DOI QR코드

DOI QR Code

Caffeine attenuates lipid accumulation via activation of AMP-activated protein kinase signaling pathway in HepG2 cells

  • Quan, Hai Yan (Department of Pharmacology and Clinical Pharmacy, College of Pharmacy, Kyung Hee University) ;
  • Kim, Do Yeon (Department of Pharmacology and Clinical Pharmacy, College of Pharmacy, Kyung Hee University) ;
  • Chung, Sung Hyun (Department of Pharmacology and Clinical Pharmacy, College of Pharmacy, Kyung Hee University)
  • Received : 2012.07.27
  • Accepted : 2012.10.22
  • Published : 2013.04.30

Abstract

The main purpose of this study is to examine the effect of caffeine on lipid accumulation in human hepatoma HepG2 cells. Significant decreases in the accumulation of hepatic lipids, such as triglyceride (TG), and cholesterol were observed when HepG2 cells were treated with caffeine as indicated. Caffeine decreased the mRNA level of lipogenesis-associated genes (SREBP1c, SREBP2, FAS, SCD1, HMGR and LDLR). In contrast, mRNA level of CD36, which is responsible for lipid uptake and catabolism, was increased. Next, the effect of caffeine on AMP-activated protein kinase (AMPK) signaling pathway was examined. Phosphorylation of AMPK and acetyl-CoA carboxylase were evidently increased when the cells were treated with caffeine as indicated for 24 h. These effects were all reversed in the presence of compound C, an AMPK inhibitor. In summary, these data indicate that caffeine effectively depleted TG and cholesterol levels by inhibition of lipogenesis and stimulation of lipolysis through modulating AMPK-SREBP signaling pathways.

Keywords

INTRODUCTION

The liver plays an integral role in the coordination of fuel homeostasis since it is the major site for storage and release of glucose and lipid. Accumulations of lipid within liver are closely linked with the development of insulin resistance and non-alcoholic fatty liver disease and are subject to nutritional influence (1). AMP-activated protein kinase (AMPK) belongs to the serine/threonine kinase family and is a key enzyme that regulates glucose and lipid metabolism in the liver. AMPK is classically activated by an increase in the intracellular AMP:ATP ratio, and phosphorylates a wide range of downstream targets, having the predominant effect of stimulating energy-producing pathways (fatty acid oxidation and glucose utilization), and inhibiting energy-consuming pathways (lipogenesis and gluconeogenesis) (2). In the liver, activation of AMPK phosphorylates and inactivates the rate-limiting enzymes of lipogenesis, such as acetyl-CoA carboxylase (ACC) (3). It is well documented that AMPK phosphorylation inhibits sterol regulatory element-binding protein-1 (SREBP-1), the key transcription factor responsible for fatty acid synthesis (4). Conversely, repressed AMPK activates anabolic pathways and inhibits catabolic pathways. In studies performed with hepatocytes and in the livers of ethanol-fed mice, You et al. demonstrated that the inhibition of AMPK leads to the activation of SREBP-1 mediated lipogenesis (5). AMPK positively regulated fatty acid oxidation by activating peroxisome proliferator-activated receptor-α (PPARα) and PPARγ coactivator (PGC)-1 (6). Thus, activation of AMPK by chemicals may contribute to the inhibition of intracellular accumulation of lipids.

Caffeine, a major ingredient found in a number of the most widely consumed non-alcoholic beverages worldwide (7,8), is highlighted as a potent dietary-component associated with reduced risk of several chronic disease, including type 2 diabetes and its complication (9,10). Several substances other than caffeine, e.g. chlorogenic acid and magnesium, have been suggested as being responsible for the protective effect of coffee in reducing the risk of type 2 diabetes (11). Although the inhibitory effects of caffeine alone or combination with other chemicals on intracellular lipid accumulation were reported in 3T3-L1 adipocytes (12) and in HepG2 cells (13), respectively, the effects of caffeine on triglyceride and cholesterol levels in liver cells have not been investigated yet. Here, we examined whether caffeine affects lipid metabolism via modulation of AMPK-SREBP signaling pathways in human hepatoma HepG2 cells.

 

RESULTS

Effects of caffeine on lipid levels and transcription factors in hepatocytes

To examine the anti-hyperlipidemic effects of caffeine, its effect on lipid contents in human hepatoma HepG2 cells was investigated. HepG2 cells were treated with caffeine for up to 24 h. Caffeine did not show any cellular toxicity up to 8 mM, but significant cytotoxicity was shown at > 25 mM (data not shown). Triglyceride and cholesterol levels were significantly decreased by caffeine in concentration-dependent manners. Compared to the basal level, triglyceride and cholesterol levels were decreased by 40% and 35%, respectively, upon treatment of cells with 4 mM caffeine (Fig. 1A and B). ER-bound SREBPs regulate lipid homeostasis by controlling the expression of genes required for cholesterol, triglyceride and phospholipid (14). Enzymatic triglyceride and cholesterol syntheses in the liver have been known to be under the control of SREBP1c and SREBP2 transcription factors, respectively. Therefore, we investigated the effects of caffeine on the gene expression of transcription factors responsible for lipid biosynthesis. As shown in Fig. 1C-E, caffeine markedly attenuated gene expression of SREBP1c in both time- and concentration- dependent manners. Lipogenic genes such as fatty acid synthase (FAS) and stearoyl-CoA desaturase 1 (SCD1), well known target enzymes of SREBP1c, were also marginally suppressed in time- and concentration-dependent manners. In contrast, gene expression of CD36, responsible for inducing lipid uptake and catabolism, was increased in a concentrationand time-dependent manner. Next, the effect of caffeine on gene expression of SREBP2 was examined. Caffeine also suppressed gene expression of SREBP2 in a time- and concentration- dependent manner (Fig. 1F-H). In addition, genes for hydroxymethyl glutaryl CoA reductase (HMGR) and low density lipoprotein receptor (LDLR), known target molecules of SREBP2, were both suppressed in time- and concentration- dependent manners. HMGR and LDLR are associated with the formation of cholesterol precursors and cholesterol uptake into cell, respectively. These results suggest that caffeine attenuates TG and cholesterol storage through the regulation of gene expression responsible for lipid metabolism in liver cells.

Fig. 1.Effects of caffeine on lipid accumulation and lipogenic or lipolytic gene expressions in HepG2 cells. HepG2 cells were treated with different concentrations (0.5-4 mM) of caffeine for 24 h. Triglyceride (TG) and cholesterol levels were measured (A, B). Each bar represents the mean ± SEM of triplicate determinations. *P < 0.05 compared to control (0 concentration in this figure). Cells were treated with 2 mM caffeine for the indicated times (C, F) or treated with different concentrations of caffeine for 24 h (D, G). (E and H) Bar graphs are densitometric determinants for panels (D and G) respectively. *P < 0.05, ***P < 0.001 compared to control.

Effects of caffeine on protein expression of AMPK, upstream kinase for SREBPs

Next, we investigated whether phosphorylation of AMPK is induced by caffeine since AMPK plays a key role in regulating carbohydrate and fat metabolism in the liver. According to studies of the fatty liver mice model, it is known that a high-fat diet reduces the basal activity of AMPK, suggesting that a certain level of AMPK basal activity may be necessary to prevent fat accumulation (15). Of further relevance is the role of AMPK in reducing cholesterol synthesis, which occurs by the direct inhibition of HMGR, a rate-limiting enzyme in hepatic cholesterol biosynthesis (16,17). As shown in Fig. 2, caffeine significantly induced AMPK and ACC phosphorylation in timeand concentration-dependent manners. These effects were reversed in the presence of compound C, a selective AMPK inhibitor (Fig. 3A and B). In addition, the caffeine induced-gene suppressions of SREBPs and their target molecules were also reversed by treatment with compound C (Fig. 3C and D). Caffeine-induced suppressions of TG and cholesterol levels in liver cells were also blunted when the cells were pretreated with compound C (Fig. 3E and F). These results strongly suggest that caffeine inhibits hepatic triglyceride and cholesterol levels via modulation of AMPK-SREBP signaling pathways.

 

DISCUSSION

Coffee is a complex mixture of more than a thousand substances, including caffeine (primary source), phenolic compounds (chlorogenic acid and quinides -primary source), minerals and vitamins (magnesium, potassium manganese, chromium, niacin), and fiber (18), and several of these coffee constituent have a possible role in lipid and glucose metabolism. Recently, Nakabayashi et al. reported that caffeine reduces the accumulation of lipid in murine 3T3-L1 adipocytes (12). In addition, Murosaki et al. reported that a combination of caffeine, arginine, soy isoflavones, and L-carnitine enhances both lipolysis and fatty acid oxidation in 3T3-L1 and HepG2 cell, and in KK mice (13). However, the specific lipid lowering effect of caffeine in the liver cell has not yet been reported. Here, we examined whether caffeine affects lipid metabolism in human hepatoma HepG2 cells. Fatty acids in the liver are esterified into triglyceride or oxidized to produce energy. Stores in the form of triglyceride are inert and harmless to the cells. However, an excess of triglyceride reduces insulin sensitivity (19) and is hydrolyzed back to fatty acyl-CoA at a rate that exceeds the oxidative needs of the cells, resulting in the un-oxidized excess being subject to more harmful pathways of fatty acid metabolism, such as de novo ceramide formation (20).

Fig. 2.Effects of caffeine on protein expressions of AMPK and ACC in HepG2 cells. HepG2 cells were treated with 2 mM caffeine for different times (A), or were treated with caffeine at the indicated concentrations for 24 h (B). After immunoblotting, total and phospho-protein levels of AMPK and ACC were identified by their specific antibodies. Actin was used as an internal control to evaluate the relative expression of protein. Bar graph (C) represents the densitometric determinant for panel (B). *P < 0.05, **P < 0.01 compared to control.

SREBPs are known as transcription factors that are conserved from fission yeast to man, and regulate the expression of genes required to maintain cellular lipid homeostasis. In mammals there are two SREBP genes, SREBP1 and 2. Most data suggest that the two SREBP1a and 1c primarily regulate fatty acid metabolism, and that SREBP2 is the main regulator of cholesterol metabolism (21). SREBP1c is the predominant isoform in most adult non-dividing metabolic tissues, such as liver and adipose. As an inhibitor of triglyceride and cholesterol accumulation in the liver cell (Fig. 1A and B), the effects of caffeine on gene expression of SREBP1c and 2 were examined in HepG2 cells. The expressions of both SREBPs and their target molecules were significantly suppressed or enhanced by caffeine (Fig. 1C-H). These data could result from alterations in the synthesis and/or uptake of fatty acids. It is well documented that AMPK phosphorylation inhibits SREBP-1 through the mammalian target of rapamycin (mTOR) and liver X receptor-α (LXRα) (4). Regulation of hepatic SREBPs in vivo is largely dependent on nutritional status. Under conditions of fasting, the activation of AMPK reduces lipogenesis in the liver by suppressing SREBPs activity. Conversely, activation of LXR increases SREBP expression under insulin-stimulated conditions and leads to hepatic lipogenesis. Thus, identifying pharmacological agents that inhibit the activity of LXR or stimulate AMPK activity in hepatocytes may provide effective treatment options for fatty liver disease. The effect of caffeine on phosphorylation of AMPK and ACC was examined. AMPK inhibits the accumulation of fat by modulating downstream-signaling components like ACC. ACC is a rate-controlling enzyme for the synthesis of malonyl-CoA, which is a critical precursor in the biosynthesis of fatty acids and a potent inhibitor of mitochondrial fatty acid oxidation (22,23). Activation and inhibition of AMPK and ACC activities were experimentally proven by enhancement of phosphorylated forms for both proteins, and these results were confirmed through the presence of the AMPK inhibitor, compound C (Fig. 3). We have not yet determined the mechanism through which caffeine activates the AMPK signaling pathway in HepG2 cells. The activation of AMPK by caffeine either directly or indirectly through modulation of the AMP:ATP ratio in mitochondria is a legitimate possibility, and deserves further investigation.

Fig. 3.Effects of compound C on AMPK protein, lipogenic and lipolytic gene expression in HepG2 cells. HepG2 cells were pretreated for 2 h with compound C, and were then treated with 2 mM caffeine for 24 h. The phosphorylation levels of AMPK and ACC were analyzed by immunoblotting using the corresponding antibodies (A), and densitometric analysis (B), and lipogenic or lipolytic genes were subjected to RT-PCR (C, D) as described in Materials and Methods. Hepatic TG and cholesterol contents were determined in the absence and presence of 10 μM compound C (E and F). *P < 0.05, **P < 0.01, ***P < 0.001 compared to control.

In conclusion, caffeine, a major component of coffee, plays a significant role in reducing hepatic lipid accumulation by modulating AMPK-SREBP signaling pathways. These results broaden our understanding of how caffeine shows anti-hyperlipidemic activity in the liver, and caffeine itself or caffeine-containing beverages could represent a promising dietary supplement to prevent fatty liver disease and hypercholesterolemia.

 

MATERIALS AND METHODS

Chemicals

Caffeine was purchased from Sigma (St. Louis, MO, USA) and triglyceride and cholesterol measuring kits were from ASAN Pharmaceutical (Seoul, Korea). Antibodies against AMPK, phospho-AMPK, ACC, phospho-ACC were from Cell Signaling Technology (Beverly, MA, USA) and anti-actin was from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Reverse transcriptase and Taq polymerase were supplied by Promega (Madison, WI, USA), and compound C was from Calbiochem (Darmstadt, Germany). Protein extraction, EASY-BLUE total RNA extraction and ECL-reagent kits were from Intron Biotechnology Inc. (Beverly, MA, USA) and the protein assay kit was from Bio-Rad (Hercules, CA, USA). Other reagents and chemicals were of analytical grade.

Cell culture and viability assay

Human hepatoma HepG2 cell line was purchased from Korean Cell Line Bank (Seoul, Korea). HepG2 cells were grown in DMEM (GibcoBL, Grand Island, NY, USA) supplemented with 10% fetal bovine serum and antibiotics (100 unit/ml penicillin and 100 μg/ml streptomycin). Cells were maintained at sub-confluent conditions in a humidified incubator at 37℃, with ambient oxygen and 5% CO2. For the cytotoxicity test, HepG2 cells were cultured in 96-well culture plates, and were treated with the indicated concentrations of caffeine for 24 h. The cytotoxicity of caffeine was determined by CellTiter 96 AQueous One solution Cell Proliferation Assay kit (Promega, Madison, WI, USA).

Determination of TG and cholesterol

TG and cholesterol levels were determined in cell lysates using a colorimetric assay, and were expressed as μg of lipid per mg of cellular protein. The levels of TG and cholesterol in cell lysates were measured according to the instructions of the manufacturer’s of InfinityTM TG/cholesterol reagents.

Western blot

Cells were washed with ice-cold phosphate buffed saline (PBS) and were lysed in a protein extraction kit. Insoluble protein was removed by centrifugation at 15,000 rpm for 20 min and soluble protein concentrations were measured using a Bio-Rad protein assay kit. Equal amounts of protein (40 μg/lane) were resolved by 8% SDS-polyacrylamide gel electrophoreses (SDS-PAGE) and transferred to polyvinylidene difluoride membranes (Millipore, Massachusetts, MA, USA), and hybridized with primary antibodies (diluted 1:1,000) overnight at 4℃. After incubation with a 1:2,000 dilution of horseradish peroxidase-conjugated secondary antibody for 2 h at room temperature, protein bands were detected using an enhance(diluted 1:1,000)d chemiluminescence Western blot detection kit (Amersham, Uppsala, Sweden), exposed to X-ray film, and then quantified by densitometric analysis.

RNA extraction and RT-PCR

Total RNAs from HepG2 cells were prepared by using EASY-BLUE total RNA extraction kit. mRNAs were reversely transcribed into cDNA, and were amplified with PCR and normalized to the endogenous GAPDH. The primers used in this experiment were as follows: SREBP1c sense 5'-GTGGCGGCTGCATTGAGAGTGAAG-, antisense 5'-AGGTACCCGAGGGCATCCGAGAAT-3'; SREBP2 sense 5'-CGCCACCTGCCCCTCTCCTTCC-3’, antisense 5'-TGCCCTGCCACCTATCCTCTCACG-3’; FAS sense 5'-CAAGAACTGCACGGAGGTGT-3', antisense 5'-AGCTGCCAGAGTCGGAGAAC-3'; SCD1 sense 5'-TTGCCAGCTCTAGCCTTTAAATTC-3’, antisense 5'-TCCTGGTAGCATTATTCAGTAGTT-3'; CD36 sense 5'-GGGCTATAGGGATCCATTTTTG-3', antisense 5'-CCTTTCAGATTAACGTCGGATTC-3'; HMGR sense 5'-TACCATGTCAGGGGTTACGTC-3’, antisense 5'-CAAGCCTAGAGACATAATCATC-3’; LDLR sense 5'-CCCCGCAGATCAAACCCCCACC-3’, antisense 5'-AGACCCCCAGGCAAAGGAGACGA-3’; GAPDH sense 5’-TCCACCACCCTGTTGCTGTA-3', antisense 5'-ACCACAGTCCATGCCATCAC-3'. PCR was performed at 95℃ for 30 sec, followed by 50℃ (FAS, HMGR), 52℃ (GAPDH), 56℃ (SCD1,CD36,LDLR,SREBP2) or 64℃ (SREBP1c) for 30 sec, and 72℃ for 1 min. The last cycle was followed by a final extension step at 72℃ for 10 min. The RT-PCR products were electrophoresed in 0.8% agarose gels under 100 V, and were stained with 0.5 μg/ml ethidium bromide. Scanning densitometry was performed with an i-MAXTM Gel Image Analysis System (Core-Bio, Seoul, Korea).

Statistical analysis

All data were expressed as a mean values ± standard error (S.E.) and differences between groups were analyzed using Student’s t-test. Mean values were considered significantly different when P < 0.05.

References

  1. Marchesini, G., Brizi, M., Bianchi, G., Tomassetti, S., Buqianesi, E., Lenzi, M., McCullough, A., Natale, S., Forlani, G. and Melchionda, N. (2001) Nonalcoholic fatty liver disease: a feature of the metabolic syndrome. Diabetes 50, 1884-1850.
  2. Hardie, D. G. (2008) AMPK: a key regulator of energy balance in the single cell and the whole organism. Int. J. Obes. 32, S7-S12. https://doi.org/10.1038/ijo.2008.116
  3. Hardie, D. G., Corton, J., Ching, Y. P., Davies, S. P. and Hawley, S. (1997) Regulation of lipid metabolism by the AMP-activated protein kinase. Biochem. Soc. Trans. 25, 1229-1231. https://doi.org/10.1042/bst0251229
  4. Porstmann, T., Santos, C. R., Griffiths, B., Cully, M., Wu, M., Leevers, S., Griffiths, J. R., Chung, Y. L. and Schulze, A. (2008) SREBP activity is regulated by mTORC1 and contributes to Akt-dependent cell growth. Cell Metab. 8, 224-236. https://doi.org/10.1016/j.cmet.2008.07.007
  5. You, M., Matsumoto, M., Pacold, C. M., Cho, W. K. and Crabb, D. W. (2004) The role of AMP-activated protein kinase in the action of ethanol in the liver. Gastroenterology 127, 1798-1808. https://doi.org/10.1053/j.gastro.2004.09.049
  6. Lee, W. J. (2006) AMPK activation increases fatty acid oxidation in skeletal muscle by activating PPARa and PGC-1. Biochem. Biophys. Res. Commun. 340, 281-295.
  7. Keijzers, G. B., De Galan, B. E., Tack, C. J. and Smits P. (2002) Caffeine can decrease insulin sensitivity in humans. Diabetes Care 25, 364-369. https://doi.org/10.2337/diacare.25.2.364
  8. Duffey, K. J. and Popkin, B. M. (2007) Shifts in patterns and consumption of beverages between 1965 and 2002. Obesity 15, 2739-2747. https://doi.org/10.1038/oby.2007.326
  9. Paynter, N. P., Yeh, H. C., Voutilainen, S., Schmidt, M. I., Heiss, G., Folsom, A. R., Brancati, F. L. and Kao, W. H. (2006) Coffee and sweetened beverage consumption and the risk of type 2 diabetes mellitus: the atherosclerosis risk in communities study. Am. J. Epidemiol. 164, 1075-1084. https://doi.org/10.1093/aje/kwj323
  10. Tuomilehto, J., Hu, G., Bidel, S., Lindstrom, J. and Jousilahti, P. (2004) Coffee consumption and risk of type 2 diabetes mellitus among middle-aged Finnish men and women. JAMA 291, 1213-1219. https://doi.org/10.1001/jama.291.10.1213
  11. McCarty, M. F. (2005) A chlorogenic acid-induced increase in GLP-1 production may mediate the impact of heavy coffee consumption on diabetes risk. Med. Hypotheses 64, 848-853. https://doi.org/10.1016/j.mehy.2004.03.037
  12. Nakabayashi, H., Hashimoto, T., Ashida, H., Nishiumi, S. and Kanazawa, K. (2008) Inhibitory effects of caffeine and its metabolites on intracellular lipid accumulation in murine 3T3-L1 adipocytes. Biofactors 34, 293-302. https://doi.org/10.1002/biof.5520340405
  13. Murosaki, S., Lee, T. R., Muroyama, K., Shin, E. S., Cho, S. Y., Yamamoto, Y. and Lee, S. J. (2007) A combination of caffeine, arginine, soy isoflavones, and L-carnitine enhances both lipolysis and fatty acid oxidation in 3T3-L1 and HepG2 cells in vitro and in KK mice in vivo. J. Nutr. 137, 2252-2257. https://doi.org/10.1093/jn/137.10.2252
  14. Eberle, D., Hegarty, B., Bossard, P., Ferré, P. and Foufelle, F. (2004) SREBP transcription factors: master regulators of lipid homeostasis. Biochimi. 86, 839-848. https://doi.org/10.1016/j.biochi.2004.09.018
  15. Kim, S. J., Jung, J. Y., Kim, H. W. and Park, T. (2008) Anti-obesity effects of Juniperus chinensis extract are associated with increased AMP-activated protein kinase expression and phosphorylation in the visceral adipose tissue of rats. Biol. Pharm. Bull. 31, 415-421.
  16. Ching, Y. P., Davies, S. P. and Hardie, D. G. (1996) Analysis of the specificity of the AMP-activated protein kinase by site-directed mutagenesis of bacterially expressed 3-hydroxy 3-methylglutaryl-CoA reductase, using a single primer variant of the unique-site-elimination method. Eur. J. Biochem. 237, 800-808. https://doi.org/10.1111/j.1432-1033.1996.0800p.x
  17. Pallottini, V., Montanari, L., Cavallini, G., Bergamini, E., Gori, Z. and Trentalance, A. (2004) Mechanisms underlying the impaired regulation of 3-hydroxy-3-methylglutaryl coenzyme A reductase in aged rat liver. Mech. Ageing Dev. 125, 633-639. https://doi.org/10.1016/j.mad.2004.08.001
  18. Department of Agriculture Agricultural Research Service: USDA. National Nutrient Database for Standard Reference (2007).
  19. Nagle, C. A., Klett, E. L. and Coleman, R. A. (2009) Hepatic triacylglycerol accumulation and insulin resistance. J. Lipid Res. 50, S74-79. https://doi.org/10.1194/jlr.R800053-JLR200
  20. Summers, S. A. (2006) Ceramide in insulin resistance and lipotoxicity. Prog. Lipid Res. 45, 42-72. https://doi.org/10.1016/j.plipres.2005.11.002
  21. Horton, J. D., Goldstein, J. L. and Brown, M. S. (2002) SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. J. Clin. Invest. 109, 1125-1131. https://doi.org/10.1172/JCI0215593
  22. Saha, A. K. and Ruderman, N. B. (2003) Malonyl-CoA and AMP-activated protein kinase: an expanding partnership. Mol. Cell Biochem. 253, 65-70. https://doi.org/10.1023/A:1026053302036
  23. Hardie, D. G. and Pan, D. A. (2002) Regulation of fatty acid synthesis and oxidation by the AMP-activated protein kinase. Biochem. Soc. Trans. 30, 1064-1070. https://doi.org/10.1042/bst0301064

Cited by

  1. Caffeine attenuates liver fibrosis via defective adhesion of hepatic stellate cells in cirrhotic model vol.28, pp.12, 2013, https://doi.org/10.1111/jgh.12317
  2. Caffeine reduces hepatic lipid accumulation through regulation of lipogenesis and ER stress in zebrafish larvae vol.22, pp.1, 2015, https://doi.org/10.1186/s12929-015-0206-3
  3. Citrus junos Tanaka peel ameliorates hepatic lipid accumulation in HepG2 cells and in mice fed a high-cholesterol diet vol.16, pp.1, 2016, https://doi.org/10.1186/s12906-016-1460-y
  4. Selected Literature Watch vol.3, pp.2, 2013, https://doi.org/10.1089/jcr.2013.1230
  5. Caffeine intake increases plasma ketones: an acute metabolic study in humans vol.95, pp.4, 2017, https://doi.org/10.1139/cjpp-2016-0338
  6. Increased DNA methylation of scavenger receptor class B type I contributes to inhibitory effects of prenatal caffeine ingestion on cholesterol uptake and steroidogenesis in fetal adrenals vol.285, pp.2, 2015, https://doi.org/10.1016/j.taap.2015.03.028
  7. A Combination of Glucosyl Hesperidin and Caffeine Exhibits an Anti-obesity Effect by Inhibition of Hepatic Lipogenesis in Mice vol.29, pp.2, 2015, https://doi.org/10.1002/ptr.5258
  8. Gymnaster koraiensis and its major components, 3,5-di-O-caffeoylquinic acid and gymnasterkoreayne B, reduce oxidative damage induced by tert-butyl hydroperoxide or acetaminophen in HepG2 cells vol.46, pp.10, 2013, https://doi.org/10.5483/BMBRep.2013.46.10.037
  9. Caffeine is responsible for the bloodglucose-lowering effects of green tea and Puer tea extractsin BALB/c mice vol.13, pp.8, 2015, https://doi.org/10.1016/S1875-5364(15)30055-8
  10. Caffeine intake antagonizes salt sensitive hypertension through improvement of renal sodium handling vol.6, pp.1, 2016, https://doi.org/10.1038/srep25746
  11. A comprehensive overview of hepatoprotective natural compounds: mechanism of action and clinical perspectives vol.90, pp.1, 2016, https://doi.org/10.1007/s00204-015-1580-z
  12. Over-expression of BvMTSH, a fusion gene for maltooligosyltrehalose synthase and maltooligosyltrehalose trehalohydrolase, enhances drought tolerance in transgenic rice vol.47, pp.1, 2014, https://doi.org/10.5483/BMBRep.2014.47.1.064
  13. BHBA Influences Bovine Hepatic Lipid Metabolism via AMPK Signaling Pathway vol.116, pp.6, 2015, https://doi.org/10.1002/jcb.25062
  14. Irisin Inhibits Hepatic Cholesterol Synthesis via AMPK-SREBP2 Signaling vol.6, 2016, https://doi.org/10.1016/j.ebiom.2016.02.041
  15. Hypolipidemic Property of a New Fermented Tea Made with Third Crop Green Tea (Camellia sinensis) Leaves and Unripe Satsuma Mandarin (Citrus unshiu) Fruits vol.21, pp.1, 2015, https://doi.org/10.3136/fstr.21.77
  16. Lipid Accumulation in HepG2 Cells Is Attenuated by Strawberry Extract through AMPK Activation vol.9, pp.6, 2017, https://doi.org/10.3390/nu9060621
  17. Metabolic effects of physiological levels of caffeine in myotubes vol.74, pp.1, 2018, https://doi.org/10.1007/s13105-017-0601-1
  18. Caffeine affects HFD-induced hepatic steatosis by multifactorial intervention vol.37, pp.9, 2018, https://doi.org/10.1177/0960327117747026
  19. Coenzyme Q10 attenuates high-fat diet-induced non-alcoholic fatty liver disease through activation of the AMPK pathway vol.10, pp.2, 2019, https://doi.org/10.1039/C8FO01236A