DOI QR코드

DOI QR Code

Ginsenoside Rb1 ameliorates liver fat accumulation by upregulating perilipin expression in adipose tissue of db/db obese mice

  • Yu, Xizhong (Medical Research Center, First College of Clinical Medicine, Nanjing University of Chinese Medicine) ;
  • Ye, Lifang (Department of Endocrinology, the Affiliated Hospital of Nanjing University of Chinese Medicine) ;
  • Zhang, Hao (Medical Research Center, First College of Clinical Medicine, Nanjing University of Chinese Medicine) ;
  • Zhao, Juan (Medical Research Center, First College of Clinical Medicine, Nanjing University of Chinese Medicine) ;
  • Wang, Guoqiang (Medical Research Center, First College of Clinical Medicine, Nanjing University of Chinese Medicine) ;
  • Guo, Chao (Medical Research Center, First College of Clinical Medicine, Nanjing University of Chinese Medicine) ;
  • Shang, Wenbin (Medical Research Center, First College of Clinical Medicine, Nanjing University of Chinese Medicine)
  • Received : 2014.07.17
  • Accepted : 2014.11.24
  • Published : 2015.07.15

Abstract

Background: Ginsenoside Rb1 (G-Rb1), the major active constituent of ginseng, improves insulin sensitivity and exerts antidiabetic effects. We tested whether the insulin-sensitizing and antidiabetic effects of G-Rb1 results from a reduction in ectopic fat accumulation, mediated by inhibition of lipolysis in adipocytes. Methods: Obese and diabetic db/db mice were treated with daily doses of 20 mg/kg G-Rb1 for 14 days. Hepatic fat accumulation was evaluated by measuring liver weight and triglyceride content. Levels of blood glucose and serum insulin were used to evaluate insulin sensitivity in db/db mice. Lipolysis in adipocytes was evaluated by measuring plasma-free fatty acids and glycerol release from 3T3-L1 adipocytes treated with G-Rb1. The expression of relevant genes was analyzed by western blotting, quantitative real-time polymerase chain reaction, and enzyme-linked immunosorbent assay kit. Results: G-Rb1 increased insulin sensitivity and alleviated hepatic fat accumulation in obese diabetic db/db mice, and these effects were accompanied by reduced liver weight and hepatic triglyceride content. Furthermore, G-Rb1 lowered the levels of free fatty acids in obese mice, which may contribute to a decline in hepatic lipid accumulation. Corresponding to these results, G-Rb1 significantly suppressed lipolysis in 3T3-L1 adipocytes and upregulated the perilipin expression in both 3T3-L1 adipocytes and mouse epididymal fat pads. Moreover, G-Rb1 increased the level of adiponectin and reduced that of tumor necrosis factor-${\alpha}$ in obese mice, and these effects were confirmed in 3T3-L1 adipocytes. Conclusion: G-Rb1 may improve insulin sensitivity in obese and diabetic db/db mice by reducing hepatic fat accumulation and suppressing adipocyte lipolysis; these effects may be mediated via the upregulation of perilipin expression in adipocytes.

Keywords

References

  1. Attele AS, Wu JA, Yuan CS. Ginseng pharmacology: multiple constituents and multiple actions. Biochem Pharmacol 1999;58:1685-93. https://doi.org/10.1016/S0006-2952(99)00212-9
  2. Yuan HD, Kim JT, Kim SH, Chung SH. Ginseng and diabetes: the evidences from in vitro, animal and human studies. J Ginseng Res 2012;36:27-39. https://doi.org/10.5142/jgr.2012.36.1.27
  3. Lim W, Mudge KW, Vermeylen F. Effects of population, age, and cultivation methods on ginsenoside content of wild American ginseng (Panax quinquefolium). J Agric Food Chem 2005;53:8498-505. https://doi.org/10.1021/jf051070y
  4. Washida D, Kitanaka S. Determination of polyacetylenes and ginsenosides in Panax species using high performance liquid chromatography. Chem Pharm Bull (Tokyo) 2003;51:1314-7. https://doi.org/10.1248/cpb.51.1314
  5. Shang W, Yang Y, Zhou L, Jiang B, Jin H, Chen M. Ginsenoside Rb1 stimulates glucose uptake through insulin-like signaling pathway in 3T3-L1 adipocytes. J Endocrinol 2008;198:561-9. https://doi.org/10.1677/JOE-08-0104
  6. Shang W, Yang Y, Jiang B, Jin H, Zhou L, Liu S, Chen M. Ginsenoside Rb1 promotes adipogenesis in 3T3-L1 cells by enhancing PPARgamma2 and C/ EBPalpha gene expression. Life Sci 2007;80:618-25. https://doi.org/10.1016/j.lfs.2006.10.021
  7. Shang W, Yang Y, Zhou L, Jiang B, Jin H, Chen M. Ginsenoside Rb1 facilitates adipocyte differentiation and inhibits lipolysis in 3T3-L1 adipocytes. Chin J Endocrinol Metab 2007;23:6-9.
  8. Xiong Y, Shen L, Liu KJ, Tso P, Wang G, Woods SC, Liu M. Antiobesity and antihyperglycemic effects of ginsenoside Rb1 in rats. Diabetes 2010;59: 2505-12. https://doi.org/10.2337/db10-0315
  9. Shen L, Xiong Y, Wang DQ, Howles P, Basford JE, Wang J, Xiong YQ, Hui DY, Woods SC, Liu M. Ginsenoside Rb1 reduces fatty liver by activating AMPactivated protein kinase in obese rats. J Lipid Res 2013;54:1430-8. https://doi.org/10.1194/jlr.M035907
  10. Shang W, Yu X, Wang G, Zhao J. Effect of ginsenoside Rb1 in ameliorating insulin resistance and ectopic fat deposition in obese mice induced by high fat diet. Zhongguo Zhong Yao Za Zhi 2013;38:4119-23 [Article in Chinese].
  11. Johnson AM, Olefsky JM. The origins and drivers of insulin resistance. Cell 2013;152:673-84. https://doi.org/10.1016/j.cell.2013.01.041
  12. Ebbert JO, Jensen MD. Fat depots, free fatty acids, and dyslipidemia. Nutrients 2013;5:498-508. https://doi.org/10.3390/nu5020498
  13. Fabbrini E, Magkos F, Mohammed BS, Pietka T, Abumrad NA, Patterson BW, Okunade A, Klein S. Intrahepatic fat, not visceral fat, is linked with metabolic complications of obesity. Proc Natl Acad Sci U S A 2009;106:15430-5. https://doi.org/10.1073/pnas.0904944106
  14. Sato F, Tamura Y, Watada H, Kumashiro N, Igarashi Y, Uchino H, Maehara T, Kyogoku S, Sunayama S, Sato H, et al. Effects of diet-induced moderate weight reduction on intrahepatic and intramyocellular triglycerides and glucose metabolism in obese subjects. J Clin Endocrinol Metab 2007;92:3326-9. https://doi.org/10.1210/jc.2006-2384
  15. Bajaj M, Suraamornkul S, Pratipanawatr T, Hardies LJ, Pratipanawatr W, Glass L, Cersosimo E, Miyazaki Y, DeFronzo RA. Pioglitazone reduces hepatic fat content and augments splanchnic glucose uptake in patients with type 2 diabetes. Diabetes 2003;52:1364-70. https://doi.org/10.2337/diabetes.52.6.1364
  16. Yki-Järvinen H. Ectopic fat accumulation: an important cause of insulin resistance in humans. J R Soc Med 2002;95(Suppl. 42):39-45. https://doi.org/10.1258/jrsm.95.1.39
  17. Brasaemle DL. Thematic review series: adipocyte biology. The perilipin family of structural lipid droplet proteins: stabilization of lipid droplets and control of lipolysis. J Lipid Res 2007;48:2547-59. https://doi.org/10.1194/jlr.R700014-JLR200
  18. Sztalryd C, Xu G, Dorward H, Tansey JT, Contreras JA, Kimmel AR, Londos C. Perilipin A is essential for the translocation of hormone-sensitive lipase during lipolytic activation. J Cell Biol 2003;161:1093-103. https://doi.org/10.1083/jcb.200210169
  19. Granneman JG, Moore HP, Granneman RL, Greenberg AS, Obin MS, Zhu Z. Analysis of lipolytic protein trafficking and interactions in adipocytes. J Biol Chem 2007;282:5726-35. https://doi.org/10.1074/jbc.M610580200
  20. Souza SC, Yamamoto MT, Franciosa MD, Lien P, Greenberg AS. BRL 49653 blocks the lipolytic actions of tumor necrosis factor-alpha: a potential new insulinsensitizing mechanism for thiazolidinediones. Diabetes 1998;47:691-5. https://doi.org/10.2337/diabetes.47.4.691
  21. Tansey JT, Sztalryd C, Gruia-Gray J, Roush DL, Zee JV, Gavrilova O, Reitman ML, Deng CX, Li C, Kimmel AR, et al. Perilipin ablation results in a lean mouse with aberrant adipocyte lipolysis, enhanced leptin production, and resistance to diet-induced obesity. Proc Natl Acad Sci U S A 2001;98:6494-9. https://doi.org/10.1073/pnas.101042998
  22. Matthews DR, Hosker JP, Rudenski AS, Naylor BA, Treacher DF, Turner RC. Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia 1985;28:412-9. https://doi.org/10.1007/BF00280883
  23. Salmon DM, Flatt JP. Effect of dietary fat content on the incidence of obesity among ad libitum fed mice. Int J Obes 1985;9:443-9.
  24. Ramírez-Zacarias JL, Castro-Muñozledo F, Kuri-Harcuch W. Quantitation of adipose conversion and triglycerides by staining intracytoplasmic lipids with Oil red O. Histochemistry 1992;97:493-7. https://doi.org/10.1007/BF00316069
  25. Perrini S, Natalicchio A, Laviola L, Belsanti G, Montrone C, Cignarelli A, Minielli V, Grano M, De Pergola G, Giorgino R, et al. Dehydroepiandrosterone stimulates glucose uptake in human and murine adipocytes by inducing GLUT1 and GLUT4 translocation to the plasma membrane. Diabetes 2004;53: 41-52. https://doi.org/10.2337/diabetes.53.1.41
  26. Samuel VT, Shulman GI. Mechanisms for insulin resistance: common threads and missing links. Cell 2012;148:852-71. https://doi.org/10.1016/j.cell.2012.02.017
  27. Mook S, Cj Cj Halkes, Bilecen S, Cabezas MC. In vivo regulation of plasma free fatty acids in insulin resistance. Metabolism 2004;53:1197-201. https://doi.org/10.1016/j.metabol.2004.02.023
  28. Yamaguchi T, Omatsu N, Morimoto E, Nakashima H, Ueno K, Tanaka T, Satouchi K, Hirose F, Osumi T. CGI-58 facilitates lipolysis on lipid droplets but is not involved in the vesiculation of lipid droplets caused by hormonal stimulation. J Lipid Res 2007;48:1078-89. https://doi.org/10.1194/jlr.M600493-JLR200
  29. Nielsen TS, Kampmann U, Nielsen RR, Jessen N, Orskov L, Pedersen SB, Jorgensen JO, Lund S, Moller N. Reduced mRNA and protein expression of perilipin A and G0/G1 switch gene 2 (G0S2) in human adipose tissue in poorly controlled type 2 diabetes. J Clin Endocrinol Metab 2012;97:E1348-52. https://doi.org/10.1210/jc.2012-1159
  30. Arimura N, Horiba T, Imagawa M, Shimizu M, Sato R. The peroxisome proliferator- activated receptor gamma regulates expression of the perilipin gene in adipocytes. J Biol Chem 2004;279:10070-6. https://doi.org/10.1074/jbc.M308522200
  31. Nagai S, Shimizu C, Umetsu M, Taniguchi S, Endo M, Miyoshi H, Yoshioka N, Kubo M, Koike T. Identification of a functional peroxisome proliferator-activated receptor responsive element within the murine perilipin gene. Endocrinology 2004;145:2346-56. https://doi.org/10.1210/en.2003-1180
  32. Kim HJ, Jung TW, Kang ES, Kim DJ, Ahn CW, Lee KW, Lee HC, Cha BS. Depotspecific regulation of perilipin by rosiglitazone in a diabetic animal model. Metabolism 2007;56:676-85. https://doi.org/10.1016/j.metabol.2006.12.017
  33. Bays H, Mandarino L, DeFronzo RA. Role of the adipocyte, free fatty acids, and ectopic fat in pathogenesis of type 2 diabetes mellitus: peroxisomal proliferator- activated receptor agonists provide a rational therapeutic approach. J Clin Endocrinol Metab 2004;89:463-78. https://doi.org/10.1210/jc.2003-030723
  34. Ryden M, Arvidsson E, Blomqvist L, Perbeck L, Dicker A, Arner P. Targets for TNF-alpha-induced lipolysis in human adipocytes. Biochem Biophys Res Commun 2004;318:168-75. https://doi.org/10.1016/j.bbrc.2004.04.010
  35. Duncan RE, Ahmadian M, Jaworski K, Sarkadi-Nagy E, Sul HS. Regulation of lipolysis in adipocytes. Annu Rev Nutr 2007;27:79-101. https://doi.org/10.1146/annurev.nutr.27.061406.093734
  36. Kadowaki T, Yamauchi T, Kubota N, Hara K, Ueki K, Tobe K. Adiponectin and adiponectin receptors in insulin resistance, diabetes, and the metabolic syndrome. J Clin Invest 2006;116:1784-92. https://doi.org/10.1172/JCI29126
  37. Qiao L, Kinney B, Schaack J, Shao J. Adiponectin inhibits lipolysis in mouse adipocytes. Diabetes 2011;60:1519-27. https://doi.org/10.2337/db10-1017

Cited by

  1. Rhinacanthus nasutus leaf improves metabolic abnormalities in high-fat diet-induced obese mice vol.6, pp.1, 2015, https://doi.org/10.1016/j.apjtb.2015.10.004
  2. Anti-obesity effects of zeaxanthin on 3T3-L1 preadipocyte and high fat induced obese mice vol.8, pp.9, 2015, https://doi.org/10.1039/c7fo00486a
  3. Ginsenoside Rg5 Inhibits Succinate-Associated Lipolysis in Adipose Tissue and Prevents Muscle Insulin Resistance vol.8, pp.None, 2017, https://doi.org/10.3389/fphar.2017.00043
  4. Ginsenoside Rb1 Enhances Atherosclerotic Plaque Stability by Improving Autophagy and Lipid Metabolism in Macrophage Foam Cells vol.8, pp.None, 2015, https://doi.org/10.3389/fphar.2017.00727
  5. Ginsenoside Rb1 inhibits free fatty acids-induced oxidative stress and inflammation in 3T3-L1 adipocytes vol.16, pp.6, 2015, https://doi.org/10.3892/mmr.2017.7710
  6. Bridging Type 2 Diabetes and Alzheimer's Disease: Assembling the Puzzle Pieces in the Quest for the Molecules With Therapeutic and Preventive Potential vol.38, pp.1, 2015, https://doi.org/10.1002/med.21440
  7. Ginsenoside Rb1 enhances atherosclerotic plaque stability by skewing macrophages to the M2 phenotype vol.22, pp.1, 2018, https://doi.org/10.1111/jcmm.13329
  8. Ginsenoside Rb1 Induces Beta 3 Adrenergic Receptor-Dependent Lipolysis and Thermogenesis in 3T3-L1 Adipocytes and db/db Mice vol.10, pp.None, 2019, https://doi.org/10.3389/fphar.2019.01154
  9. Ginsenoside Rb1 as an Anti-Diabetic Agent and Its Underlying Mechanism Analysis vol.8, pp.3, 2019, https://doi.org/10.3390/cells8030204
  10. Ginsenoside Rk3 ameliorates high-fat-diet/streptozocin induced type 2 diabetes mellitus in mice via the AMPK/Akt signaling pathway vol.10, pp.5, 2019, https://doi.org/10.1039/c9fo00095j
  11. New Insights into Bioactive Compounds of Traditional Chinese Medicines for Insulin Resistance Based on Signaling Pathways vol.16, pp.9, 2019, https://doi.org/10.1002/cbdv.201900176
  12. Black Ginseng and Ginsenoside Rb1 Promote Browning by Inducing UCP1 Expression in 3T3-L1 and Primary White Adipocytes vol.11, pp.11, 2015, https://doi.org/10.3390/nu11112747
  13. Ginsenoside-Rb1 Improved Diabetic Cardiomyopathy through Regulating Calcium Signaling by Alleviating Protein O-GlcNAcylation vol.67, pp.51, 2019, https://doi.org/10.1021/acs.jafc.9b05706
  14. Potential Impact of the Multi-Target Drug Approach in the Treatment of Some Complex Diseases vol.14, pp.None, 2020, https://doi.org/10.2147/dddt.s257494
  15. Effectiveness and Safety of Panax ginseng Extract on Hepatic Dysfunction: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial vol.2020, pp.None, 2020, https://doi.org/10.1155/2020/2689565
  16. Ginsenoside Rb1 Alleviated High-Fat-Diet-Induced Hepatocytic Apoptosis via Peroxisome Proliferator-Activated Receptor γ vol.2020, pp.None, 2015, https://doi.org/10.1155/2020/2315230
  17. Herbal Medicine in the Treatment of Non-Alcoholic Fatty Liver Diseases-Efficacy, Action Mechanism, and Clinical Application vol.11, pp.None, 2015, https://doi.org/10.3389/fphar.2020.00601
  18. Ginsenoside Rb1, salvianolic acid B and their combination modulate gut microbiota and improve glucolipid metabolism in high-fat diet induced obese mice vol.9, pp.None, 2015, https://doi.org/10.7717/peerj.10598
  19. Effect of Panax notoginseng Saponins and Major Anti-Obesity Components on Weight Loss vol.11, pp.None, 2021, https://doi.org/10.3389/fphar.2020.601751
  20. Differentially Expressed Functional LncRNAs in Human Subjects With Metabolic Syndrome Reflect a Competing Endogenous RNA Network in Circulating Extracellular Vesicles vol.8, pp.None, 2015, https://doi.org/10.3389/fmolb.2021.667056
  21. Plants Secondary Metabolites as Blood Glucose-Lowering Molecules vol.26, pp.14, 2015, https://doi.org/10.3390/molecules26144333
  22. Ginsenoside Rb1 alleviates liver injury induced by 3‐chloro‐1,2‐propanediol by stimulating autophagic flux vol.86, pp.12, 2021, https://doi.org/10.1111/1750-3841.15968