1
|
Abbas K, Mustafa M, Alam M, Habib S, Ahmad W, Adnan M, Hassan MI, Usmani N. Multi-target approach to Alzheimer's disease prevention and treatment: antioxidant, anti-inflammatory, and amyloid- modulating mechanisms. Neurogenetics 2025; 26:39. [PMID: 40167826 DOI: 10.1007/s10048-025-00821-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease (AD) is characterized by amyloid-β (Aβ) plaque accumulation, neurofibrillary tangles, neuroinflammation, and progressive cognitive decline, posing a significant global health challenge. Growing evidence suggests that dietary polyphenols may reduce the risk and progression of AD through multifaceted neuroprotective mechanisms. Polyphenols regulate amyloid proteostasis by inhibiting β/γ-secretase activity, preventing Aβ aggregation, and enhancing clearance pathways. Their strong antioxidant properties neutralize reactive oxygen species, chelate redox-active metals, and activate cytoprotective enzymes via Nrf2 signaling. This review examines the potential therapeutic targets, signaling pathways, and molecular mechanisms by which dietary polyphenols exert neuroprotective effects in AD, focusing on their roles in modulating amyloid proteostasis, oxidative stress, neuroinflammation, and cerebrovascular health. Polyphenols mitigate neuroinflammation by suppressing NF-κB signaling and upregulating brain-derived neurotrophic factor, supporting neuroplasticity and neurogenesis. They also enhance cerebrovascular health by improving cerebral blood flow, maintaining blood-brain barrier integrity, and modulating angiogenesis. This review examines the molecular and cellular pathways through which polyphenols exert neuroprotective effects, focusing on their antioxidant, anti-inflammatory, and amyloid-modulating roles. We also discuss their influence on key AD pathologies, including Aβ deposition, tau hyperphosphorylation, oxidative stress, and neuroinflammation. Insights from clinical and preclinical studies highlight the potential of polyphenols in preventing or slowing AD progression. Future research should explore personalized dietary strategies that integrate genetic and lifestyle factors to optimize the neuroprotective effects of polyphenols.
Collapse
Affiliation(s)
- Kashif Abbas
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Mohd Mustafa
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Mudassir Alam
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Safia Habib
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Waleem Ahmad
- Department of Medicine, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'Il, Ha'il, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Nazura Usmani
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
2
|
Shah AJ, Dar MY, Adnan M, Varma T, Agarwal D, Garg P, Mir RH, Meena R, Masoodi MH. Integration of phytochemical profiling and computational approaches to evaluate the neuroprotective potential of Nardostachys jatamansi in Alzheimer's disease. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2025; 45:e00881. [PMID: 40027104 PMCID: PMC11872466 DOI: 10.1016/j.btre.2025.e00881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/01/2025] [Accepted: 02/05/2025] [Indexed: 03/05/2025]
Abstract
Despite broad spectrum utility of Nardostachys jatamansi (D. Don) DC, little is known about the molecular processes that underlie its anti-Alzheimer action. To investigate the molecular targets and therapeutic potential of N. jatamansi for Alzheimer's disease (AD), we used Gas Chromatography-Mass Spectrometry (GC-MS), ADMET analysis, network pharmacology, differential gene expression analysis, molecular docking, and molecular dynamics (MD) simulations. The STITCH database was used for network creation and protein-protein interaction analysis, while Cytoscape was used for network visualization and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment and Gene Ontology (GO) for term enrichment. Additionally, to investigate the intermolecular interactions between the active chemicals and target proteins, molecular docking experiments were conducted using the Blind docking on the Achilles server. The stability of the PS1 gene complex with Spirojatamol, was further evaluated using MD simulations. With Spirojatamol showing the highest binding energy scores against PS1 (-6.9 kcal/mol), molecular docking confirmed the activity of this metabolite against AD targets PS1 and Spirojatamol formed a stable complex at 100 nanoseconds, according to additional investigation using MD simulations. Significant ligand-protein interactions were verified by binding free energy calculations using the MM/GBSA technique. The PS1-Spirojatamol complex had a binding energy of ΔG: -36.95 ± 5.00 kcal/mol. By focusing on several genes and pathways, involved in AD, this work reveals the molecular underpinnings behind N. jatamansi possible use in the treatment of AD.
Collapse
Affiliation(s)
- Abdul Jalil Shah
- Pharmaceutical Chemistry Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar-190006, Jammu and Kashmir, India
| | - Mohammad Younis Dar
- Drug Standardization Research Unit, Regional Research Institute of Unani medicine (CCRUM), Naseem Bagh campus, University of Kashmir, Srinagar, Jammu and Kashmir, India. 190006
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'il, Ha'il, P.O. Box 2440, Saudi Arabia
| | - Tanmaykumar Varma
- National Institute of Pharmaceutical Education and Research, S.A.S. Nagar Mohali, 160062, Punjab India
| | - Dhairiya Agarwal
- National Institute of Pharmaceutical Education and Research, S.A.S. Nagar Mohali, 160062, Punjab India
| | - Prabha Garg
- National Institute of Pharmaceutical Education and Research, S.A.S. Nagar Mohali, 160062, Punjab India
| | - Reyaz Hassan Mir
- Pharmaceutical Chemistry Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar-190006, Jammu and Kashmir, India
| | - Rampratap Meena
- Central Council for Research in Unani medicine (CCRUM), 61-65, opp. D-Block, Institutional Area, Janakpuri, New Delhi,110058, India
| | - Mubashir Hussain Masoodi
- Pharmaceutical Chemistry Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar-190006, Jammu and Kashmir, India
| |
Collapse
|
3
|
Cantón-Suárez A, Sánchez-Valdeón L, Bello-Corral L, Cuevas MJ, Estébanez B. Understanding the Molecular Impact of Physical Exercise on Alzheimer's Disease. Int J Mol Sci 2024; 25:13576. [PMID: 39769339 PMCID: PMC11677557 DOI: 10.3390/ijms252413576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Alzheimer's disease is one of the most common neurodegenerative diseases, characterized by a wide range of neurological symptoms that begin with personality changes and psychiatric symptoms, progress to mild cognitive impairment, and eventually lead to dementia. Physical exercise is part of the non-pharmacological treatments used in Alzheimer's disease, as it has been shown to delay the neurodegenerative process by improving the redox state in brain tissue, providing anti-inflammatory effects or stimulating the release of the brain-derived neurotrophic factor that enhances the brain structure and cognitive performance. Here, we reviewed the results obtained from studies conducted in both animal models and human subjects to comprehend how physical exercise interventions can exert changes in the molecular mechanisms underlying the pathophysiological processes in Alzheimer's disease: amyloid β-peptide pathology, tau pathology, neuroglial changes, mitochondrial dysfunction, and oxidative stress. Physical exercise seems to have a protective effect against Alzheimer's disease, since it has been shown to induce positive changes in some of the biomarkers related to the pathophysiological processes of the disease. However, additional studies in humans are necessary to address the current lack of conclusive evidence.
Collapse
Affiliation(s)
| | - Leticia Sánchez-Valdeón
- Health Research Nursing Group (GREIS), University of Leon, 24071 Leon, Spain; (L.S.-V.); (L.B.-C.)
- Department of Nursing and Physiotherapy, University of Leon, 24071 Leon, Spain
| | - Laura Bello-Corral
- Health Research Nursing Group (GREIS), University of Leon, 24071 Leon, Spain; (L.S.-V.); (L.B.-C.)
- Department of Nursing and Physiotherapy, University of Leon, 24071 Leon, Spain
| | - María J. Cuevas
- Institute of Biomedicine (IBIOMED), University of León, 24071 Leon, Spain;
| | - Brisamar Estébanez
- Institute of Biomedicine (IBIOMED), University of León, 24071 Leon, Spain;
| |
Collapse
|
4
|
Wu W, Alexander JS, Booth JL, Miller CA, Metcalf JP, Drevets DA. Influenza virus infection exacerbates gene expression related to neurocognitive dysfunction in brains of old mice. Immun Ageing 2024; 21:39. [PMID: 38907247 PMCID: PMC11191167 DOI: 10.1186/s12979-024-00447-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 06/11/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Age > 65 years is a key risk factor for poor outcomes after human influenza infection. Specifically, in addition to respiratory disease, non-neurotropic influenza A virus (IAV) causes neuro-cognitive complications, e.g. new onset depression and increases the risk of dementia after hospitalization. This study aimed to identify potential mechanisms of these effects by determining differences between young and old mice in brain gene expression in a mouse model of non-neurotropic IAV infection. METHODS Young (12 weeks) and old (70 weeks) C57Bl/6J mice were inoculated intranasally with 200 PFU H1N1 A/PR/34/8 (PR8) or sterile PBS (mock). Gene expression in lung and brain was measured by qRT-PCR and normalized to β-actin. Findings were confirmed using the nCounter Mouse Neuroinflammation Array (NanoString) and analyzed with nSolver 4.0 and Ingenuity Pathway Analysis (IPA, Qiagen). RESULTS IAV PR8 did not invade the central nervous system. Young and old mice differed significantly in brain gene expression at baseline and during non-neurotropic IAV infection. Expression of brain Ifnl, Irf7, and Tnf mRNAs was upregulated over baseline control at 3 days post-infection (p.i.) only in young mice, but old mice expressed more Ifnl than young mice 7 days p.i. Gene arrays showed down-regulation of the Epigenetic Regulation, Insulin Signaling, and Neurons and Neurotransmission pathways in old mice 3 days p.i. while young mice demonstrated no change or induction of these pathways at the same time point. IPA revealed marked baseline differences between old and young mice. Gene expression related to Cognitive Impairment, Memory Deficits and Learning worsened in old mice relative to young mice during IAV infection. Aged mice demonstrate more severe changes in gene expression related to memory loss and cognitive dysfunction by IPA. CONCLUSIONS These data suggest the genes and pathways related to learning and cognitive performance that were worse at baseline in old mice were further worsened by IAV infection, similar to old patients. Early events in the brain triggered by IAV infection portend downstream neurocognitive pathology in old adults.
Collapse
Affiliation(s)
- Wenxin Wu
- Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Room 425, RP1 800 N. Research Pkwy, Oklahoma City, OK, 73104, USA
| | - Jeremy S Alexander
- Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Room 425, RP1 800 N. Research Pkwy, Oklahoma City, OK, 73104, USA
| | - J Leland Booth
- Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Room 425, RP1 800 N. Research Pkwy, Oklahoma City, OK, 73104, USA
| | - Craig A Miller
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Oklahoma State University, Stillwater, OK, USA
| | - Jordan P Metcalf
- Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Room 425, RP1 800 N. Research Pkwy, Oklahoma City, OK, 73104, USA.
- Veterans Affairs Medical Center, Oklahoma City, OK, USA.
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Douglas A Drevets
- Infectious Diseases, Department of Medicine, University of Oklahoma Health Sciences Center, 800 Stanton L. Young, Suite 7300, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
5
|
Chen J, Chen J, Lei Z, Zhang F, Zeng LH, Wu X, Li S, Tan J. Amyloid precursor protein facilitates SARS-CoV-2 virus entry into cells and enhances amyloid-β-associated pathology in APP/PS1 mouse model of Alzheimer's disease. Transl Psychiatry 2023; 13:396. [PMID: 38104129 PMCID: PMC10725492 DOI: 10.1038/s41398-023-02692-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/17/2023] [Accepted: 11/27/2023] [Indexed: 12/19/2023] Open
Abstract
Although there are indications of a trend towards less severe acute respiratory symptoms and a decline in overall lethality from the novel Coronavirus Disease 2019 (COVID-19) caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), more and more attention has been paid to the long COVID, including the increased risk of Alzheimer's disease (AD) in COVID-19 patients. In this study, we aim to investigate the involvement of N-terminal amyloid precursor protein (APP) in SARS-CoV-2-induced amyloid-β (Aβ) pathology. Utilizing both in vitro and in vivo methodologies, we first investigated the interaction between the spike protein of SARS-CoV-2 and N-terminal APP via LSPR and CoIP assays. The in vitro impacts of APP overexpression on virus infection were further evaluated in HEK293T/ACE2 cells, SH-SY5Y cells, and Vero cells. We also analyzed the pseudovirus infection in vivo in a mouse model overexpressing human wild-type APP. Finally, we evaluated the impact of APP on pseudovirus infection within human brain organoids and assessed the chronic effects of pseudovirus infection on Aβ levels. We reported here for the first time that APP, the precursor of the Aβ of AD, interacts with the Spike protein of SARS-CoV-2. Moreover, both in vivo and in vitro data further indicated that APP promotes the cellular entry of the virus, and exacerbates Aβ-associated pathology in the APP/PS1 mouse model of AD, which can be ameliorated by N-terminal APP blockage. Our findings provide experimental evidence to interpret APP-related mechanisms underlying AD-like neuropathology in COVID-19 patients and may pave the way to help inform risk management and therapeutic strategies against diseases accordingly.
Collapse
Grants
- This study was supported by the High-level Talent Foundation of Guizhou Medical University (YJ19017, HY2020, J.T.), Anyu Biopharmaceutics, Inc., Hangzhou (06202010204, J.T.), and Zhejiang Provincial Natural Science foundation (LY19HH090013, ZW),
- Scientific Research Project of higher education Institutions in Guizhou Province [192(2022), J.C.], Science and Technology Program of Guizhou Province [ZK(2023), General 301, J.C.].
Collapse
Affiliation(s)
- Jiang Chen
- Department of Pharmacology, Zhejiang University School of Medicine, 310058, Hangzhou, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, 310015, Hangzhou, Zhejiang, China
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, 550025, Guiyang, Guizhou, China
| | - Junsheng Chen
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, 550025, Guiyang, Guizhou, China
| | - Zhifeng Lei
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, 550025, Guiyang, Guizhou, China
| | - Fengning Zhang
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, 550025, Guiyang, Guizhou, China
| | - Ling-Hui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, 310015, Hangzhou, Zhejiang, China
| | - Ximei Wu
- Department of Pharmacology, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Song Li
- First Affiliated Hospital of Dalian Medical University, 116021, Dalian, Liaoning, China.
| | - Jun Tan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, 310015, Hangzhou, Zhejiang, China.
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, 550025, Guiyang, Guizhou, China.
| |
Collapse
|