DOI QR코드

DOI QR Code

Restoration of the adipogenic gene expression by naringenin and naringin in 3T3-L1 adipocytes

  • Dayarathne, Lakshi A. (College of Veterinary Medicine, Jeju National University) ;
  • Ranaweera, Sachithra S. (College of Veterinary Medicine, Jeju National University) ;
  • Natraj, Premkumar (College of Veterinary Medicine, Jeju National University) ;
  • Rajan, Priyanka (College of Veterinary Medicine, Jeju National University) ;
  • Lee, Young Jae (College of Veterinary Medicine, Jeju National University) ;
  • Han, Chang-Hoon (College of Veterinary Medicine, Jeju National University)
  • 투고 : 2021.04.13
  • 심사 : 2021.06.20
  • 발행 : 2021.07.31

초록

Background: Naringenin and its glycoside naringin are well known citrus flavonoids with several therapeutic benefits. Although the anti-adipogenic effects of naringenin and naringin have been reported previously, the detailed mechanism underlying their anti-adipogenesis effects is poorly understood. Objectives: This study examined the anti-adipogenic effects of naringenin and naringin by determining differential gene expression patterns in these flavonoids-treated 3T3-L1 adipocytes. Methods: Lipid accumulation and triglyceride (TG) content were determined by Oil red O staining and TG assay. Glucose uptake was measured using a 2-[N-(7-Nitrobenz-2-oxa-1,3-diazol-4-yl)amino]-2-deoxy-d-glucose fluorescent d-glucose analog. The phosphorylation levels of AMP-activated protein kinase (AMPK) and acetyl Co-A carboxylase (ACC) were observed via Western blot analysis. Differential gene expressions in 3T3-L1 adipocytes were evaluated via RNA sequencing analysis. Results: Naringenin and naringin inhibited both lipid accumulation and TG content, increased phosphorylation levels of both AMPK and ACC and decreased the expression level of 3-hydroxy-3-methylglutaryl CoA reductase (HMGCR) in 3T3-L1 adipocytes. RNA sequencing analysis revealed that 32 up-regulated (> 2-fold) and 17 down-regulated (< 0.6-fold) genes related to lipid metabolism, including Acaca, Fasn, Scd1, Mogat1, Dgat, Lipin1, Cpt1a, and Lepr, were normalized to the control level in naringenin-treated adipocytes. In addition, 25 up-regulated (> 2-fold) and 25 down-regulated (< 0.6-fold) genes related to lipid metabolism, including Acaca, Fasn, Fabp5, Scd1, Srebf1, Hmgcs1, Cpt1c, Lepr, and Lrp1, were normalized to the control level by naringin. Conclusions: The results indicate that naringenin and naringin have anti-adipogenic potentials that are achieved by normalizing the expression levels of lipid metabolism-related genes that were perturbed in differentiated 3T3-L1 cells.

키워드

과제정보

This work was supported by Korean Institute of Planning and Evaluation for Technology in Food, Agriculture and Forestry (IPET) through Innovational Food Technology Development Program, funded by Ministry of Agriculture, Food and Rural Affairs (MAFRA) (grant No. 11901303).

참고문헌

  1. Sun NN, Wu TY, Chau CF. Natural dietary and herbal products in anti-obesity treatment. Molecules. 2016;21(10):1351. https://doi.org/10.3390/molecules21101351
  2. Park J, Kim HL, Jung Y, Ahn KS, Kwak HJ, Um JY. Bitter orange (Citrus aurantium Linne) improves obesity by regulating adipogenesis and thermogenesis through AMPK activation. Nutrients. 2019;11(9):1988. https://doi.org/10.3390/nu11091988
  3. Xu L, Li Y, Dai Y, Peng J. Natural products for the treatment of type 2 diabetes mellitus: Pharmacology and mechanisms. Pharmacol Res. 2018;130:451-465. https://doi.org/10.1016/j.phrs.2018.01.015
  4. Kang SW, Kang SI, Shin HS, Yoon SA, Kim JH, Ko HC, et al. Sasa quelpaertensis Nakai extract and its constituent p-coumaric acid inhibit adipogenesis in 3T3-L1 cells through activation of the AMPK pathway. Food Chem Toxicol. 2013;59:380-385. https://doi.org/10.1016/j.fct.2013.06.033
  5. Hardie DG, Pan DA. Regulation of fatty acid synthesis and oxidation by the AMP-activated protein kinase. Biochem Soc Trans. 2002;30(Pt 6):1064-1070. https://doi.org/10.1042/bst0301064
  6. Richard AJ, Amini-Vaughan Z, Ribnicky DM, Stephens JM. Naringenin inhibits adipogenesis and reduces insulin sensitivity and adiponectin expression in adipocytes. Evid Based Complement Alternat Med. 2013;2013:549750.
  7. Mahmoud AM, Hernandez Bautista RJ, Sandhu MA, Hussein OE. Beneficial effects of citrus flavonoids on cardiovascular and metabolic health. Oxid Med Cell Longev. 2019;2019:5484138. https://doi.org/10.1155/2019/5484138
  8. Claussnitzer M, Skurk T, Hauner H, Daniel H, Rist MJ. Effect of flavonoids on basal and insulin-stimulated 2-deoxyglucose uptake in adipocytes. Mol Nutr Food Res. 2011;55 Suppl 1:S26-S34. https://doi.org/10.1002/mnfr.201000372
  9. Alam MA, Subhan N, Rahman MM, Uddin SJ, Reza HM, Sarker SD. Effect of citrus flavonoids, naringin and naringenin, on metabolic syndrome and their mechanisms of action. Adv Nutr. 2014;5(4):404-417. https://doi.org/10.3945/an.113.005603
  10. Rizzatti V, Boschi F, Pedrotti M, Zoico E, Sbarbati A, Zamboni M. Lipid droplets characterization in adipocyte differentiated 3T3-L1 cells: size and optical density distribution. Eur J Histochem. 2013;57(3):e24.
  11. Ranaweera SS, Dissanayake CY, Natraj P, Lee YJ, Han CH. Anti-inflammatory effect of sulforaphane on LPS-stimulated RAW 264.7 cells and ob/ob mice. J Vet Sci. 2020;21(6):e91. https://doi.org/10.4142/jvs.2020.21.e91
  12. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods. 2012;9(4):357-359. https://doi.org/10.1038/nmeth.1923
  13. Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, et al. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 2004;5(10):R80. https://doi.org/10.1186/gb-2004-5-10-r80
  14. Guo X, Liu J, Cai S, Wang O, Ji B. Synergistic interactions of apigenin, naringin, quercetin and emodin on inhibition of 3T3-L1 preadipocyte differentiation and pancreas lipase activity. Obes Res Clin Pract. 2016;10(3):327-339. https://doi.org/10.1016/j.orcp.2015.08.004
  15. Goldwasser J, Cohen PY, Yang E, Balaguer P, Yarmush ML, Nahmias Y. Transcriptional regulation of human and rat hepatic lipid metabolism by the grapefruit flavonoid naringenin: role of PPARalpha, PPARgamma and LXRalpha. PLoS One. 2010;5(8):e12399. https://doi.org/10.1371/journal.pone.0012399
  16. Lien DN, Quynh NT, Phuc DV, Phong VC, Huong PT. Effect of pomelo (citrus grandis (l). osbeck) peel extract on lipid-carbohydrate metabolic enzymes and blood lipid, glucose parameters in experimental obese and diabetic mice. VNU J Sci Nat Sci Technol. 2010;26(4):224-232.
  17. Ha T, Trung TN, Phuong TT, Yim N, Chen QC, Bae K. The selected flavonol glycoside derived from Sophorae Flos improves glucose uptake and inhibits adipocyte differentiation via activation AMPK in 3T3-L1 cells. Bioorg Med Chem Lett. 2010;20(20):6076-6081. https://doi.org/10.1016/j.bmcl.2010.08.054
  18. Fang XK, Gao J, Zhu DN. Kaempferol and quercetin isolated from Euonymus alatus improve glucose uptake of 3T3-L1 cells without adipogenesis activity. Life Sci. 2008;82(11-12):615-622. https://doi.org/10.1016/j.lfs.2007.12.021
  19. Rajappa R, Sireesh D, Salai MB, Ramkumar KM, Sarvajayakesavulu S, Madhunapantula SV. Treatment with naringenin elevates the activity of transcription factor Nrf2 to protect pancreatic β-cells from streptozotocin-induced diabetes in vitro and in vivo. Front Pharmacol. 2019;9:1562. https://doi.org/10.3389/fphar.2018.01562
  20. Constantin RP, Constantin RP, Bracht A, Yamamoto NS, Ishii-Iwamoto EL, Constantin J. Molecular mechanisms of citrus flavanones on hepatic gluconeogenesis. Fitoterapia. 2014;92:148-162. https://doi.org/10.1016/j.fitote.2013.11.003
  21. Pu P, Gao DM, Mohamed S, Chen J, Zhang J, Zhou XY, et al. Naringin ameliorates metabolic syndrome by activating AMP-activated protein kinase in mice fed a high-fat diet. Arch Biochem Biophys. 2012;518(1):61-70. https://doi.org/10.1016/j.abb.2011.11.026
  22. Ceddia RB. The role of AMP-activated protein kinase in regulating white adipose tissue metabolism. Mol Cell Endocrinol. 2013;366(2):194-203. https://doi.org/10.1016/j.mce.2012.06.014
  23. Lee SH, Park YB, Bae KH, Bok SH, Kwon YK, Lee ES, et al. Cholesterol-lowering activity of naringenin via inhibition of 3-hydroxy-3-methylglutaryl coenzyme A reductase and acyl coenzyme A:cholesterol acyltransferase in rats. Ann Nutr Metab. 1999;43(3):173-180. https://doi.org/10.1159/000012783
  24. Jung UJ, Lee MK, Park YB, Kang MA, Choi MS. Effect of citrus flavonoids on lipid metabolism and glucose-regulating enzyme mRNA levels in type-2 diabetic mice. Int J Biochem Cell Biol. 2006;38(7):1134-1145. https://doi.org/10.1016/j.biocel.2005.12.002
  25. Matsumoto H, Sasaki K, Bessho T, Kobayashi E, Abe T, Sasazaki S, et al. The SNPs in the ACACA gene are effective on fatty acid composition in Holstein milk. Mol Biol Rep. 2012;39(9):8637-8644. https://doi.org/10.1007/s11033-012-1718-5
  26. Hillgartner FB, Salati LM, Goodridge AG. Physiological and molecular mechanisms involved in nutritional regulation of fatty acid synthesis. Physiol Rev. 1995;75(1):47-76. https://doi.org/10.1152/physrev.1995.75.1.47
  27. Jiang Z, Michal JJ, Tobey DJ, Daniels TF, Rule DC, Macneil MD. Significant associations of stearoyl-CoA desaturase (SCD1) gene with fat deposition and composition in skeletal muscle. Int J Biol Sci. 2008;4(6):345-351.
  28. Hall AM, Kou K, Chen Z, Pietka TA, Kumar M, Korenblat KM, et al. Evidence for regulated monoacylglycerol acyltransferase expression and activity in human liver. J Lipid Res. 2012;53(5):990-999. https://doi.org/10.1194/jlr.P025536
  29. Harris CA, Haas JT, Streeper RS, Stone SJ, Kumari M, Yang K, et al. DGAT enzymes are required for triacylglycerol synthesis and lipid droplets in adipocytes. J Lipid Res. 2011;52(4):657-667. https://doi.org/10.1194/jlr.M013003
  30. Smith SJ, Cases S, Jensen DR, Chen HC, Sande E, Tow B, et al. Obesity resistance and multiple mechanisms of triglyceride synthesis in mice lacking Dgat. Nat Genet. 2000;25(1):87-90. https://doi.org/10.1038/75651
  31. Peterfy M, Phan J, Xu P, Reue K. Lipodystrophy in the fld mouse results from mutation of a new gene encoding a nuclear protein, lipin. Nat Genet. 2001;27(1):121-124. https://doi.org/10.1038/83685
  32. Bonnefont JP, Djouadi F, Prip-Buus C, Gobin S, Munnich A, Bastin J. Carnitine palmitoyltransferases 1 and 2: biochemical, molecular and medical aspects. Mol Aspects Med. 2004;25(5-6):495-520. https://doi.org/10.1016/j.mam.2004.06.004
  33. Gao XF, Chen W, Kong XP, Xu AM, Wang ZG, Sweeney G, et al. Enhanced susceptibility of Cpt1c knockout mice to glucose intolerance induced by a high-fat diet involves elevated hepatic gluconeogenesis and decreased skeletal muscle glucose uptake. Diabetologia. 2009;52(5):912-920. https://doi.org/10.1007/s00125-009-1284-0
  34. Bjorbaek C. Central leptin receptor action and resistance in obesity. J Investig Med. 2009;57(7):789-794. https://doi.org/10.2310/jim.0b013e3181bb0d49
  35. Morton GJ, Niswender KD, Rhodes CJ, Myers MG Jr, Blevins JE, Baskin DG, et al. Arcuate nucleus-specific leptin receptor gene therapy attenuates the obesity phenotype of Koletsky (fa(k)/fa(k)) rats. Endocrinology. 2003;144(5):2016-2024. https://doi.org/10.1210/en.2002-0115
  36. Rebello CJ, Greenway FL, Lau FH, Lin Y, Stephens JM, Johnson WD, et al. Naringenin promotes thermogenic gene expression in human white adipose tissue. Obesity (Silver Spring). 2019;27(1):103-111. https://doi.org/10.1002/oby.22352
  37. Mauvoisin D, Mounier C. Hormonal and nutritional regulation of SCD1 gene expression. Biochimie. 2011;93(1):78-86. https://doi.org/10.1016/j.biochi.2010.08.001
  38. Guaita-Esteruelas S, Guma J, Masana L, Borras J. The peritumoural adipose tissue microenvironment and cancer. The roles of fatty acid binding protein 4 and fatty acid binding protein 5. Mol Cell Endocrinol. 2018;462(Pt B):107-118. https://doi.org/10.1016/j.mce.2017.02.002
  39. Eberle D, Clement K, Meyre D, Sahbatou M, Vaxillaire M, Le Gall A, et al. SREBF-1 gene polymorphisms are associated with obesity and type 2 diabetes in French obese and diabetic cohorts. Diabetes. 2004;53(8):2153-2157. https://doi.org/10.2337/diabetes.53.8.2153
  40. Quintana AM, Hernandez JA, Gonzalez CG. Functional analysis of the zebrafish ortholog of HMGCS1 reveals independent functions for cholesterol and isoprenoids in craniofacial development. PLoS One. 2017;12(7):e0180856. https://doi.org/10.1371/journal.pone.0180856
  41. Actis Dato V, Chiabrando GA. The role of low-density lipoprotein receptor-related protein 1 in lipid metabolism, glucose homeostasis and inflammation. Int J Mol Sci. 2018;19(6):1780. https://doi.org/10.3390/ijms19061780
  42. Hsiu SL, Huang TY, Hou YC, Chin DH, Chao PDL. Comparison of metabolic pharmacokinetics of naringin and naringenin in rabbits. Life Sci. 2002;70(13):1481-1489. https://doi.org/10.1016/S0024-3205(01)01491-6
  43. Kumar S, Pandey AK. Chemistry and biological activities of flavonoids: an overview. Sci World J. 2013;2013:162750. https://doi.org/10.1155/2013/162750
  44. Veitch NC, Grayer RJ. Flavonoids and their glycosides, including anthocyanins. Nat Prod Rep. 2011;28(10):1626-1695. https://doi.org/10.1039/c1np00044f
  45. Morand C, Manach C, Crespy V, Remesy C. Respective bioavailability of quercetin aglycone and its glycosides in a rat model. Biofactors. 2000;12(1-4):169-174. https://doi.org/10.1002/biof.5520120127
  46. Kren V. Glycoside vs. aglycon: the role of glycosidic residue in biological activity. In: Fraser-Reid BO, Tatsuta K, Thiem J, editors. Glycoscience. Berlin: Springer; 2008, 2589-2644.
  47. Havsteen BH. The biochemistry and medical significance of the flavonoids. Pharmacol Ther. 2002;96(2-3):67-202. https://doi.org/10.1016/S0163-7258(02)00298-X
  48. Xiao J. Dietary flavonoid aglycones and their glycosides: Which show better biological significance? Crit Rev Food Sci Nutr. 2017;57(9):1874-1905.
  49. Ribeiro IA, Ribeiro MHL. Naringin and naringenin determination and control in grapefruit juice by a validated HPLC method. Food Control. 2008;19(4):432-438. https://doi.org/10.1016/j.foodcont.2007.05.007
  50. Ameer B, Weintraub RA, Johnson JV, Yost RA, Rouseff RL. Flavanone absorption after naringin, hesperidin, and citrus administration. Clin Pharmacol Ther. 1996;60(1):34-40. https://doi.org/10.1016/S0009-9236(96)90164-2