Catalpol alleviates amyloid- generation and neuronal oxidative stress injury via activating the Keap1-Nrf2/ARE signaling pathway in the immortalized lymphocytes from patients with late-onset Alzheimer’s disease and SKNMC cells co-culture model

Document Type : Original Article

Authors

1 Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China

2 Cent Hosp Enshi Tujia & Miao Autonomous Prefectur, 158 Wuyang Ave, Enshi 445000, Hubei, Peoples R China

3 Department of Neurology, Shenzhen Traditional Chinese Medicine Hospital

10.22038/ijbms.2024.78543.16982

Abstract

Objective(s): To assess the effect of catalpol, the major bioactive constituents of Rehmannia glutinosa, on our Alzheimer’s disease (AD) in vitro model.
Materials and Methods: We employed the immortalized lymphocytes (lymphoblastoid cell line, LCL) from late-onset AD patients and co-cultured “them” to mimic the pathological process of late-onset AD and investigated the effect of catalpol on our AD in vitro model.
Results: In the co-culture model, AD-derived LCL triggered excessive Aβ1-42 in SKNMC cells due to its high levels of oxidative stress and resulted in neuronal oxidative stress injury through inhibiting Keap1-Nrf2/ARE signaling pathway. Treatment with catalpol and N-acetylcysteine (NAC), an antioxidant, prevented the AD LCL-induced Aβ1-42 overproduction and reduced the level of β-site amyloid precursor protein cleaving enzyme-1 (BACE1) and amyloid precursor protein (APP)-C99. Catalpol and NAC also enhanced the antioxidant capacity and reduced apoptosis in SKNMC cells co-cultured with AD LCL. The anti-oxidative effect of catalpol was antagonized by ML385, the Nrf2 inhibitor. Therefore, we speculate that the antioxidant and anti-apoptotic effects of catalpol are mediated by activating the Keap1-Nrf2/ARE signaling pathway.
Conclusion: Catalpol affects the anti-Aβ generation and the antioxidative and antiapoptotic properties in the AD co-cultured model. So, it might be a novel natural drug and offer a potential therapeutic approach for AD.

Keywords

Main Subjects


1. Wilson RS, Segawa E, Boyle PA, Anagnos SE, Hizel LP, Bennett DA. The natural history of cognitive decline in Alzheimer’s disease. Psychol Aging 2012; 27: 1008-1017.
2. Barker WW, Luis CA, Kashuba A, Luis M, Harwood DG, Loewenstein D, et al. Relative frequencies of Alzheimer disease, Lewy body, vascular and frontotemporal dementia, and hippocampal sclerosis in the State of Florida Brain Bank. Alzheimer Dis Assoc Disord 2002; 16: 203-212.
3. Alzheimer’s Association. 2016 Alzheimer’s disease facts and figures. Alzheimers Dement 2016;12: 459-509.
4. Adav SS, Sze SK. Insight of brain degenerative protein modifications in the pathology of neurodegeneration and dementia by proteomic profiling. Mol Brain 2016; 9: 92-111.
5. Simpson DSA, Oliver PL. ROS generation in microglia: Understanding oxidative stress and inflammation in neurodegenerative disease. Antioxidants 2020; 9: 743-759.
6. Dezfulian M. A new Alzheimer’s disease cell model using B cells to induce beta amyloid plaque formation and increase TNF alpha expression. Int Immunopharmacol 2018; 59: 106-12.
7. Bettcher BM, Tansey MG, Dorothée G, Heneka MT. Peripheral and central immune system crosstalk in Alzheimer disease - a research prospectus. Nat Rev Neurol 2021; 17: 689-701.
8. Wang J, Xiong S, Xie C, Markesbery WR, Lovell MA. Increased oxidative damage in nuclear and mitochondrial DNA in Alzheimer’s disease. J Neurochem 2005; 93: 953-962.
9. Perluigi M, Sultana R, Cenini G, Di Domenico F, Memo M, Pierce WM, et al. Redox proteomics identification of 4-hydroxynonenal-modified brain proteins in Alzheimer’s disease: Role of lipid peroxidation in Alzheimer’s disease pathogenesis. Proteomics Clin Appl 2009; 3: 682-693.
10. Ansari MA, Scheff SW. Oxidative stress in the progression of Alzheimer disease in the frontal cortex. J Neuropathol Exp Neurol 2010; 69: 155-167.
11. Sultana R, Mecocci P, Mangialasche F, Cecchetti R, Baglioni M, Butterfield DA. Increased protein and lipid oxidative damage in mitochondria isolated from lymphocytes from patients with Alzheimer’s disease: Insights into the role of oxidative stress in alzheimer’s disease and initial investigations into a potential biomarker for this dementing disorder. J Alzheimers Dis 2011; 24: 77-84.
12. Leandro GS, Lobo RR, Oliveira D, Moriguti JC, Sakamoto-Hojo ET. Lymphocytes of patients with Alzheimer’s disease display different DNA damage repair kinetics and expression profiles of DNA repair and stress response genes. Int J Mol Sci 2013; 14: 12380-12400.
13. Buizza L, Cenini G, Lanni C, Ferrari-Toninelli G, Prandelli C, Govoni S, et al. Conformational altered p53 as an early marker of oxidative stress in Alzheimer’s disease. PloS One 2012; 7: e29789-29799.
14. Li YN, Xi MM, Guo Y, Hai CX, Yang WL, Qin XJ. NADPH oxidase-mitochondria axis-derived ROS mediate arsenite-induced HIF-1α stabilization by inhibiting prolyl hydroxylases activity. Toxicol Lett 2014; 224: 165-174.
15. Salminen A, Kauppinen A, Kaarniranta K. Hypoxia/ischemia activate processing of amyloid precursor protein: impact of vascular dysfunction in the pathogenesis of Alzheimer’s disease. J Neurochem 2017; 140: 536-549.
16. Saadipour K, Tiberi A, Lombardo S, Grajales E, Montroull L, Mañucat-Tan NB, et al. Regulation of BACE1 expression after injury is linked to the p75 neurotrophin receptor. Mol Cell Neurosci 2019; 99: 103395-103426.
17. Austin SA, d’Uscio LV, Katusic ZS. Supplementation of nitric oxide attenuates AβPP and BACE1 protein in cerebral microcirculation of eNOS-deficient mice. J Alzheimers Dis 2013; 33: 29-33.
18. Biswas SC, Buteau J, Greene LA. Glucagon-like peptide-1 (GLP-1) diminishes neuronal degeneration and death caused by NGF deprivation by suppressing Bim induction. Neurochem Res 2008; 33: 1845-1851.
19. Zhang RX, Li MX, Jia ZP. Rehmannia glutinosa: Review of botany, chemistry and pharmacology. J Ethnopharmacol 2008; 117: 199-214.
20. Bi J, Jiang B, Zorn A, Zhao RG, Liu P, An LJ. Catalpol inhibits LPS plus IFN-γ-induced inflammatory response in astrocytes primary cultures. Toxicol in vitro 2013;27: 543-550.
21. Chen C, Chen Z, Xu F, Zhu C, Fang F, Shu S, et al. Radio-protective effect of catalpol in cultured cells and mice. J Radiat Res 2013; 54: 76-82.
22. Yang S, Deng H, Zhang Q, Xie J, Zeng H, Jin X, et al. Amelioration of diabetic mouse nephropathy by catalpol correlates with down-regulation of Grb10 expression and activation of insulin-like growth factor 1/insulin-like growth factor 1 receptor signaling. PLoS One 2016; 11: e0151857-151868.
23. Wei M, Lu Y, Liu D, Ru W. Ovarian failure-resistant effects of catalpol in aged female rats. Biol Pharm Bull 2014; 37: 1444-1449.
24. Wang Z, Huang X, Zhao P, Zhao L, Wang Z-Y. Catalpol inhibits amyloid-beta generation through promoting alpha-cleavage of APP in swedish mutant APP overexpressed N2a cells. Front Aging Neurosci 2018;10: 66-76.
25. Seo HW, Cheon SM, Lee M-H, Kim HJ, Jeon H, Cha DS. Catalpol modulates lifespan via DAF-16/FOXO and SKN-1/Nrf2 activation in caenorhabditis elegans. Evid Based Complement Alternat Med 2015; 2015: 524878-524887.
26. Dong W, Xian Y, Yuan W, Huifeng Z, Tao W, Zhiqiang L, et al. Catalpol stimulates VEGF production via the JAK2/STAT3 pathway to improve angiogenesis in rats’ stroke model. J Ethnopharmacol 2016;191:169-79.
27. Liu C, Chen K, Lu Y, Fang Z, Yu G. Catalpol provides a protective effect on fibrillary Aβ(1-42) -induced barrier disruption in an in vitro model of the blood-brain barrier. Phytother Res  2018; 32: 1047-1055.
28. Huang JZ, Wu J, Xiang S, Sheng S, Jiang Y, Yang Z, et al. Catalpol preserves neural function and attenuates the pathology of Alzheimer’s disease in mice. Mol Med Rep 2016; 13:491-496.
29. Recuero M, Serrano E, Bullido MJ, Valdivieso F. Abeta production as consequence of cellular death of a human neuroblastoma overexpressing APP. FEBS Lett 2004 ;570:114-118.
30. Zhang P, Wang X, Peng Q, Jin Y, Shi G, Fan Z, et al. Four-octyl itaconate protects chondrocytes against H2O2-induced oxidative injury and attenuates osteoarthritis progression by activating Nrf2 signaling. Oxid Med Cell Longev 2022; 2022: 2206167.
31. Yao Y, Li R, Liu D, Long LH, He N. Rosmarinic acid alleviates acetaminophen-induced hepatotoxicity by targeting Nrf2 and NEK7-NLRP3 signaling pathway. Ecotoxicol Environ Saf 2022;241: 113773.
32. Siddiqui MA, Farshori NN, Al-Oqail MM, Pant AB, Al-Khedhairy AA. Neuroprotective effects of withania somnifera on 4-hydroxynonenal induced cell death in human neuroblastoma SH-SY5Y cells through ROS inhibition and apoptotic mitochondrial pathway. Neurochem Res 2021; 46: 171-182.
33. Li F, Wu X, Liu H, Liu M, Yue Z, Wu Z, et al. Copper depletion strongly enhances ferroptosis via mitochondrial perturbation and reduction in antioxidative mechanisms. Antioxidants (Basel) 2022; 11: 2084-2100.
34. Lu Y, Hao R, Hu Y, Wei Y, Xie Y, Shen Y, et al. Harpagide alleviate neuronal apoptosis and blood-brain barrier leakage by inhibiting TLR4/MyD88/NF-kappaB signaling pathway in Angiotensin II-induced microglial activation in vitro. Chem Biol Interact 2021; 348: 109653.
35. Gan L, Johnson JA. Oxidative damage and the Nrf2-ARE pathway in neurodegenerative diseases. Biochim Biophys Acta 2014; 1842: 1208-1218.
36. Kim K, Wang X, Ragonnaud E, Bodogai M, Illouz T, DeLuca M, et al. Therapeutic B-cell depletion reverses progression of Alzheimer’s disease. Nat commun 2021; 12: 2185-2195.
37. Wiltfang J, Esselmann H, Bibl M, Smirnov A, Otto M, Paul S, et al. Highly conserved and disease-specific patterns of carboxyterminally truncated Abeta peptides 1-37/38/39 in addition to 1-40/42 in Alzheimer’s disease and in patients with chronic neuroinflammation. J Neurochem 2002; 81: 481-496.
38. Schieber M, Chandel NS. ROS function in redox signaling and oxidative stress. Curr Biol 2014; 24: R453-462.
39. Kruman I, Bruce-Keller AJ, Bredesen D, Waeg G, Mattson MP. Evidence that 4-hydroxynonenal mediates oxidative stress-induced neuronal apoptosis. J Neurosci 1997; 17: 5089-6100.
40. Simon HU, Haj-Yehia A, Levi-Schaffer F. Role of reactive oxygen species (ROS) in apoptosis induction. Apoptosis 2000; 5: 415-418.
41. Fukui M, Song JH, Choi J, Choi HJ, Zhu BT. Mechanism of glutamate-induced neurotoxicity in HT22 mouse hippocampal cells. Eur J Pharmacol 2009; 617: 1-11.
42. Sinha K, Das J, Pal PB, Sil PC. Oxidative stress: The mitochondria-dependent and mitochondria-independent pathways of apoptosis. Arch Toxicol 2013; 87: 1157-1180.
43. Xie K, Li X. The signal pathway of apoptosis. J Shandong Agricultural Univ Nat Sci 2015; 46: 514-518.
44. Estaquier J, Vallette F, Vayssiere JL, Mignotte B. The Mitochondrial Pathways of Apoptosis. In: Scatena R, Bottoni P, Giardina B, editors. Advances in Mitochondrial Medicine. Advances in Experimental Medicine and Biology. 9422012. p. 157-183.
45. Moratilla-Rivera I, Sánchez M, Valdés-González JA, Gómez-Serranillos MP. Natural products as modulators of Nrf2 signaling pathway in neuroprotection. Int J Mol Sci 2023; 24: 3748-3773.
46. Yamamoto M, Kensler TW, Motohashi hr. The KEAP1-NRF2 system: A thiol-based sensor-effector apparatus for maintaining redox homeostasis. Physiol Rev 2018; 98: 1169-1203.
47. Fakhri S, Pesce M, Patruno A, Moradi SZ, Iranpanah A, Farzaei MH, et al. Attenuation of Nrf2/Keap1/ARE in Alzheimer’s disease by plant secondary metabolites: A mechanistic review. Molecules (Basel) 2020; 25: 4926-4950.