Adiponectin receptor agonist AdipoRon suppresses adipogenesis in C3H10T1/2 cells through the adenosine monophosphate‑activated protein kinase signaling pathway

  • Authors:
    • Shu‑Juan Wang
    • Wen‑Yi Lu
    • Kai‑Yan Liu
  • View Affiliations

  • Published online on: September 8, 2017     https://doi.org/10.3892/mmr.2017.7450
  • Pages: 7163-7169
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to investigate the effects of AdipoRon, an adiponectin receptor agonist, on adipogenesis in C3H10T1/2 cells and to explore the underlying mechanisms. C3H10T1/2 cells were treated with increasing doses of AdipoRon for 8 days, and Oil Red O staining was used to assess lipid accumulation. The protein and mRNA expression levels of adipogenic transcription factors and adipocyte‑specific genes were examined by western blotting and reverse transcription quantitative polymerase chain reaction, respectively. AdipoRon treatment inhibited lipid accumulation in C3H10T1/2 cells in a dose‑dependent manner and significantly suppressed the expression of adipogenic transcription factors, including peroxisome proliferator‑activated receptor γ, CAAT/enhancer binding protein (C/EBP)‑β and C/EBPα. In addition, cells treated with AdipoRon exhibited a significant decrease in the expression of adipocyte‑specific genes, including fatty acid binding protein 4, fatty acid synthase, leptin, adiponectin, and stearoyl‑CoA desaturase‑1. Notably, AdipoRon significantly increased the phosphorylation of adenosine monophosphate‑activated protein kinase (AMPK) and acetyl‑CoA carboxylase (ACC). The results indicated that AdipoRon exerted an inhibitory effect on adipogenesis in C3H10T1/2 cells by downregulating the expression of adipogenic transcription factors and adipocyte‑specific genes and by promoting the phosphorylation of AMPK and ACC, which suggested that AdipoRon may be a potential drug to prevent and treat diseases caused by abnormal adipogenesis, such as obesity.

Introduction

Obesity is a common public health problem and is a significant risk factor for a number of metabolic disorders, including type 2 diabetes, hypertension and cardiovascular disease (1). The development of obesity is characterized by an increase in the number and size of mature adipocytes that are produced by differentiation and mitogenesis (2). Adipogenesis is controlled by many transcription factors, including peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer binding proteins (C/EBPs) (35). At the initial stage of adipogenesis, C/EBPβ is activated, followed by the sequential activation of C/EBPα and PPARγ, two major late-adipogenic transcription factors. Activation of C/EBPα and PPARγ subsequently leads to the expression of a number of genes involved in adipocyte differentiation, including genes encoding lipid metabolizing enzymes [such as fatty acid binding protein (FABP)-4 and fatty acid synthase (FAS)] and adipokines (such as adiponectin and leptin) (6).

Adenosine monophosphate-activated protein kinase (AMPK) is an important cellular energy sensor, and its activation is closely related to the balance between lipid accumulation and carbohydrate metabolism (7). AMPK phosphorylation inhibits metabolic enzymes involved in fatty acid synthesis, leading to suppressed adipogenesis (8,9).

Adiponectin is an adipokine that is mainly produced by adipocytes. Unlike other adipokines, plasma adiponectin levels are reduced in obese subjects (10). Adiponectin binds to the adiponectin receptors, AdipoR1 and AdipoR2, and reduces obesity-related insulin resistance (11). AdipoRon, a newly discovered, orally active small molecule, is an adiponectin receptor agonist that binds to and activates both AdipoR1 and AdipoR2, and has been previously reported to improve obesity-related insulin resistance and type 2 diabetes by activating AMPK and PPARα pathways (12). However, the effects of adiponectin on obesity itself and on adipogenesis are controversial. Adiponectin may promote adipogenesis induced by weight gain (11,13). By contrast, elevated adiponectin levels were recently reported to reduce lipid content and lipid metabolism in 3T3-L1 cells (14). However, the effects of AdipoRon on adipogenesis have not yet been investigated. The present study investigated the effects of AdipoRon on adipogenesis and explored the underlying mechanism in C3H10T1/2 cells.

Materials and methods

Reagents

AdipoRon, 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside (AICAR), dexamethasone (Dex), 3-isobutyl-1-methylxanthine (IBMX), indomethacin, insulin and Oil Red O were purchased from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany). Minimum essential Eagle's medium with Earle's Balanced Salts (MEM-EBSS) was purchased from HyClone (GE Healthcare Life Sciences, Logan, UT, USA). Fetal bovine serum (FBS) was purchased from Gibco (Thermo Fisher Scientific, Inc., Waltham, MA, USA). Cell Counting Kit-8 (CCK-8) was purchased from Dojindo Molecular Technologies, Inc. (Kumamoto, Japan). Monoclonal antibodies, including rabbit anti-PPARγ (cat no. 2443), rabbit anti-FABP4 (cat no. 3544), rabbit anti-C/EBPβ (cat no. 3087), rabbit anti-C/EBPα (cat no. 2295), rabbit anti-AMPKα (cat no. 5831), rabbit anti-phosphorylated-acetyl-CoA carboxylase (p-ACC; cat no. 11818), rabbit anti-adiponectin (cat no. 2789) and rabbit anti-GAPDH (cat no. 5174) were purchased from Cell Signaling Technology (Danvers, MA, USA). Rabbit polyclonal anti-p-AMPKα1/2 antibody (cat no. sc-33524) and goat polyclonal anti-AdipoR1 antibody (cat no. sc-46748) and anti-AdipoR2 antibody (cat no. sc-46751) were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA).

Cell culture and adipocyte differentiation

The C3H10T1/2 mouse embryonic mesenchymal stem cell line was purchased from the Chinese Academy of Medical Sciences (Beijing, China). Cells were cultured in MEM-EBSS supplemented with 10% FBS at 37°C in a 5% CO2 atmosphere until adipocyte differentiation. Two days after reaching confluence (day 0), the C3H10T1/2 cells were cultured in differentiation medium (DM; MEM/EBSS containing 10% FBS, 1 µM Dex, 0.5 mM IBMX, 50 mM indomethacin and 1 µg/ml insulin) for 2 days to induce differentiation. Following incubation, MEM/EBSS supplemented with 10% FBS and 1 µg/ml insulin was added for 2 days as a differentiation medium and was changed every 2 days.

Cell viability assay

Cell viability was detected by using a CCK-8 kit, according to the manufacturer's instructions. Briefly, C3H10T1/2 cells were seeded in 96-well plates at a density of 5×104 cells/ml at 37°C in a humidified 5% CO2 atmosphere and treated with increasing doses of AdipoRon (1, 5, 10, 20 and 40 µM) for 24, 48 or 72 h. Subsequently, 10 µl of kit reagent was added to each well and incubated at 37°C for 2 h. The plates were scanned with a microplate reader (Benchmark; Bio-Rad Laboratories, Inc., Hercules, CA, USA) at 450 nm.

Oil Red O (ORO) staining

Cells were cultured in 24-well plates with a seeding density of 5×104 cells/ml and differentiated as described above. On day 8, the cells were stained with ORO as previously described (2). Briefly, the cells were fixed with 10% formalin (Solarbio, Beijing, China) at 4°C for 30 min and stained with ORO at room temperature for 30 min following washes with phosphate-buffered saline. Oil droplets in the cells were observed under an inverted microscope (Olympus-CKX41; Olympus Corporation, Tokyo, Japan). The ORO-stained plates were then washed and treated with 100% isopropanol (Beijing Solarbio Science & Technology Co., Ltd., Beijing, China), and lipid accumulation was detected by measuring the absorbance at 490 nm using a Benchmark microplate reader (Bio-Rad Laboratories, Inc.).

Western blot analysis

Cells were cultured in 6-well plates with a seeding density of 5×104 cells/ml and differentiated as described above. Cells were lysed in radioimmunoprecipitation assay buffer (Beyotime Institute of Biotechnology, Shanghai, China) supplemented with a protease inhibitor cocktail and phosphatase inhibitors (Roche Molecular Diagnostics, Pleasanton, CA, USA) for 20 min and centrifuged at 12,000 × g for 20 min at 4°C. Protein concentration was determined using the Bicinchoninic Acid Protein Assay reagent kit (Beijing Solarbio Science & Technology Co., Ltd.). A total of 30 µg of protein lysates from each sample were separated on 10 or 12% SDS-PAGE and transferred to a polyvinylidene difluoride membrane (EMD Millipore, Billerica, MA, USA). The membrane was incubated with 5% bovine serum albumin in Tris-buffered saline with 0.1% Tween-20 (TBST) (both from Beijing Solarbio Science & Technology Co., Ltd.) for 1 h at room temperature, followed by hybridization with primary antibodies against PPARγ (1:1,000), C/EBPβ (1:1,000), C/EBPα (1:1,000), FABP4 (1:2,000), adiponectin (1:500), AMPKα (1:1,000), p-ACC (1:500), GAPDH (1:1,000), p-AMPKα1/2 (1:200), AdipoR1 (1:200) and AdipoR2 (1:200) overnight at 4°C. Following three washes with TBST, the membrane was incubated with a horseradish peroxidase-conjugated goat anti-rabbit IgG secondary antibody (cat no. sc2004; 1:10,000; Santa Cruz Biotechnology, Inc.) or a horseradish peroxidase-conjugated rabbit anti-goat IgG secondary antibody (cat no. ZB-2306; 1:10,000; Beijing Zhongshan Jinqiao Biotechnology Co., Ltd., Beijing, China) for 1 h at room temperature. Following three washes with TBST, the bands were visualized with an Enhanced Chemiluminescence Substrate (Thermo Fisher Scientific, Inc.). Densitometry of the western blot bands was performed using ImageJ software version 1.31 (National Institutes of Health, Bethesda, ML, USA).

RNA preparation and reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Cells were cultured in 6-well plates with a seeding density of 5×104 cells/ml and differentiated as described above. RNA was extracted from C3H10T1/2 cells on day 8 using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.). A High Capacity cDNA Reverse Transcription kit (Applied Biosystems; Thermo Fisher Scientific, Inc.) was used to synthesize the cDNA, according to the manufacturer's protocol. mRNA expression levels were determined using an ABI 7500 Fast real-time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.) and a SYBR Green PCR Master Mix (Applied Biosystems; Thermo Fisher Scientific, Inc.). Primers were obtained from Genotech (Shanghai, China) and the sequences are provided in Table I. GAPDH was used as the internal control. qPCR was performed using the following previous described cycling conditions (15): Initial denaturation at 95°C for 10 min, followed by 40 cycles at 95°C for 15 sec and 60°C for 1 min. Each experiment was performed in triplicate. Gene transcript levels were calculated using the 2−ΔΔCq method (16).

Table I.

Primers used for reverse transcription quantitative polymerase chain reaction.

Table I.

Primers used for reverse transcription quantitative polymerase chain reaction.

GenePrimer sequence (5′→3′)
C/EBPαF: CGGGAACGCAACAACATCGC
C/EBPαR: CGGTCATTGTCACTGGTCAACTC
C/EBPβF: GTTTCGGGAGTTGATGCAATC
C/EBPβR: AACAACCCCGCAGGAACAT
PPARγF: CATTCGCATTCCTTTGAC
PPARγR: CGCACTTTGGTATTCTTGGAG
FASF: CAAGTGTCCACCAACAAGCG
FASR: GGAGCGCAGGATAGACTCAC
FABP4F: TGTGCGAAACTGAATTTCCTGC
FABP4R: GAGATCGGTCCTGAGCCAGC
AdiponectinF: GCCGCTTATGTGTATCGCTCAG
AdiponectinR: GCCAGTGCTGCCGTCATAATG
LeptinF: TCGCCTTTCTCCTGATGACG
LeptinR: GCAATCACACGGATGGCTTC
SCD-1F: CGCTGGCACATCAACTTCAC
SCD-1R: AGGAACTCAGAAGCCCAAAGC
GAPDHF: TCAATGACAACTTTGTCAAGCTCA
GAPDHR: GTGGGTGGTCCAGGGTTTCTTACT

[i] C/EBP, CAAT/enhancer binding protein; FABP, fatty acid binding protein; FAS, fatty acid synthase; PPAR, peroxisome proliferator-activated receptor; SCD-1, stearoyl-CoA desaturase 1.

Statistical analysis

All experiments were performed at least three times. The results are expressed as the mean ± standard deviations (SD). The data were analyzed using Student's t-test or one-way analysis of variance followed by least significance difference test. All analyses were performed with GraphPad Prism version 5.01 (GraphPad Software, Inc., La Jolla, CA, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

AdipoRon suppresses lipid accumulation in C3H10T1/2 cells without cytotoxicity

Following treatment for 24, 48 or 72 h, AdipoRon exhibited no significant effects on C3H10T1/2 cell viability at concentrations ≤40 µM (Fig. 1A). Therefore, 5–20 µM AdipoRon was used in all subsequent experiments. The antiadipogenic effects of AdipoRon treatment were not due to cytotoxicity. C3H10T1/2 cells were induced into adipocytes for 8 days in the presence or absence of various doses of AdipoRon (0–20 µM) to investigate the effects of AdipoRon on adipocyte differentiation. Based on the results of ORO staining, lipid accumulation in C3H10T1/2 cells was dose-dependently decreased by AdipoRon (Fig. 1B and C). Protein expression of the adiponectin receptors, AdipoR1 and AdipoR2, were detected in C3H10T1/2 cells (Fig. 1D).

Effects of AdipoRon on adipogenic transcription factors expression in C3H10T1/2 cells

Adipogenesis is controlled by numerous transcription factors; the present study examined the effects of various concentrations of AdipoRon (0–20 µM) on the mRNA expression levels C/EBPβ, C/EBPα and PPARγ, to investigate whether AdipoRon inhibits adipogenesis by downregulating the expression of these adipogenic transcription factors. AdipoRon treatment significantly reduced the expression levels of the C/EBPβ, C/EBPα and PPARγ mRNAs in a dose-dependent manner (Fig. 2A-C). In addition, treatment with 20 µM AdipoRon was used to examine the expression levels of the C/EBPβ, C/EBPα and PPARγ proteins. The expression levels of the C/EBPβ, C/EBPα and PPARγ proteins were decreased by 20 µM AdipoRon compared with those of the controls (Fig. 2D).

Effects of AdipoRon on the expression levels of adipogenesis-related genes in C3H10T1/2 cells

As AdipoRon treatment significantly downregulated the expression of adipogenic transcription factors, the effects of various concentrations of AdipoRon on the expression levels of adipogenesis-related genes were also examined. AdipoRon exposure dose-dependently suppressed the mRNA expression of FAS, leptin, stearoyl-CoA desaturase (SCD)-1, adiponectin and FABP4 (Fig. 3A-E). Consistent with the changes in mRNA expression levels, 20 µM AdipoRon decreased the expression levels of the adiponectin and FABP4 proteins (Fig. 3F).

Effects of AdipoRon on AMPK phosphorylation in C3H10T1/2 cells

AdipoRon treatment significantly increased the levels of p-AMPK and p-ACC, which is the downstream target of AMPK (Fig. 4A-C). The AMPK activator AICAR was used as a positive control to further explore the association between the activation of AMPK signaling and the antiadipogenic effects mediated by AdipoRon. As shown in Fig. 4D and E, both AICAR (1 mM) and AdipoRon (10 and 20 µM) were able to activate the AMPK signaling pathway. Both AICAR and AdipoRon treatments were also able to significantly reduce lipid accumulation and the expression of adipogenesis-related genes, PPARγ and adiponectin, in C3H10T1/2 cells (Fig. 4F-H).

Discussion

Obesity and its associated complications have become common problems worldwide. AdipoRon, an adiponectin receptor agonist, is a promising new orally administered drug for the treatment of obesity-related diabetes (12). However, its effects on obesity itself and adipogenesis have not yet been investigated. Results from the present study demonstrated that AdipoRon treatment inhibited the differentiation of C3H10T1/2 cells into adipocytes by downregulating the expression of adipogenic transcription factors and regulating the AMPK signaling pathway.

Abnormal adipogenesis and lipid accumulation have previously been associated with the development of obesity (2). A potential treatment strategy for obesity is to inhibit adipogenesis. The presents study results indicated that AdipoRon dose-dependently inhibited lipid accumulation in C3H10T1/2 cells without cytotoxicity. Adipogenesis is coordinated by many transcription factors, such as C/EBPs and PPARs; AdipoRon exposure downregulated the expression levels of the C/EBPβ mRNA, which subsequently suppressed the expression of the C/EBPα and PPARγ mRNAs. PPARγ and C/EBPα are major regulators of adipogenesis by activating the transcription of terminal adipocyte differentiation marker genes, including FABP4, FAS, leptin, SCD-1 and adiponectin (35); the expression levels of these mRNAs were demonstrated to be significantly decreased by AdipoRon in the present study. Therefore, AdipoRon treatment suppressed the expression of transcription factors during adipogenesis, which then inhibited adipocyte differentiation of C3H10T1/2 cells.

Obesity occurs when total energy intake exceeds total energy expenditure. AMPK is a metabolic energy sensor that senses nutritional stress for the purpose of regulating glucose and lipid metabolism and may be a potential target for the treatment of obesity (17). AMPK has been reported to serve an important role in inhibiting adipogenesis in 3T3-L1 cells that have been induced by a number of different natural compounds (2,6,8,1820). Activated AMPK inhibits the differentiation of adipocytes and the expression of pro-adipogenesis transcription factors PPARγ and C/EBPα (21). AMPK phosphorylation induces the phosphorylation of its substrate, ACC, which functions to strictly regulate the enzymes that are involved in the fatty acid synthesis pathway that produces malonyl-CoA. Notably, p-ACC lacks the ability to synthesize fatty acids (22). AdipoRon was previously reported to activate AMPK signaling in muscle and liver of obese mice (12). To elucidate whether AdipoRon inhibited adipogenesis through the AMPK signaling pathway, the present study measured the levels of p-AMPK and p-ACC; AdipoRon treatment significantly increased the phosphorylation of AMPK and ACC. In addition, the AMPK activator AICAR was used as a positive control to further elucidate the association between the anti-adipogenesis ability of AdipoRon and the activation of the AMPK signaling pathway. AICAR was previously reported to inhibit adipogenesis in 3T3-L1 adipocytes and restore metabolic alterations in a diet-induced mouse model of obesity (2325). Results from the present study revealed that both AICAR and AdipoRon were able to activate the AMPK signaling pathway and markedly reduce lipid accumulation in C3H10T1/2 cells, which indicated that the anti-adipogenic effects of AdipoRon may be related to the activation of the AMPK signaling pathway. As C3H10T1/2 is a mouse embryonic mesenchymal stem cell line, the anti-adipogenesis effects of AdipoRon require further validation in human-sourced mesenchymal stem cell line.

In conclusion, AdipoRon, an adiponectin receptor agonist, suppressed adipocyte differentiation of C3H10T1/2 cells, probably by activating the AMPK signaling pathway and downregulating the expression of key regulators of adipogenesis. Therefore, AdipoRon may be a drug with the potential to prevent and treat obesity.

Acknowledgements

This study was supported by grants from the National Natural Science Foundation of China (grant nos. 81670098 and 81270572) and the National Basic Research 973 Program (grant no. 2013CB733701).

References

1 

Despres JP and Lemieux I: Abdominal obesity and metabolic syndrome. Nature. 444:881–887. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Ko SC, Lee M, Lee JH, Lee SH, Lim Y and Jeon YJ: Dieckol, a phlorotannin isolated from a brown seaweed, Ecklonia cava, inhibits adipogenesis through AMP-activated protein kinase (AMPK) activation in 3T3-L1 preadipocytes. Environ Toxicol Pharmacol. 36:1253–1260. 2013. View Article : Google Scholar : PubMed/NCBI

3 

White UA and Stephens JM: Transcriptional factors that promote formation of white adipose tissue. Mol Cell Endocrinol. 318:10–14. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Rosen ED and Spiegelman BM: PPARgamma: A nuclear regulator of metabolism, differentiation, and cell growth. J Biol Chem. 276:37731–37734. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Wu Z, Rosen ED, Brun R, Hauser S, Adelmant G, Troy AE, McKeon C, Darlington GJ and Spiegelman BM: Cross-regulation of C/EBP alpha and PPAR gamma controls the transcriptional pathway of adipogenesis and insulin sensitivity. Mol Cell. 3:151–158. 1999. View Article : Google Scholar : PubMed/NCBI

6 

Lee JH, Kim T, Lee JJ, Lee KJ, Kim HK, Yun B, Jeon J, Kim SK and Ma JY: The herbal medicine KBH-1 inhibits fat accumulation in 3T3-L1 adipocytes and reduces high fat diet-induced obesity through regulation of the AMPK pathway. PLoS One. 10:e01420412015. View Article : Google Scholar : PubMed/NCBI

7 

Lin JT, Chen HM, Chiu CH and Liang YJ: AMP-activated protein kinase activators in diabetic ulcers: From animal studies to phase II drugs under investigation. Expert Opin Investig Drugs. 23:1253–1265. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Kim SK and Kong CS: Anti-adipogenic effect of dioxinodehydroeckol via AMPK activation in 3T3-L1 adipocytes. Chem Biol Interact. 186:24–29. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Ceddia RB: The role of AMP-activated protein kinase in regulating white adipose tissue metabolism. Mol Cell Endocrinol. 366:194–203. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Hotta K, Funahashi T, Arita Y, Takahashi M, Matsuda M, Okamoto Y, Iwahashi H, Kuriyama H, Ouchi N, Maeda K, et al: Plasma concentrations of a novel, adipose-specific protein, adiponectin, in type 2 diabetic patients. Arterioscler Thromb Vasc Biol. 20:1595–1599. 2000. View Article : Google Scholar : PubMed/NCBI

11 

Yamauchi T, Kamon J, Waki H, Terauchi Y, Kubota N, Hara K, Mori Y, Ide T, Murakami K, Tsuboyama-Kasaoka N, et al: The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat Med. 7:941–946. 2001. View Article : Google Scholar : PubMed/NCBI

12 

Okada-Iwabu M, Yamauchi T, Iwabu M, Honma T, Hamagami K, Matsuda K, Yamaguchi M, Tanabe H, Kimura-Someya T, Shirouzu M, et al: A small-molecule AdipoR agonist for type 2 diabetes and short life in obesity. Nature. 503:493–499. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Holland WL and Scherer PE: Cell Biology. Ronning after the adiponectin receptors. Science. 342:1460–1461. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Lazra Y, Falach A, Frenkel L, Rozenberg K, Sampson S and Rosenzweig T: Autocrine/paracrine function of globular adiponectin: Inhibition of lipid metabolism and inflammatory response in 3T3-L1 adipocytes. J Cell Biochem. 116:754–766. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Poudel B, Nepali S, Xin M, Ki HH, Kim YH, Kim DK and Lee YM: Flavonoids from Triticum aestivum inhibit adipogenesis in 3T3-L1 cells by upregulating the insig pathway. Mol Med Rep. 12:3139–3145. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Cantó C, Gerhart-Hines Z, Feige JN, Lagouge M, Noriega L, Milne JC, Elliott PJ, Puigserver P and Auwerx J: AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity. Nature. 458:1056–1060. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Liou CJ, Lai XY, Chen YL, Wang CL, Wei CH and Huang WC: Ginkgolide C suppresses adipogenesis in 3T3-L1 adipocytes via the AMPK signaling pathway. Evid Based Complement Alternat Med. 2015:2986352015. View Article : Google Scholar : PubMed/NCBI

19 

Lee YK, Lee WS, Hwang JT, Kwon DY, Surh YJ and Park OJ: Curcumin exerts antidifferentiation effect through AMPKalpha-PPAR-gamma in 3T3-L1 adipocytes and antiproliferatory effect through AMPKalpha-COX-2 in cancer cells. J Agric Food Chem. 57:305–310. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Ahn J, Lee H, Kim S, Park J and Ha T: The anti-obesity effect of quercetin is mediated by the AMPK and MAPK signaling pathways. Biochem Biophys Res Commun. 373:545–549. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Chen S, Li Z, Li W, Shan Z and Zhu W: Resveratrol inhibits cell differentiation in 3T3-L1 adipocytes via activation of AMPK. Can J Physiol Pharmacol. 89:793–799. 2011.PubMed/NCBI

22 

Daval M, Foufelle F and Ferré P: Functions of AMP-activated protein kinase in adipose tissue. J Physiol. 574:55–62. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Giri S, Rattan R, Haq E, Khan M, Yasmin R, Won JS, Key L, Singh AK and Singh I: AICAR inhibits adipocyte differentiation in 3T3L1 and restores metabolic alterations in diet-induced obesity mice model. Nutr Metab (Lond). 3:312006. View Article : Google Scholar : PubMed/NCBI

24 

Habinowski SA and Witters LA: The effects of AICAR on adipocyte differentiation of 3T3-L1 cells. Biochem Biophys Res Commun. 286:852–856. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Lee H, Kang R, Bae S and Yoon Y: AICAR, an activator of AMPK, inhibits adipogenesis via the WNT/β-catenin pathway in 3T3-L1 adipocytes. Int J Mol Med. 28:65–71. 2011.PubMed/NCBI

Related Articles

Journal Cover

November-2017
Volume 16 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang SJ, Lu WY and Liu KY: Adiponectin receptor agonist AdipoRon suppresses adipogenesis in C3H10T1/2 cells through the adenosine monophosphate‑activated protein kinase signaling pathway. Mol Med Rep 16: 7163-7169, 2017
APA
Wang, S., Lu, W., & Liu, K. (2017). Adiponectin receptor agonist AdipoRon suppresses adipogenesis in C3H10T1/2 cells through the adenosine monophosphate‑activated protein kinase signaling pathway. Molecular Medicine Reports, 16, 7163-7169. https://doi.org/10.3892/mmr.2017.7450
MLA
Wang, S., Lu, W., Liu, K."Adiponectin receptor agonist AdipoRon suppresses adipogenesis in C3H10T1/2 cells through the adenosine monophosphate‑activated protein kinase signaling pathway". Molecular Medicine Reports 16.5 (2017): 7163-7169.
Chicago
Wang, S., Lu, W., Liu, K."Adiponectin receptor agonist AdipoRon suppresses adipogenesis in C3H10T1/2 cells through the adenosine monophosphate‑activated protein kinase signaling pathway". Molecular Medicine Reports 16, no. 5 (2017): 7163-7169. https://doi.org/10.3892/mmr.2017.7450