DOI QR코드

DOI QR Code

Prophylactic role of Korean Red Ginseng in astrocytic mitochondrial biogenesis through HIF-1α

  • Park, Jinhong (Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University) ;
  • Lee, Minjae (Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University) ;
  • Kim, Minsu (Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University) ;
  • Moon, Sunhong (Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University) ;
  • Kim, Seunghee (Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University) ;
  • Kim, Sueun (Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University) ;
  • Koh, Seong-Ho (Department of Neurology, Hanyang University Guri Hospital) ;
  • Kim, Young-Myeong (Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University) ;
  • Choi, Yoon Kyung (Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University)
  • Received : 2021.03.02
  • Accepted : 2021.07.07
  • Published : 2022.05.01

Abstract

Background: Korean Red Ginseng extract (KRGE) has been used as a health supplement and herbal medicine. Astrocytes are one of the key cells in the central nervous system (CNS) and have bioenergetic potential as they stimulate mitochondrial biogenesis. They play a critical role in connecting the brain vasculature and nerves in the CNS. Methods: Brain samples from KRGE-administered mice were tested using immunohistochemistry. Treatment of human brain astrocytes with KRGE was subjected to assays such as proliferation, cytotoxicity, Mitotracker, ATP production, and O2 consumption rate as well as western blotting to demonstrate the expression of proteins related to mitochondria functions. The expression of hypoxia-inducible factor-1α (HIF-1α) was diminished utilizing siRNA transfection. Results: Brain samples from KRGE-administered mice harbored an increased number of GFAP-expressing astrocytes. KRGE triggered the proliferation of astrocytes in vitro. Enhanced mitochondrial biogenesis induced by KRGE was detected using Mitotracker staining, ATP production, and O2 consumption rate assays. The expression of proteins related to mitochondrial electron transport was increased in KRGE-treated astrocytes. These effects were blocked by HIF-1α knockdown. The factors secreted from KRGE-treated astrocytes were determined, revealing the expression of various cytokines and growth factors, especially those related to angiogenesis and neurogenesis. KRGE-treated astrocyte conditioned media enhanced the differentiation of adult neural stem cells into mature neurons, increasing the migration of endothelial cells, and these effects were reduced in the background of HIF-1α knockdown. Conclusion: Our findings suggest that KRGE exhibits prophylactic potential by stimulating astrocyte mitochondrial biogenesis through HIF-1α, resulting in improved neurovascular function.

Keywords

Acknowledgement

Active form of HIF-1α overexpressing vector (pcDNA3.1/HIF-1αP402A/P564G) was thankfully provided by Dr. Gregg L. Semenza (Johns Hopkins University, USA).

References

  1. Kim HJ, Kim P, Shin CY. A comprehensive review of the therapeutic and pharmacological effects of ginseng and ginsenosides in central nervous system. J Ginseng Res 2013;37(1):8-29. https://doi.org/10.5142/jgr.2013.37.8.
  2. Abbott NJ, Ronnback L, Hansson E. Astrocyte-endothelial interactions at the blood-brain barrier. Nature Reviews Neuroscience 2006;7(1):41-53. https://doi.org/10.1038/nrn1824
  3. Hayakawa K, Esposito E, Wang X, Terasaki Y, Liu Y, Xing C, et al. Transfer of mitochondria from astrocytes to neurons after stroke. Nature 2016;535(7613):551-5. https://doi.org/10.1038/nature18928.
  4. Oberheim NA, Takano T, Han X, He W, Lin JH, Wang F, et al. Uniquely hominid features of adult human astrocytes. J Neurosci 2009;29(10):3276-87. https://doi.org/10.1523/JNEUROSCI.4707-08.
  5. Hettiarachchi NT, Boyle JP, Dallas ML, Al-Owais MM, Scragg JL, Peers C. Heme oxygenase-1 derived carbon monoxide suppresses Abeta1-42 toxicity in astrocytes. Cell Death Dis 2017;8(6):e2884. https://doi.org/10.1038/cddis.2017.276.
  6. Chen-Roetling J, Benvenisti-Zarom L, Regan RF. Cultured astrocytes from heme oxygenase-1 knockout mice are more vulnerable to heme-mediated oxidative injury. J Neurosci Res 2005;82(6):802-10. https://doi.org/10.1002/jnr.20681.
  7. Lee BS, Heo J, Kim YM, Shim SM, Pae HO, Chung HT. Carbon monoxide mediates heme oxygenase 1 induction via Nrf2 activation in hepatoma cells. Biochem Biophys Res Commun 2006;343(3):965-72. https://doi.org/10.1016/j.bbrc.2006.03.058
  8. Tudor C, Lerner-Marmarosh N, Engelborghs Y, Gibbs PE, Maines MD. Biliverdin reductase is a transporter of haem into the nucleus and is essential for regulation of HO-1 gene expression by haematin. Biochem J 2008;413(3): 405-16. https://doi.org/10.1042/BJ20080018.
  9. Ryter SW, Choi AM. Targeting heme oxygenase-1 and carbon monoxide for therapeutic modulation of inflammation. Transl Res 2016;167(1):7-34. https://doi.org/10.1016/j.trsl.2015.06.011.
  10. Choi YK, Park JH, Baek YY, Won MH, Jeoung D, Lee H, et al. Carbon monoxide stimulates astrocytic mitochondrial biogenesis via L-type Ca2+ channel-mediated PGC-1alpha/ERRalpha activation. Biochem Biophys Res Commun 2016;479(2):297-304. https://doi.org/10.1016/j.bbrc.2016.09.063.
  11. Choi YK, Park JH, Yun JA, Cha JH, Kim Y, Won MH, et al. Heme oxygenase metabolites improve astrocytic mitochondrial function via a Ca2+-dependent HIF-1alpha/ERRalpha circuit. PLoS One 2018;13(8):e0202039. https://doi.org/10.1371/journal.pone.0202039.
  12. Badawi Y, Ramamoorthy P, Shi H. Hypoxia-inducible factor 1 protects hypoxic astrocytes against glutamate toxicity. ASN Neuro 2012;4(4):231-41. https://doi.org/10.1042/AN20120006.
  13. Choi SH, Bylykbashi E, Chatila ZK, Lee SW, Pulli B, Clemenson GD, et al. Combined adult neurogenesis and BDNF mimic exercise effects on cognition in an Alzheimer's mouse model. Science 2018;361(6406). https://doi.org/10.1126/science.aan8821.
  14. Min JK, Kim JH, Cho YL, Maeng YS, Lee SJ, Pyun BJ, et al. 20(S)-Ginsenoside Rg3 prevents endothelial cell apoptosis via inhibition of a mitochondrial caspase pathway. Biochem Biophys Res Commun 2006;349(3):987-94. https://doi.org/10.1016/j.bbrc.2006.08.129.
  15. Tian J, Fu F, Geng M, Jiang Y, Yang J, Jiang W, et al. Neuroprotective effect of 20(S)-ginsenoside Rg3 on cerebral ischemia in rats. Neurosci Lett 2005;374(2):92-7. https://doi.org/10.1016/j.neulet.2004.10.030.
  16. Chaban Y, Boekema EJ, Dudkina NV. Structures of mitochondrial oxidative phosphorylation supercomplexes and mechanisms for their stabilisation. Biochim Biophys Acta 2014;1837(4):418-26. https://doi.org/10.1016/j.bbabio.2013.10.004.
  17. Schubert D, Soucek T, Blouw B. The induction of HIF-1 reduces astrocyte activation by amyloid beta peptide. Eur J Neurosci 2009;29(7):1323-34. https://doi.org/10.1111/j.1460-9568.2009.06712.x.
  18. Hwang M, Song SH, Chang MS, Koh SH. Glia-like cells from human mesenchymal stem cells protect neural stem cells in an in vitro model of alzheimer's disease by reducing NLRP-3 inflammasome. Dement Neurocogn Disord 2021;20(1):1-8. https://doi.org/10.12779/dnd.2021.20.1.1.
  19. Choi YK, Maki T, Mandeville ET, Koh SH, Hayakawa K, Arai K, et al. Dual effects of carbon monoxide on pericytes and neurogenesis in traumatic brain injury. Nat Med 2016;22(11):1335-41. https://doi.org/10.1038/nm.4188.
  20. Tint I, Jean D, Baas PW, Black MM. Doublecortin associates with microtubules preferentially in regions of the axon displaying actin-rich protrusive structures. J Neurosci 2009;29(35):10995-1010. https://doi.org/10.1523/JNEUROSCI.3399-09.
  21. Na HJ, Hwang JY, Lee KS, Choi YK, Choe J, Kim JY, et al. TRAIL negatively regulates VEGF-induced angiogenesis via caspase-8-mediated enzymatic and non-enzymatic functions. Angiogenesis 2014;17(1):179-94. https://doi.org/10.1007/s10456-013-9387-0.
  22. Semenza GL. Life with oxygen. Science 2007;318(5847):62-4. https://doi.org/10.1126/science.1147949
  23. Choi YK, Kim CK, Lee H, Jeoung D, Ha KS, Kwon YG, et al. Carbon monoxide promotes VEGF expression by increasing HIF-1alpha protein level via two distinct mechanisms, translational activation and stabilization of HIF-1alpha protein. J Biol Chem 2010;285(42):32116-25. https://doi.org/10.1074/jbc.M110.131284
  24. Chan DA, Sutphin PD, Yen SE, Giaccia AJ. Coordinate regulation of the oxygen-dependent degradation domains of hypoxia-inducible factor 1 alpha. Mol Cell Biol 2005;25(15):6415-26. https://doi.org/10.1128/MCB.25.15.6415-6426.
  25. Ivan M, Kondo K, Yang H, Kim W, Valiando J, Ohh M, et al. HIFalpha targeted for VHL-mediated destruction by proline hydroxylation: implications for O2 sensing. Science 2001;292(5516):464-8. https://doi.org/10.1126/science.1059817
  26. Arenberg DA, Keane MP, DiGiovine B, Kunkel SL, Morris SB, Xue YY, et al. Epithelial-neutrophil activating peptide (ENA-78) is an important angiogenic factor in non-small cell lung cancer. J Clin Invest 1998;102(3):465-72. https://doi.org/10.1172/JCI3145.
  27. Caunt M, Hu L, Tang T, Brooks PC, Ibrahim S, Karpatkin S. Growth-regulated oncogene is pivotal in thrombin-induced angiogenesis. Cancer Res 2006;66(8):4125-32. https://doi.org/10.1158/0008-5472.CAN-05-2570.
  28. Chen Y, Zhong M, Liang L, Gu F, Peng H. Interleukin-17 induces angiogenesis in human choroidal endothelial cells in vitro. Invest Ophthalmol Vis Sci 2014;55(10):6968-75. https://doi.org/10.1167/iovs.14-15029.
  29. Enzmann V, Lecaude S, Kruschinski A, Vater A. CXCL12/SDF-1-Dependent retinal migration of endogenous bone marrow-derived stem cells improves visual function after pharmacologically induced retinal degeneration. Stem Cell Rev 2017;13(2):278-86. https://doi.org/10.1007/s12015-016-9706-0.
  30. Sun Y, Jin K, Xie L, Childs J, Mao XO, Logvinova A, et al. VEGF-induced neuroprotection, neurogenesis, and angiogenesis after focal cerebral ischemia. J Clin Invest 2003;111(12):1843-51. https://doi.org/10.1172/JCI17977.
  31. During MJ, Cao L. VEGF, a mediator of the effect of experience on hippocampal neurogenesis. Curr Alzheimer Res 2006;3(1):29-33. https://doi.org/10.2174/156720506775697133
  32. Cheng X, Wang H, Zhang X, Zhao S, Zhou Z, Mu X, et al. The role of SDF-1/CXCR4/CXCR7 in neuronal regeneration after cerebral ischemia. Front Neurosci 2017;11:590. https://doi.org/10.3389/fnins.2017.00590.
  33. Lin Y, Zhang JC, Yao CY, Wu Y, Abdelgawad AF, Yao SL, et al. Critical role of astrocytic interleukin-17 A in post-stroke survival and neuronal differentiation of neural precursor cells in adult mice. Cell Death Dis 2016;7(6):e2273. https://doi.org/10.1038/cddis.2015.284.
  34. Braun SM, Jessberger S. Adult neurogenesis and its role in neuropsychiatric disease, brain repair and normal brain function. Neuropathol Appl Neurobiol 2014;40(1):3-12. https://doi.org/10.1111/nan.12107.