Planta Med 2019; 85(09/10): 692-700
DOI: 10.1055/a-0853-7754
Biological and Pharmacological Activity
Original Papers
Georg Thieme Verlag KG Stuttgart · New York

Neuroprotective Effects of Dammarane-Type Saponins from Panax notoginseng on Glutamate-Induced Cell Damage in PC12 Cells

Bao-Bao Zhang
State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Peopleʼs Republic of China
,
Xiao-Long Hu
State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Peopleʼs Republic of China
,
Yu-Yan Wang
State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Peopleʼs Republic of China
,
Jun-Yan Li
State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Peopleʼs Republic of China
,
Thi-Anh Pham
State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Peopleʼs Republic of China
,
Hao Wang
State Key Laboratory of Natural Medicines, Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Peopleʼs Republic of China
› Author Affiliations
Further Information

Publication History

received 05 November 2018
revised 20 January 2019

accepted 03 February 2019

Publication Date:
21 February 2019 (online)

Abstract

Dammarane-type saponins, the main active ingredients of Panax notoginseng, have substantial neuroprotective effects in different animal models of neurodegenerative diseases. However, because these compounds have different structures, the level of protection provided by individual compounds varies, and highly active compounds can be selected based on structure-activity relationships. Glutamate is a major excitatory neurotransmitter that plays an important role in synaptic response development. However, excessive extracellular glutamate levels lead to neuronal dysfunctions in the central nervous system. Herein, we investigated the neuroprotective effects of nine saponins (compounds 1 – 9) on glutamate-treated PC12 cells in the concentration range of 0.1 – 10 µM. The MTT assay revealed that these compounds increased cell viability to 65.6, 69.8, 76.9, 91.7, 74.4, 63.3, 59.9, 64.7, and 59.9%, respectively, compared with the glutamate-treated cells (44.6%). Protopanaxatriol (compound 4) was the most neuroprotective compound, and subsequent experiments revealed that pretreatment with compound 4 significantly reverses mitochondrial membrane potential collapse, increases superoxide dismutase activity, and decreases lactate dehydrogenase leakage, malondiadehyde levels, reactive oxygen species generation, and cell apoptosis. Compound 4 also decreased the Bax/Bcl-2 ratio, cleaved caspase-3, N-methyl-D-aspartic receptor 1, and Ca2+-/calmodulin-dependent protein kinase II expression, and inhibited glutamate-induced cytochrome C release and phosphorylation of apoptosis signal-regulating kinase 1, c-Jun N-terminal kinase, and p38. Overall, the results indicate that protopanaxatriol has significant neuroprotective effects, and might be a promising neuroprotective agent for preventing and treating neurodegenerative diseases.

Supporting Information

 
  • References

  • 1 Bondy SC. Anthropogenic pollutants may increase the incidence of neurodegenerative disease in an aging population. Toxicology 2016; 341: 41-46
  • 2 Cannon JR, Greenamvre JT. The role of environmental exposures in neurodegeneration and neurodegenerative diseases. Toxicol Sci 2011; 124: 225-250
  • 3 Garden GA, La Spada AR. Intercellular (mis)communication in neurodegenerative disease. Neuron 2012; 73: 886-901
  • 4 Rama Rao KV, Kielian T. Neuron-astrocyte interactions in neurodegenerative diseases: Role of neuroinflammation. Clin Exp Neuroimmunol 2015; 6: 245-263
  • 5 Bae JR, Kim SH. Synapses in neurodegenerative diseases. BMB Rep 2017; 50: 237-246
  • 6 Neal M, Richardson JR. Time to get personal: a framework for personalized targeting of oxidative stress in neurotoxicity and neurodegenerative disease. Curr Opin Toxicol 2018; 7: 127-132
  • 7 Xue XY, Guo XH, Li M, Luo HM. Advances in the pathogenesis of neurodegenerative diseases. Chin J Gerontol 2015; 35: 3149-3152
  • 8 Flanagan B, McDaid L, Wade J, Wong-Lin K, Harkin J. A computational study of astrocytic glutamate influence on post-synaptic neuronal excitability. PLoS Comput Biol 2018; 14: e1006040
  • 9 Tokarshi K, Bobula B, Wabno J, Hess G. Repeated administration of imipramine attenuates glutamatergic transmission in rat frontal cortex. Neuroscience 2008; 153: 789-795
  • 10 Olatunji OJ, Feng Y, Olatunji OO, Tang J, Wei Y, Ouyang Z, Su Z. Polysaccharides purified from Cordyceps cicadae protects PC12 cells against glutamate-induced oxidative damage. Carbohydr Polym 2016; 153: 187-195
  • 11 Aman TK, Maki BA, Ruffino TJ, Kasperek EM, Popescu GK. Separate intramolecular targets for protein kinase A control N-methyl-D-aspartate receptor gating and Ca2+ permeability. J Biol Chem 2014; 289: 18805-18817
  • 12 Min D, Guo F, Zhu S, Xu X, Mao X, Cao Y, Lv X, Gao Q, Wang L, Chen T, Shaw C, Hao L, Cai J. The alterations of Ca2+/calmodulin/CaMKIICaV1.2 signaling in experimental models of Alzheimerʼs disease and vascular dementia. Neurosci Lett 2013; 538: 60-65
  • 13 Maher P, Leyen VK, Dey PN, Honrath B, Dolga A, Methner A. The role of Ca2+ in cell death caused by oxidative glutamate toxicity and ferroptosis. Cell Calcium 2017; 70: 47-55
  • 14 Kim JJ, Kang YJ, Shin SA, Bak DH, Lee JW, Lee KB, Yoo YC, Kim DK, Lee BH, Kim DW, Lee J, Jo EK, Yuk JM. Phlorofucofuroeckol improves glutamate-induced neurotoxicity through modulation of oxidative stress-mediated mitochondrial dysfunction in PC12 cells. PLoS One 2016; 11: e0163444
  • 15 Tu L, Wang Y, Chen D, Xiang P, Shen JJ, Li YB, Wang SL. Protective effects of notoginsenoside R1 via regulation of the PI3K-Akt-mTOR/JNK pathway in neonatal cerebral hypoxic-ischemic brain injury. Neurochem Res 2018; 43: 1210-1226
  • 16 Zhou S, Zhang MX, Feng LM, Zhou YF, Li L, Ban LL. Protective effects of notoginsenoside R1 on cerebral ischemia-reperfusion injury in rats. Exp Ther Med 2017; 14: 6012-6016
  • 17 Ghaeminia M, Rajkumar R, Koh HL, Dawe GS, Tan CH. Ginsenoside Rg1 modulates medial prefrontal cortical firing and suppresses the hippocampo- medial prefrontal cortical long-term potentiation. J Ginseng Res 2018; 42: 298-303
  • 18 Zhang Y, Hu W, Zhang B, Yin Y, Zhang J, Huang D, Huang R, Li W, Li W. Ginsenoside Rg1 protects against neuronal degeneration induced by chronic dexamethasone treatment by inhibiting NLRP-1 inflammasome in mice. Int J Mol Med 2017; 40: 1134-1142
  • 19 Shi Y, Miao W, Teng J, Zhang L. Ginsenoside Rb1 protects the brain from damage induced by epileptic seizure via Nrf2/ARE signaling. Cell Physiol Biochem 2018; 45: 212-225
  • 20 Zhang YL, Liu Y, Kang XP, Dou CY, Zhuo RG, Huang SQ, Peng L, Wen L. Ginsenoside Rb1 confers neuroprotection via promotion of glutamate transporters in a mouse model of Parkinsonʼs disease. Neuropharmacology 2018; 131: 223-227
  • 21 Kwok HH, Ng WY, Yang MS, Mak NK, Wong RN, Yue PY. The ginsenoside protopanaxatriol protects endothelial cells from hydrogen peroxide-induced cell injury and cell death by modulating intracellular redox status. Free Radic Biol Med 2010; 48: 437-445
  • 22 Zhang Y, Yu L, Cai W, Fan S, Feng L, Ji G, Huang C. Protopanaxatriol, a novel PPARγ antagonist from Panax ginseng, alleviates steatosis in mice. Sci Rep 2014; 4: 7375
  • 23 Oh HA, Kim DE, Choi HJ, Kim NJ, Kim DH. Anti-stress effects of 20(S)-protopanaxadiol and 20(S)-protopanaxatriol in immobilized mice. Biol Pharm Bull 2015; 38: 331-335
  • 24 Lu C, Lv J, Dong L, Jiang N, Wang Y, Wang Q, Li Y, Chen S, Fan B, Wang F, Liu X. Neuroprotective effects of 20(S)-protopanaxatriol (PPT) on scopolamine-induced cognitive deficits in mice. Phytother Res 2018; 32: 1056-1063
  • 25 Radi E, Formichi P, Battisti C, Federico A. Apoptosis and oxidative stress in neurodegenerative diseases. J Alzheimers Dis 2014; 42: S125-S152
  • 26 Xiao X, Liu J, Hu J, Zhu X, Yang H, Wang C, Zhang Y. Protective effects of protopine on hydrogen peroxide-induced oxidative injury of PC12 cells and Ca(2+) antagonism and antioxidant mechanisms. Eur J Pharmacol 2008; 591: 21-27
  • 27 Satpute RM, Kashyap RS, Deopujari JY, Purohit HJ, Taori GM, Daginawala HF. Protection of PC12 cells from chemical ischemia induced oxidative stress by Fagonia arabica. Food Chem Toxicol 2009; 47: 2689-2695
  • 28 Ghasemi M, Mayasi Y, Hannoun A, Eslami SM, Caradang R. Nitric oxide and mitochondrial function in neurological diseases. Neuroscience 2018; 376: 48-71
  • 29 Gao J, Deng Y, Yin C, Liu Y, Zhang W, Shi J, Gong Q. Icariside II, a novel phosphodiesterase 5 inhibitor, protects against H2O2-induced PC12 cells death by inhibiting mitochondria-mediated autophagy. J Cell Mol Med 2017; 21: 375-386
  • 30 Yuan Y, Wang Y, Hu FF, Jiang CY, Zhang YJ, Zhao SW, Gu JH, Liu XZ, Bian JC, Liu ZP. Cadmium activates reactive oxygen species-dependent AKT/mTOR and mitochondrial apoptotic pathways in neuronal cells. Biomed Environ Sci 2016; 29: 117-126
  • 31 Rebola N, Srikumar BN, Mulle C. Activity-dependent synaptic plasticity of NMDA receptors. J Physiol 2010; 588: 93-99
  • 32 Qu JJ, Zhou YT, Li C, Chen ZJ, Li HC, Fang M, Zhu C, Huo CY, Yung KKL, Li J, Luo CH, Mo ZX. Sinomenine protects against morphine dependence through the NMDAR1/CAMKII/CREB pathway: a possible role of astrocyte-derived exosomes. Molecules 2018; 23: E2370
  • 33 Jiang G, Wu H, Hu Y, Li J, Li Q. Gastrodin inhibits glutamate-induced apoptosis of PC12 cells via inhibition of CaMKII/ASK-1/p38 MAPK/p53 signaling cascade. Cell Mol Neurobiol 2014; 34: 591-602
  • 34 Chen WN, Li XM, Jia LQ, Wang J, Zhang L, Hou DD, Wang JY, Ren L. Neuroprotective activities of cataipol against CaMKII-dependent apoptosis induced by LPS in PC12 cells. Br J Pharmacol 2013; 169: 1140-1152