DOI QR코드

DOI QR Code

Effect of Tartary Buckwheat Sprout on Non-Alcoholic Fatty Liver Disease through Anti-Histone Acetyltransferase Activity

쓴메밀 새싹 추출물의 히스톤 아세틸화 효소 활성 저해에 의한 비알코올성 지방간 억제 효능

  • Received : 2016.11.10
  • Accepted : 2017.01.09
  • Published : 2017.02.28

Abstract

Non-alcoholic fatty liver disease (NAFLD) is caused by chronic lipid accumulation due to dysregulation of lipid metabolism in the liver, and it is associated with various human diseases such as obesity, dyslipidemia, hypertension, and diabetes. Histone acetylation is a representative epigenetic mechanism regulated by histone acetyltransferases (HATs) and deacetylases. We observed that tartary buckwheat sprout (TBS) suppressed lipid accumulation in HepG2 cells through its anti-HAT activity. We showed that TBS was a novel HAT inhibitor with specificity for the major HAT enzyme p300. Importantly, TBS reduced acetylation of total and histone proteins, H3K9, H3K36, and H4K8, resulting in decreased transcriptional activities of sterol regulatory element-binding protein 1c, ATP citrate lyase, and fatty acid synthase. These results suggest that TBS inhibits the NAFLD transcription-modulating activity of lipogenesis-related genes through modification of histone acetylation.

본 연구에서는 쓴메밀 새싹 추출물(TBS)을 대상으로 histone acetyltransferase(HAT) 활성 저해능을 평가하고 oleic acid와 palmitic acid(OPA)를 이용하여 HepG2 세포에서 비알코올성 지방간을 유도하여 그 효과를 검토하였다. HeLa 세포의 nuclear extract(NE)를 HAT의 source로 하여 in vitro에서 TBS에 의한 HAT 활성 저해능을 평가한 결과 추출물의 처리에 의하여 HAT 활성이 억제됨을 관찰할 수 있었다. 또한, 대표적인 HAT 단백질인 p300과 CBP를 이용하여 동일한 방식으로 HAT 억제능을 평가한 결과 TBS 처리에 의하여 두 단백질 모두 활성이 감소하였으며, 특히 TBS는 p300의 활성을 특이적으로 저해함을 확인할 수 있었다. 그뿐만 아니라 HepG2 세포에 $400{\mu}M$의 oleic acid 및 $100{\mu}M$의 palmitic acid와 함께 $200{\mu}g/mL$, $500{\mu}g/mL$의 TBS를 처리한 후 NE를 이용하여 세포 내 HAT 활성을 측정한 결과 역시 추출물 처리에 의하여 세포 내 HAT 활성이 저해되어 있음이 관찰되었다. TBS에 의한 HAT 활성의 억제는 세포 내 다양한 단백질들의 아세틸화 저해와 지질축적에 의하여 아세틸화 변형을 일으키는 것으로 알려진 histone H3K9, H4K8의 아세틸화 및 H3K36의 아세틸화를 감소시켰으며, 세포 내 지질합성과 관련된 대표적 유전자인 SREBP1c, ACLY, FAS의 전사 활성 역시 저해함을 관찰하였다. 이와 같은 변화를 통하여 OPA에 의하여 HepG2 세포 내에 축적되었던 지질은 TBS의 처리에 의하여 효과적으로 감소하였으며 이때 처리된 OPA와 소재에 의한 세포 내 독성은 관찰되지 않았다. 그러므로 이러한 결과는 TBS에 의한 HAT 활성의 저해가 히스톤 단백질의 아세틸활 변형을 억제하고 이를 통하여 지방 합성 관련 유전자들의 전사 활성을 감소시켜 결과적으로 세포 내 지질축적을 방지하는 것으로 생각되며, TBS은 비알코올성 지방간질환의 예방에 좋은 천연물 소재로 활용될 수 있을 것이라 여겨진다.

Keywords

References

  1. Browning JD, Szczepaniak LS, Dobbins R, Nuremberg P, Horton JD, Cohen JC, Grundy SM, Hobbs HH. 2004. Prevalence of hepatic steatosis in an urban population in the United States: impact of ethnicity. Hepatology 40: 1387-1395. https://doi.org/10.1002/hep.20466
  2. Byrne CD, Targher G. 2015. NAFLD: a multisystem disease. J Hepatol 62: S47-S64. https://doi.org/10.1016/j.jhep.2014.12.012
  3. Jeong EH, Jun DW, Cho YK, Choe YG, Ryu S, Lee SM, Jang EC. 2013. Regional prevalence of non-alcoholic fatty liver disease in Seoul and Gyeonggi-do, Korea. Clin Mol Hepatol 19: 266-272. https://doi.org/10.3350/cmh.2013.19.3.266
  4. Armstrong MJ, Adams LA, Canbay A, Syn WK. 2014. Extrahepatic complications of nonalcoholic fatty liver disease. Hepatology 59: 1174-1197. https://doi.org/10.1002/hep.26717
  5. Tannapfel A, Denk H, Dienes HP, Langner C, Schirmacher P, Trauner M, Flott-Rahmel B. 2011. Histopathological diagnosis of non-alcoholic and alcoholic fatty liver disease. Virchows Arch 458: 511-523. https://doi.org/10.1007/s00428-011-1066-1
  6. Burt AD, Mutton A, Day CP. 1998. Diagnosis and interpretation of steatosis and steatohepatitis. Semin Diagn Pathol 15: 246-258.
  7. Xu JY, Zhang L, Li ZP, Ji G. 2015. Natural products on nonalcoholic fatty liver disease. Curr Drug Targets 16: 1347-1355. https://doi.org/10.2174/1389450116666150531155711
  8. Jin J, Iakova P, Breaux M, Sullivan E, Jawanmardi N, Chen D, Jiang Y, Medrano EM, Timchenko NA. 2013. Increased expression of enzymes of triglyceride synthesis is essential for the development of hepatic steatosis. Cell Rep 3: 831-843. https://doi.org/10.1016/j.celrep.2013.02.009
  9. Bricambert J, Miranda J, Benhamed F, Girard J, Postic C, Dentin R. 2010. Salt-inducible kinase 2 links transcriptional coactivator p300 phosphorylation to the prevention of ChREBP-dependent hepatic steatosis in mice. J Clin Invest 120: 4316-4331. https://doi.org/10.1172/JCI41624
  10. Sun Z, Miller RA, Patel RT, Chen J, Dhir R, Wang H, Zhang D, Graham MJ, Unterman TG, Shulman GI, Sztalryd C, Bennett MJ, Ahima RS, Birnbaum MJ, Lazar MA. 2012. Hepatic Hdac3 promotes gluconeogenesis by repressing lipid synthesis and sequestration. Nat Med 18: 934-942. https://doi.org/10.1038/nm.2744
  11. Denechaud PD, Dentin R, Girard J, Postic C. 2008. Role of ChREBP in hepatic steatosis and insulin resistance. FEBS Lett 528: 68-73.
  12. Jin J, Valanejad L, Nguyen TP, Lewis K, Wright M, Cast A, Stock L, Timchenko L, Timchenko NA. 2016. Activation of CDK4 triggers development of non-alcoholic fatty liver disease. Cell Rep 16: 744-756. https://doi.org/10.1016/j.celrep.2016.06.019
  13. Bhaskara S, Knutson SK, Jiang G, Chandrasekharan MB, Wilson AJ, Zheng S, Yenamandra A, Locke K, Yuan JL, Bonine-Summers AR, Wells CE, Kaiser JF, Washington MK, Zhao Z, Wagner FF, Sun ZW, Xia F, Holson EB, Khabele D, Hiebert SW. 2010. Hdac3 is essential for the maintenance of chromatin structure and genome stability. Cancer Cell 18: 436-447. https://doi.org/10.1016/j.ccr.2010.10.022
  14. Tian Y, Wong VW, Chan HL, Cheng AS. 2013. Epigenetic regulation of hepatocellular carcinoma in non-alcoholic fatty liver disease. Semin Cancer Biol 23: 471-482. https://doi.org/10.1016/j.semcancer.2013.08.010
  15. Herranz D, Serrano M. 2010. SIRT1: recent lessons from mouse models. Nat Rev Cancer 10: 819-823. https://doi.org/10.1038/nrc2962
  16. Escande C, Chini CC, Nin V, Dykhouse KM, Novak CM, Levine J, van Deursen J, Gores GJ, Chen J, Lou Z, Chini EN. 2010. Deleted in breast cancer-1 regulates SIRT1 activity and contributes to high-fat diet-induced liver steatosis in mice. J Clin Invest 120: 545-558. https://doi.org/10.1172/JCI39319
  17. Purushotham A, Schug TT, Xu Q, Surapureddi S, Guo X, Li X. 2009. Hepatocyte-specific deletion of SIRT1 alters fatty acid metabolism and results in hepatic steatosis and inflammation. Cell Metab 9: 327-338. https://doi.org/10.1016/j.cmet.2009.02.006
  18. Kim SL, Kim SK, Park CH. 2004. Introduction and nutritional evaluation of buckwheat sprouts as a new vegetable. Food Res Int 37: 319-327. https://doi.org/10.1016/j.foodres.2003.12.008
  19. Liu CL, Chen YS, Yang JH, Chiang BH. 2007. Antioxidant activity of tartary (Fagopyrum tataricum (L.) Gaertn.) and common (Fagopyrum esculentum moench) buckwheat sprouts. J Agric Food Chem 56: 173-178.
  20. Nam TG, Lee SM, Park JH, Kim DO, Baek NI, Eom SH. 2015. Flavonoid analysis of buckwheat sprouts. Food Chem 170: 97-101. https://doi.org/10.1016/j.foodchem.2014.08.067
  21. Kumar S, Pandey AK. 2013. Chemistry and biological activities of flavonoids: An overview. Sci World J 2013: 162750.
  22. Kim JK, Kim SK. 2005. Compositions and pasting properties of Fagopyrum esculentum and Fagopyrum tartaricum endosperm flour. Korean J Food Sci Technol 37: 149-153.
  23. Lee DG, Jang IS, Yang KE, Yoon SJ, Baek S, Lee JY, Suzuki T, Chung KY, Woo SH, Choi JS. 2016. Effect of rutin from tartary buckwheat sprout on serum glucose-lowering in animal model of type 2 diabetes. Acta Pharm 66: 297-302. https://doi.org/10.1515/acph-2016-0021
  24. Christa K, Soral-Smietana M. 2008. Buckwheat grains and buckwheat products-Nutritional and prophylactic value of their components: A review. Czech J Food Sci 26: 153-162. https://doi.org/10.17221/1602-CJFS
  25. Li D, Li X, Gao C, Chen X. 2009. Tartary buckwheat, Fagopyrum tataricum (Linn) Gaertn: A functional food ingredient from Eastern Asia. Eur J Plant Sci Biotechnol 3: 45-48.
  26. Choi KC, Lee YH, Jung MG, Kwon SH, Kim MJ, Jun WJ, Lee J, Lee JM, Yoon HG. 2009. Gallic acid suppresses lipopolysaccharide-induced nuclear factor-${\kappa}B$ signaling by preventing RelA acetylation in A549 lung cancer cells. Mol Cancer Res 7: 2011-2021. https://doi.org/10.1158/1541-7786.MCR-09-0239
  27. Choi KC, Jung MG, Lee YH, Yoon JC, Kwon SH, Kang HB, Kim MJ, Cha JH, Kim YJ, Jun WJ, Lee JM, Yoon HG. 2009. Epigallocatechin-3-gallate, a histone acetyltransferase inhibitor, inhibits EBV-induced B lymphocyte transformation via suppression of RelA acetylation. Cancer Res 69: 583-592. https://doi.org/10.1158/0008-5472.CAN-08-2442
  28. Saadat N, Gupta SV. 2012. Potential role of garcinol as an anticancer agent. J Oncol 2012: 647206.
  29. Mikula M, Majewska A, Ledwon JK, Dzwonek A, Ostrowski J. 2014. Obesity increases histone H3 lysine 9 and 18 acetylation at Tnfa and Ccl2 genes in mouse liver. Int J Mol Med 34: 1647-1654. https://doi.org/10.3892/ijmm.2014.1958
  30. Galdieri L, Zhang T, Rogerson D, Lleshi R, Vancura A. 2014. Protein acetylation and acetyl coenzyme a metabolism in budding yeast. Eukaryot Cell 13: 1472-1483. https://doi.org/10.1128/EC.00189-14
  31. Chow JD, Lawrence RT, Healy ME, Dominy JE, Liao JA, Breen DS, Byrne FL, Kenwood BM, Lackner C, Okutsu S, Mas VR, Caldwell SH, Tomsig JL, Cooney GJ, Puigserver PB, Turner N, James DE, Villen J, Hoehn KL. 2014. Genetic inhibition of hepatic acetyl-CoA carboxylase activity increases liver fat and alters global protein acetylation. Mol Metab 3: 419-431. https://doi.org/10.1016/j.molmet.2014.02.004
  32. Hebbes TR, Thorne AW, Crane-Robinson C. 1988. A direct link between core histone acetylation and transcriptionally active chromatin. EMBO J 7: 1397-1402.
  33. Hong L, Schroth GP, Matthews H, Yau P, Bradbury EM. 1993. Studies of the DNA binding properties of histone H4 amino terminus. Thermal denaturation studies reveal that acetylation markedly reduces the binding constant of the H4 "tail" to DNA. J Biol Chem 268: 305-314.
  34. Vettese-Dadey M, Grant PA, Hebbes TR, Crane-Robinson C, Allis CD, Workman JL. 1996. Acetylation of histone H4 plays a primary role in enhancing transcription factor binding to nucleosomal DNA in vitro. EMBO J 15: 2508-2518.

Cited by

  1. Comparative Study of the Effects of Light Controlled Germination Conditions on Saponarin Content in Barley Sprouts and Lipid Accumulation Suppression in HepG2 Hepatocyte and 3T3-L1 Adipocyte Cells Usi vol.25, pp.22, 2020, https://doi.org/10.3390/molecules25225349