17β-estradiol induces an interaction between adenosine monophosphate-activated protein kinase and the insulin signaling pathway in 3T3-L1 adipocytes

  • Authors:
    • Ju-Young Kim
    • Kyung-Jin Jo
    • Byung-Joon Kim
    • Haing-Woon Baik
    • Seong-Kyu Lee
  • View Affiliations

  • Published online on: July 18, 2012     https://doi.org/10.3892/ijmm.2012.1070
  • Pages: 979-985
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Estrogen (17β-estradiol) has been implicated in maintaining insulin sensitivity. It is thought to act predominantly through genomic pathways and regulate the expression of various genes via binding to estrogen receptors (ERs)-α and -β. 17β-estradiol has been reported to simultaneously stimulate protein kinase B (Akt) and adenosine monophosphate-activated protein kinase (AMPK) in ex vivo skeletal muscle. Since data regarding the interaction between AMPK and the insulin receptor substrate-1 (IRS-1)/Akt pathway are controversial, the correlation between AMPK activation and insulin signaling remains unclear. In this study, we examined whether 17β-estradiol simultaneously stimulates the activation of AMPK and IRS-1/Akt in 3T3-L1 adipocytes as well as the 17β-estradiol-ER-induced interaction between the AMPK and IRS-1/Akt pathway in 3T3-L1 adipocytes not exposed to insulin. 17β-estradiol (10-7 M) rapidly activated AMPK and IRS-1/Akt in 3T3-L1 adipocytes, while the ER-α/β non-specific antagonist, ICI 182.780 (10 µM), and the AMPK antagonist compound C (20 µM) reversed the estrogen-induced activation of AMPK and tyrosine (Tyr)-IRS-1/Akt in these cells. Moreover, 17β-estradiol increased the expression of the peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α), adiponectin, uncoupling protein 2 (UCP2) and glucose transporter 4 (GLUT4) genes 24 h after treatment, whereas the ER-α/β non-specific antagonist, ICI 182.780 (10 µM), and the AMPK antagonist compound C (20 µM) reversed the estrogen-induced increase in the expression of these genes. These results indicate that 17β-estradiol activates AMPK through an ER and activates Akt through AMPK activation in 3T3-L1 adipocytes, despite the absence of insulin. Furthermore, 17β-estradiol regulates the expression of genes related to glucose metabolism through ER-AMPK activation in these cells.

Introduction

Type 2 diabetes and metabolic syndrome, both characterized by a combination of risk factors such as abdominal obesity, insulin resistance, glucose intolerance, dyslipidemia and hypertension, are more prevalent in postmenopausal women who suffer from estrogen deficiency compared to premenopausal women or men (13). It has been shown that long-term hormone therapy prevents diabetes and that short-term hormone therapy does not provide complete benefits in postmenopausal women (4). It has also been suggested that the effect of long-term hormone therapy in postmenopausal women may be due to altered body fat distribution and insulin sensitivity (5). Long-term estrogen treatment protects against obesity, glucose intolerance and insulin resistance in obese and insulin-resistant rodents with prolonged estrogen deficiency in the ovariectomized rodent (an animal model of menopause) (610). However, the molecular mechanisms underlying the short-term direct metabolic actions of estrogen on insulin sensitivity and glucose metabolism in non-obese rodents with short-term ovariectomy have not yet been clearly elucidated.

Estrogen (17β-estradiol) is considered to act predominantly through genomic pathways and regulate the expression of a number of genes via binding to estrogen receptors (ERs)-α and -β (11). Recently, the activation of non-genomic or rapid signaling pathways in response to 17β-estradiol has received increasing attention (12,13). Previous studies using ER knockout mouse models have demonstrated the role that estrogen and its receptors play in obesity and glucose tolerance in rodents. ER-α knockout mice develop adipocyte hyperplasia and hypertrophy, insulin resistance and glucose intolerance (14). ER-α knockout male mice display an obesity phenotype after sexual maturation (15). These previous studies support the idea that 17β-estradiol plays a role in obesity, insulin sensitivity and glucose metabolism via ER-α.

Common features of the metabolic abnormalities observed in metabolic syndrome or type 2 diabetes are the dysregulation of adenosine monophosphate-activated protein kinase (AMPK) (16) and insulin resistance involving changes in the molecules of the insulin signaling cascade, such as the insulin receptor, insulin receptor substrate-1 (IRS-1), phosphatidylinositol 3-kinase (PI3K) and protein kinase B (Akt) (17,18). Previous studies have shown that 17β-estradiol activates AMPK in rodent muscle (10,19,20) and C2C12 myotubes (19,21) and that 17β-estradiol upregulates PI3K/Akt signaling in human breast cancer cells through the activation of ER-α (22). It has been reported that 17β-estradiol simultaneously stimulates Akt and AMPK in ex vivo skeletal muscle, although 17β-estradiol does not stimulate ex vivo skeletal muscle glucose uptake (20). Another study has demonstrated that in vivo stimulation of ER-α in rats increases the activation of AMPK phosphorylation and insulin signaling proteins, such as phospho-Akt (pAkt), pAkt substrate (PAS) and insulin-stimulated skeletal muscle glucose uptake in rats (23). However, Rogers et al (20) suggested that 17β-estradiol does not stimulate Akt and AMPK in skeletal muscle cells, but instead stimulates these proteins in the non-skeletal muscle cells found within skeletal muscle. Since data regarding the interaction between AMPK and the IRS-1/Akt pathway are controversial, the correlation between AMPK activation and insulin signaling remains unclear.

In this study, we investigated whether 17β-estradiol simultaneously stimulates AMPK and IRS-1/Akt activation as well as the 17β-estradiol-ER-induced interaction between AMPK and the IRS-1/Akt pathway in 3T3-L1 adipocytes not exposed to insulin. Moreover, we examined whether 17β-estradiol regulates the expression of genes related to glucose metabolism through ER-AMPK activation in these cells.

Materials and methods

Materials

17β-estradiol, dexamethasone, 3-isobutyl-1-methylxanthine (IBMX) and β-actin antibody were purchased from Sigma (St. Louis, MO, USA). Recombinant human insulin was obtained from Eli Lilly and Company Lilly Corporate Center (Indianapolis, IN, USA). ICI 182.780 (ER-α/β non-specific antagonist) and compound C (AMPK inhibitor) were obtained from Tocris (Ellisville, MO, USA) and Calbiochem (San Diego, CA, USA), respectively. Dulbecco’s modified Eagle’s medium (DMEM), fetal bovine serum (FBS), trypsin-EDTA and penicillin/streptomycin were obtained from Gibco BRL (Grand Island, NY, USA). Anti-acetyl CoA carboxylase (ACC), anti-phospho Ser79 ACC, anti-IRS-1 and anti-phospho Ser789 IRS-1 monoclonal antibodies and anti-AMPKα and anti-phospho Thr172 AMPKα monoclonal antibodies were purchased from Cell Signaling Technology, Inc. (Beverly, MA, USA). Anti-phosphorylated tyrosine (pTyr)-IRS-1 was obtained from Santa Cruz Biotechnology Inc. (Santa Cruz, CA, USA). Anti-Akt and anti-phospho Ser473 Akt antibodies were purchased from Upstate Biotechnology, Inc. (Lake Placid, NY, USA). Horseradish peroxidase-conjugated sheep anti-mouse and donkey anti-rabbit immunoglobulin antibodies were obtained from Amersham Pharmacia Biotech, Inc. (Arlington Heights, IL, USA). The reagents for electrophoresis were obtained from Bio-Rad Laboratories, Inc. (Hercules, CA, USA). All other reagents were of analytical grade or complied with the standards required for cell culture experiments.

Cell culture and differentiation

3T3-L1 fibroblasts were obtained from the American Type Culture Collection (Manassas, VA, USA) and were grown in DMEM supplemented with 10% heat-inactivated FBS, penicillin (100 U/ml) and streptomycin (100 μg/ml). Two days after confluence, 3T3-L1 fibroblasts were used for differentiation (day 0). The differentiation medium, phenol red-free DMEM, contained 10% charcoal-stripped FBS (CS-FBS), 0.5 mM IBMX, 1 μM dexamethasone and 5 μg/ml insulin. After day 3, the differentiation medium was replaced with a post-differentiation medium containing 10% CS-FBS and 5 μg/ml insulin. After day 5, the post-differentiation medium was replaced with phenol red-free DMEM supplemented with 10% CS-FBS. The medium was changed every 3 days until the cells were used for experiments. Eight days after the induction of differentiation, >80–90% of the cells featured the morphological and biochemical properties of adipocytes. Before their use in experiments, the cells were starved for 12 h in phenol red-free DMEM containing 1% CS-FBS and then incubated with 10 μM ICI 182.780, 20 μM compound C, or 10−7 M 17β-estradiol alone or in combinations. ICI 182.780 and compound C were added 30 min prior to 17β-estradiol treatment for the indicated time and dose.

Quantitative real-time reverse transcription-polymerase chain reaction (RT-PCR)

Total RNA was isolated from the 3T3-L1 adipocytes using the TRIzol reagent (Invitrogen Life Technologies, Carlsbad, CA, USA), according to the manufacturer’s instructions. Equal amounts of total RNA were reverse-transcribed into cDNA using the ImProm-II™ reverse transcriptase system (Promega Corp., Madison, WI, USA) and an oligo(dT)15 primer (Promega Corp.). The peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α), adiponectin, uncoupling protein 2 (UCP2) and glucose transporter 4 (GLUT4) genes were selected for the experiments as the genes related to glucose metabolism. The primers used were as follows: PGC1α, 5′-TATGGTTTCATCACCTACCG-3′ and 5′-CGTCCACAAAAGTACAGCTC-3′; adiponectin, 5′-CCGTTCTCTTCACCTACGACC-3′ and 5′-GGGCTAT GGGTAGTTGCAGTC-3′; UCP2, 5′-AGATACATGAACT CTGCCTTGGG-3′ and 5′-GGCAGAGGATGAAGAAA AAGAC-3′; GLUT4, 5′-GTGTGTGAGCGAGTGCTTTCC-3′ and 5′-TGGAGACTGATGCGCTCTAACC-3′ and β-actin, 5′-ACGGCCAGGTCATCACTATT-3′ and 5′-AATGTAG TTTCATGGATGCC-3′. RT-PCR was conducted using the DNA Engine Opticon System (MJ Research, Inc., South San Francisco, CA, USA) in a 20-μl reaction mixture containing 10 μl of SYBR Premix Ex Taq (Takara Bio, Inc., Otsu, Japan), 10 pmol of forward primer, 10 pmol of reverse primer and 1 μg of cDNA. Amplification parameters consisted of an initial denaturation at 95°C for 5 min and 40 cycles of 3-step PCR (denaturation at 95°C for 1 min, annealing at 52–57°C for 30 sec and extension at 72°C for 1 min). Data were analyzed using a comparative critical threshold (Ct) method with the amount of the target gene normalized to the average of the control gene (β-actin). The percentage difference was calculated by 2−ΔΔCt. Each experiment was performed at least 4 times.

Western blot analysis

Whole cell lysates were prepared by lysing cells in Pro-prep protein extraction solution (Intron Biotechnology, Seoul, Korea) containing 10 mM sodium phosphate (pH 7), 1% Triton X-100, 0.1% SDS, 2 mM EDTA, 150 mM NaCl, 50 mM NaF, 0.1 mM sodium vanadate, 4 μg/ml leupeptin and 1 mM PMSF. The protein concentration of the lysates was measured using a Bio-Rad protein assay kit (Bio-Rad Laboratories, Inc.). Equal amounts of protein (20 μg/lane) were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) (4–20% gel) and transferred by electroblotting onto nitrocellulose membranes (Invitrogen Life Technologies). The membranes were then blocked with 5% non-fat dry milk and probed with primary antibodies. After being washed with phosphate-buffered saline (PBS) containing 0.1% Tween-20, the membranes were incubated for 1 h with horseradish peroxidase-conjugated sheep anti-mouse or donkey anti-rabbit immunoglobulin antibodies (1:500) (Amersham Pharmacia Biotech, Inc.). After washing with PBS containing 0.1% Tween-20, the specific signals were detected with an enhanced chemiluminescence detection system (Amersham Pharmacia Biotech, Inc.). The band densities were quantified using ImageJ software (http://rsb.info.nih.gov/ij/). Each experiment was performed at least 3 times.

Statistical analyses

Data are presented as the means ± standard deviation (SD). All statistical analyses were performed using the Statistical Package for the Social Sciences software (SPSS; Korean version 20.0). The statistical differences were analyzed using one-way analysis of variance (ANOVA) followed by Tukey’s test. Statistical significance was defined as P<0.05.

Results

17β-estradiol rapidly activates AMPK and IRS-1/Akt in 3T3-L1 adipocytes

17β-estradiol (10−7 M) phosphorylated AMPK and its substrate, ACC, within 10 min in 3T3-L1 adipocytes which were not exposed to insulin (Fig. 1A). 17β-estradiol (10−7 M) also pTyr-IRS-1 and Akt and decreased the phosphorylation of Ser789-IRS-1, whose level increases with insulin resistance (18), within 10 min in 3T3-L1 adipocytes not exposed to insulin (Fig. 1B). These results indicate that 17β-estradiol rapidly activates AMPK and IRS-1/Akt in 3T3-L1 adipocytes without insulin-induced signaling activation.

17β-estradiol activates AMPK and IRS-1/Akt through an ER in 3T3-L1 adipocytes

17β-estradiol (10−7 M) phosphorylated AMPK, its substrate ACC, Tyr-IRS-1 and Akt and decreased the phosphorylation of Ser789-IRS-1 10 min after treatment in 3T3-L1 adipocytes not exposed to insulin (Fig. 2). However, the ER-α/β non-specific antagonist, ICI 182.780 (10 μM), reversed the estrogen-induced activation of AMPK, ACC and Tyr-IRS-1/Akt and the estrogen-induced decrease in Ser789-IRS-1 phosphorylation in 3T3-L1 adipocytes not exposed to insulin (Fig. 2). These results indicate that 17β-estradiol activates AMPK and IRS-1/Akt through an ER in 3T3-L1 adipocytes without insulin-induced signaling activation.

17β-estradiol activates IRS-1/Akt through AMPK activation in 3T3-L1 adipocytes

17β-estradiol (10−7 M) phosphorylated AMPK, its substrate ACC, Tyr-IRS-1 and Akt and decreased Ser789-IRS-1 phosphorylation 10 min after treatment in 3T3-L1 adipocytes not exposed to insulin (Fig. 3). By contrast, the AMPK antagonist compound C (20 μM) reversed the estrogen-induced activation of AMPK, ACC and Tyr-IRS-1/Akt and the estrogen-induced decrease in Ser789-IRS-1 phosphorylation in 3T3-L1 adipocytes not exposed to insulin (Fig. 3). These results indicate that 17β-estradiol activates IRS-1/Akt through AMPK activation in 3T3-L1 adipocytes without insulin-induced signaling activation.

17β-estradiol regulates genes related to glucose metabolism through ER-AMPK activation in 3T3-L1 adipocytes

17β-estradiol (10−7 M) increased the expression of PGC1α, the adiponectin gene, UCP2 and GLUT4 at 24 h after treatment in 3T3-L1 adipocytes not exposed to insulin (Fig. 4). However, the ER-α/β non-specific antagonist, ICI 182.780 (10 μM), reversed the estrogen-induced increase in PGC1α, the adiponectin gene, UCP2 and GLUT4 expression 24 h after 17β-estradiol (10−7 M) treatment in 3T3-L1 adipocytes not exposed to insulin. In addition, the AMPK antagonist compound C (20 μM) also reversed the estrogen-induced increase in PGC1α, the adiponectin gene, UCP2 and GLUT4 expression 24 h after 17β-estradiol (10−7 M) treatment in 3T3-L1 adipocytes not exposed to insulin. These results indicate that 17β-estradiol regulates genes related to glucose metabolism through ER-AMPK activation in 3T3-L1 adipocytes without insulin-induced signaling activation.

Discussion

In the present study, we demonstrate that 17β-estradiol simultaneously stimulates the activation of IRS-1/Akt and AMPK and induces an interaction between AMPK and the IRS-1/Akt signaling pathway via an ER in 3T3-L1 adipocytes, despite the absence of insulin. Our data also demonstrate that an estrogen/ER-induced activation of AMPK functions upstream of IRS-1/Akt signaling and estrogen regulates the expression of genes related to glucose metabolism through 17β-estradiol-ER-AMPK activation in 3T3-L1 adipocytes in the absence of insulin-induced signaling activation.

Given that data concerning the simultaneous stimulation of IRS-1/Akt and AMPK signaling and the interaction between AMPK and IRS-1/Akt pathway remain controversial, this type of interaction may be highly cell type-dependent. The present study demonstrates that 17β-estradiol simultaneously stimulates the activation of IRS-1/Akt and AMPK and induces an interaction between AMPK and the IRS-1/Akt pathway without insulin-induced signaling activation in 3T3-L1 adipocytes. In addition, we show that AMPK functions upstream of Akt signaling in 3T3-L1 adipocytes, consistent with a previous study using human umbilical vein endothelium cells (HUVECs) (24). However, this result is in direct contrast with the results of another study showing that AMPK functions downstream of the PI3K pathway in vascular endothelial cells (25). Further research in various cell types or tissues is required to clarify this phenomenon.

Although we show that estrogen simultaneously activates AMPK and IRS-1/Akt via ER 10 min after treatment, the mechanism involved remains unclear. In a previous study, ER-α was shown to regulate insulin sensitivity through IRS-1 tyrosine phosphorylation in mature 3T3-L1 adipocytes (26). Additionally, a previous study demonstrated that ER-α regulates insulin sensitivity and glucose metabolism (15,27). On the basis of these observations, we postulated that 17β-estradiol may activate AMPK and IRS-1/Akt through ER-α. Further research on the activation of AMPK and IRS-1/Akt through the G protein-coupled receptor 30 (GPR30) or G protein-coupled ER (GPER) by 17β-estradiol is required as these receptors have recently been implicated in mediating several of the non-genomic or rapid signaling effects of 17β-estradiol (13,28,29). In the present study, 17β-estradiol (10−7 M) increased the expression of the glucose metabolism-related genes, PGC1α, the adiponectin gene, UCP2 and GLUT4, 24 h after treatment through 17β-estradiol-ER-AMPK activation in 3T3-L1 adipocytes without insulin-induced signaling activation. The co-activator protein, PGC1α, has been implicated in mitochondrial metabolism and biogenesis, thermogenesis, adipocyte differentiation and glucose metabolism, including gluconeogenesis and glucose uptake (30,31). Mitochondrial UCP2 has been implicated in physiological and pathological processes related to glucose and lipid metabolism (32). UCP2 is an inner mitochondrial membrane protein expressed in white adipose tissue and skeletal muscle (33) and is involved in energy expenditure (34). In skeletal muscle and adipose tissue, glucose uptake is maintained by one of the isoforms of the GLUT family (GLUT4) (35) and insulin resistance is associated with decreased glucose uptake in insulin-sensitive tissues, such as skeletal muscle and adipose tissue (36). The adipocytokine, adiponectin, reverses insulin resistance associated with both lipoatrophy and obesity (37). On the basis of these findings, PGC1α, UCP2, GLUT4 and the adiponectin gene were selected as the glucose metabolism-related genes for the present study. We demonstrated that 17β-estradiol increased the expression of PGC1α, UCP2, GLUT4 and the adiponectin gene in 3T3-L1 adipocytes, suggesting that estrogen possibly regulates insulin sensitivity, glucose metabolism and energy expenditure through the regulation of the expression levels of these genes. By contrast, an ER-α/β non-specific antagonist and an AMPK antagonist reversed the estrogen-induced increase of PGC1α, the adiponectin gene, UCP2 and GLUT4 expression, indicating that 17β-estradiol regulates the expression levels of genes related to glucose metabolism through 17β-estradiol-ER induced AMPK activation in 3T3-L1 adipocytes. Further studies are required in order to determine whether this phenomenon occurs in other cell types or tissues.

In conclusion, 17β-estradiol activates AMPK through an ER (possibly ER-α) and then activates Akt through AMPK activation in 3T3-L1 adipocytes, despite the absence of insulin. Moreover, 17β-estradiol also regulates the expression levels of genes related to glucose metabolism through ER-AMPK activation in 3T3-L1 adipocytes, despite the absence of insulin. Further research is required to elucidate the precise mechanisms underlying the estrogen-induced interaction between AMPK and the IRS-1/Akt pathway and the regulation of gene expression related to glucose metabolism.

Acknowledgements

This study was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education, Science and Technology of the Korean Government (NRF-2009-0076470).

References

1. 

S LiuF Mauvais-JarvisMinireview: estrogenic protection of β-cell failure in metabolic diseasesEndocrinology151859864201019966178

2. 

JG SchneiderC TompkinsRS BlumenthalS MoraThe metabolic syndrome in womenCardiol Rev14286291200610.1097/01.crd.0000233757.15181.6717053375

3. 

YW ParkS ZhuL PalaniappanS HeshkaMR CarnethonSB HeymsfieldThe metabolic syndrome: prevalence and associated risk factor findings in the US population from the Third National Health and Nutrition Examination Survey, 1988–1994Arch Intern Med1634274362003

4. 

K PenttiMT TuppurainenR HonkanenL SandiniH KrögerE AlhavaS SaarikoskiHormone therapy protects from diabetes: the Kuopio osteoporosis risk factor and prevention studyEur J Endocrinol160979983200910.1530/EJE-09-015119321660

5. 

L RichardsDiabetes: Postmenopausal hormone therapy prevents diabetesNat Rev Endocrinol5352200910.1038/nrendo.2009.92

6. 

YQ LiangM AkishitaS KimJ AkoM HashimotoK IijimaY OhikeT WatanabeN SudohK TobaEstrogen receptor beta is involved in the anorectic action of estrogenInt J Obes Relat Metab Disord2611031109200210.1038/sj.ijo.080205412119576

7. 

ML LiuX XuWQ RangYJ LiHP SongInfluence of ovariectomy and 17beta-estradiol treatment on insulin sensitivity, lipid metabolism and post-ischemic cardiac functionInt J Cardiol97485493200410.1016/j.ijcard.2003.11.04615561337

8. 

A PaquetteM ShinodaR Rabasa LhoretD Prud’hommeJM LavoieTime course of liver lipid infiltration in ovariectomized rats: impact of a high-fat dietMaturitas58182190200710.1016/j.maturitas.2007.08.00217889461

9. 

V SaengsirisuwanS PongseedaM PrasannarongK VichaiwongC ToskulkaoModulation of insulin resistance in ovariectomized rats by endurance exercise training and estrogen replacementMetabolism583847200910.1016/j.metabol.2008.08.00419059529

10. 

E RiantA WagetH CogoJF ArnalR BurcelinP GourdyEstrogens protect against high-fat diet-induced insulin resistance and glucose intolerance in miceEndocrinology15021092117200910.1210/en.2008-097119164473

11. 

SC HewittJC HarrellKS KorachLessons in estrogen biology from knockout and transgenic animalsAnnu Rev Physiol67285308200510.1146/annurev.physiol.67.040403.11591415709960

12. 

ER LevinRapid signaling by steroid receptorsAm J Physiol Regul Integr Comp Physiol295R1425R1430200810.1152/ajpregu.90605.200818784332

13. 

ER ProssnitzJB ArterburnHO SmithTI OpreaLA SklarHJ HathawayEstrogen signaling through the transmembrane G protein-coupled receptor GPR30Annu Rev Physiol70165190200810.1146/annurev.physiol.70.113006.10051818271749

14. 

PA HeineJA TaylorGA IwamotoDB LubahnPS CookeIncreased adipose tissue in male and female estrogen receptor-α knockout miceProc Natl Acad Sci USA9712729127342000

15. 

C OhlssonN HellbergP PariniO VidalM Bohlooly-YM RudlingMK LindbergM WarnerB AngelinJA GustafssonObesity and disturbed lipoprotein profile in estrogen receptor-alpha-deficient male miceBiochem Biophys Res Commun278640645200010.1006/bbrc.2000.382711095962

16. 

R LageC DiéguezA Vidal-PuigM LópezAMPK: a metabolic gauge regulating whole-body energy homeostasisTrends Mol Med14539549200810.1016/j.molmed.2008.09.00718977694

17. 

AR SaltielCR KahnInsulin signaling and the regulation of glucose and lipid metabolismNature414799806200110.1038/414799a11742412

18. 

N HorikeH TakemoriY KatohJ DoiL MinT AsanoXJ SunH YamamotoS KasayamaM MuraokaAdipose-specific expression, phosphorylation of Ser794 in insulin receptor substrate-1, and activation in diabetic animals of salt-inducible kinase-2J Biol Chem2871844018447200310.1074/jbc.M21177020012624099

19. 

TM D’EonSC SouzaM AronovitzMS ObinSK FriedAS GreenbergEstrogen regulation of adiposity and fuel partitioning. Evidence of genomic and non-genomic regulation of lipogenic and oxidative pathwaysJ Biol Chem2803598335991200516109719

20. 

NH RogersCA WitczakMF HirshmanLJ GoodyearAS GreenbergEstradiol stimulates Akt, AMP-activated protein kinase (AMPK) and TBC1D1/4, but not glucose uptake in rat soleusBiochem Biophys Res Commun382646650200910.1016/j.bbrc.2009.02.15419265681

21. 

TM D’EonNH RogersZS StanchevaAS GreenbergEstradiol and the estradiol metabolite, 2-hydroxyestradiol, activate AMP-activated protein kinase in C2C12 myotubesObesity (Silver Spring)1612841288200818421261

22. 

YR LeeJ ParkHN YuJS KimHJ YounSH JungUp-regulation of PI3K/Akt signaling by 17beta-estradiol through activation of estrogen receptor-alpha, but not estrogen receptor-beta, and stimulates cell growth in breast cancer cellsBiochem Biophys Res Commun33612211226200510.1016/j.bbrc.2005.08.25616169518

23. 

BK GorresGL BomhoffJK MorrisPC GeigerIn vivo stimulation of oestrogen receptor α increases insulin-stimulated skeletal muscle glucose uptakeJ Physiol589204120542011

24. 

N OuchiH KobayashiS KiharaM KumadaK SatoT InoueT FunahashiK WalshAdiponectin stimulates angiogenesis by promoting cross-talk between AMP-activated protein kinase and Akt signaling in endothelial cellsJ Biol Chem27913041309200410.1074/jbc.M31038920014557259

25. 

MH ZouSS KirkpatrickBJ DavisJS NelsonWG Wiles IVU SchlattnerD NeumannM BrownleeMB FreemanMH GoldmanActivation of the AMP-activated protein kinase by the anti-diabetic drug metformin in vivo. Role of mitochondrial reactive nitrogen speciesJ Biol Chem2794394043951200410.1074/jbc.M40442120015265871

26. 

K MurakiS OkuyaY TanizawaEstrogen receptor alpha regulates insulin sensitivity through IRS-1 tyrosine phosphorylation in mature 3T3-L1 adipocytesEndocr J53841851200610.1507/endocrj.K06-00517001108

27. 

A NaazM ZakroczymskiP HeineJ TaylorP SaundersD LubahnPS CookeEffect of ovariectomy on adipose tissue of mice in the absence of estrogen receptor alpha (ER alpha): a potential role for estrogen receptor beta (ER beta)Horm Metab Res34758763200210.1055/s-2002-3825912660895

28. 

G SharmaER ProssnitzMechanisms of estradiol-induced insulin secretion by the G protein-coupled estrogen receptor GPR30/GPER in pancreatic beta-cellsEndocrinology15230303039201110.1210/en.2011-009121673097

29. 

E FilardoJ QuinnY PangC GraeberS ShawJ DongP ThomasActivation of the novel estrogen receptor G protein-coupled receptor 30 (GPR30) at the plasma membraneEndocrinology14832363245200710.1210/en.2006-160517379646

30. 

P PuigserverBM SpiegelmanPeroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1 alpha): transcriptional coactivator and metabolic regulatorEndocr Rev247890200310.1210/er.2002-0012

31. 

D KnuttiA KralliPGC-1, a versatile coactivatorTrends Endocrinol Metab12360365200110.1016/S1043-2760(01)00457-X11551810

32. 

S DianoTL HorvathMitochondrial uncoupling protein 2 (UCP2) in glucose and lipid metabolismTrends Mol Med185258201210.1016/j.molmed.2011.08.00321917523

33. 

MC SalehMB WheelerCB ChanUncoupling protein-2: evidence for its function as a metabolic regulatorDiabetologia45174187200210.1007/s00125-001-0737-x

34. 

C PecqueurE CouplanF BouillaudD RicquierGenetic and physiological analysis of the role of uncoupling proteins in human energy homeostasisJ Mol Med (Berl)794856200110.1007/s00109000015011327103

35. 

JW RyderM GilbertJR ZierathSkeletal muscle and insulin sensitivity: pathophysiological alterationsFront Biosci6D154D163200110.2741/Ryder11171554

36. 

GI BellKS PolonskyDiabetes mellitus and genetically programmed defects in beta-cell functionNature414788791200110.1038/414788a11742410

37. 

T YamauchiJ KamonH WakiY TerauchiN KubotaK HaraY MoriT IdeK MurakamiN Tsuboyama-KasaokaThe fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesityNat Med7941946200110.1038/9098411479627

Related Articles

Journal Cover

October 2012
Volume 30 Issue 4

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kim J, Jo K, Kim B, Baik H and Lee S: 17β-estradiol induces an interaction between adenosine monophosphate-activated protein kinase and the insulin signaling pathway in 3T3-L1 adipocytes. Int J Mol Med 30: 979-985, 2012
APA
Kim, J., Jo, K., Kim, B., Baik, H., & Lee, S. (2012). 17β-estradiol induces an interaction between adenosine monophosphate-activated protein kinase and the insulin signaling pathway in 3T3-L1 adipocytes. International Journal of Molecular Medicine, 30, 979-985. https://doi.org/10.3892/ijmm.2012.1070
MLA
Kim, J., Jo, K., Kim, B., Baik, H., Lee, S."17β-estradiol induces an interaction between adenosine monophosphate-activated protein kinase and the insulin signaling pathway in 3T3-L1 adipocytes". International Journal of Molecular Medicine 30.4 (2012): 979-985.
Chicago
Kim, J., Jo, K., Kim, B., Baik, H., Lee, S."17β-estradiol induces an interaction between adenosine monophosphate-activated protein kinase and the insulin signaling pathway in 3T3-L1 adipocytes". International Journal of Molecular Medicine 30, no. 4 (2012): 979-985. https://doi.org/10.3892/ijmm.2012.1070