Journal of Obesity & Metabolic Syndrome

Search

Article

J Obes Metab Syndr 2024; 33(2): 177-188

Published online June 30, 2024 https://doi.org/10.7570/jomes23048

Copyright © Korean Society for the Study of Obesity.

Alpha-Lipoic Acid Induces Adipose Tissue Browning through AMP-Activated Protein Kinase Signaling in Vivo and in Vitro

Shieh-Yang Huang1, Ming-Ting Chung2, Ching-Wen Kung3, Shu-Ying Chen4, Yi-Wen Chen5, Tong Pan5, Pao-Yun Cheng6, Hsin-Hsueh Shen5,* , Yen-Mei Lee5,*

1Department of Pharmacy, Kaohsiung Armed Forces General Hospital, Kaohsiung; 2Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Chi-Mei Medical Center, Tainan; 3Department of Nursing, Tzu Chi University of Science and Technology, Hualien; 4Department of Nursing, Hung Kuang University, Taichung; 5Department of Pharmacology and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei; 6Department of Physiology & Biophysics, National Defense Medical Center, Taipei, Taiwan

Correspondence to:
Yen-Mei Lee
https://orcid.org/0000-0002-5855-5346
Department Pharmacology and Institute of Pharmacology, National Defense Medical Center, No. 161, Section 6, Min-Chuan East Road, Taipei 114, Taiwan
Tel: +886-2-87927877
Fax: +886-2-87927877
E-mail: ymlee@mail.ndmctsgh.edu.tw

Hsin-Hsueh Shen
https://orcid.org/0000-0002-5937-3472
Department Pharmacology and Institute of Pharmacology, National Defense Medical Center, No. 161, Section 6, Min-Chuan East Road, Taipei 114, Taiwan
Tel: +886-2-87927877
Fax: +886-2-87927877
E-mail: hsshen@mail.ndmctsgh.edu.tw

The first two authors contributed equally to this study.

Received: August 4, 2023; Reviewed : October 3, 2023; Accepted: December 19, 2023

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Background: AMP-activated protein kinase (AMPK) is a key enzyme for cellular energy homeostasis and improves metabolic disorders. Brown and beige adipose tissues exert thermogenesis capacities to dissipate energy in the form of heat. Here, we investigated the beneficial effects of the antioxidant alpha-lipoic acid (ALA) in menopausal obesity and the underlying mechanisms.
Methods: Female Wistar rats (8 weeks old) were subjected to bilateral ovariectomy (Ovx) and divided into four groups: Sham (n=8), Ovx (n=11), Ovx+ALA2 (n=10), and Ovx+ALA3 (n=6) (ALA 200 and 300 mg/kg/day, respectively; gavage) for 8 weeks. 3T3-L1 cells were used for in vitro study.
Results: Rats receiving ALA2 and ALA3 treatment showed significantly lower levels of body weight and white adipose tissue (WAT) mass than those of the Ovx group. ALA improved plasma lipid profiles including triglycerides, total cholesterol, low-density lipoprotein cholesterol, and high-density lipoprotein cholesterol. Hematoxylin & eosin staining of inguinal WAT showed that ALA treatment reduced Ovx-induced adipocyte size and enhanced uncoupling protein 1 (UCP1) expression. Moreover, plasma levels of irisin were markedly increased in ALA-treated Ovx rats. Protein expression of brown fat-specific markers including UCP1, PRDM16, and CIDEA was downregulated by Ovx but markedly increased by ALA. Phosphorylation of AMPK, its downstream acetyl-CoA carboxylase, and its upstream LKB1 were all significantly increased by ALA treatment. In 3T3-L1 cells, administration of ALA (100 and 250 μM) reduced lipid accumulation and enhanced oxygen consumption and UCP1 protein expression, while inhibition of AMPK by dorsomorphin (5 μM) significantly reversed these effects.
Conclusion: ALA improves estrogen deficiency-induced obesity via browning of WAT through AMPK signaling.

Keywords: Thioctic acid, Menopause, Obesity, Adipose tissue browning, AMP-activated protein kinases

Menopause, the loss of ovary function, results in the increased prevalence of obesity and metabolic syndromes including insulin resistance, type 2 diabetes mellitus, and cardiovascular diseases.1 Despite estrogen exerting beneficial effects for menopause-induced metabolic abnormalities, long-term hormonal replacement therapy imparts risk of breast and cervical cancer and is not the primary strategy.2 Dietary supplementation for counteracting obesity and metabolic risks requires further investigation for the management of postmenopausal health.

In mammals, three types of adipose tissue have been identified based on their functions and characteristics: white adipose tissue (WAT), brown adipose tissue (BAT), and beige adipose tissue.3 WAT stores energy in the form of triglyceride (TG) and regulates whole-body energy homeostasis, while BAT dissipates energy as heat through uncoupling protein 1 (UCP1) and produces thermogenesis capacity.4 Recently, the recruitment of beige adipocytes has been observed in WAT in response to various stimuli such as cold exposure and pharmacological interventions.5 Several transcription factors have been reported to induce beige fat formation, including PR domain containing 16 (PRDM16), peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α), and cell death-inducing DNA fragmentation factor α-like effector A (CIDEA), as well as secretory mediators such as irisin and bone morphogenetic protein 7 (BMP7).6

AMP-activated protein kinase (AMPK) is a master regulator of energy homeostasis and is regulated by adenosine triphosphate (ATP)-depleting conditions such as physical exercise, ischemia, and glucose deprivation.7 AMPK activation is induced via phosphorylation of Thr172 by liver kinase B1 (LKB1) or Ca2+/calmodulin-dependent protein kinase kinases (CAMKII).8 Activation of AMPK results in phosphorylation and inhibition of acetyl-CoA carboxylase (ACC), which plays an important role in fatty acid (FA) synthesis to promote FA oxidation and reduce fat accumulation.9 In addition to its metabolic effects, AMPK also regulates inflammatory cascades and alleviates chronic low-grade inflammatory diseases such as insulin resistance and atherosclerosis.10 It is reported that estradiol regulates BAT thermogenesis via hypothalamic AMPK to maintain energy homeostasis.11 Thus, pharmacological and nutritional strategies to induce AMPK signaling and promote WAT browning are promising approaches in obesity management.

Alpha-lipoic acid (ALA) is a naturally occurring antioxidant that acts as a cofactor of mitochondrial respiratory chain enzymes and plays an important role in the Krebs cycle phase of mitochondrial energy metabolism.12 It has been shown that ALA dose-dependently reduces body weight (BW) by increasing energy expenditure with the upregulation of Ucp1 mRNA in BAT, which might result from the suppression of hypothalamic AMPK activity.13 Recent investigation suggests that ALA induces mitochondria biogenesis and promotes the browning of WAT in human subcutaneous adipocytes.14 However, the beneficial effects of ALA in estrogen deficiency-induced obesity and its association with AMPK have not been fully established. Here, we investigated the anti-obesity effects of ALA in ovariectomy (Ovx) rats and explored whether AMPK signaling was involved in the regulation of WAT browning by ALA.

Ethical considerations

All experimental procedures conducted in the study complied with the accepted ethical guidelines of Animal Care and Use and the Animal Research: Reporting of In Vivo Experiments (ARRIVE) guidelines and with the principles for laboratory animal use following the Guide for the Care and Use of Laboratory Animals published by the U.S. National Institutes of Health (NIH Publication No. 85-23, revised in 1996). This study was approved by the Institutional Animal Care and Use Committee of the National Defense Medical Center, Taiwan (Permission No. IACUC-16-089).

Animal preparation

Female Wistar rats (8 weeks old, 250 to 270 g) were obtained from BioLASCO Taiwan Co. Ltd. Ovx in rodents is a useful model to mimic human menopause and has been shown to cause weight gain and fat accumulation.15 In short, rats were anesthetized with sodium pentobarbital (50 mg/kg, intraperitoneal injection) and subjected to bilateral Ovx. Small incisions were made bilaterally on the back sides to expose the ovaries. The ovaries were clamped and discarded and the uterine tubes were ligated, followed by the suturing of muscle and skin. The Sham operation consisted of anesthesia, visualization of the ovaries through incisions into the abdominal cavity, and closure of the wounds as described previously.16

Experimental groups

Rats were randomly divided into four groups: (1) Sham: rats were subjected to Sham operations (n=8); (2) Ovx: rats were ovariectomized bilaterally (n=11); (3) Ovx+ALA2: Ovx rats were administered ALA (200 mg/kg/day, gavage) (Sigma-Aldrich) beginning 1 week after operation (n=10). ALA was homogeneously suspended in 0.8% carboxymethylcellulose; and (4) Ovx+ALA3: rats were administered ALA (300 mg/kg/day, gavage) beginning 1 week after operation (n=6). After 8 weeks of treatment, rats were euthanized with pentobarbital (60 mg/kg, intraperitoneally); adipose tissues were dissected, weighed, immediately frozen in liquid nitrogen, and stored at −80 °C for further analysis.

Measurement of plasma levels of lipid profiles

Whole blood samples (1 mL) were collected by cardiac puncture and centrifuged at 12,000 g for 5 minutes at 4 °C. Plasma levels of TG, total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C) were detected by the Fuji DRI-CHEM 3030 analyzer (Fuji Photo Film).

Enzyme-linked immunosorbent assay for plasma irisin levels

Determination of plasma levels of irisin was performed using a commercial enzyme-linked immunosorbent assay (ELISA) kit (BioVision Inc.) according to the manufacturer’s instructions.

Histological analysis and immunohistochemistry staining

Browning of WAT mostly occurs in subcutaneous WAT depots; in rodents, the inguinal fat depot is normally used.17 Thus, inguinal WAT (iWAT) was dissected and fixed in 10% paraformaldehyde, embedded in paraffin blocks, sectioned, and serially stained with hematoxylin & eosin for histological analysis. For immunohistochemistry (IHC) staining, sections of iWAT were deparaffinized, boiled in antigen-retrieval solution, and treated with anti-UCP1 primary antibody (1:500, Abcam). The secondary antibody (1:1,000, anti-rabbit horseradish peroxidase [HRP]-conjugated) was incubated for 1 hour at room temperature. An enzymatic reaction was performed with 3,3'-diaminobenzidine tetrahydrochloride (HRP-DAB). The images were captured under a light microscope (Nikon) and photographed at ×200 magnification. Cell morphology and size were analyzed in 10 random fields per section using ImageJ software version 1.53b (National Institutes of Health).

Western blot analysis

Frozen iWAT was prepared in 200 μL of radioimmunoprecipitation assay buffer (Millipore), supplemented with 5% Protease Inhibitor Cocktail (Millipore) at 4 °C by bead homogenizer. Cellular debris was removed by centrifugation at 14,000 ×g for 20 minutes at 4 °C. The protein concentration of the supernatant was determined using a bicinchoninic acid assay kit (Thermo Scientific) following the manufacturer’s instructions. Aliquots (20 μg) of protein extract were loaded and separated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred to nitrocellulose membranes. The membranes were blocked with 5% bovine serum albumin in Tris‐buffered saline containing 0.1% Tween 20 (TBST) for 1 hour at room temperature. Primary antibodies against phosphorylated AMPK (p-AMPK), AMPK, phosphorylated ACC (p-ACC), ACC, LKB1 (1:1,000, Cell Signaling Technology), and β-actin (1:5,000, Cell Signaling Technology) and UCP1, PRDM16, and CIDEA (1:1,000, Abcam) were incubated at 4 °C overnight. The membranes were then probed with corresponding secondary antibodies conjugated with HRP (1:3,000, Cell Signaling Technology). The density of the individual protein bands was quantified by densitometric scanning using ImageJ software version 1.53b.

Cell culture and differentiation

3T3-L1 mouse embryo fibroblasts (American Type Culture Collection [ATCC]) were cultured as previously described.16 Cell differentiation was induced by the addition of differentiation cocktail, including 10 μg/mL insulin, 1 μM dexamethasone, and 0.5 mM 3-isobutyl-1-methylxanthine (IBMX). After incubation for 3 days, the culture medium was replaced with fresh Dulbecco’s Modified Eagle’s Medium containing 10% fetal bovine serum and insulin (10 μg/mL) every 3 days. The cells were fully differentiated into mature adipocytes on day 9. During the differentiation process (day 0 to 9), cells were treated with ALA (100 μM) in the presence or absence of AMPK inhibitor dorsomorphin (DOR; 5 μM).

Cell viability

Cell viability was measured by a cell counting kit-8 (CCK-8) assay following the manufacturer’s instructions. Briefly, 104 cells/well were placed into 96-well plates and treated with various concentrations of ALA (0, 50, 100, 250, and 400 μM) for 9 days. Cell viability was determined by colorimetry using the CCK-8 assay. Insoluble formazan crystals were dissolved in dimethyl sulfoxide, and the absorbance was measured at 490 nm.

Oil Red O staining

At the end of differentiation, cells were stained by Oil Red O (Sigma-Aldrich) for determination of lipid accumulation and visualized using bright-field microscopy. Oil droplets were eluted by administration with 100% isopropanol, and optical density was measured using a spectrophotometer at 520 nm.

Measurement of oxygen consumption

The rate of oxygen consumption (OCR) was determined using the CLARIOstar Plus microplate reader (BMG Labtech). Cells were plated in culture plates at a density of 2×103 cells. OCR was measured after the addition of a phosphorescent oxygen probe and mineral oil (Cayman Chemical).

Statistical analysis

Statistical analysis was performed with GraphPad Prism version 8 Software (GraphPad). These data are presented as mean±standard error of the mean. A two-way analysis of variance (ANOVA) and least significant difference post hoc comparison test were used to compare the means of BW and food intake by group and time. Statistical evaluation of other data was performed with one-way ANOVA followed by the Newman-Keuls comparison method. Normality of the data was analyzed by the Shapiro-Wilk method. Differences with P<0.05 were considered statistically significant.

Availability of data and materials

The datasets generated and/or analyzed during the current study are available in the figshare repository (https://doi.org/10.6084/m9.figshare.14233997).

Effects of ALA on BW, adiposity, and food intake in Ovx rats

Nine weeks after Ovx, the BW of the Ovx group (374.3±8.2 g) was significantly increased compared to that of the Sham group (296.6±1.21 g) (P<0.05). Interestingly, the Ovx+ALA2 (333.1±7.4 g) and Ovx+ALA3 (309.3±3.7 g) groups showed significantly lower BW than those of the Ovx group (P<0.05) (Fig. 1A). A similar result was observed in BW gain (Fig. 1B). Fig. 1C shows that food intake was not significantly affected during 8 weeks of ALA treatment. Total visceral fat pad mass consists of peri-renal, retroperitoneal, and mesenteric WAT weight. Fig. 1D shows a significant increase in total visceral fat pad in the Ovx group (19.0±1.1 g) compared with the Sham group (11.2±1.4 g) (P<0.05). The increase in visceral fat pad mass in Ovx rats was not observed in the Ovx+ALA2 (16.0±1.7 g) and Ovx+ALA3 (7.9±0.6 g) groups (P<0.05), indicating that ALA drastically attenuated fat pad mass in a dose-dependent manner.

Effects of ALA treatment in plasma lipid profiles of Ovx rats

We next evaluated the effects of ALA on the lipid profiles of Ovx rats. As shown in Fig. 1E, plasma levels of TG in the Ovx group were significantly increased compared to the Sham group (P<0.05). The increase in plasma TG in Ovx rats was markedly attenuated by the administration of ALA 300 mg/kg (P<0.05) in a dose-dependent manner. Moreover, ALA supplementation markedly decreased Ovx-induced elevation of TC and LDL-C (P<0.05) (Fig. 1F and G). However, HDL-C was drastically elevated by ALA treatment in Ovx rats (P<0.05) (Fig. 1H). These results indicate that ALA has beneficial effects on lipid profiles.

Effects of ALA treatment on adipocyte size and UCP1 protein expression in iWAT of Ovx rats

Assessment of adipocyte cross-sectional area in iWAT showed that Ovx resulted in an increase in adipocyte size, and the administration of ALA reduced adipocyte hypertrophy and increased the appearance of small multilocular adipocytes (P<0.05) (Fig. 2A and B). Subsequently, IHC staining of UCP1 showed enhanced UCP1 expression in both ALA2- and ALA3-treated Ovx rats (Fig. 2C), suggesting that ALA-induced browning of iWAT produces thermogenic beige adipocytes.

ALA increased biomarkers of beige fat development in iWAT of Ovx rats

To further examine whether ALA-induced beige fat formation, we assessed the protein expression of beige fat-specific biomarkers. Western blot analysis showed that UCP1 protein expression was significantly lower in iWAT of the Ovx group than that of the Sham group (Fig. 3A). ALA treatment significantly upregulated UCP1 protein expression compared to the Ovx group (P<0.05). In addition, transcription factors that regulate the trans-differentiation of white into beige adipocytes, including PRDM16 (Fig. 3B) and CIDEA (Fig. 3C), were significantly upregulated in the Ovx+ALA2 and Ovx+ALA3 groups compared to the Ovx group. Collectively, these results indicate the potential of ALA in the conversion of white to beige fat in iWAT of Ovx rats.

Effects of ALA treatment on plasma levels of irisin in Ovx rats

Irisin is a myokine secreted from skeletal muscle to promote energy expenditure through UCP1-mediated thermogenesis and induction of WAT browning.18,19 We investigated whether ALA treatment affected the level of circulating irisin in Ovx rats. As shown in Fig. 4A, plasma irisin levels were significantly decreased in the Ovx group compared to the Sham group (P<0.05) Additionally, ALA treatment brought about a significant increase in plasma irisin levels in Ovx rats, suggesting that ALA-induced myokine secretion promotes WAT browning.

ALA-induced the activation of AMPK signaling in iWAT of Ovx rats

AMPK is a master energy switch regulator and plays an important role in mediating the browning of WAT.20 We examined whether ALA treatment affected phosphorylation levels of AMPK in iWAT of Ovx rats. As shown in Fig. 4B, the expression of p-AMPK/AMPK was significantly lower in the Ovx group than in the Sham group. ALA supplementation markedly increased the p-AMPK/AMPK ratio in Ovx rats (P<0.05). It is well established that AMPK activation phosphorylates and inhibits ACC, an essential enzyme in promoting FA oxidation and reducing fat accumulation.21 Fig. 4C further indicates that the p-ACC/ACC ratio was dramatically increased by ALA administration in Ovx rats. Moreover, AMPK is mainly activated by phosphorylation of the α subunit at Thr172 by LKB1 under energy stress.8 Intriguingly, ALA markedly increased the expression of LKB1 in Ovx rats (Fig. 4D). These results suggest that ALA induces activation of the LKB1-AMPK-ACC axis to mediate the browning of WAT.

Effects of ALA administration on lipid accumulation, OCR, and UCP1 protein expression in 3T3-L1 cells

Cell viability of ALA is shown in Fig. 5A. No significant cytotoxicity of ALA was observed below the concentration of 250 μM. Thus, 100 and 250 μM of ALA were assessed on lipid droplet accumulation using Oil Red O stain. Results indicated that both ALA 100 and ALA 250 groups had significantly reduced lipid accumulation in adipocytes (Fig. 5B). Furthermore, ALA (100 μM) treatment markedly increased OCR, and the addition of AMPK inhibitor DOR (5 μM) significantly reversed the increase of OCR by ALA (Fig. 5C). As shown in Fig. 5D, UCP1 protein expression in the ALA group was significantly higher than that of the control (CTL) group. In the ALA+DOR group, the increase of UCP1 expression was significantly reduced compared to that of ALA, indicating that AMPK plays an important role in mediating the metabolic effects of ALA.

In the present study, we showed that administration of ALA significantly suppresses BW gain and fat pad mass in Ovx rats. We also found that plasma levels of TG, TC, and LDL-C were higher and HDL-C was lower in Ovx rats, but these detrimental effects were improved by ALA supplementation. In addition, ALA treatment reduced adipocyte size and exhibited unique staining of UCP1 of iWAT. Myokine irisin secretion as well as expression of thermogenic proteins such as UCP1, PRDM16, and CIDEA were upregulated by ALA, suggesting that ALA exerts browning capacities. We further demonstrated that ALA treatment increased LKB1 and stimulated the phosphorylation of AMPK and its downstream target ACC, revealing that the beneficial effects were mediated at least in part by activation of the LKB1/AMPK pathway. Intriguingly, inhibition of AMPK reversed the induction of UCP1 caused by ALA in 3T3-L1 cells. To the best of our knowledge, this is the first study to examine the effects of ALA on estrogen deficiency-induced metabolic alterations and WAT browning via activation of LKB1/AMPK.

It is well established that oxidative stress during menopause can aggravate the inflammatory status that contributes to the detrimental effects of obesity and metabolic syndromes.15 Ovx suppresses expression of the mitochondrial proteins involved in ATP synthesis and thermogenic UCP1 in BAT and subcutaneous WAT. This contributes to impairment of mitochondrial function and heat generation, reducing energy expenditure.22 Estrogen exerts antioxidant and anti-inflammatory effects in the modulation of insulin action.23 Moreover, reduced energy expenditure during the menopause transition may predispose to obesity and dyslipidemia.24 Thus, antioxidant treatment has been proposed as a therapeutic strategy to prevent obesity and associated comorbidities.25 In line with our report, Delgobo et al.26 showed that ALA improved antioxidant defenses and alleviated oxidative stress, inflammation, and lipid profiles in estrogen-independent mechanisms. The enhancement of energy expenditure in BAT and the trans-differentiation of white to beige adipocytes are gaining attention for their potential to prevent the metabolic complications of obesity.27 The development of beige adipocytes in WAT enhances energy expenditure and switches adipocytes from an energy storage state to an energy dissipation state via induction of UCP14 to reduce the risk of metabolic diseases. We demonstrated that major transcriptional regulators of WAT browning such as PRDM16, PGC-1α, and CIDEA were upregulated by ALA (Fig. 3). PGC-1α promotes UCP1 to activate thermogenesis in these two distinct types of adipocytes,28 while PRDM16 plays a critical role during the differentiation of BAT and trans-differentiation of white into beige adipocytes. Furthermore, irisin is secreted as a myokine and an adipokine into circulation following the cleavage of fibronectin type III domain-containing protein 5 (FNDC5) to induce the browning of WAT.29 Emerging evidence supports the weak affect of irisin on classical BAT isolated from the interscapular depot, suggesting that activation of the thermogenic program in response to irisin is a selective characteristic of beige cells.30 We found that Ovx decreased the browning of iWAT and irisin secretion, leading to weight gain after estrogen depletion (Fig. 4A), in line with our previous study.5 Sul et al.31 further indicated that exogenous administration of E2 enhanced browning in vivo and in vitro, which might result from the antioxidant activity of E2. ALA supplementation increased the plasma levels of irisin, suggesting that the browning effects of ALA are partly mediated via irisin production. The lower weight gain in ALA-treated Ovx rats was consistent with a previous report, which showed that the anti-obesity effects of ALA are due to the enhancement of energy expenditure.13 ALA supplementation reduced fat depots and the appearance of multilocular adipocytes within the iWAT, which may be attributed to the upregulation of lipolysis32 and the downregulation of adipogenesis.33

Estrogen deficiency-induced oxidative stress alters lipid metabolism, resulting in excessive TG, TC, and LDL-C accumulation, which are common risk factors for the pathological progression of metabolic disorders.34 The current study showed that ALA treatment reduced TG, TC, and LDL-C and increased HDL-C levels in Ovx rats. These results were in accordance with a previous report showing that ALA reduced TG accumulation in skeletal muscles, pancreatic islets, and adipose tissue in diabetes-prone obese rats and increased FA oxidation.35 The improvement in lipid profiles by ALA may result from its antioxidative and anti-inflammatory properties.36 Moreover, BAT and beige adipocytes generate heat using TG-derived FA from the circulation in a UCP1-dependent process, leading to improvement of lipid profiles and prevention of atherosclerosis progression.37 Thus, the beneficial effects of ALA in lowering TG and cholesterol might be a consequence of the induction of browning in iWAT.

Activation of AMPK inhibits the synthesis of FA and enhances its oxidation by phosphorylation of its downstream target ACC,38 which directs FA toward degradation. Inhibition of AMPK activity in WAT has been widely observed in obese and diabetic rodents as well as in human subjects with insulin resistance.39 Deletion of AMPKα1 impaired the thermogenic program in BAT, possibly due to the defective unwinding of the DNA of the promoter region of the PRDM16,40 suggesting that AMPK coordinates thermogenic BAT and beige adipocyte functions. In the present study, we showed that Ovx has inhibitory effects in AMPK signaling cascades, spanning from the upstream LKB1 to the downstream ACC, whereas ALA treatment reverses these changes.

As depicted in Fig. 6, ALA ameliorated E2 deficiency-induced obesity and metabolic disorders via the reprogramming of white to thermogenic beige adipocytes. Activation of AMPK may contribute to the beneficial effects of ALA. Thus, ALA provides a potential preventive strategy for the improvement of metabolic dysfunctions after menopause.

The work had a preprint posting in Research Square (DOI: 10. 21203/rs.3.rs-338317/v1) and was licensed under a Creative Commons Attribution 4.0 International License. This work was supported by research grants from the Ministry of National Defense (MAB-109-006 and MAB-109-007), Kaohsiung Armed Forces General Hospital (109-051), and Chi-Mei Hospital (CMNDMC10910), Republic of China, Taiwan.

Study concept and design: YWC; acquisition of data: YWC and TP; analysis and interpretation of data: YWC and TP; drafting of the manuscript: SYH and MTC; critical revision of the manuscript: HHS and YML; statistical analysis: YWC; obtained funding: SYH and MTC; administrative, technical, or material support: CWK, SYC, and PYC; and study supervision: HHS and YML.

Fig. 1. Effects of treatment with ALA on body weight and fat pad mass in ovariectomized rats. (A) Changes of body weight; (B) body weight gain; (C) food intake; (D) total visceral fat pad mass; plasma levels of (E) triglycerides, (F) total cholesterol, (G) low-density lipoprotein cholesterol (LDL-C), and (H) high-density lipoprotein cholesterol (HDL-C) in ovariectomized rats. Data are expressed as mean±standard error of the mean. *P<0.05 vs. Sham; P<0.05 vs. ovariectomy (Ovx); P<0.05 vs. Ovx+ALA2 (n=6–11). ALA2, alpha-lipoic acid (200 mg/kg/day, gavage); ALA3, alphalipoic acid (300 mg/kg/day, gavage) for 8 weeks.
Fig. 2. Effects of treatment with ALA on adipocyte size and uncoupling protein 1 (UCP1) expression in inguinal white adipose tissue of ovariectomized rats. (A) Representative photomicrographs of cross sections using hematoxylin & eosin (H&E) and (B) the quantification of adipocyte size; (C) representative staining for UCP1. The images were captured at 200× magnification. Arrows indicate UCP1 protein expression. Data are expressed as mean±standard error of the mean. *P<0.05 vs. Sham; P<0.05 vs. ovariectomy (Ovx) (n=6–11). ALA2, alpha-lipoic acid treatment (200 mg/kg/day, gavage); ALA3, alpha-lipoic acid (300 mg/kg/day, gavage) for 8 weeks.
Fig. 3. Effects of treatment with ALA on the markers of white adipose tissue (WAT) browning in inguinal WAT of ovariectomized rats. Representative protein expression and quantification of (A) uncoupling protein 1 (UCP1), (B) PR domain containing 16 (PRDM16), and (C) cell death-inducing DNA fragmentation factor α-like effector A (CIDEA). Data are expressed as mean±standard error of the mean. *P<0.05 vs. Sham; P<0.05 vs. ovariectomy (Ovx) (n=6–11). ALA2, alpha-lipoic acid treatment (200 mg/kg/day, gavage); ALA3, alpha-lipoic acid (300 mg/kg/day, gavage) for 8 weeks.
Fig. 4. Effects of treatment with ALA on (A) plasma levels of irisin and protein expression and quantification of (B) phosphorylated AMP-activated protein kinase (p-AMPK), (C) phosphorylated acetyl-CoA carboxylase (p-ACC), and (D) liver kinase B1 (LKB1) in inguinal white adipose tissue of ovariectomized rats. Data are expressed as mean±standard error of the mean. *P<0.05 vs. Sham; P<0.05 vs. ovariectomy (Ovx) (n=6–11). ALA2, alpha-lipoic acid treatment (200 mg/kg/day, gavage); ALA3, alpha-lipoic acid (300 mg/kg/day, gavage) for 8 weeks.
Fig. 5. Effects of treatment with alpha-lipoic acid (ALA) on lipid accumulation and uncoupling protein 1 (UCP1) protein expression in 3T3-L1 cells. (A) Cell viability of 3T3-L1 cells treated with ALA (0 to 400 μM); (B) lipid accumulation in differentiated 3T3-L1 cells stained by Oil Red O; (C) oxygen consumption; and (D) representative protein expression and quantification of UCP1 in ALA (100 μM) and cotreatment of ALA with dorsomorphin (DOR; 5 μM, an AMP-activated protein kinase inhibitor) in 3T3-L1 cells. *P<0.05 vs. control (CTL); P<0.05 vs. ALA (n=4). ME, 0.1 % methanol, the vehicle of ALA.
Fig. 6. Schematic depiction showing that alpha-lipoic acid (ALA) activates AMP-activated protein kinase (AMPK) signaling and irisin secretion with concurrent induction of uncoupling protein 1 (UCP1), PR domain containing 16 (PRDM16), and cell death-inducing DNA fragmentation factor α-like effector A (CIDEA) to coordinate the conversion of white adipocytes to the beige phenotype, leading to improvement in estrogen deficiency-induced obesity.
  1. Carr MC. The emergence of the metabolic syndrome with menopause. J Clin Endocrinol Metab 2003;88:2404-11.
    Pubmed CrossRef
  2. Stubbins RE, Holcomb VB, Hong J, Núñez NP. Estrogen modulates abdominal adiposity and protects female mice from obesity and impaired glucose tolerance. Eur J Nutr 2012;51:861-70.
    Pubmed CrossRef
  3. Giralt M, Villarroya F. White, brown, beige/brite: different adipose cells for different functions?. Endocrinology 2013;154:2992-3000.
    Pubmed CrossRef
  4. Harms M, Seale P. Brown and beige fat: development, function and therapeutic potential. Nat Med 2013;19:1252-63.
    Pubmed CrossRef
  5. Shen HH, Huang SY, Kung CW, Chen SY, Chen YF, Cheng PY, et al. Genistein ameliorated obesity accompanied with adipose tissue browning and attenuation of hepatic lipogenesis in ovariectomized rats with high-fat diet. J Nutr Biochem 2019;67:111-22.
    Pubmed CrossRef
  6. Rosen ED, Spiegelman BM. What we talk about when we talk about fat. Cell 2014;156:20-44.
    Pubmed KoreaMed CrossRef
  7. Hardie DG. AMPK: a key regulator of energy balance in the single cell and the whole organism. Int J Obes (Lond) 2008;32 Suppl 4:S7-12.
    Pubmed CrossRef
  8. Woods A, Johnstone SR, Dickerson K, Leiper FC, Fryer LG, Neumann D, et al. LKB1 is the upstream kinase in the AMP-activated protein kinase cascade. Curr Biol 2003;13:2004-8.
    Pubmed CrossRef
  9. Fullerton MD, Galic S, Marcinko K, Sikkema S, Pulinilkunnil T, Chen ZP, et al. Single phosphorylation sites in Acc1 and Acc2 regulate lipid homeostasis and the insulin-sensitizing effects of metformin. Nat Med 2013;19:1649-54.
    Pubmed KoreaMed CrossRef
  10. Day EA, Ford RJ, Steinberg GR. AMPK as a therapeutic target for treating metabolic diseases. Trends Endocrinol Metab 2017;28:545-60.
    Pubmed CrossRef
  11. Martínez de Morentin PB, González-García I, Martins L, Lage R, Fernández-Mallo D, Martínez-Sánchez N, et al. Estradiol regulates brown adipose tissue thermogenesis via hypothalamic AMPK. Cell Metab 2014;20:41-53.
    Pubmed KoreaMed CrossRef
  12. Li N, Yan W, Hu X, Huang Y, Wang F, Zhang W, et al. Effects of oral α-lipoic acid administration on body weight in overweight or obese subjects: a crossover randomized, double-blind, placebo-controlled trial. Clin Endocrinol (Oxf) 2017;86:680-7.
    Pubmed CrossRef
  13. Kim MS, Park JY, Namkoong C, Jang PG, Ryu JW, Song HS, et al. Anti-obesity effects of alpha-lipoic acid mediated by suppression of hypothalamic AMP-activated protein kinase. Nat Med 2004;10:727-33.
    Pubmed CrossRef
  14. Fernández-Galilea M, Pérez-Matute P, Prieto-Hontoria PL, Houssier M, Burrell MA, Langin D, et al. α-Lipoic acid treatment increases mitochondrial biogenesis and promotes beige adipose features in subcutaneous adipocytes from overweight/obese subjects. Biochim Biophys Acta 2015;1851:273-81.
    Pubmed CrossRef
  15. Lizcano F, Guzmán G. Estrogen deficiency and the origin of obesity during menopause. Biomed Res Int 2014;2014:757461.
    Pubmed KoreaMed CrossRef
  16. Shen HH, Yang CY, Kung CW, Chen SY, Wu HM, Cheng PY, et al. Raloxifene inhibits adipose tissue inflammation and adipogenesis through Wnt regulation in ovariectomized rats and 3 T3-L1 cells. J Biomed Sci 2019;26:62.
    Pubmed KoreaMed CrossRef
  17. Nedergaard J, Cannon B. The browning of white adipose tissue: some burning issues. Cell Metab 2014;20:396-407.
    Pubmed CrossRef
  18. Boström P, Wu J, Jedrychowski MP, Korde A, Ye L, Lo JC, et al. A PGC1-α-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature 2012;481:463-8.
    Pubmed KoreaMed CrossRef
  19. Roca-Rivada A, Castelao C, Senin LL, Landrove MO, Baltar J, Belén Crujeiras A, et al. FNDC5/irisin is not only a myokine but also an adipokine. PLoS One 2013;8:e60563.
    Pubmed KoreaMed CrossRef
  20. O'Neill HM, Holloway GP, Steinberg GR. AMPK regulation of fatty acid metabolism and mitochondrial biogenesis: implications for obesity. Mol Cell Endocrinol 2013;366:135-51.
    Pubmed CrossRef
  21. Saha AK, Ruderman NB. Malonyl-CoA and AMP-activated protein kinase: an expanding partnership. Mol Cell Biochem 2003;253:65-70.
    Pubmed CrossRef
  22. Gupte AA, Pownall HJ, Hamilton DJ. Estrogen: an emerging regulator of insulin action and mitochondrial function. J Diabetes Res 2015;2015:916585.
    Pubmed KoreaMed CrossRef
  23. Ribas V, Nguyen MT, Henstridge DC, Nguyen AK, Beaven SW, Watt MJ, et al. Impaired oxidative metabolism and inflammation are associated with insulin resistance in ERalpha-deficient mice. Am J Physiol Endocrinol Metab 2010;298:E304-19.
    Pubmed KoreaMed CrossRef
  24. Lovejoy JC, Champagne CM, de Jonge L, Xie H, Smith SR. Increased visceral fat and decreased energy expenditure during the menopausal transition. Int J Obes (Lond) 2008;32:949-58.
    Pubmed KoreaMed CrossRef
  25. Zhang S, Xu M, Zhang W, Liu C, Chen S. Natural polyphenols in metabolic syndrome: protective mechanisms and clinical applications. Int J Mol Sci 2021;22:6110.
    Pubmed KoreaMed CrossRef
  26. Delgobo M, Agnes JP, Gonçalves RM, Dos Santos VW, Parisotto EB, Zamoner A, et al. N-acetylcysteine and alpha-lipoic acid improve antioxidant defenses and decrease oxidative stress, inflammation and serum lipid levels in ovariectomized rats via estrogen-independent mechanisms. J Nutr Biochem 2019;67:190-200.
    Pubmed CrossRef
  27. Bartelt A, Heeren J. Adipose tissue browning and metabolic health. Nat Rev Endocrinol 2014;10:24-36.
    Pubmed CrossRef
  28. Barbera MJ, Schluter A, Pedraza N, Iglesias R, Villarroya F, Giralt M. Peroxisome proliferator-activated receptor alpha activates transcription of the brown fat uncoupling protein-1 gene: a link between regulation of the thermogenic and lipid oxidation pathways in the brown fat cell. J Biol Chem 2001;276:1486-93.
    Pubmed CrossRef
  29. Zhang Y, Li R, Meng Y, Li S, Donelan W, Zhao Y, et al. Irisin stimulates browning of white adipocytes through mitogen-activated protein kinase p38 MAP kinase and ERK MAP kinase signaling. Diabetes 2014;63:514-25.
    Pubmed CrossRef
  30. Wu J, Boström P, Sparks LM, Ye L, Choi JH, Giang AH, et al. Beige adipocytes are a distinct type of thermogenic fat cell in mouse and human. Cell 2012;150:366-76.
    Pubmed KoreaMed CrossRef
  31. Sul OJ, Hyun HJ, Rajasekaran M, Suh JH, Choi HS. Estrogen enhances browning in adipose tissue by M2 macrophage polarization via heme oxygenase-1. J Cell Physiol 2021;236:1875-88.
    Pubmed CrossRef
  32. Fernández-Galilea M, Pérez-Matute P, Prieto-Hontoria PL, Martinez JA, Moreno-Aliaga MJ. Effects of lipoic acid on lipolysis in 3T3-L1 adipocytes. J Lipid Res 2012;53:2296-306.
    Pubmed KoreaMed CrossRef
  33. Hahm JR, Noh HS, Ha JH, Roh GS, Kim DR. Alpha-lipoic acid attenuates adipocyte differentiation and lipid accumulation in 3T3-L1 cells via AMPK-dependent autophagy. Life Sci 2014;100:125-32.
    Pubmed CrossRef
  34. Signorelli SS, Neri S, Sciacchitano S, Pino LD, Costa MP, Marchese G, et al. Behaviour of some indicators of oxidative stress in postmenopausal and fertile women. Maturitas 2006;53:77-82.
    Pubmed CrossRef
  35. Lee WJ, Song KH, Koh EH, Won JC, Kim HS, Park HS, et al. Alpha-lipoic acid increases insulin sensitivity by activating AMPK in skeletal muscle. Biochem Biophys Res Commun 2005;332:885-91.
    Pubmed CrossRef
  36. Zhang Y, Han P, Wu N, He B, Lu Y, Li S, et al. Amelioration of lipid abnormalities by α-lipoic acid through antioxidative and anti-inflammatory effects. Obesity (Silver Spring) 2011;19:1647-53.
    Pubmed CrossRef
  37. Khedoe PP, Hoeke G, Kooijman S, Dijk W, Buijs JT, Kersten S, et al. Brown adipose tissue takes up plasma triglycerides mostly after lipolysis. J Lipid Res 2015;56:51-9.
    Pubmed KoreaMed CrossRef
  38. Janovská A, Hatzinikolas G, Staikopoulos V, McInerney J, Mano M, Wittert GA. AMPK and ACC phosphorylation: effect of leptin, muscle fibre type and obesity. Mol Cell Endocrinol 2008;284:1-10.
    Pubmed CrossRef
  39. Xu XJ, Gauthier MS, Hess DT, Apovian CM, Cacicedo JM, Gokce N, et al. Insulin sensitive and resistant obesity in humans: AMPK activity, oxidative stress, and depot-specific changes in gene expression in adipose tissue. J Lipid Res 2012;53:792-801.
    Pubmed KoreaMed CrossRef
  40. Yang Q, Liang X, Sun X, Zhang L, Fu X, Rogers CJ, et al. AMPK/α-ketoglutarate axis dynamically mediates DNA demethylation in the Prdm16 promoter and brown adipogenesis. Cell Metab 2016;24:542-54.
    Pubmed KoreaMed CrossRef