DOI QR코드

DOI QR Code

Dieckol Attenuates Microglia-mediated Neuronal Cell Death via ERK, Akt and NADPH Oxidase-mediated Pathways

  • Cui, Yanji (Department of Physiology, Jeju National University School of Medicine) ;
  • Park, Jee-Yun (Department of Physiology, Jeju National University School of Medicine) ;
  • Wu, Jinji (Department of Physiology, Jeju National University School of Medicine) ;
  • Lee, Ji Hyung (Department of Physiology, Jeju National University School of Medicine) ;
  • Yang, Yoon-Sil (Department of Physiology, Jeju National University School of Medicine) ;
  • Kang, Moon-Seok (Department of Physiology, Jeju National University School of Medicine) ;
  • Jung, Sung-Cherl (Department of Physiology, Jeju National University School of Medicine) ;
  • Park, Joo Min (Department of Physiology, Jeju National University School of Medicine) ;
  • Yoo, Eun-Sook (Department of Pharmacology, Jeju National University School of Medicine) ;
  • Kim, Seong-Ho (BotaMedi Inc. 307 Jeju Bio-industry Center) ;
  • Ahn Jo, Sangmee (Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine and Department of Pharmacology, Dankook University) ;
  • Suk, Kyoungho (Department of Pharmacology, Kyungpook National University School of Medicine) ;
  • Eun, Su-Yong (Department of Physiology, Jeju National University School of Medicine)
  • Received : 2014.11.10
  • Accepted : 2015.02.23
  • Published : 2015.05.01

Abstract

Excessive microglial activation and subsequent neuroinflammation lead to synaptic loss and dysfunction as well as neuronal cell death, which are involved in the pathogenesis and progression of several neurodegenerative diseases. Thus, the regulation of microglial activation has been evaluated as effective therapeutic strategies. Although dieckol (DEK), one of the phlorotannins isolated from marine brown alga Ecklonia cava, has been previously reported to inhibit microglial activation, the molecular mechanism is still unclear. Therefore, we investigated here molecular mechanism of DEK via extracellular signal-regulated kinase (ERK), Akt and nicotinamide adenine dinuclelotide phosphate (NADPH) oxidase-mediated pathways. In addition, the neuroprotective mechanism of DEK was investigated in microglia-mediated neurotoxicity models such as neuron-microglia co-culture and microglial conditioned media system. Our results demonstrated that treatment of anti-oxidant DEK potently suppressed phosphorylation of ERK in lipopolysaccharide (LPS, $1{\mu}g/ml$)-stimulated BV-2 microglia. In addition, DEK markedly attenuated Akt phosphorylation and increased expression of $gp91^{phox}$, which is the catalytic component of NADPH oxidase complex responsible for microglial reactive oxygen species (ROS) generation. Finally, DEK significantly attenuated neuronal cell death that is induced by treatment of microglial conditioned media containing neurotoxic secretary molecules. These neuroprotective effects of DEK were also confirmed in a neuron-microglia co-culture system using enhanced green fluorescent protein (EGFP)-transfected B35 neuroblastoma cell line. Taken together, these results suggest that DEK suppresses excessive microglial activation and microglia-mediated neuronal cell death via downregulation of ERK, Akt and NADPH oxidase-mediated pathways.

Keywords

References

  1. Aschner M, Allen JW, Kimelberg HK, LoPachin RM, Streit WJ. Glial cells in neurotoxicity development. Annu Rev Pharmacol Toxicol. 1999;39:151-173. https://doi.org/10.1146/annurev.pharmtox.39.1.151
  2. Cui Y, Wu J, Jung SC, Park DB, Maeng YH, Hong JY, Kim SJ, Lee SR, Kim SJ, Kim SJ, Eun SY. Anti-neuroinflammatory activity of nobiletin on suppression of microglial activation. Biol Pharm Bull. 2010;33:1814-1821. https://doi.org/10.1248/bpb.33.1814
  3. Ock J, Han HS, Hong SH, Lee SY, Han YM, Kwon BM, Suk K. Obovatol attenuates microglia-mediated neuroinflammation by modulating redox regulation. Br J Pharmacol. 2010;159: 1646-1662. https://doi.org/10.1111/j.1476-5381.2010.00659.x
  4. Lee JW, Cheong IY, Kim HS, Lee JJ, Lee YS, Kwon YS, Kim MJ, Lee HJ, Kim SS, Chun W. Anti-inflammatory activity of 1-docosanoyl cafferate isolated from rhus verniciflua in LPSstimulated BV2 microglial cells. Korean J Physiol Pharmacol. 2011;15:9-15. https://doi.org/10.4196/kjpp.2011.15.1.9
  5. Cheret C, Gervais A, Lelli A, Colin C, Amar L, Ravassard P, Mallet J, Cumano A, Krause KH, Mallat M. Neurotoxic activation of microglia is promoted by a nox1-dependent NADPH oxidase. J Neurosci. 2008;28:12039-12051. https://doi.org/10.1523/JNEUROSCI.3568-08.2008
  6. Cooney SJ, Bermudez-Sabogal SL, Byrnes KR. Cellular and temporal expression of NADPH oxidase (NOX) isotypes after brain injury. J Neuroinflammation. 2013;10:155. https://doi.org/10.1186/1742-2094-10-155
  7. Babior BM. NADPH oxidase: an update. Blood. 1999;93: 1464-1476.
  8. DeLeo FR, Quinn MT. Assembly of the phagocyte NADPH oxidase: molecular interaction of oxidase proteins. J Leukoc Biol. 1996;60:677-691. https://doi.org/10.1002/jlb.60.6.677
  9. Oh YT, Lee JY, Lee J, Kim H, Yoon KS, Choe W, Kang I. Oleic acid reduces lipopolysaccharide-induced expression of iNOS and COX-2 in BV2 murine microglial cells: possible involvement of reactive oxygen species, p38 MAPK, and IKK/NF-kappaB signaling pathways. Neurosci Lett. 2009;464:93-97. https://doi.org/10.1016/j.neulet.2009.08.040
  10. Sun HN, Kim SU, Lee MS, Kim SK, Kim JM, Yim M, Yu DY, Lee DS. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-dependent activation of phosphoinositide 3-kinase and p38 mitogen-activated protein kinase signal pathways is required for lipopolysaccharide-induced microglial phagocytosis. Biol Pharm Bull. 2008;31:1711-1715. https://doi.org/10.1248/bpb.31.1711
  11. Zhang L, Wu C, Zhao S, Yuan D, Lian G, Wang X, Wang L, Yang J. Demethoxycurcumin, a natural derivative of curcumin attenuates LPS-induced pro-inflammatory responses through down-regulation of intracellular ROS-related MAPK/NF-kappaB signaling pathways in N9 microglia induced by lipopolysaccharide. Int Immunopharmacol. 2010;10:331-338. https://doi.org/10.1016/j.intimp.2009.12.004
  12. Kim AR, Shin TS, Lee MS, Park JY, Park KE, Yoon NY, Kim JS, Choi JS, Jang BC, Byun DS, Park NK, Kim HR. Isolation and identification of phlorotannins from Ecklonia stolonifera with antioxidant and anti-inflammatory properties. J Agric Food Chem. 2009;57:3483-3489. https://doi.org/10.1021/jf900820x
  13. Ryu B, Li Y, Qian ZJ, Kim MM, Kim SK. Differentiation of human osteosarcoma cells by isolated phlorotannins is subtly linked to COX-2, iNOS, MMPs, and MAPK signaling: implication for chronic articular disease. Chem Biol Interact. 2009;179: 192-201. https://doi.org/10.1016/j.cbi.2009.01.006
  14. Jung WK, Heo SJ, Jeon YJ, Lee CM, Park YM, Byun HG, Choi YH, Park SG, Choi IW. Inhibitory effects and molecular mechanism of dieckol isolated from marine brown alga on COX-2 and iNOS in microglial cells. J Agric Food Chem. 2009; 57:4439-4446. https://doi.org/10.1021/jf9003913
  15. Li Y, Qian ZJ, Ryu B, Lee SH, Kim MM, Kim SK. Chemical components and its antioxidant properties in vitro: an edible marine brown alga, Ecklonia cava. Bioorg Med Chem. 2009; 17:1963-1973. https://doi.org/10.1016/j.bmc.2009.01.031
  16. Eun SY, Hong YH, Kim EH, Jeon H, Suh YH, Lee JE, Jo C, Jo SA, Kim J. Glutamate receptor-mediated regulation of c-fos expression in cultured microglia. Biochem Biophys Res Commun. 2004;325:320-327. https://doi.org/10.1016/j.bbrc.2004.10.035
  17. Bocchini V, Mazzolla R, Barluzzi R, Blasi E, Sick P, Kettenmann H. An immortalized cell line expresses properties of activated microglial cells. J Neurosci Res. 1992;31:616-621. https://doi.org/10.1002/jnr.490310405
  18. Breyer A, Elstner M, Gillessen T, Weiser D, Elstner E. Glutamate-induced cell death in neuronal HT22 cells is attenuated by extracts from St. John's wort (Hypericum perforatum L.). Phytomedicine. 2007;14:250-255. https://doi.org/10.1016/j.phymed.2007.02.001
  19. Maher P, Davis JB. The role of monoamine metabolism in oxidative glutamate toxicity. J Neurosci. 1996;16:6394-6401. https://doi.org/10.1523/JNEUROSCI.16-20-06394.1996
  20. Schubert D, Heinemann S, Carlisle W, Tarikas H, Kimes B, Patrick J, Steinbach JH, Culp W, Brandt BL. Clonal cell lines from the rat central nervous system. Nature. 1974;249:224-227. https://doi.org/10.1038/249224a0
  21. Cui Y, Wu J, Jung SC, Kim GO, Kyeong Ko R, Lee HJ, Yoo ES, Kang HK, Suk K, Eun SY. Neuroprotective effect of methyl lucidone against microglia-mediated neurotoxicity. Eur J Pharmacol. 2012;690:4-12. https://doi.org/10.1016/j.ejphar.2012.05.041
  22. Park GH, Jeon SJ, Ko HM, Ryu JR, Lee JM, Kim HY, Han SH, Kang YS, Park SH, Shin CY, Ko KH. Activation of microglial cells via protease-activated receptor 2 mediates neuronal cell death in cultured rat primary neuron. Nitric Oxide. 2010;22:18-29. https://doi.org/10.1016/j.niox.2009.10.008
  23. Wang S, Wang H, Guo H, Kang L, Gao X, Hu L. Neuroprotection of Scutellarin is mediated by inhibition of microglial inflammatory activation. Neuroscience. 2011;185:150-160. https://doi.org/10.1016/j.neuroscience.2011.04.005
  24. Dohi K, Ohtaki H, Nakamachi T, Yofu S, Satoh K, Miyamoto K, Song D, Tsunawaki S, Shioda S, Aruga T. Gp91phox (NOX2) in classically activated microglia exacerbates traumatic brain injury. J Neuroinflammation. 2010;7:41. https://doi.org/10.1186/1742-2094-7-41
  25. Zhao S, Zhang L, Lian G, Wang X, Zhang H, Yao X, Yang J, Wu C. Sildenafil attenuates LPS-induced pro-inflammatory responses through down-regulation of intracellular ROS-related MAPK/NF- B signaling pathways in N9 microglia. Int Immunopharmacol. 2011;11:468-474. https://doi.org/10.1016/j.intimp.2010.12.017
  26. Tsai HH, Lee WR, Wang PH, Cheng KT, Chen YC, Shen SC. Propionibacterium acnes-induced iNOS and COX-2 protein expression via ROS-dependent NF- B and AP-1 activation in macrophages. J Dermatol Sci. 2013;69:122-131. https://doi.org/10.1016/j.jdermsci.2012.10.009
  27. Zhang H, Wang ZW, Wu HB, Li Z, Li LC, Hu XP, Ren ZL, Li BJ, Hu ZP. Transforming growth factor- 1 induces matrix metalloproteinase-9 expression in rat vascular smooth muscle cells via ROS-dependent ERK-NF- B pathways. Mol Cell Biochem. 2013;375:11-21.
  28. Ko HM, Koppula S, Kim BW, Kim IS, Hwang BY, Suk K, Park EJ, Choi DK. Inflexin attenuates proinflammatory responses and nuclear factor-kappaB activation in LPS-treated microglia. Eur J Pharmacol. 2010;633:98-106. https://doi.org/10.1016/j.ejphar.2010.02.011
  29. Lin HY, Tang CH, Chen YH, Wei IH, Chen JH, Lai CH, Lu DY. Peptidoglycan enhances proinflammatory cytokine expression through the TLR2 receptor, MyD88, phosphatidylinositol 3-kinase/AKT and NF-kappaB pathways in BV-2 microglia. Int Immunopharmacol. 2010;10:883-891. https://doi.org/10.1016/j.intimp.2010.04.026
  30. Karunakaran S, Ravindranath V. Activation of p38 MAPK in the substantia nigra leads to nuclear translocation of NFkappaB in MPTP-treated mice: implication in Parkinson's disease. J Neurochem. 2009;109:1791-1799. https://doi.org/10.1111/j.1471-4159.2009.06112.x
  31. Manea A, Tanase LI, Raicu M, Simionescu M. Transcriptional regulation of NADPH oxidase isoforms, Nox1 and Nox4, by nuclear factor-kappaB in human aortic smooth muscle cells. Biochem Biophys Res Commun. 2010;396:901-907. https://doi.org/10.1016/j.bbrc.2010.05.019
  32. Ago T, Kuroda J, Kamouchi M, Sadoshima J, Kitazono T. Pathophysiological roles of NADPH oxidase/nox family proteins in the vascular system. -Review and perspective-. Circ J. 2011;75:1791-1800. https://doi.org/10.1253/circj.CJ-11-0388
  33. Beckman JS, Koppenol WH. Nitric oxide, superoxide, and peroxynitrite: the good, the bad, and ugly. Am J Physiol. 1996; 271:C1424-1437. https://doi.org/10.1152/ajpcell.1996.271.5.C1424
  34. Gibbons HM, Dragunow M. Microglia induce neural cell death via a proximity-dependent mechanism involving nitric oxide. Brain Res. 2006;1084:1-15. https://doi.org/10.1016/j.brainres.2006.02.032
  35. Rhee SG, Chang TS, Bae YS, Lee SR, Kang SW. Cellular regulation by hydrogen peroxide. J Am Soc Nephrol. 2003;14(8 Suppl 3):S211-215. https://doi.org/10.1097/01.ASN.0000077404.45564.7E
  36. Hattori K, Naguro I, Runchel C, Ichijo H. The roles of ASK family proteins in stress responses and diseases. Cell Commun Signal. 2009;7:9. https://doi.org/10.1186/1478-811X-7-9
  37. Huo Y, Rangarajan P, Ling EA, Dheen ST. Dexamethasone inhibits the Nox-dependent ROS production via suppression of MKP-1-dependent MAPK pathways in activated microglia. BMC Neurosci. 2011;12:49. https://doi.org/10.1186/1471-2202-12-49
  38. Yang CS, Shin DM, Lee HM, Son JW, Lee SJ, Akira S, Gougerot-Pocidalo MA, El-Benna J, Ichijo H, Jo EK. ASK1-p38 MAPK-p47phox activation is essential for inflammatory responses during tuberculosis via TLR2-ROS signalling. Cell Microbiol. 2008;10:741-754. https://doi.org/10.1111/j.1462-5822.2007.01081.x
  39. Yang CS, Lee HM, Lee JY, Kim JA, Lee SJ, Shin DM, Lee YH, Lee DS, El-Benna J, Jo EK. Reactive oxygen species and p47phox activation are essential for the Mycobacterium tuberculosis-induced pro-inflammatory response in murine microglia. J Neuroinflammation. 2007;4:27. https://doi.org/10.1186/1742-2094-4-27
  40. Tanga FY, Nutile-McMenemy N, DeLeo JA. The CNS role of Toll-like receptor 4 in innate neuroimmunity and painful neuropathy. Proc Natl Acad Sci U S A. 2005;102:5856-5861. https://doi.org/10.1073/pnas.0501634102
  41. Cao CX, Yang QW, Lv FL, Cui J, Fu HB, Wang JZ. Reduced cerebral ischemia-reperfusion injury in Toll-like receptor 4 deficient mice. Biochem Biophys Res Commun. 2007;353:509-514. https://doi.org/10.1016/j.bbrc.2006.12.057
  42. Boivin A, Pineau I, Barrette B, Filali M, Vallieres N, Rivest S, Lacroix S. Toll-like receptor signaling is critical for Wallerian degeneration and functional recovery after peripheral nerve injury. J Neurosci. 2007;27:12565-12576. https://doi.org/10.1523/JNEUROSCI.3027-07.2007
  43. Kim D, Kim MA, Cho IH, Kim MS, Lee S, Jo EK, Choi SY, Park K, Kim JS, Akira S, Na HS, Oh SB, Lee SJ. A critical role of toll-like receptor 2 in nerve injury-induced spinal cord glial cell activation and pain hypersensitivity. J Biol Chem. 2007;282:14975-14983. https://doi.org/10.1074/jbc.M607277200

Cited by

  1. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  2. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  3. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  4. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  5. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  6. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  7. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  8. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  9. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  10. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  11. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  12. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  13. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  14. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  15. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  16. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  17. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  18. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  19. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  20. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  21. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  22. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  23. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  24. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  25. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  26. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  27. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  28. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  29. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  30. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  31. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  32. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  33. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  34. Algae as Functional Foods for the Elderly vol.7, pp.12, 2016, https://doi.org/10.4236/fns.2016.712107
  35. Recent advances in pharmacological research on Ecklonia species: a review vol.40, pp.9, 2017, https://doi.org/10.1007/s12272-017-0948-4
  36. Pristimerin Inhibits LPS-Triggered Neurotoxicity in BV-2 Microglia Cells Through Modulating IRAK1/TRAF6/TAK1-Mediated NF-κB and AP-1 Signaling Pathways In Vitro vol.33, pp.2, 2015, https://doi.org/10.1007/s12640-017-9837-3
  37. Marine-derived protein kinase inhibitors for neuroinflammatory diseases vol.17, pp.None, 2018, https://doi.org/10.1186/s12938-018-0477-5
  38. HDAC8 Inhibitor WK2-16 Therapeutically Targets Lipopolysaccharide-Induced Mouse Model of Neuroinflammation and Microglial Activation vol.20, pp.2, 2019, https://doi.org/10.3390/ijms20020410
  39. Anti-Neuroinflammatory Property of Phlorotannins from Ecklonia cava on Aβ 25-35 -Induced Damage in PC12 Cells vol.17, pp.1, 2015, https://doi.org/10.3390/md17010007
  40. Radioprotective efficacy of dieckol against gamma radiation-induced cellular damage in hepatocyte cells vol.392, pp.8, 2015, https://doi.org/10.1007/s00210-019-01652-z
  41. Conformational Isomerism for Eckol and its Skeleton: Theoretical Study vol.40, pp.10, 2015, https://doi.org/10.1002/bkcs.11852
  42. Oleanolic Acid Exerts a Neuroprotective Effect Against Microglial Cell Activation by Modulating Cytokine Release and Antioxidant Defense Systems vol.9, pp.11, 2015, https://doi.org/10.3390/biom9110683
  43. Dieckol: an algal polyphenol attenuates urban fine dust-induced inflammation in RAW 264.7 cells via the activation of anti-inflammatory and antioxidant signaling pathways vol.32, pp.4, 2015, https://doi.org/10.1007/s10811-019-01964-w
  44. MicroRNA-138 promotes neuroblastoma SH-SY5Y cell apoptosis by directly targeting DEK in Alzheimer’s disease cell model vol.21, pp.1, 2015, https://doi.org/10.1186/s12868-020-00579-z
  45. Chemical Analysis of the Ingredients of 20% Aqueous Ethanol Extract of Nardostachys jatamansi through Phytochemical Study and Evaluation of Anti-Neuroinflammatory Component vol.2021, pp.None, 2015, https://doi.org/10.1155/2021/5901653
  46. Dieckol Ameliorates Aβ Production via PI3K/Akt/GSK-3β Regulated APP Processing in SweAPP N2a Cell vol.19, pp.3, 2015, https://doi.org/10.3390/md19030152
  47. Antineuroinflammatory Effects of 7,3',4'-Trihydroxyisoflavone in Lipopolysaccharide-Stimulated BV2 Microglial Cells through MAPK and NF-κB Signaling Suppression vol.29, pp.2, 2015, https://doi.org/10.4062/biomolther.2020.093
  48. In Vitro and In Silico Characterization of G-Protein Coupled Receptor (GPCR) Targets of Phlorofucofuroeckol-A and Dieckol vol.19, pp.6, 2015, https://doi.org/10.3390/md19060326
  49. Dieckol: a brown algal phlorotannin with biological potential vol.142, pp.None, 2015, https://doi.org/10.1016/j.biopha.2021.111988