DOI QR코드

DOI QR Code

Ginsenoside Rb1 and compound K improve insulin signaling and inhibit ER stress-associated NLRP3 inflammasome activation in adipose tissue

  • Chen, Weijie (Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University) ;
  • Wang, Junlian (Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University) ;
  • Luo, Yong (Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University) ;
  • Wang, Tao (Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University) ;
  • Li, Xiaochun (Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University) ;
  • Li, Aiyun (Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University) ;
  • Li, Jia (Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University) ;
  • Liu, Kang (Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University) ;
  • Liu, Baolin (Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University)
  • Received : 2015.06.17
  • Accepted : 2015.11.18
  • Published : 2016.10.15

Abstract

Background: This study was designed to investigate whether ginsenoside Rb1 (Rb1) and compound K (CK) ameliorated insulin resistance by suppressing endoplasmic reticulum (ER) stress-induced inflammation in adipose tissue. Methods: To induce ER stress, epididymal adipose tissue from mice or differentiated 3T3 adipocytes were exposed to high glucose. The effects of Rb1 and CK on reactive oxygen species production, ER stress, TXNIP/NLRP3 inflammasome activation, inflammation, insulin signaling activation, and glucose uptake were detected by western blot, emzyme-linked immunosorbent assay, or fluorometry. Results: Rb1 and CK suppressed ER stress by dephosphorylation of $IRE1{\alpha}$ and PERK, thereby reducing TXNIP-associated NLRP3 inflammasome activation in adipose tissue. As a result, Rb1 and CK inhibited IL-$1{\beta}$ maturation and downstream inflammatory factor IL-6 secretion. Inflammatory molecules induced insulin resistance by upregulating phosphorylation of insulin receptor substrate-1 at serine residues and impairing insulin PI3K/Akt signaling, leading to decreased glucose uptake by adipocytes. Rb1 and CK reversed these changes by inhibiting ER stress-induced inflammation and ameliorating insulin resistance, thereby improving the insulin IRS-1/PI3K/Akt-signaling pathway in adipose tissue. Conclusion: Rb1 and CK inhibited inflammation and improved insulin signaling in adipose tissue by suppressing ER stress-associated NLRP3 inflammation activation. These findings offered novel insight into the mechanism by which Rb1 and CK ameliorate insulin resistance in adipose tissue.

Keywords

References

  1. Hager SR, Jochen AL, Kalkhoff RK. Insulin resistance in normal rats infused with glucose for 72 h. Am J Physiol 1991;260:E353-62.
  2. Rossetti L. Glucose toxicity: the implications of hyperglycemia in the pathophysiology of diabetes mellitus. Clin Invest Med 1995;18:255-60.
  3. Haber CA, Lam TK, Yu Z, Gupta N, Goh T, Bogdanovic E, Giacca A, Fantus IG. N-acetylcysteine and taurine prevent hyperglycemia-induced insulin resistance in vivo: possible role of oxidative stress. Am J Physiol Endocrinol Metab 2003;285:E744-53. https://doi.org/10.1152/ajpendo.00355.2002
  4. Dedoussis GV, Kaliora AC, Panagiotakos DB. Genes, diet and type 2 diabetes mellitus: a review. Rev Diabet Stud 2007;4:13-24. https://doi.org/10.1900/RDS.2007.4.13
  5. Eizirik DL, Cardozo AK, Cnop M. The role for endoplasmic reticulum stress in diabetes mellitus. Endocr Rev 2008;29:42-61. https://doi.org/10.1210/er.2007-0015
  6. Marciniak SJ, Ron D. Endoplasmic reticulum stress signaling in disease. Physiol Rev 2006;86:1133-49. https://doi.org/10.1152/physrev.00015.2006
  7. Ringseis R, Eder K, Mooren FC, Kruger K. Metabolic signals and innate immune activation in obesity and exercise. Exerc Immunol Rev 2015;21:58-68.
  8. Schroder K, Tschopp J. The inflammasomes. Cell 2010;140:821-32. https://doi.org/10.1016/j.cell.2010.01.040
  9. Oslowski CM, Hara T, O'Sullivan-Murphy B, Kanekura K, Lu S, Hara M, Ishigaki S, Zhu LJ, Hayashi E, Hui ST, et al. Thioredoxin-interacting protein mediates ER stress-induced ${\beta}$ cell death through initiation of the inflammasome. Cell Metab 2012;16:265-73. https://doi.org/10.1016/j.cmet.2012.07.005
  10. Gual P, Le Marchand-Brustel Y, Tanti JF. Positive and negative regulation of insulin signaling through IRS-1 phosphorylation. Biochimie 2005;87:99-109. https://doi.org/10.1016/j.biochi.2004.10.019
  11. Zhou R, Tardivel A, Thorens B, Choi I, Tschopp J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat Immunol 2010;11:136-40. https://doi.org/10.1038/ni.1831
  12. Gao D, Madi M, Ding C, Fok M, Steele T, Ford C, Hunter L, Bing C. Interleukin-$1{\beta}$ mediates macrophage-induced impairment of insulin signaling in human primary adipocytes. Am J Physiol Endocrinol Metab 2014;307:E289-304. https://doi.org/10.1152/ajpendo.00430.2013
  13. Maedler K, Dharmadhikari G, Schumann DM, Storling J. Interleukin-1 beta targeted therapy for type 2 diabetes. Expert Opin Biol Ther 2009;9:1177-88. https://doi.org/10.1517/14712590903136688
  14. Akao T, Kida H, Kanaoka M, Hattori M, Kobashi K. Intestinal bacterial hydrolysis is required for the appearance of compound K in rat plasma after oral administration of ginsenoside Rb1 from Panax ginseng. J Pharm Pharmacol 1998;50:1155-60. https://doi.org/10.1111/j.2042-7158.1998.tb03327.x
  15. Xiong Y, Shen L, Liu KJ, Tso P, Xiong Y, 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
  16. 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
  17. Jiang S, Ren D, Li J, Yuan G, Li H, Xu G, Han X, Du P, An L. Effects of compound K on hyperglycemia and insulin resistance in rats with type 2 diabetes mellitus. Fitoterapia 2014;95:58-64. https://doi.org/10.1016/j.fitote.2014.02.017
  18. Park EK, Shin YW, Lee HU, Kim SS, Lee YC, Lee BY, Kim DH. Inhibitory effect of ginsenoside Rb1 and compound K on NO and prostaglandin E2 biosyntheses of RAW264.7 cells induced by lipopolysaccharide. Biol Pharm Bull 2005;28:652-6. https://doi.org/10.1248/bpb.28.652
  19. Joh EH, Lee IA, Jung IH, Kim DH. Ginsenoside Rb1 and its metabolite compound K inhibit IRAK-1 activation-the key step of inflammation. Biochem Pharmacol 2011;82:278-86. https://doi.org/10.1016/j.bcp.2011.05.003
  20. Kim S, Na JY, Song KB, Choi DS, Kim JH, Kwon YB, Kwon J. Protective effect of ginsenoside Rb1 on hydrogen peroxide-induced oxidative stress in rat articular chondrocytes. J Ginseng Res 2012;36:161-8. https://doi.org/10.5142/jgr.2012.36.2.161
  21. Roberts CK, Hevener AL, Barnard RJ. Metabolic syndrome and insulin resistance: underlying causes and modification by exercise training. Compr Physiol 2013;3:1-58.
  22. Cao SS, Kaufman RJ. Endoplasmic reticulum stress and oxidative stress in cell fate decision and human disease. Antioxid Redox Signal 2014;21:396-413. https://doi.org/10.1089/ars.2014.5851
  23. Zhang K, Kaufman RJ. From endoplasmic-reticulum stress to the inflammatory response. Nature 2008;454:455-62. https://doi.org/10.1038/nature07203
  24. Lambeth JD, Neish AS. Nox enzymes and new thinking on reactive oxygen: a double-edged sword revisited. Annu Rev Pathol 2014;9:119-45. https://doi.org/10.1146/annurev-pathol-012513-104651
  25. Shao X, Li N, Zhan J, Sun H, An L, Du P. Protective effect of compound K on diabetic rats. Nat Prod Commun 2015;10:243-5.
  26. Mohamed IN, Hafez SS, Fairaq A, Ergul A, Imig JD, EI-Remessy AB. Thioredoxin-interacting protein is required for endothelial NLRP3 inflammasome activation and cell death in a rat model of high-fat diet. Diabetologia 2014;57:413-23. https://doi.org/10.1007/s00125-013-3101-z
  27. Mittal M, Siddiqui MR, Tran K, Reddy SP, Malik AB. Reactive oxygen species in inflammation and tissue injury. Antioxid Redox Signal 2014;20:1126-67. https://doi.org/10.1089/ars.2012.5149
  28. Aguirre V, Werner ED, Giraud J, Lee YH, Shoelson SE, White MF. Phosphorylation of Ser307 in insulin receptor substrate-1 blocks interactions with the insulin receptor and inhibits insulin action. J Biol Chem 2002;277:1531-7. https://doi.org/10.1074/jbc.M101521200

Cited by

  1. 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
  2. Attenuation of TNF-α-Induced Inflammatory Injury in Endothelial Cells by Ginsenoside Rb1 via Inhibiting NF-κB, JNK and p38 Signaling Pathways vol.8, pp.None, 2016, https://doi.org/10.3389/fphar.2017.00464
  3. Ginsenoside Rg1 Inhibits Glucagon-Induced Hepatic Gluconeogenesis through Akt-FoxO1 Interaction vol.7, pp.16, 2016, https://doi.org/10.7150/thno.18788
  4. The NLPR3 inflammasome and obesity‐related kidney disease vol.22, pp.1, 2018, https://doi.org/10.1111/jcmm.13333
  5. Anti-inflammatory effects of a novel compound, MPQP, through the inhibition of IRAK1 signaling pathways in LPS-stimulated RAW 264.7 macrophages vol.51, pp.6, 2016, https://doi.org/10.5483/bmbrep.2018.51.6.064
  6. Can Topical Use of Ginseng or Ginsenosides Accelerate Wound Healing? vol.21, pp.11, 2016, https://doi.org/10.1089/jmf.2018.29000.com
  7. Potential Dissociative Glucocorticoid Receptor Activity for Protopanaxadiol and Protopanaxatriol vol.20, pp.1, 2019, https://doi.org/10.3390/ijms20010094
  8. Accelerated and Severe Lupus Nephritis Benefits From M1, an Active Metabolite of Ginsenoside, by Regulating NLRP3 Inflammasome and T Cell Functions in Mice vol.10, pp.None, 2016, https://doi.org/10.3389/fimmu.2019.01951
  9. Endoplasmic reticulum stress and NLRP3 inflammasome: Crosstalk in cardiovascular and metabolic disorders vol.234, pp.9, 2016, https://doi.org/10.1002/jcp.28275
  10. Ginsenoside Rb1 Ameliorates Age-Related Myocardial Dysfunction by Regulating the NF- $ \kappa $ B Signaling Pathway vol.48, pp.6, 2020, https://doi.org/10.1142/s0192415x20500676
  11. Ginsenoside Rb1 Alleviated High-Fat-Diet-Induced Hepatocytic Apoptosis via Peroxisome Proliferator-Activated Receptor γ vol.2020, pp.None, 2016, https://doi.org/10.1155/2020/2315230
  12. Role of Inflammasomes in Kidney Diseases via Both Canonical and Non-canonical Pathways vol.8, pp.None, 2016, https://doi.org/10.3389/fcell.2020.00106
  13. Effects of compound K, a metabolite of ginsenosides, on memory and cognitive dysfunction in db/db mice involve the inhibition of ER stress and the NLRP3 inflammasome pathway vol.11, pp.5, 2016, https://doi.org/10.1039/c9fo02602a
  14. Nutraceutical Compounds Targeting Inflammasomes in Human Diseases vol.21, pp.14, 2020, https://doi.org/10.3390/ijms21144829
  15. Effects of ginsenoside Rb1 on serum brain natriuretic peptide level and caspase-3 protein expression in cardiomyocytes of rats with chronic heart failure vol.17, pp.74, 2016, https://doi.org/10.4103/pm.pm_561_19
  16. Red ginseng extracts as an adjunctive therapeutic for gout: preclinical and clinical evidence vol.32, pp.1, 2021, https://doi.org/10.1080/09540105.2020.1854189
  17. Anti-NLRP3 Inflammasome Natural Compounds: An Update vol.9, pp.2, 2021, https://doi.org/10.3390/biomedicines9020136
  18. Role of Thioredoxin-Interacting Protein in Diseases and Its Therapeutic Outlook vol.22, pp.5, 2016, https://doi.org/10.3390/ijms22052754
  19. Plants Secondary Metabolites as Blood Glucose-Lowering Molecules vol.26, pp.14, 2016, https://doi.org/10.3390/molecules26144333
  20. Ginsenoside CK inhibits obese insulin resistance by activating PPARγ to interfere with macrophage activation vol.157, pp.None, 2016, https://doi.org/10.1016/j.micpath.2021.105002
  21. Medicinal plants and bioactive natural products as inhibitors of NLRP3 inflammasome vol.35, pp.9, 2016, https://doi.org/10.1002/ptr.7118
  22. Exploring the Use of Medicinal Plants and Their Bioactive Derivatives as Alveolar NLRP3 Inflammasome Regulators during Mycobacterium tuberculosis Infection vol.22, pp.17, 2016, https://doi.org/10.3390/ijms22179497
  23. Pyroptosis in diabetic nephropathy vol.523, pp.None, 2016, https://doi.org/10.1016/j.cca.2021.09.003
  24. Antidiabetic effects of quercetin and liraglutide combination through modulation of TXNIP/IRS‐1/PI3K pathway vol.40, pp.1, 2016, https://doi.org/10.1002/cbf.3678