 282: 2038-2046.CrossRefPubMed Sakai T, Sakaue H, Nakamura T, Okada M, Matsuki Y, Watanabe E, Hiramatsu R, Nakayama K, Nakayama KI, Kasuga M: Skp2 controls adipocyte proliferation during the development of obesity. J Biol Chem. 2007, 282: 2038-2046.CrossRefPubMed
22.
Zurück zum Zitat Shimabukuro M, Ohneda M, Lee Y, Unger RH: Role of nitric oxide in obesity-induced beta cell disease. J Clin Invest. 1997, 100: 290-5. 10.1172/JCI119534.PubMedCentralCrossRefPubMed Shimabukuro M, Ohneda M, Lee Y, Unger RH: Role of nitric oxide in obesity-induced beta cell disease. J Clin Invest. 1997, 100: 290-5. 10.1172/JCI119534.PubMedCentralCrossRefPubMed
23.
Zurück zum Zitat Yaffe D, Saxel O: Serial passaging and differentiation of myogenic cells isolated from dystrophic mouse muscle. Nature. 1977, 270: 725-727. 10.1038/270725a0.CrossRefPubMed Yaffe D, Saxel O: Serial passaging and differentiation of myogenic cells isolated from dystrophic mouse muscle. Nature. 1977, 270: 725-727. 10.1038/270725a0.CrossRefPubMed
24.
Zurück zum Zitat Iwabu M, Yamauchi T, Okada-Iwabu M: Adiponectin and AdipoR1 regulate PGC-1α and mitochondria by Ca2+ and AMPK/SIRT1. Nature. 2010, 464: 1313-1319. 10.1038/nature08991.CrossRefPubMed Iwabu M, Yamauchi T, Okada-Iwabu M: Adiponectin and AdipoR1 regulate PGC-1α and mitochondria by Ca2+ and AMPK/SIRT1. Nature. 2010, 464: 1313-1319. 10.1038/nature08991.CrossRefPubMed
25.
Zurück zum Zitat Qiao L, Lee B, Kinney B, Yoo HS, Shao J: Energy intake and adiponectin gene expression. Am J Physiol Endocrinol Metab. 2011, 300: E809-816. 10.1152/ajpendo.00004.2011.PubMedCentralCrossRefPubMed Qiao L, Lee B, Kinney B, Yoo HS, Shao J: Energy intake and adiponectin gene expression. Am J Physiol Endocrinol Metab. 2011, 300: E809-816. 10.1152/ajpendo.00004.2011.PubMedCentralCrossRefPubMed
26.
Zurück zum Zitat Caton PW, Kieswich J, Yaqoob MM, Holness MJ, Sugden MC: Metformin opposes impaired AMPK and SIRT1 function and deleterious changes in core clock protein expression in white adipose tissue of genetically-obese db/db mice. Diabetes Obes Metab. 2011, 13: 1097-1104. 10.1111/j.1463-1326.2011.01466.x.CrossRefPubMed Caton PW, Kieswich J, Yaqoob MM, Holness MJ, Sugden MC: Metformin opposes impaired AMPK and SIRT1 function and deleterious changes in core clock protein expression in white adipose tissue of genetically-obese db/db mice. Diabetes Obes Metab. 2011, 13: 1097-1104. 10.1111/j.1463-1326.2011.01466.x.CrossRefPubMed
27.
Zurück zum Zitat Clark SJ, Falchi M, Olsson B: Association of sirtuin 1 (SIRT1) gene SNPs and transcript expression levels with severe obesity. Obesity. 2012, 20: 178-185. 10.1038/oby.2011.200.PubMedCentralCrossRefPubMed Clark SJ, Falchi M, Olsson B: Association of sirtuin 1 (SIRT1) gene SNPs and transcript expression levels with severe obesity. Obesity. 2012, 20: 178-185. 10.1038/oby.2011.200.PubMedCentralCrossRefPubMed
28.
Zurück zum Zitat Lee GH, Proenca R, Montez JM: Abnormal splicing of the leptin receptor in diabetic mice. Nature. 1996, 379: 632-635. 10.1038/379632a0.CrossRefPubMed Lee GH, Proenca R, Montez JM: Abnormal splicing of the leptin receptor in diabetic mice. Nature. 1996, 379: 632-635. 10.1038/379632a0.CrossRefPubMed
29.
Zurück zum Zitat Rong JX, Qiu Y, Hansen MK: Adipose mitochondrial biogenesis is suppressed in db/db and high-fat diet-fed mice and improved by rosiglitazone. Diabetes. 2007, 56: 1751-1760. 10.2337/db06-1135.CrossRefPubMed Rong JX, Qiu Y, Hansen MK: Adipose mitochondrial biogenesis is suppressed in db/db and high-fat diet-fed mice and improved by rosiglitazone. Diabetes. 2007, 56: 1751-1760. 10.2337/db06-1135.CrossRefPubMed
30.
Zurück zum Zitat Nishida Y, Takahashi Y, Nakayama T, Soma M, Asai S: Comparative effect of olmesartan and candesartan on lipid metabolism and renal function in patients with hypertension: a retrospective observational study. Cardiovasc Diabetol. 2011, 10: 74-10.1186/1475-2840-10-74.PubMedCentralCrossRefPubMed Nishida Y, Takahashi Y, Nakayama T, Soma M, Asai S: Comparative effect of olmesartan and candesartan on lipid metabolism and renal function in patients with hypertension: a retrospective observational study. Cardiovasc Diabetol. 2011, 10: 74-10.1186/1475-2840-10-74.PubMedCentralCrossRefPubMed
31.
Zurück zum Zitat Liang F, Kume S, Koya D: SIRT1 and insulin resistance. Nature Rev Endocrinol. 2009, 5: 367-373. 10.1038/nrendo.2009.101.CrossRef Liang F, Kume S, Koya D: SIRT1 and insulin resistance. Nature Rev Endocrinol. 2009, 5: 367-373. 10.1038/nrendo.2009.101.CrossRef
32.
Zurück zum Zitat Cantó C, Auwerx J: PGC-1α, SIRT1 and AMPK, an energy sensing network that controls energy expenditure. Curr Opin Lipidol. 2009, 20: 98-105. 10.1097/MOL.0b013e328328d0a4.PubMedCentralCrossRefPubMed Cantó C, Auwerx J: PGC-1α, SIRT1 and AMPK, an energy sensing network that controls energy expenditure. Curr Opin Lipidol. 2009, 20: 98-105. 10.1097/MOL.0b013e328328d0a4.PubMedCentralCrossRefPubMed
33.
Zurück zum Zitat Banks AS, Kon N, Knight C: SirT1 gain of function increases energy efficiency and prevents diabetes in mice. Cell Metab. 2008, 8: 333-341. 10.1016/j.cmet.2008.08.014.PubMedCentralCrossRefPubMed Banks AS, Kon N, Knight C: SirT1 gain of function increases energy efficiency and prevents diabetes in mice. Cell Metab. 2008, 8: 333-341. 10.1016/j.cmet.2008.08.014.PubMedCentralCrossRefPubMed
34.
Zurück zum Zitat Bordone L, Cohen D, Robinson A: SIRT1 transgenic mice show phenotypes resembling calorie restriction. Aging Cell. 2007, 6: 759-767. 10.1111/j.1474-9726.2007.00335.x.CrossRefPubMed Bordone L, Cohen D, Robinson A: SIRT1 transgenic mice show phenotypes resembling calorie restriction. Aging Cell. 2007, 6: 759-767. 10.1111/j.1474-9726.2007.00335.x.CrossRefPubMed
35.
Zurück zum Zitat Milne JC, Lambert PD, Schenk S: Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes. Nature. 2007, 450: 712-716. 10.1038/nature06261.PubMedCentralCrossRefPubMed Milne JC, Lambert PD, Schenk S: Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes. Nature. 2007, 450: 712-716. 10.1038/nature06261.PubMedCentralCrossRefPubMed
36.
Zurück zum Zitat Baggio LL, Drucker DJ: Biology of incretins: GLP-1 and GIP. Gastroenterology. 2007, 132: 2131-2157. 10.1053/j.gastro.2007.03.054.CrossRefPubMed Baggio LL, Drucker DJ: Biology of incretins: GLP-1 and GIP. Gastroenterology. 2007, 132: 2131-2157. 10.1053/j.gastro.2007.03.054.CrossRefPubMed
37.
Zurück zum Zitat Westerink J, Visseren FLJ: Pharmacological and non-pharmacological interventions to influence adipose tissue function. Cardiovasc Diabetol. 2011, 10: 13-10.1186/1475-2840-10-13.PubMedCentralCrossRefPubMed Westerink J, Visseren FLJ: Pharmacological and non-pharmacological interventions to influence adipose tissue function. Cardiovasc Diabetol. 2011, 10: 13-10.1186/1475-2840-10-13.PubMedCentralCrossRefPubMed
38.
Zurück zum Zitat Bourron O, Daval M, Hainault I, Hajduch E, Servant JM, Gautier JF: Biguanides and thiazolidinediones inhibit stimulated lipolysis in human adipocytes through activation of AMP-activated protein kinase. Diabetologia. 2010, 53: 768-778. 10.1007/s00125-009-1639-6.CrossRefPubMed Bourron O, Daval M, Hainault I, Hajduch E, Servant JM, Gautier JF: Biguanides and thiazolidinediones inhibit stimulated lipolysis in human adipocytes through activation of AMP-activated protein kinase. Diabetologia. 2010, 53: 768-778. 10.1007/s00125-009-1639-6.CrossRefPubMed
39.
Zurück zum Zitat Saitoh Y, Hongwei W, Ueno H, Mizuta M, Nakazato M: Telmisartan attenuates fatty-acid-induced oxidative stress and NAD(P)H oxidase activity in pancreatic beta-cells. Diabetes Metab. 2009, 35: 392-397. 10.1016/j.diabet.2009.04.005.CrossRefPubMed Saitoh Y, Hongwei W, Ueno H, Mizuta M, Nakazato M: Telmisartan attenuates fatty-acid-induced oxidative stress and NAD(P)H oxidase activity in pancreatic beta-cells. Diabetes Metab. 2009, 35: 392-397. 10.1016/j.diabet.2009.04.005.CrossRefPubMed
40.
Zurück zum Zitat Yuan L, Li X, Li J, Li HL, Cheng SS: Effects of renin-angiotensin system blockade on the islet morphology and function in rats with long-term high-fat diet. Acta Diabetol. 2010, 10.1007/s00592-010-0210-8. Yuan L, Li X, Li J, Li HL, Cheng SS: Effects of renin-angiotensin system blockade on the islet morphology and function in rats with long-term high-fat diet. Acta Diabetol. 2010, 10.1007/s00592-010-0210-8.
41.
Metadaten
Titel
Telmisartan ameliorates insulin sensitivity by activating the AMPK/SIRT1 pathway in skeletal muscle of obese db/db mice
verfasst von
Asuka Shiota
Michio Shimabukuro
Daiju Fukuda
Takeshi Soeki
Hiromi Sato
Etsuko Uematsu
Yoichiro Hirata
Hirotsugu Kurobe
Norikazu Maeda
Hiroshi Sakaue
Hiroaki Masuzaki
Iichiro Shimomura
Masataka Sata
Publikationsdatum
01.12.2012
Verlag
BioMed Central
Erschienen in
Cardiovascular Diabetology / Ausgabe 1/2012
Elektronische ISSN: 1475-2840
DOI
https://doi.org/10.1186/1475-2840-11-139

Weitere Artikel der Ausgabe 1/2012

Cardiovascular Diabetology 1/2012 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Perioperative Checkpointhemmer-Therapie verbessert NSCLC-Prognose

28.05.2024 NSCLC Nachrichten

Eine perioperative Therapie mit Nivolumab reduziert das Risiko für Rezidive und Todesfälle bei operablem NSCLC im Vergleich zu einer alleinigen neoadjuvanten Chemotherapie um über 40%. Darauf deuten die Resultate der Phase-3-Studie CheckMate 77T.

Positiver FIT: Die Ursache liegt nicht immer im Dickdarm

27.05.2024 Blut im Stuhl Nachrichten

Immunchemischer Stuhltest positiv, Koloskopie negativ – in solchen Fällen kann die Blutungsquelle auch weiter proximal sitzen. Ein Forschungsteam hat nachgesehen, wie häufig und in welchen Lokalisationen das der Fall ist.

GLP-1-Agonisten können Fortschreiten diabetischer Retinopathie begünstigen

24.05.2024 Diabetische Retinopathie Nachrichten

Möglicherweise hängt es von der Art der Diabetesmedikamente ab, wie hoch das Risiko der Betroffenen ist, dass sich sehkraftgefährdende Komplikationen verschlimmern.

Update Innere Medizin

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