DOI QR코드

DOI QR Code

Notoginseng leaf triterpenes ameliorates mitochondrial oxidative injury via the NAMPT-SIRT1/2/3 signaling pathways in cerebral ischemic model rats

  • Weijie, Xie (Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences) ;
  • Ting, Zhu (Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences) ;
  • Ping, Zhou (Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences) ;
  • Huibo, Xu (Jilin Academy of Chinese Medicine) ;
  • Xiangbao, Meng (Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences) ;
  • Tao, Ding (Jilin Academy of Chinese Medicine) ;
  • Fengwei, Nan (Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences) ;
  • Guibo, Sun (Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences) ;
  • Xiaobo, Sun (Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences)
  • Received : 2020.05.09
  • Accepted : 2020.11.16
  • Published : 2023.03.02

Abstract

Background: Due to the interrupted blood supply in cerebral ischemic stroke (CIS), ischemic and hypoxia results in neuronal depolarization, insufficient NAD+, excessive levels of ROS, mitochondrial damages, and energy metabolism disorders, which triggers the ischemic cascades. Currently, improvement of mitochondrial functions and energy metabolism is as a vital therapeutic target and clinical strategy. Hence, it is greatly crucial to look for neuroprotective natural agents with mitochondria protection actions and explore the mediated targets for treating CIS. In the previous study, notoginseng leaf triterpenes (PNGL) from Panax notoginseng stems and leaves was demonstrated to have neuroprotective effects against cerebral ischemia/reperfusion injury. However, the potential mechanisms have been not completely elaborate. Methods: The model of middle cerebral artery occlusion and reperfusion (MCAO/R) was adopted to verify the neuroprotective effects and potential pharmacology mechanisms of PNGL in vivo. Antioxidant markers were evaluated by kit detection. Mitochondrial function was evaluated by ATP content measurement, ATPase, NAD and NADH kits. And the transmission electron microscopy (TEM) and pathological staining (H&E and Nissl) were used to detect cerebral morphological changes and mitochondrial structural damages. Western blotting, ELISA and immunofluorescence assay were utilized to explore the mitochondrial protection effects and its related mechanisms in vivo. Results: In vivo, treatment with PNGL markedly reduced excessive oxidative stress, inhibited mitochondrial injury, alleviated energy metabolism dysfunction, decreased neuronal loss and apoptosis, and thus notedly raised neuronal survival under ischemia and hypoxia. Meanwhile, PNGL significantly increased the expression of nicotinamide phosphoribosyltransferase (NAMPT) in the ischemic regions, and regulated its related downstream SIRT1/2/3-MnSOD/PGC-1α pathways. Conclusion: The study finds that the mitochondrial protective effects of PNGL are associated with the NAMPT-SIRT1/2/3-MnSOD/PGC-1α signal pathways. PNGL, as a novel candidate drug, has great application prospects for preventing and treating ischemic stroke.

Keywords

Acknowledgement

This work was supported by the National Natural Science Foundation of China (No. 81773938), the National Key Research and Development Project (No. 2017YFC1702504), the Key Laboratory Construction Project of Chinese Academy of Medical Sciences (No. 2018PT35030), the National Natural Science Foundation of China (No. 81891012), and the Clinical Research Project of Chinese Medicine "Qiye Tongmai Capsule" (No. 2015ZX09101020). We express our sincere gratitude to the foundations. In additions, it was appreciated that PNGL samples were provided by Jilin Academy of Chinese Medicine.

References

  1. Eckert B. Acute stroke therapy 1981-2009*. Clinical Neuroradiology 2009;19(1):8.
  2. Feigin VL, Forouzanfar MH, Krishnamurthi R, Mensah GA, Connor M, Bennett DA, Moran AE, Sacco RL, Anderson L, Truelsen T, et al. Global and regional burden of stroke during 1990-2010: findings from the global burden of disease study 2010. The Lancet 2014;383(9913):245-55. https://doi.org/10.1016/S0140-6736(13)61953-4
  3. Turley KR, Toledo-Pereyra LH, Kothari RU. Molecular mechanisms in the pathogenesis and treatment of acute ischemic stroke. J Invest Surg 2005;18(4):207-18. https://doi.org/10.1080/08941930591004449
  4. Bolanos JP, Moro MA, Lizasoain I, Almeida A. Mitochondria and reactive ox- ~ ygen and nitrogen species in neurological disorders and stroke: therapeutic implications. Advanced Drug Delivery Reviews 2009;61(14):1299-315. https://doi.org/10.1016/j.addr.2009.05.009
  5. Chamorro A, Dirnagl U, Urra X, Planas AM. Neuroprotection in acute stroke: targeting excitotoxicity, oxidative and nitrosative stress, and inflammation. The Lancet Neurology 2016;15(8):869-81. https://doi.org/10.1016/S1474-4422(16)00114-9
  6. Tuttolomondo A, Di SR, Di RD, Pedone C, La PS, Pinto A, Licata G. Effects of clinical and laboratory variables and of pretreatment with cardiovascular drugs in acute ischaemic stroke: a retrospective chart review from the GIFA study. International Journal of Cardiology 2011;151(3):318-22. https://doi.org/10.1016/j.ijcard.2010.06.005
  7. Woodruff TM, Thundyil J, Tang S-C, Sobey CG, Taylor SM, Arumugam TV. Pathophysiology, treatment, and animal and cellular models of human ischemic stroke. Molecular Neurodegeneration 2011;6(1):11.
  8. Katsyuba E, Romani M, Hofer D, Auwerx J. NAD+ homeostasis in health and disease. Nature Metabolism 2020;2(1):9-31. https://doi.org/10.1038/s42255-019-0161-5
  9. Stein LR, Imai S. The dynamic regulation of NAD metabolism in mitochondria. Trends Endocrinol Metab 2012;23(9):420-8. https://doi.org/10.1016/j.tem.2012.06.005
  10. Liu F, Lu J, Manaenko A, Tang J, Hu Q. Mitochondria in ischemic stroke: new insight and implications. Aging Dis 2018;9(5):924-37. https://doi.org/10.14336/AD.2017.1126
  11. Walters AM, Porter Jr GA, Brookes PS. Mitochondria as a drug target in ischemic heart disease and cardiomyopathy. Circ Res 2012;111(9):1222-36. https://doi.org/10.1161/CIRCRESAHA.112.265660
  12. Siegel CS, McCullough LD. NAD+ and nicotinamide: sex differences in cerebral ischemia. Neuroscience 2013;237:223-31. https://doi.org/10.1016/j.neuroscience.2013.01.068
  13. Yamamoto T, Byun J, Zhai P, Ikeda Y, Oka S, Sadoshima J. Nicotinamide mononucleotide, an intermediate of NAD+ synthesis, protects the heart from ischemia and reperfusion. PLoS One 2014;9(6):-98972.
  14. Liu D, Gharavi R, Pitta M, Gleichmann M, Mattson MP. Nicotinamide prevents NAD+ depletion and protects neurons against excitotoxicity and cerebral ischemia: NAD+ consumption by SIRT1 may endanger energetically compromised neurons. Neuromolecular Med 2009;11(1):28-42. https://doi.org/10.1007/s12017-009-8058-1
  15. Wang X, Zhang Z, Zhang N, Li H, Zhang L, Baines CP, Ding S. Subcellular NAMPT-mediated NAD(+) salvage pathways and their roles in bioenergetics and neuronal protection after ischemic injury. J Neurochem 2019;151(6): 732-48. https://doi.org/10.1111/jnc.14878
  16. Crack PJ, Taylor JM. Reactive oxygen species and the modulation of stroke. Free Radical Biology and Medicine 2005;38(11):1433-44. https://doi.org/10.1016/j.freeradbiomed.2005.01.019
  17. Imai SI, Guarente L. It takes two to tango: NAD(+) and sirtuins in aging/ longevity control. NPJ Aging Mech Dis 2016;2:16017.
  18. Wang P, Miao CY. NAMPT as a therapeutic target against stroke. Trends Pharmacol Sci 2015;36(12):891-905. https://doi.org/10.1016/j.tips.2015.08.012
  19. Tan CL, Chin T, Tan CYR, Rovito HA, Quek LS, Oblong JE, Bellanger S. Nicotinamide metabolism modulates the proliferation/differentiation balance and senescence of human primary keratinocytes. J Invest Dermatol 2019;139(8): 1638-1647 -3.
  20. Zhao Y, Liu XZ, Tian WW, Guan YF, Wang P, Miao CY. Extracellular visfatin has nicotinamide phosphoribosyltransferase enzymatic activity and is neuroprotective against ischemic injury. CNS Neurosci Ther 2014;20(6):539-47. https://doi.org/10.1111/cns.12273
  21. Zou XD, Guo SQ, Hu ZW, Li WL. NAMPT protects against 6-hydroxydopamineinduced neurotoxicity in PC12 cells through modulating SIRT1 activity. Mol Med Rep 2016;13(5):4058-64. https://doi.org/10.3892/mmr.2016.5034
  22. Xie W, Zhu T, Dong X, Nan F, Meng X, Zhou P, Sun G, Sun X. HMGB1-triggered inflammation inhibition of notoginseng leaf triterpenes against cerebral ischemia and reperfusion injury via MAPK and NF-kB signaling pathways. Biomolecules 2019;9(10):512.
  23. Cui HM, Zhang CG, Lin H, Lu WL, Cheng HP, Wang J. [Determination of effective components in different positions of Panax notoginseng by HPLC]. Zhong Yao Cai 2009;32(12):1810-3.
  24. Jiang B, Wang C, Han Y, Hu X, Zheng L, Zhao Y. [Isolation and identification of minor bioactive saponins from the leaves of Panax notoginseng]. Zhong Yao Cai 2004;27(7):489-91.
  25. Yu Y, Sun G, Luo Y, Wang M, Chen R, Zhang J, Ai Q, Xing N, Sun X. Cardioprotective effects of Notoginsenoside R1 against ischemia/reperfusion injuries by regulating oxidative stress- and endoplasmic reticulum stressrelated signaling pathways. Sci Rep 2016;6:21730.
  26. Ma B, Meng X, Wang J, Sun J, Ren X, Qin M, Sun J, Sun G, Sun X. Notoginsenoside R1 attenuates amyloid-beta-induced damage in neurons by inhibiting reactive oxygen species and modulating MAPK activation. Int Immunopharmacol 2014;22(1):151-9. https://doi.org/10.1016/j.intimp.2014.06.018
  27. Meng X, Xie W, Xu Q, Liang T, Xu X, Sun G, Sun X. Neuroprotective effects of radix scrophulariae on cerebral ischemia and reperfusion injury via MAPK pathways. Molecules 2018;23(9).
  28. Xie W, Meng X, Zhai Y, Ye T, Zhou P, Nan F, Sun G, Sun X. Antidepressant-like effects of the Guanxin Danshen formula via mediation of the CaMK II-CREBBDNF signalling pathway in chronic unpredictable mild stress-induced depressive rats. Annals of Translational Medicine 2019.
  29. Zhou P, Xie W, Meng X, Zhai Y, Dong X, Zhang X, Sun G, Sun X. Notoginsenoside R1 ameliorates diabetic retinopathy through PINK1-dependent activation of mitophagy. Cells 2019;8(3).
  30. Guaragnella N, Antonacci L, Passarella S, Marra E, Giannattasio S. Achievements and perspectives in yeast acetic acid-induced programmed cell death pathways. Biochem Soc Trans 2011;39(5):1538-43. https://doi.org/10.1042/BST0391538
  31. Agarwal B, Stowe DF, Dash RK, Bosnjak ZJ, Camara AK. Mitochondrial targets for volatile anesthetics against cardiac ischemia-reperfusion injury. Front Physiol 2014;5:341.
  32. Chen SD, Yang DI, Lin TK, Shaw FZ, Liou CW, Chuang YC. Roles of oxidative stress, apoptosis, PGC-1alpha and mitochondrial biogenesis in cerebral ischemia. Int J Mol Sci 2011;12(10):7199-215. https://doi.org/10.3390/ijms12107199
  33. Fu B, Zhao J, Peng W, Wu H, Zhang Y. Resveratrol rescues cadmium-induced mitochondrial injury by enhancing transcriptional regulation of PGC-1alpha and SOD2 via the Sirt3/FoxO3a pathway in TCMK-1 cells. Biochem Biophys Res Commun 2017;486(1):198-204. https://doi.org/10.1016/j.bbrc.2017.03.027
  34. Sidorova-Darmos E, Sommer R, Eubanks JH. The role of SIRT3 in the brain under physiological and pathological conditions. Front Cell Neurosci 2018;12: 196.
  35. Wang Q, Li L, Li CY, Pei Z, Zhou M, Li N. SIRT3 protects cells from hypoxia via PGC-1α- and MnSOD-dependent pathways. Neuroscience 2015;286:109-21. https://doi.org/10.1016/j.neuroscience.2014.11.045
  36. Xie W, Zhou P, Sun Y, Meng X, Dai Z, Sun G, Sun X. Protective effects and target network analysis of ginsenoside Rg1 in cerebral ischemia and reperfusion injury: a comprehensive overview of experimental studies. Cells 2018;7(12).
  37. Rouble AN, Storey KB. Characterization of the SIRT family of NAD+-dependent protein deacetylases in the context of a mammalian model of hibernation, the thirteen-lined ground squirrel. Cryobiology 2015;71(2):334-43. https://doi.org/10.1016/j.cryobiol.2015.08.009
  38. Khoury N, Koronowski KB, Young JI, Perez-Pinzon MA. The NAD+-Dependent family of sirtuins in cerebral ischemia and preconditioning. Antioxidants & Redox Signaling 2018;28(8):691-710. https://doi.org/10.1089/ars.2017.7258
  39. Wang P, Xu TY, Guan YF, Tian WW, Viollet B, Rui YC, Zhai QW, Su DF, Miao CY. Nicotinamide phosphoribosyltransferase protects against ischemic stroke through SIRT1-dependent adenosine monophosphate-activated kinase pathway. Ann Neurol 2011;69(2):360-74. https://doi.org/10.1002/ana.22236
  40. Zhao D, Sun X, Lv S, Sun M, Guo H, Zhai Y, Wang Z, Dai P, Zheng L, Ye M, et al. Salidroside attenuates oxidized lowdensity lipoproteininduced endothelial cell injury via promotion of the AMPK/SIRT1 pathway. Int J Mol Med 2019;43(6):2279-90.
  41. Li X, Liu M, Sun R, Zeng Y, Chen S, Zhang P. Protective approaches against myocardial ischemia reperfusion injury. Exp Ther Med 2016;12(6):3823-9. https://doi.org/10.3892/etm.2016.3877
  42. Garten A, Grohmann T, Kluckova K, Lavery GG, Kiess W, Penke M. Sorafenibinduced apoptosis in hepatocellular carcinoma is reversed by SIRT1. Int J Mol Sci 2019;20(16).
  43. Wang SN, Miao CY. Targeting NAMPT as a therapeutic strategy against stroke. Stroke Vasc Neurol 2019;4(2):83-9. https://doi.org/10.1136/svn-2018-000199
  44. Yu A, Zhou R, Xia B, Dang W, Yang Z, Chen X. NAMPT maintains mitochondria content via NRF2-PPARα/AMPKα pathway to promote cell survival under oxidative stress. Cellular Signalling 2019;66:109496.
  45. Lin JB, Kubota S, Ban N, Yoshida M, Santeford A, Sene A, Nakamura R, Zapata N, Kubota M, Tsubota K, et al. NAMPT-mediated NAD(+) biosynthesis is essential for vision in mice. Cell Rep 2016;17(1):69-85. https://doi.org/10.1016/j.celrep.2016.08.073
  46. Verma R, Ritzel RM, Crapser J, Friedler BD, McCullough LD. Evaluation of the neuroprotective effect of Sirt3 in experimental stroke. Transl Stroke Res 2019;10(1):57-66. https://doi.org/10.1007/s12975-017-0603-x
  47. Libert S, Cohen D, Guarente L. Neurogenesis directed by Sirt1. Nat Cell Biol 2008;10(4):373-4. https://doi.org/10.1038/ncb0408-373
  48. Zhang L, Chen C-L, Kang PT, Jin Z, Chen Y-R. Differential protein acetylation assists import of excess SOD2 into mitochondria and mediates SOD2 aggregation associated with cardiac hypertrophy in the murine SOD2-tg heart. Free Radical Biology and Medicine 2017;108:595-609. https://doi.org/10.1016/j.freeradbiomed.2017.04.022
  49. Potenza MA, Sgarra L, Nacci C, Leo V, De Salvia MA, Montagnani M. Activation of AMPK/SIRT1 axis is required for adiponectin-mediated preconditioning on myocardial ischemia-reperfusion (I/R) injury in rats. PLoS One 2019;14(1): e0210654.
  50. Song SB, Jang SY, Kang HT, Wei B, Jeoun UW, Yoon GS, Hwang ES. Modulation of mitochondrial membrane potential and ROS generation by nicotinamide in a manner independent of SIRT1 and mitophagy. Mol Cells 2017;40(7): 503-14. https://doi.org/10.14348/molcells.2017.0081
  51. Huang X, Sun J, Chen G, Niu C, Wang Y, Zhao C, Sun J, Huang H, Huang S, Liang Y, et al. Resveratrol promotes diabetic wound healing via SIRT1-FOXO1- c-myc signaling pathway-mediated angiogenesis. Front Pharmacol 2019;10: 421.
  52. Olmos Y, Sanchez-Gomez FJ, Wild B, Garcia-Quintans N, Cabezudo S, Lamas S, Monsalve M. SirT1 regulation of antioxidant genes is dependent on the formation of a FoxO3a/PGC-1alpha complex. Antioxid Redox Signal 2013;19(13): 1507-21. https://doi.org/10.1089/ars.2012.4713
  53. Song H, Zhang JJ, Wang Z, Chen-Yang DU, Zheng H, Shen ZY. Effects of FOXO3a on regulating mitophagy in hepatic ischemia reperfusion injury. Tianjin Medical Journal 2017.
  54. She DT, Wong LJ, Baik S-H, Arumugam TV. SIRT2 inhibition confers neuroprotection by downregulation of FOXO3a and MAPK signaling pathways in ischemic stroke. Molecular Neurobiology 2018;55(12):9188-203. https://doi.org/10.1007/s12035-018-1058-0
  55. Zhao W, Zhang L, Chen R, Lu H, Sui M, Zhu Y, Zeng L. SIRT3 protects against acute kidney injury via AMPK/mTOR-Regulated autophagy. Frontiers in Physiology 2018;9.