Browse > Article
http://dx.doi.org/10.9718/JBER.2022.43.1.1

Effects of Micro-current Stimulation on lipid metabolism in Oleic Acid-Induced Non-Alcoholic Fatty Liver disease in FL83B cells  

Lee, Hana (Department of Biomedical Engineering, Yonsei University)
Lee, Minjoo (Department of Biomedical Engineering, Yonsei University)
Kim, Han Sung (Department of Biomedical Engineering, Yonsei University)
Publication Information
Journal of Biomedical Engineering Research / v.43, no.1, 2022 , pp. 1-10 More about this Journal
Abstract
Non-alcoholic fatty liver disease(NAFLD) is excessive hepatic lipid accumulation mainly caused by obesity. This study aimed to evaluate whether micro-current stimulation(MCS) could modulate lipid metabolism regarding the Sirt1/AMPK pathway, fatty acid β-oxidation pathway, and lipolysis and lipogenesis-related factors in FL83B cells. For the NAFLD cell model, FL83B cells were treated with oleic acid for lipid accumulation. MCS were stimulated for 1 hr and used frequency 10 Hz, duty cycle 50%, and biphasic rectangular current pulse. The intensity of MCS was divided into 50, 100, 200, and 400 ㎂. Through the results of Oil red O staining, it was confirmed that MCSs with the intensity of 200 ㎂ and 400 ㎂ significantly reduced the degree of lipid droplet formation. Thus, these MCS intensities were applied to western blot analysis. Western blot analysis was performed to analyze the effects of MCS on lipid metabolism. MCS with the intensity of 400 ㎂ showed that significantly activated the Sirt1/AMPK pathway, a key pathway for regulating lipid metabolism in hepatocytes, and fatty acid β-oxidation-related transcription factors. Moreover, it activated the lipolysis pathway and suppressed lipogenesis-related transcription factors such as SREBP-1c, FAS, and PPARγ. In the case of MCS with the intensity of 200 ㎂, only PGC1α and SREBP-1c showed significant differences compared to cells treated only with oleic acid. Taken together, these results suggested that MCS with the intensity of 400 ㎂ could alleviate hepatic lipid accumulation by modulating lipid metabolism in hepatocytes.
Keywords
Micro-current stimulation; Lipid metabolism; Hepatic lipid accumulation; Non-alcoholic fatty liver disease; FL83B cells;
Citations & Related Records
Times Cited By KSCI : 1  (Citation Analysis)
연도 인용수 순위
1 Hazlehurst JM, Woods C, Marjot T, Cobbold JF, Tomlinson JW. Non-alcoholic fatty liver disease and diabetes. Metabolism 2016;65(8):1096-1108.   DOI
2 Ducharme NA, Bickel PE. Minireview: lipid droplets in lipogenesis and lipolysis. Endocrinology 2008;149(3):942-949.   DOI
3 Staels B, Fruchart JC. Therapeutic roles of peroxisome proliferator-activated receptor agonists. Diabetes 2005;54(8):2460-2470.   DOI
4 Day EA, Ford RJ, Steinberg GR. AMPK as a therapeutic target for treating metabolic diseases. Trends in Endocrinology & Metabolism 2017;28(8):545-560.   DOI
5 Wang Z, Pini M, Yao T, Zhou Z, Sun C, Fantuzzi G, Song Z. Homocysteine suppresses lipolysis in adipocytes by activating the AMPK pathway. American journal of physiology-endocrinology and metabolism 2011;301(4):E703-E712.   DOI
6 Brunt EM. Nonalcoholic fatty liver disease: pros and cons of histologic systems of evaluation. International journal of molecular sciences 2016;17(1):97.   DOI
7 Evans RM, Barish GD, Wang YX. PPARs and the complex journey to obesity. Nature medicine 2004;10(4):355-361.   DOI
8 Choi SS, Diehl AM. Hepatic triglyceride synthesis and nonalcoholic fatty liver disease. Current opinion in lipidology 2008;19(3):295-300.   DOI
9 Finck BN, Kelly DP. PGC-1 coactivators: inducible regulators of energy metabolism in health and disease. The Journal of clinical investigation 2006;116(3):615-622.   DOI
10 Smith BK, Marcinko K, Desjardins EM, Lally JS, Ford RJ, Steinberg GR. Treatment of nonalcoholic fatty liver disease: role of AMPK. American Journal of Physiology-Endocrinology and Metabolism 2016;311(4):E730-E740.   DOI
11 Gui Y, Pan Q, Chen X, Xu S, Luo X, Chen L. The association between obesity related adipokines and risk of breast cancer: a meta-analysis. Oncotarget 2017;8(43):75389.   DOI
12 Postic C, Girard J. Contribution of de novo fatty acid synthesis to hepatic steatosis and insulin resistance: lessons from genetically engineered mice. The Journal of clinical investigation 2008;118(3):829-838.   DOI
13 Tian J, Goldstein JL, Brown MS. Insulin induction of SREBP-1c in rodent liver requires LXRα-C/EBPβ complex. Proceedings of the National Academy of Sciences 2016;113(29):8182-8187.   DOI
14 Kim CS, Cho EH, Choe SY, Kang MS, Yu R. Inhibitory effect of isorhamnetin on lipid accumulation in free fatty acid-induced steatotic hepatocytes through the PPARα pathway. Journal of the Korean Society of Food Science and Nutrition 2018;47(7):703-709.   DOI
15 Raghow R, Yellaturu C, Deng X, Park EA, Elam MB. SREBPs: the crossroads of physiological and pathological lipid homeostasis. Trends in endocrinology & metabolism 2008;19(2):65-73.   DOI
16 Browning JD, Horton JD. Molecular mediators of hepatic steatosis and liver injury. The Journal of clinical investigation 2004;114(2):147-152.   DOI
17 Long YC, Zierath JR. AMP-activated protein kinase signaling in metabolic regulation. The Journal of clinical investigation 2006;116(7):1776-1783.   DOI
18 Foretz M, Viollet B. Regulation of hepatic metabolism by AMPK. Journal of Hepatology 2011;54(4):827-9.   DOI
19 Cherkaoui-Malki M, Surapureddi S, I El Hajj H, Vamecq J, Andreoletti P. Hepatic steatosis and peroxisomal fatty acid beta-oxidation. Current Drug Metabolism 2012;13(10):1412-1421.   DOI
20 Szkudelski T, Szkudelska K. Effects of AMPK activation on lipolysis in primary rat adipocytes: studies at different glucose concentrations. Archives of physiology and biochemistry 2017;123(1):43-49.   DOI
21 Reccia I, Kumar J, Akladios C, Virdis F, Pai M, Habib N, Spalding D. Non-alcoholic fatty liver disease: a sign of systemic disease. Metabolism 2017;72:94-108.   DOI
22 Cho MS, Park RJ, Park SH, Cho YH, Cheng GA. The effect of microcurrent-inducing shoes on fatigue and pain in middle-aged people with plantar fascitis. Journal of Physical Therapy Science 2007;19(2):165-170.   DOI
23 Oh HJ, Kim JY, Park RJ. The effects of microcurrent stimulation on recovery of function and pain in chronic low back pain. Journal of the Korean Society of Physical Medicine 2008;3(1):47-56.
24 김한성 외, 세포 전기자극 시스템 및 방법, 대한민국 특허청 등록특허공보 등록번호 10-2101372, 2020.
25 Yoo KD, Jun DW. Nonalcoholic Fatty Liver Disease: Lifestyle Modification. Korean Journal of Medicine 2014;86(4):416-424.   DOI
26 Lin HTV, Tsou YC, Chen YT, Lu WJ, Hwang PA. Effects of low-molecular-weight fucoidan and high stability fucoxanthin on glucose homeostasis, lipid metabolism, and liver function in a mouse model of type II diabetes. Marine drugs 2017;15(4):113.   DOI
27 He XX, Wu XL, Chen RP, Chen C, Liu XG, Wu BJ, Huang ZM. Effectiveness of omega-3 polyunsaturated fatty acids in non-alcoholic fatty liver disease: a meta-analysis of randomized controlled trials. PLoS One 2016;11(10):e0162368.   DOI
28 Yeh YT, Cho YY, Hsieh SC, Chiang AN. Chinese olive extract ameliorates hepatic lipid accumulation in vitro and in vivo by regulating lipid metabolism. Scientific reports 2018;8(1):1-13.
29 Yoo A, Narayan VP, Hong EY, Whang WK, Park T. Scopolin ameliorates high-fat diet induced hepatic steatosis in mice: potential involvement of SIRT1-mediated signaling cascades in the liver. Scientific reports 2017;7(1):1-11.   DOI
30 Chen H, Liu X, Chen H, Cao J, Zhang L, Hu X, Wang J. Role of SIRT1 and AMPK in mesenchymal stem cells differentiation. Ageing research reviews 2014;13:55-64.   DOI
31 Peng IC, Chen Z, Sun W, Li YS, Marin TL, Hsu PH, Shyy JY. Glucagon regulates ACC activity in adipocytes through the CAMKKβ/AMPK pathway. American Journal of Physiology-Endocrinology and Metabolism 2012;302(12):E1560-E1568.   DOI
32 Huang WC, Chang WT, Wu SJ, Xu PY, Ting NC, Liou CJ. Phloretin and phlorizin promote lipolysis and inhibit inflammation in mouse 3T3-L1 cells and in macrophage-adipocyte co-cultures. Molecular nutrition & food research 2013;57(10):1803-1813.   DOI
33 Houten SM, Violante S, Ventura FV, Wanders RJ. The biochemistry and physiology of mitochondrial fatty acid β-oxidation and its genetic disorders. Annual review of physiology 2016;78:23-44.   DOI
34 Fruhbeck G, Mendez-Gimenez L, Fernandez-Formoso JA, Fernandez S, Rodriguez A. Regulation of adipocyte lipolysis. Nutrition research reviews 2014;27(1):63-93.   DOI
35 Malandrino MI, Fucho R, Weber M, Calderon-Dominguez M, Mir JF, Valcarcel L, Herrero L. Enhanced fatty acid oxidation in adipocytes and macrophages reduces lipid-induced triglyceride accumulation and inflammation. American Journal of Physiology-Endocrinology and Metabolism 2015;308(9):E756-E769.   DOI
36 Duncan RE, Ahmadian M, Jaworski K, Sarkadi-Nagy E, Sul HS. Regulation of lipolysis in adipocytes. Annu. Rev. Nutr. 2007;27:79-101.   DOI
37 Heindel JJ, Newbold R, Schug TT. Endocrine disruptors and obesity. Nature Reviews Endocrinology 2015;11(11):653-661.   DOI
38 Li YU, Xu S, Mihaylova MM, Zheng B, Hou X, Jiang B, Zang M. AMPK phosphorylates and inhibits SREBP activity to attenuate hepatic steatosis and atherosclerosis in diet-induced insulin-resistant mice. Cell metabolism 2011;13(4):376-388.   DOI
39 Malandrino MI, Fucho R, Weber M, Calderon-Dominguez M, Mir JF, Valcarcel L, Herrero L. Enhanced fatty acid oxidation in adipocytes and macrophages reduces lipid-induced triglyceride accumulation and inflammation. American Journal of Physiology-Endocrinology and Metabolism 2015;308(9):E756-E769.   DOI
40 Wang Y, Viscarra J, Kim SJ, Sul HS. Transcriptional regulation of hepatic lipogenesis. Nature reviews Molecular cell biology 2015;16(11):678-689.   DOI
41 Liou CJ, Wei CH, Chen YL, Cheng CY, Wang CL, Huang WC. Fisetin protects against hepatic steatosis through regulation of the Sirt1/AMPK and fatty acid β-oxidation signaling pathway in high-fat diet-induced obese mice. Cellular physiology and biochemistry 2018;49(5):1870-1884.   DOI
42 Herbrechter R, Ziemba PM, Hoffmann KM, Hatt H, Werner M, Gisselmann, G. Identification of Glycyrrhiza as the rikkunshito constituent with the highest antagonistic potential on heterologously expressed 5-HT3A receptors due to the action of flavonoids. Frontiers in pharmacology 2015;6:130.
43 Gaidhu MP, Fediuc S, Anthony NM, So M, Mirpourian M, Perry RL, Ceddia RB. Prolonged AICAR-induced AMPkinase activation promotes energy dissipation in white adipocytes: novel mechanisms integrating HSL and ATGL. Journal of lipid research 2009;50(4):704-715.   DOI
44 Hwang D, Lee H, Lee M, Cho S, Kim HS. The Micro-Current Stimulation Inhibits Adipogenesis by Activating Wnt/ β-Catenin Signaling. Journal of Biomedical Engineering Research 2020;41(6):235-246.   DOI
45 Kitade H, Chen G, Ni Y, Ota T. Nonalcoholic fatty liver disease and insulin resistance: new insights and potential new treatments. Nutrients 2017;9(4):387.   DOI
46 Lim CT, Kola B, Korbonits M. AMPK as a mediator of hormonal signalling. Journal of molecular endocrinology 2010;44(2):87.
47 Lampidonis AD, Rogdakis E, Voutsinas GE, Stravopodis DJ. The resurgence of Hormone-Sensitive Lipase (HSL) in mammalian lipolysis. Gene 2011;477(1-2):1-11.   DOI
48 Li Y, Zhu W, Li J, Liu M, Wei M. Resveratrol suppresses the STAT3 signaling pathway and inhibits proliferation of high glucose-exposed HepG2 cells partly through SIRT1. Oncology reports 2013;30(6):2820-2828.   DOI