Home Evaluating the effectiveness of pterostilbene in improving memory and offering neuroprotection in a rat model of Alzheimer’s disease induced by aluminum chloride
Article
Licensed
Unlicensed Requires Authentication

Evaluating the effectiveness of pterostilbene in improving memory and offering neuroprotection in a rat model of Alzheimer’s disease induced by aluminum chloride

  • Ameer Raheem Waheed ORCID logo EMAIL logo and Bahir Abdul Razzaq Mshimesh ORCID logo
Published/Copyright: July 31, 2025

Abstract

Background and Objectives

Alzheimer’s disease (AD) is a neurodegenerative disease and is the predominant etiology of dementia. We hypothesize that the naturally occurring pterostilbene (PTE) at doses of 50 and 100 mg/kg would yield dose-dependent neuroprotective effects, reducing cognitive deficits and pathological hallmarks by modulating biomarkers (Amyloid Beta protein (Aβ), Phosphorylated tau protein (P-tau), Brain-Derived Neurotrophic Factor (BDNF), acetylcholinesterase (ACHE), glutamate (GLU)) and a novel synaptic marker neurogranin (NRGN) in rats induced by aluminum chloride (AlCl3). This current research aims to evaluate the neuroprotective effects of pterostilbene (PTE) against neurobehavioral and pathological alterations induced by aluminum chloride (AlCl3) in rats with Alzheimer’s.

Methods

40 rats were divided into five groups, eight in each group. They received 70 mg/kg of body weight AlCl3 intraperitoneally for 30 days, followed by oral administration of PTE at 50 mg/kg or 100 mg/kg, or donepezil at 1 mg/kg for 14 days. The Y-maze and novel object recognition tests were used for the neurobehavioral evaluation of the rats. This was followed by a biochemical evaluation using ELISA kits to demonstrate the impact of PTE on the levels of Aβ, P-tau, BDNF, NRGN, AChE, and GLU. Additional validations were conducted through histopathological evaluation of the cortex and basal ganglia in the rat brain. Using GraphPad Prism 10, statistical data were obtained by ANOVA and Tukey’s multiple comparisons test. The histopathologic score system was determined using the non-parametric Kruskal-Wallis one-way ANOVA k-samples (all pairwise) test.

Results

PTE at 50 mg/kg significantly increases spontaneous alternation percentage (SAP) by 35.7 % and discrimination index (DI) by 79.7 %, while also considerably lowering Aβ by 70.6 %, P-tau by 33.9 %, BDNF by 59.7 %, NRGN by 40 %, ACHE by 28.8 %, and GLU by 28.4 %. Moreover, PTE at 100 mg/kg significantly increases SAP by 42.9 % and DI by 83.4 %, and substantially decreases Aβ by 83.8 %, P-tau by 45.5 %, BDNF by 69 %, NRGN by 42.5 %, ACHE by 69 %, and GLU by 50.9 % compared to the AlCl3 group. Histopathological evaluation of the cortex and basal ganglia in AlCl3-induced rat brains revealed pathological alterations absent in rats treated with PTE.

Conclusions

This study supports the hypothesis that PTE can reverse memory loss and pathological markers.


Corresponding author: Ameer Raheem Waheed, Department of Pharmacology and Toxicology, 108487 Mustansiriyah University, College of Pharmacy , Baghdad, Iraq, E-mail:

Acknowledgment

Authors would like to express their gratitude to the College of Pharmacy/ Mustansiriyah University (www.uomustansiriyiah.edu.iq), Baghdad, Iraq, for its support with this work.

  1. Research ethics: This study was conducted in accordance with the Declaration of Helsinki (as revised in 2013) and approved by the Institutional Review Board Approval no.41 on 10/10/2024.

  2. Informed consent: Not applicable.

  3. Author contributions: All authors contributed to the study, accepted responsibility for the entire content of this manuscript, and approved its submission.

  4. Use of Large Language Models, AI and Machine Learning Tools: Not applicable.

  5. Conflict of interest: The authors state no conflict of interest.

  6. Research funding: None declared.

  7. Data availability: The raw data can be obtained at the request of the corresponding author.

References

1. Breijyeh, Z, Karaman, R. Comprehensive review on alzheimer’s disease: causes and treatment. Molecules 2020;25. https://doi.org/10.3390/molecules25245789.Search in Google Scholar PubMed PubMed Central

2. Ratan, Y, Rajput, A, Maleysm, S, Pareek, A, Jain, V, Pareek, A, et al.. An insight into cellular and molecular mechanisms underlying the pathogenesis of neurodegeneration in Alzheimer’s disease. Biomedicines 2023;11:1398. Available from: https://www.mdpi.com/2227-9059/11/5/1398/html.10.3390/biomedicines11051398Search in Google Scholar PubMed PubMed Central

3. Alzheimer’s disease facts and figures. Alzheimer’s Dement 2021;17:327–406. Available from: https://pubmed.ncbi.nlm.nih.gov/33756057/.10.1002/alz.12328Search in Google Scholar PubMed

4. Arvanitakis, Z, Shah, RC, Bennett, DA. Diagnosis and management of dementia: review. JAMA 2019;322:1589–99. Available from: https://pubmed.ncbi.nlm.nih.gov/31638686/.10.1001/jama.2019.4782Search in Google Scholar PubMed PubMed Central

5. Aranda, MP, Kremer, IN, Hinton, L, Zissimopoulos, J, Whitmer, RA, Hummel, CH, et al.. Impact of dementia: health disparities, population trends, care interventions, and economic costs. J Am Geriatr Soc 2021;69:1774. Available from: https://pmc/articles/PMC8608182/.10.1111/jgs.17345Search in Google Scholar PubMed PubMed Central

6. Sehar, U, Rawat, P, Reddy, AP, Kopel, J, Reddy, PH. Amyloid beta in aging and alzheimer’s disease. Int J Mol Sci 2022;23. Available from: https://pubmed.ncbi.nlm.nih.gov/36361714/.10.3390/ijms232112924Search in Google Scholar PubMed PubMed Central

7. Du, X, Wang, X, Geng, M. Alzheimer’s disease hypothesis and related therapies. Transl Neurodegener 2018;7. Available from: https://pmc/articles/PMC5789526/.10.1186/s40035-018-0107-ySearch in Google Scholar PubMed PubMed Central

8. Seo, Doh, Holtzman, DM. Current understanding of the Alzheimer’s disease-associated microbiome and therapeutic strategies. Exp Mol Med 2023;56:86–94. Available from: https://www.nature.com/articles/s12276-023-01146-2.10.1038/s12276-023-01146-2Search in Google Scholar PubMed PubMed Central

9. Flores-Muñoz, C, Gómez, B, Mery, E, Mujica, P, Gajardo, I, Córdova, C, et al.. Acute pannexin 1 blockade mitigates early synaptic plasticity defects in a mouse model of alzheimer’s disease. Front Cell Neurosci 2020;14:502969. Available from: www.frontiersin.org.10.3389/fncel.2020.00046Search in Google Scholar PubMed PubMed Central

10. Agnello, L, Gambino, CM, Lo Sasso, B, Bivona, G, Milano, S, Ciaccio, AM, et al.. Neurogranin as a novel biomarker in alzheimer’s disease. Lab Med 2021;52:188–96. https://doi.org/10.1093/labmed/lmaa062.Search in Google Scholar PubMed

11. Numakawa, T, Kajihara, R. The role of brain-derived neurotrophic factor as an essential mediator in neuronal functions and the therapeutic potential of its mimetics for neuroprotection in neurologic and psychiatric disorders. Molecules 2025;30:848. https://doi.org/10.3390/molecules30040848.Search in Google Scholar PubMed PubMed Central

12. Yang, X, Du, W, Zhang, Y, Wang, H, He, M. Neuroprotective effects of higenamine against the alzheimer’s disease via amelioration of cognitive impairment, Aβ burden, apoptosis and regulation of Akt/GSK3β signaling pathway. Dose Response 2020;18. https://doi.org/10.1177/1559325820972205.Search in Google Scholar PubMed PubMed Central

13. Xu, QQ, Yang, W, Zhong, M, Lin, ZX, Gray, NE, Xian, YF, et al.. Animal models of alzheimer’s disease: preclinical insights and challenges. Acta Materia Medica 2023;2:192–215. https://doi.org/10.15212/amm-2023-0001.Search in Google Scholar

14. Zhang, J, Zhang, Y, Wang, J, Xia, Y, Zhang, J, Chen, L, et al.. Recent advances in Alzheimer’s disease: mechanisms, clinical trials and new drug development strategies. Signal Transduct Targeted Ther 2024;9:1–35. [Internet]. 2024 Aug 23 [cited 2025 Mar 19] Available from: https://www.nature.com/articles/s41392-024-01911-3.10.1038/s41392-024-01911-3Search in Google Scholar PubMed PubMed Central

15. Adamu, A, Li, S, Gao, F, Xue, G. The role of neuroinflammation in neurodegenerative diseases: current understanding and future therapeutic targets. Front Aging Neurosci 2024;16:1–16.10.3389/fnagi.2024.1347987Search in Google Scholar PubMed PubMed Central

16. Wangikar, P, Chaudhari, P, Sharma, E, Godse, C, Vora, A, Nair, S. Acute and sub-chronic oral GLP toxicity of withania somnifera root extract in sprague dawley rats. Drug Metab Pers Ther 2024;39:145–58. https://doi.org/10.1515/dmpt-2024-0056.Search in Google Scholar PubMed

17. Liu, P, Tang, W, Xiang, K, Li, G. Pterostilbene in the treatment of inflammatory and oncological diseases. Front Pharmacol 2023;14:1–13.10.3389/fphar.2023.1323377Search in Google Scholar PubMed PubMed Central

18. Liu, Y, You, Y, Lu, J, Chen, X, Yang, Z. Recent advances in synthesis, bioactivity, and pharmacokinetics of pterostilbene, an important analog of resveratrol. Molecules 2020;25. https://doi.org/10.3390/molecules25215166.Search in Google Scholar PubMed PubMed Central

19. Fadhel Khalif, AA, Mshimesh, BAR, Abood, DAH, Oudah Al-Qaysi, SA. Comparative study between adenine and folic acid as optimal models to induce anemia of chronic kidney disease in Male rats; an experimental study. Immunopathologia Persa 2025:1–10. https://immunopathol.com/Inpress/ipp-43776.10.34172/ipp.2025.43776Search in Google Scholar

20. Ahmed, SA, Nasry, MN, Mohammed, WI. Carvedilol protects against cognitive dysfunction through inhibition of amyloid beta (1-42) and inflammation in aluminum chloride induced alzheimer’s disease compared to donepezil as a reference drug in wistar rats. Rec Pharm Biomedical Sci2022;6:69–85. Available from: https://rpbs.journals.ekb.eg/article_231971.html.10.21608/rpbs.2022.129511.1132Search in Google Scholar

21. Rao, YL, Ganaraja, B, Marathe, A, Manjrekar, PA, Joy, T, Ullal, S, et al.. Comparison of malondialdehyde levels and superoxide dismutase activity in resveratrol and resveratrol/donepezil combination treatment groups in Alzheimer’s disease induced rat model. 3 Biotech 2021;11:1–10. [Internet] Available from: https://link.springer.com/article/10.1007/s13205-021-02879-5.10.1007/s13205-021-02879-5Search in Google Scholar PubMed PubMed Central

22. Nieoczym, D, Socała, K, Jedziniak, P, Wyska, E, Wlaź, P. Effect of pterostilbene, a natural analog of resveratrol, on the activity of some antiepileptic drugs in the acute seizure tests in mice. Neurotox Res 2019;36:859–69. [Internet] Available from: https://link.springer.com/article/10.1007/s12640-019-00021-1.10.1007/s12640-019-00021-1Search in Google Scholar PubMed PubMed Central

23. Naik, B, Nirwane, A, Majumdar, A. Pterostilbene ameliorates intracerebroventricular streptozotocin induced memory decline in rats. Cogn Neurodyn 2017;11:35–49. https://doi.org/10.1007/s11571-016-9413-1.Search in Google Scholar PubMed PubMed Central

24. Mahdi, O, Chiroma, SM, Baharuldin, MTH, Nor, NHM, Taib, CNM, Jagadeesan, S, et al.. WIN55,212-2 attenuates cognitive impairments in AlCl3 + d-Galactose-Induced alzheimer’s disease rats by enhancing neurogenesis and reversing oxidative stress. Biomedicines 2021;9. Available from https://pubmed.ncbi.nlm.nih.gov/34572456/.10.3390/biomedicines9091270Search in Google Scholar PubMed PubMed Central

25. Galvao, J, Davis, B, Tilley, M, Normando, E, Duchen, MR, Cordeiro, MF. Unexpected low-dose toxicity of the universal solvent DMSO. FASEB J 2014;28:1317–30. https://doi.org/10.1096/fj.13-235440.Search in Google Scholar PubMed

26. Luptakova, L, Dvorcakova, S, Demcisakova, Z, Belbahri, L, Holovska, K, Petrovova, E. Dimethyl sulfoxide: morphological, histological, and molecular view on developing chicken liver. Toxics 2021;9:55. Available from: https://pmc.ncbi.nlm.nih.gov/articles/PMC8001493/.10.3390/toxics9030055Search in Google Scholar PubMed PubMed Central

27. Zhang, H, Yao, M, Morrison, RA, Chong, S. Commonly used surfactant, tween 80, improves absorption of P-glycoprotein substrate, digoxin, in rats. Arch Pharm Res (Seoul) 2003;26:768–72. [Internet] Available from: https://pubmed.ncbi.nlm.nih.gov/14560928/.10.1007/BF02976689Search in Google Scholar PubMed

28. Zhu, L, Lu, F, Zhang, X, Liu, S, Mu, P. SIRT1 is involved in the neuroprotection of pterostilbene against amyloid β 25–35-Induced cognitive deficits in mice. Front Pharmacol 2022;13:877098. [Internet] Available from: https://pmc.ncbi.nlm.nih.gov/articles/PMC9047953/.10.3389/fphar.2022.877098Search in Google Scholar PubMed PubMed Central

29. Justin-Thenmozhi, A, Dhivya Bharathi, M, Kiruthika, R, Manivasagam, T, Borah, A, Essa, MM. Attenuation of aluminum chloride-induced neuroinflammation and caspase activation through the AKT/GSK-3β pathway by hesperidin in wistar rats. Neurotox Res 2018;34:463–76. [Internet] Available from: https://link.springer.com/article/10.1007/s12640-018-9904-4.10.1007/s12640-018-9904-4Search in Google Scholar PubMed

30. Abdul Ghani, IA, Razzaq Mshimesh, BA, Mohammed, NH, Oudah, SA. Tri-chromium picolinate significantly improved the hormonal levels and CYP17A1 expression in female rats induced with polycystic ovary syndrome. Immunopathologia Persa 2024:1–12. https://immunopathol.com/inpress/ipp-41728.10.34172/ipp.2025.41728Search in Google Scholar

31. Li, P, Bartlett, MG. A review of sample preparation methods for quantitation of small-molecule analytes in brain tissue by liquid chromatography tandem mass spectrometry (LC-MS/MS). Anal Methods 2014;6:6183–207. [Internet] Available from: https://pubs.rsc.org/en/content/articlehtml/2014/ay/c4ay00915k.10.1039/C4AY00915KSearch in Google Scholar

32. Ahmed, TH, Raoof, IB, Mshemish, BA. Evaluation of Nicorandil in Treatment of induced pulmonary arterial hypertension in male Rats. Al-Mustansiriyah J Pharm Sci 2024;24:330–42.10.32947/ajps.v24i3.1077Search in Google Scholar

33. Basil, D, Razzaq, B, Abdul-Razzaq Mshimesh B. Antiproliferative activity of Brassica nigra seeds extract in liver tissue of mice exposed to phenobarbital. Al-Mustansiriyah J Pharm Sci 2022;22:8–22.10.32947/ajps.v22i1.826Search in Google Scholar

34. Charafe-Jauffret, E, Tarpin, C, Bardou, VJ, Bertucci, F, Ginestier, C, Braud, AC, et al.. Immunophenotypic analysis of inflammatory breast cancers: identification of an “inflammatory signature.”. J Pathol 2004;202:265–73. [Internet] Available from: https://pubmed.ncbi.nlm.nih.gov/14991891/.10.1002/path.1515Search in Google Scholar PubMed

35. Shunan, D, Yu, M, Guan, H, Zhou, Y. Neuroprotective effect of betalain against AlCl3-induced Alzheimer’s disease in sprague dawley rats via putative modulation of oxidative stress and nuclear factor kappa B (NF-κB) signaling pathway. Biomed Pharmacother 2021;137:111369. https://doi.org/10.1016/j.biopha.2021.111369.Search in Google Scholar PubMed

36. Al-Amin, MM, Chowdury, MIA, Saifullah, ARM, Alam, MN, Jain, P, Hossain, M, et al.. Levocarnitine improves AlCl3-Induced spatial working memory impairment in Swiss albino mice. Front Neurosci 2019;13:441320. https://doi.org/10.3389/fnins.2019.00278.Search in Google Scholar PubMed PubMed Central

37. Ali, AA, Ahmed, HI, Abuelfoutouh, K, Abu-Elfotuh, K. Modeling stages mimic alzheimer’s disease induced by different doses of aluminum in rats: focus on progression of the disease in response to time. [Internet] Available from: https://www.researchgate.net/publication/349609068.Search in Google Scholar

38. Haider, S, Liaquat, L, Ahmad, S, Batool, Z, Siddiqui, RA, Tabassum, S, et al.. Naringenin protects AlCl3/D-galactose induced neurotoxicity in rat model of AD via attenuation of acetylcholinesterase levels and inhibition of oxidative stress. PLoS One 2020;15. Available from: https://pubmed.ncbi.nlm.nih.gov/31945778/.10.1371/journal.pone.0227631Search in Google Scholar PubMed PubMed Central

39. Pang, K, Jiang, R, Zhang, W, Yang, Z, Li, LL, Shimozawa, M, et al.. An app knock-in rat model for Alzheimer’s disease exhibiting Aβ and tau pathologies, neuronal death and cognitive impairments. Cell Res 2021;32:157–75. Available from: https://www.nature.com/articles/s41422-021-00582-x.10.1038/s41422-021-00582-xSearch in Google Scholar PubMed PubMed Central

40. Alqarni, SS, Afzal, M, Alharbi, KS, Alenezi, SK, Alsahli, TG, Zaidi, S, et al.. Rosiridin protects against aluminum chloride-induced memory impairment via modulation of BDNF/NFκB/PI3K/Akt pathway in rats. Medicina 2024;60:1812. Available from: https://pmc.ncbi.nlm.nih.gov/articles/PMC11596433/.10.3390/medicina60111812Search in Google Scholar PubMed PubMed Central

41. Xu, J, Liu, J, Li, Q, Mi, Y, Zhou, D, Meng, Q, et al.. Pterostilbene alleviates Aβ1-42-Induced cognitive dysfunction via inhibition of oxidative stress by activating Nrf2 signaling pathway. Mol Nutr Food Res 2021;65. Available from: https://pubmed.ncbi.nlm.nih.gov/33280250/.10.1002/mnfr.202000711Search in Google Scholar PubMed

42. Huang, W, Cheng, P, Yu, K, Han, Y, Song, M, Li, Y. Hyperforin attenuates aluminum-induced Aβ production and tau phosphorylation via regulating Akt/GSK-3β signaling pathway in PC12 cells. Biomed Pharmacother 2017;96:1–6. Available from: https://www.sciencedirect.com/science/article/abs/pii/S0753332217330597?utm_source=chatgpt.com.10.1016/j.biopha.2017.09.114Search in Google Scholar PubMed

43. Gao, S, Zhang, H, Li, N, Zhang, L, Zhu, Z, Xu, C. Pterostilbene: a natural neuroprotective stilbene with anti-alzheimer’s disease properties. J Pharm Anal 2024;17:101043. https://doi.org/10.1016/j.jpha.2024.101043.Search in Google Scholar PubMed PubMed Central

44. Miranda, M, Morici, JF, Zanoni, MB, Bekinschtein, P. Brain-derived neurotrophic factor: a key molecule for memory in the healthy and the pathological brain. Front Cell Neurosci. 2019;13:472800, [Internet] Available from: www.frontiersin.org.10.3389/fncel.2019.00363Search in Google Scholar PubMed PubMed Central

45. Bathina, S, Das, UN. Brain-derived neurotrophic factor and its clinical implications. Arch Med Sci 2015;11:1164. [Internet] Available from: https://pmc.ncbi.nlm.nih.gov/articles/PMC4697050/.10.5114/aoms.2015.56342Search in Google Scholar PubMed PubMed Central

46. Bhuvanendran, S, Kumari, Y, Othman, I, Shaikh, MF. Amelioration ofc ognitive deficit by embelin in a scopolamine-induced Alzheimer’s disease-like condition in a rat model. Front Pharmacol 2018;9:374992. https://doi.org/10.3389/fphar.2018.00665.Search in Google Scholar PubMed PubMed Central

47. Meng, J, Chen, Y, Bi, F, Li, H, Chang, C, Liu, W. Pterostilbene attenuates amyloid-β induced neurotoxicity with regulating PDE4A-CREB-BDNF pathway. Am J Transl Res 2019;11:6356. [Internet] Available from: https://pmc.ncbi.nlm.nih.gov/articles/PMC6834512/.Search in Google Scholar

48. Saunders, T, Gunn, C, Blennow, K, Kvartsberg, H, Zetterberg, H, Shenkin, SD, et al.. Neurogranin in Alzheimer’s disease and ageing: a human post-mortem study. Neurobiol Dis 2023;177:105991. https://doi.org/10.1016/j.nbd.2023.105991.Search in Google Scholar PubMed

49. Davidsson, P, Blennow, K. Neurochemical dissection of synaptic pathology in Alzheimer’s disease. Int Psychogeriatr 1998;10:11–23. Available from: https://www.intpsychogeriatrics.org/action/showFullText?pii=S1041610224001066.10.1017/S1041610298005110Search in Google Scholar PubMed

50. Kester, MI, Teunissen, CE, Crimmins, DL, Herries, EM, Ladenson, JKH, Scheltens, P, et al.. Neurogranin as a cerebrospinal fluid biomarker for synaptic loss in symptomatic alzheimer disease. JAMA Neurol 2015;72:1275. [Internet] Available from: https://pmc.ncbi.nlm.nih.gov/articles/PMC4694558/.10.1001/jamaneurol.2015.1867Search in Google Scholar PubMed PubMed Central

51. Bhembre, N, Bonthron, C, Opazo, P. Synaptic compensatory plasticity in alzheimer’s disease. J Neurosci. 2023;43:6833, Available from: https://pmc.ncbi.nlm.nih.gov/articles/PMC10573755/.10.1523/JNEUROSCI.0379-23.2023Search in Google Scholar PubMed PubMed Central

52. Chen, X, Zhang, M, Ahmed, M, Surapaneni, KM, Veeraraghavan, VP, Arulselvan, P, et al.. Neuroprotective effects of ononin against the aluminium chloride-induced Alzheimer’s disease in rats. Saudi J Biol Sci 2021;28:4232–9. https://doi.org/10.1016/j.sjbs.2021.06.031.Search in Google Scholar PubMed PubMed Central

53. Soares, C, das Ros, LU, da Rocha, AS, Machado, LS, Bellaver, B, Zimmer, ER. The glutamatergic system in Alzheimer’s disease: a systematic review with meta-analysis. Alzheimer’s Dementia 2022;18:e064821. [Internet] Available from: https://onlinelibrary.wiley.com/doi/full/10.1002/alz.064821.10.1002/alz.064821Search in Google Scholar

54. Chang, Y, Wang, SJ. Inhibitory effect of glutamate release from rat cerebrocortical nerve terminals by resveratrol. Neurochem Int 2009;54:135–41. [Internet] [cited 2025 Apr 9] Available from: https://pubmed.ncbi.nlm.nih.gov/19059293/.10.1016/j.neuint.2008.11.001Search in Google Scholar PubMed

55. Zeng, Q, Lian, W, Wang, G, Qiu, M, Lin, L, Zeng, R. Pterostilbene induces Nrf2/HO-1 and potentially regulates NF-κB and JNK–Akt/mTOR signaling in ischemic brain injury in neonatal rats. 3 Biotech 2020;10:192. [cited 2025 Apr 9] https://pmc.ncbi.nlm.nih.gov/articles/PMC7128026/.10.1007/s13205-020-02167-8Search in Google Scholar PubMed PubMed Central

Received: 2025-04-15
Accepted: 2025-07-17
Published Online: 2025-07-31

© 2025 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 7.9.2025 from https://www.degruyterbrill.com/document/doi/10.1515/jcim-2025-0142/html
Scroll to top button