1
|
Aschner M, Skalny AV, Santamaria A, Rocha JBT, Mansouri B, Tizabi Y, Madeddu R, Lu R, Lee E, Tinkov AA. Epigenetic Mechanisms of Aluminum-Induced Neurotoxicity and Alzheimer's Disease: A Focus on Non-Coding RNAs. Neurochem Res 2024; 49:2988-3005. [PMID: 39060769 DOI: 10.1007/s11064-024-04214-9] [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: 04/12/2024] [Revised: 06/16/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024]
Abstract
Aluminum (Al) is known to induce neurotoxic effects, potentially contributing to Alzheimer's disease (AD) pathogenesis. Recent studies suggest that epigenetic modification may contribute to Al neurotoxicity, although the mechanisms are still debatable. Therefore, the objective of the present study was to summarize existing data on the involvement of epigenetic mechanisms in Al-induced neurotoxicity, especially AD-type pathology. Existing data demonstrate that Al exposure induces disruption in DNA methylation, histone modifications, and non-coding RNA expression in brains. Alterations in DNA methylation following Al exposure were shown to be mediated by changes in expression and activity of DNA methyltransferases (DNMTs) and ten-eleven translocation proteins (TETs). Al exposure was shown to reduce histone acetylation by up-regulating expression of histone deacetylases (HDACs) and impair histone methylation, ultimately contributing to down-regulation of brain-derived neurotrophic factor (BDNF) expression and activation of nuclear factor κB (NF-κB) signaling. Neurotoxic effects of Al exposure were also associated with aberrant expression of non-coding RNAs, especially microRNAs (miR). Al-induced patterns of miR expression were involved in development of AD-type pathology by increasing amyloid β (Aβ) production through up-regulation of Aβ precursor protein (APP) and β secretase (BACE1) expression (down-regulation of miR-29a/b, miR-101, miR-124, and Let-7c expression), increasing in neuroinflammation through NF-κB signaling (up-regulation of miR-9, miR-125b, miR-128, and 146a), as well as modulating other signaling pathways. Furthermore, reduced global DNA methylation, altered histone modification, and aberrant miRNA expression were associated with cognitive decline in Al-exposed subjects. However, further studies are required to evaluate the contribution of epigenetic mechanisms to Al-induced neurotoxicity and/or AD development.
Collapse
Affiliation(s)
- Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Anatoly V Skalny
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Sovetskaya Str. 14, Yaroslavl, 150000, Russia
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), Bolshaya Pirogovskaya St., 2-4, Moscow, 119146, Russia
| | - Abel Santamaria
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico
- Laboratorio de Nanotecnología y Nanomedicina, Departamento de Atención a la Salud, Universidad Autónoma Metropolitana-Xochimilco, Mexico City, 04960, Mexico
| | - Joao B T Rocha
- Departamento de Bioquímica e Biologia Molecular, CCNE, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Borhan Mansouri
- Substance Abuse Prevention Research Center, Research Institute for Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, 20059, USA
| | - Roberto Madeddu
- Department of Biomedical Sciences-Histology, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
| | - Rongzu Lu
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, People's Republic of China
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Alexey A Tinkov
- Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Sovetskaya Str. 14, Yaroslavl, 150000, Russia.
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), Bolshaya Pirogovskaya St., 2-4, Moscow, 119146, Russia.
| |
Collapse
|
2
|
Liu Q, Li J, Cheng X, Chen G, Zhang Y, Tian Z, Wang Y, Wang H, Guo X, Li H, Sun L, Hu B, Zhang D, Liang C, Sheng J, Tao F, Wang J, Yang L. APOE ε4 allele modifies the associations of toxic metals and their mixture with cognitive impairment among older adults. ENVIRONMENTAL RESEARCH 2024; 255:119148. [PMID: 38754607 DOI: 10.1016/j.envres.2024.119148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/01/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND The evidence of interactive effect of the toxic metal (TM) mixture and apolipoprotein E (APOE) ε4 gene on cognitive impairment in older adults is scarce. We aimed to explore whether the associations of single TMs and their mixture with cognitive impairment depend on APOE ε4 in Chinese community-dwelling older people. METHODS A total of 1148 older adults from a subset of the baseline survey of a cohort study were included. Blood arsenic (As), cadmium (Cd), lead (Pb), strontium (Sr), and vanadium (V) were detected by inductively coupled plasma mass spectrometry. APOE gene (rs429358, rs7412) polymorphisms were analyzed by the Polymerase Chain Reaction instrument. Mixed effects logistic regression was applied to estimate the relationships of single TMs and APOE genotype with cognitive impairment. Weighted quantile sum (WQS) and Bayesian kernel machine regression (BKMR) models were performed to examine joint impacts of the TM mixture, as well as the interaction of the TM mixture with APOE ε4 genotype on cognitive impairment. RESULTS Pb displayed a significant linear association with an increased odds of cognitive impairment after adjustment for covariates (Ptrend = 0.045). While APOE genotype did not show a significant correlation with cognitive impairment. WQS showed that the TM mixture was associated with an increased risk of cognitive impairment by 31.0% (OR=1.31, 95% CI: 0.92, 1.87) while no significance was found. BKMR exhibited a significant linear association between the TM mixture and cognitive impairment. Moreover, both WQS and BKMR indicated that Pb contributed the most to cognitive impairment within the mixture. Significant interactions of Pb or the TM mixture and APOE genotype on cognitive impairment were observed, contributing to 38.1% and 38.2% of total effects, respectively. CONCLUSIONS APOE ε4 allele amplifies the associations of single Pb or the TM mixture with cognitive impairment. These findings may help to develop precision prevention.
Collapse
Affiliation(s)
- Qiang Liu
- Department of Epidemiology and Health Statistics, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Junzhe Li
- Department of Epidemiology and Health Statistics, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Xuqiu Cheng
- Department of Epidemiology and Health Statistics, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Guimei Chen
- School of Health Services Management, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yan Zhang
- Department of Epidemiology and Health Statistics, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Ziwei Tian
- Department of Epidemiology and Health Statistics, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yuan Wang
- Department of Epidemiology and Health Statistics, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Hongli Wang
- Department of Epidemiology and Health Statistics, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Xianwei Guo
- Department of Epidemiology and Health Statistics, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Huaibiao Li
- Fuyang Center for Diseases Prevention and Control, Fuyang, 236069, Anhui, China
| | - Liang Sun
- Fuyang Center for Diseases Prevention and Control, Fuyang, 236069, Anhui, China
| | - Bing Hu
- Fuyang Center for Diseases Prevention and Control, Fuyang, 236069, Anhui, China
| | - Dongmei Zhang
- School of Health Services Management, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Chunmei Liang
- Department of Hygiene Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Jie Sheng
- Scientific Research Center in Preventive Medicine, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Fangbiao Tao
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei, 230032, Anhui, China
| | - Jun Wang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Linsheng Yang
- Department of Epidemiology and Health Statistics, Center for Big Data and Population Health of IHM, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, China.
| |
Collapse
|
3
|
Abu-Elfotuh K, Hamdan AME, Mohamed SA, Bakr RO, Ahmed AH, Atwa AM, Hamdan AM, Alanzai AG, Alnahhas RK, Gowifel AMH, Salem MA. The potential anti-Alzheimer's activity of Oxalis corniculata Linn. Methanolic extract in experimental rats: Role of APOE4/LRP1, TLR4/NF-κβ/NLRP3, Wnt 3/β-catenin/GSK-3β, autophagy and apoptotic cues. JOURNAL OF ETHNOPHARMACOLOGY 2024; 324:117731. [PMID: 38218505 DOI: 10.1016/j.jep.2024.117731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/23/2023] [Accepted: 01/06/2024] [Indexed: 01/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Oxalis corniculata (O. corniculata) is a member of Oxalidaceae family, widely distributed in Asia, Europe, America, and Africa, used extensively as food and its traditional folkloric uses include management of epilepsy, gastric disorders, and neurodegenerative diseases, together with its use in enhancing health. Numerous pharmacological benefits of O. corniculata are linked to its anti-inflammatory and antioxidant abilities. One of the most prevalent neurodegenerative disorders is Alzheimer's disease (AD) in which neuroinflammation and oxidative stress are its main pathogenic processes. AIM OF THE STUDY Our research aimed to study the neuroprotective effect of the methanolic extract of Oxalis corniculata Linn. (O. corniculata ME), compared to selenium (Se) against AlCl3-induced AD. MATERIALS AND METHODS Forty male albino rats were allocated into four groups (Gps). Gp I a control group, the rest of the animals received AlCl3 (Gp II-Gp IV). Rats in Gp III and IV were treated with Se and O. corniculata ME, respectively. RESULTS The chemical profile of O. corniculata ME was studied using ultraperformance liquid chromatography-electrospray ionization-quadrupole time-of-flight mass spectrometry, allowing the tentative identification of sixty-six compounds, including organic acids, phenolics and others, cinnamic acid and its derivatives, fatty acids, and flavonoids. AlCl3 showed deterioration in short-term memory and brain histological pictures. Our findings showed that O. corniculata ME and selenium helped to combat oxidative stress produced by accumulation of AlCl3 in the brain and in prophylaxis against AD. Thus, Selenium (Se) and O. corniculata ME restored antioxidant defense, via enhancing Nrf2/HO-1 hub, hampered neuroinflammation, via TLR4/NF-κβ/NLRP3, along with dampening apoptosis, Aβ generation, tau hyperphosphorylation, BACE1, ApoE4 and LRP1 levels. Treatments also promoted autophagy and modulated Wnt 3/β-catenin/GSK3β cue. CONCLUSIONS It was noted that O. corniculata ME showed a notable ameliorative effect compared to Se on Nrf2/HO-1, TLR4/NF-κβ/NLRP3, APOE4/LRP1, Wnt 3/β-catenin/GSK-3β and PERK axes.
Collapse
Affiliation(s)
- Karema Abu-Elfotuh
- Clinical Pharmacy Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt; Al-Ayen Iraqi University, Thi-Qar, 64001, Iraq.
| | - Ahmed M E Hamdan
- Pharmacy Practice Department, Faculty of Pharmacy, University of Tabuk, Tabuk 74191, Saudi Arabia.
| | - Shaza A Mohamed
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11754, Egypt.
| | - Riham O Bakr
- Department of Pharmacognosy, Faculty of Pharmacy, October University for Modern Sciences and Arts (MSA) University, Giza 11787, Egypt.
| | - Amal H Ahmed
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11754, Egypt.
| | - Ahmed M Atwa
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo-Suez Road, Cairo 11829, Egypt.
| | - Amira M Hamdan
- Oceanography Department, Faculty of Science, Alexandria University, Alexandria 21511, Egypt.
| | | | | | - Ayah M H Gowifel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo 11571, Egypt.
| | - Maha A Salem
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo 11571, Egypt.
| |
Collapse
|
4
|
Sun Y, Guo D, Yue S, Zhou M, Wang D, Chen F, Wang L. Afzelin protects against doxorubicin-induced cardiotoxicity by promoting the AMPKα/SIRT1 signaling pathway. Toxicol Appl Pharmacol 2023; 477:116687. [PMID: 37703929 DOI: 10.1016/j.taap.2023.116687] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/15/2023]
Abstract
BACKGROUND Doxorubicin (DOX), a chemotherapeutic drug, could relieve the progressions of various diseases. However, its clinical application is limited due to its cardiotoxicity. This study aimed to investigate the effects of afzelin (a flavonol glycoside found in Houttuynia cordata) on the cardiotoxicity induced by DOX. METHODS In ex-vivo, H9C2 cells were incubated with 20, 40, or 80 μM afzelin for 12 h, followed by the treatment with 1 μM DOX for 12 h. In vivo, C57BL/6 J mice were intraperitoneally injected with 4 mg/kg/day DOX on days 1, 7, and 14. Meanwhile, starting from day 1, mice were intragastrically administrated with 5 mg/kg/day or 10 mg/kg/day afzelin for 20 days. The cardiac function of mice was evaluated by detecting hemodynamic parameters using the M-mode echocardiography. RESULTS DOX decreased the cell survival rate, and elevated apoptotic rate, as well as induced the oxidative stress and mitochondrial dysfunction in H9C2 cells. All these changes were alleviated by afzelin treatment in a concentration-dependent manner. The results were further proven by the mitigation of cardiac injury in vivo, as evidenced by the elevation of fractional shortening, heart weight/tibia length, and the rate of the increase/decrease of left ventricular pressure in mice subjected to DOX-induced cardiotoxicity. Furthermore, afzelin upregulated the expression of p-AMP-activated protein kinase alpha (AMPKα) and sirtuin1 (SIRT1). Dorsomorphin (an AMPKα inhibitor) abrogated the anti-cardiotoxicity effects of afzelin in H9C2 cells induced by DOX. CONCLUSION Afzelin protected against DOX-induced cardiotoxicity by promoting the AMPKα/SIRT1 signaling pathway.
Collapse
Affiliation(s)
- Yixin Sun
- Department of Ultrasound, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin 150081, Heilongjiang, China
| | - Danyang Guo
- Department of Ultrasound, the Sixth Affiliated Hospital of Harbin Medical University, 57 Youyi Road, Daoli District, Harbin 150076, Heilongjiang, China
| | - Saiding Yue
- Department of Nephrology, Harbin Jing-En Nephrology Hospital, 11 Xiangbin Road, Xiangfang District, Harbin 150036, Heilongjiang, China
| | - Mingyan Zhou
- Department of Ultrasound, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin 150081, Heilongjiang, China
| | - Dongxu Wang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin 150081, Heilongjiang, China
| | - Fengjiao Chen
- Department of Ultrasound, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin 150081, Heilongjiang, China
| | - Lingling Wang
- Department of Ultrasound, the Sixth Affiliated Hospital of Harbin Medical University, 998 Aiying Avenue, Songbei District, Harbin 150027, Heilongjiang, China.
| |
Collapse
|
5
|
Wang S, Xue Y, Zhang J, Meng H, Zhang J, Li X, Zhang Z, Li H, Pan B, Lu X, Zhang Q, Niu Q. Interaction between aluminum exposure and ApoEε4 gene on cognitive function of in-service workers. CHEMOSPHERE 2023; 323:138282. [PMID: 36868418 DOI: 10.1016/j.chemosphere.2023.138282] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/28/2023] [Accepted: 02/28/2023] [Indexed: 06/18/2023]
Abstract
The occurrence and development of cognitive impairment, the early stage of AD, may be affected both by factors of environmental (aluminum exposure) and genetic (ApoEε4 gene). But whether there is an interaction between the two factors on cognitive function is still unknown. To explore the interaction between the two factors on cognitive function of in-service workers. A total of 1121 in-service workers in a large aluminum factory were investigated in Shanxi Province. Cognitive function was assessed by the Mini-mental State Examination (MMSE), the clock-drawing test (CDT), the Digit Span Test (DST, including DSFT and DSBT), the fuld object memory evaluation (FOM), and the verbal fluency task (VFT). The plasma-Al (p-Al) concentrations were measured by inductively coupled plasma-mass spectrometry (ICP-MS) as an internal exposure indicator, and the participants were divided into four Al exposure groups according to the quartile of p-Al concentrations, namely Q1, Q2, Q3, and Q4. ApoE genotype was determined by Ligase Detection Reaction (LDR). The multiplicative model was fitted using non-conditional logistic regression and additive model was fitted using crossover analysis to analyze the interaction between p-Al concentrations and the ApoEε4 gene. Finally, a dose-response relationship between p-Al concentrations and cognitive impairment was observed, with the p-Al concentrations increased, cognitive function performance gradually becomes worse (Ptrend<0.05), and the risk of cognitive impairment gradually increases (Ptrend<0.05), mainly in executive/visuospatial impairment, auditory memory impairment (particularly the working memory impairment). And ApoEε4 gene may be a risk factor for cognitive impairment, while no association between the ApoEε2 gene and cognitive impairment is observed. Additionally, an additive but no multiplicative interaction between p-Al concentrations and ApoEε4 gene is observed, and when the two factors work together, the risk of cognitive impairment further increased, of which 44.2% can be attributed to the interaction effect.
Collapse
Affiliation(s)
- Shanshan Wang
- Section of Occupational Medicine, Department of Special Medicine, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Yingjun Xue
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Jintao Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Huaxing Meng
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, 030001, China; Department of Neurology, First Hospital, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Jingsi Zhang
- Section of Occupational Medicine, Department of Special Medicine, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Xiaoyan Li
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Zhuoran Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Huan Li
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, 030001, China; Department of Occupational Health, School of Public Health, Jining Medical University, Jining, Shandong, 272000, China
| | - Baolong Pan
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, 030001, China; Sixth Hospital of Shanxi Medical University (General Hospital of Tisco), Taiyuan, Shanxi, 030001, China
| | - Xiaoting Lu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, 030001, China; Key Lab of Environmental Hazards and Health Damage of Shanxi Province, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Qinli Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, 030001, China; Key Lab of Environmental Hazards and Health Damage of Shanxi Province, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, 030001, China; Key Lab of Environmental Hazards and Health Damage of Shanxi Province, Shanxi Medical University, Taiyuan, Shanxi, 030001, China.
| |
Collapse
|
6
|
Dey M, Singh RK. Exposure of aluminium to C6 glioma cells modulates molecular and functional neurotoxic markers. J Biochem Mol Toxicol 2022; 36:e23210. [PMID: 36056780 DOI: 10.1002/jbt.23210] [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: 10/22/2021] [Revised: 06/11/2022] [Accepted: 08/12/2022] [Indexed: 11/06/2022]
Abstract
The risk of aluminium exposure to humans is very high as it may get into the human body through excessive dietary contaminants, inhalation of fine particulate matter, or through parenteral routes as a vaccine adjuvant and so forth. The increased level of aluminium in brain tissue has been shown to be associated with several neurodegenerative and neurotoxic adverse effects, including AD. However, the exact mechanism of aluminium-induced neurotoxicity is still unclear. Therefore, our study aimed to investigate the mechanism of neurotoxic and neurodegenerative effects through in vitro exposure of aluminium in rat glioma C6 cell line. The findings of our study have indicated that aluminium chloride exposure may lead to alteration in acetylcholine levels, increased oxidative imbalance and induction of molecular structural and functional markers of neuronal inflammation. This study also demonstrated that aluminium exposure may lead to the induction of caspase-3 along with apoptotic cell death and a significant increase in amyloid-beta and hyperphosphorylated tau levels in C6 cells. Thus, this study may provide a mechanistic understanding of the regulation of neuroinflammatory and neurodegenerative biomarkers due to aluminium exposure.
Collapse
Affiliation(s)
- Mangaldeep Dey
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli Transit Campus, Lucknow, Uttar Pradesh, India
| | - Rakesh Kumar Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli Transit Campus, Lucknow, Uttar Pradesh, India
| |
Collapse
|
7
|
Neuroprotective Effects of Phytochemicals against Aluminum Chloride-Induced Alzheimer’s Disease through ApoE4/LRP1, Wnt3/β-Catenin/GSK3β, and TLR4/NLRP3 Pathways with Physical and Mental Activities in a Rat Model. Pharmaceuticals (Basel) 2022; 15:ph15081008. [PMID: 36015156 PMCID: PMC9416484 DOI: 10.3390/ph15081008] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/05/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Alzheimer’s disease (AD) is a neurodegenerative disorder that is associated with abnormal cognition. AD is aided in its initiation and progression by hereditary and environmental factors. Aluminum (Al) is a neurotoxic agent that causes oxidative stress, which is linked to AD progression. Additionally, Nrf2/HO-1, APOE4/LRP1, Wnt3/β-catenin, and TLR4/NLRP3 are the main signaling pathways involved in AD pathogenesis. Several phytochemicals are promising options in delaying AD evolution. Objectives: This study aimed at studying the neuroprotective effects of some phytochemicals as morin (MOR), thymol (TML), and thymoquinone (TMQ) on physical and mental activities (PhM) in Al chloride (AlCl3)-induced AD rat model. Another objective was to determine the specificity of phytochemicals to AD signaling pathways using molecular docking. Methods: Eighty male Dawley rats were divided into eight groups. Each group received: saline (control group), AlCl3, (ALAD), PhM, either alone or with a combination of MOR, TML, and/or TMQ for five weeks. Animals were then subjected to behavioral evaluation. Brain tissues were used for histopathological and biochemical analyses to determine the extent of neurodegeneration. The effect of phytochemicals on AlCl3-induced oxidative stress and the main signaling pathways involved in AD progression were also investigated. Results: AlCl3 caused a decline in spatial learning and memory, as well as histopathological changes in the brains of rats. Phytochemicals combined with PhM restored antioxidant activities, increased HO-1 and Nrf2 levels, blocked inflammasome activation, apoptosis, TLR4 expression, amyloide-β generation, and tau hyperphophorylation. They also brought ApoE4 and LRP1 levels back to normal and regulated Wnt3/β-catenin/GSK3β signaling pathway. Conclusions: The use of phytochemicals with PhM is a promising strategy for reducing AD by modulating Nrf2/HO-1, TLR4/NLRP3, APOE4/LRP1, and Wnt3/β-catenin/GSK-3β signaling pathways.
Collapse
|
8
|
Yu Q, Zhu K, Ding Y, Han R, Cheng D. Comparative study of aluminum (Al) speciation on apoptosis-promoting process in PC12 cells: Correlations between morphological characteristics and mitochondrial kinetic disorder. J Inorg Biochem 2022; 232:111835. [DOI: 10.1016/j.jinorgbio.2022.111835] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 12/12/2022]
|
9
|
Dey M, Singh RK. Chronic oral exposure of aluminum chloride in rat modulates molecular and functional neurotoxic markers relevant to Alzheimer's disease. Toxicol Mech Methods 2022; 32:616-627. [PMID: 35341471 DOI: 10.1080/15376516.2022.2058898] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Aluminum is an environmentally abundant potential neurotoxic agent that may result in oxidative damage to a range of cellular biomarkers. The potential sources of aluminum accumulation in body include drinking water, food, medicines, vaccines, and aluminum cookware utensils etc. The accumulation of aluminum in brain is reported to be associated with cholinergic dysfunction, oxidative stress and neuronal damage, that may ultimately cause Alzheimer's disease. Since chronic exposure of aluminum leads to its accumulation in brain, so this study was done by a long-term (24 weeks) low dose (20 mg/kg) oral exposure of aluminum chloride in rats. In this chronic model, we have evaluated the major hallmarks of Alzheimer's disease including amyloid beta (Aβ1-42) and phosphorylated-tau (p231-tau) protein in brain tissue. Furthermore, we evaluated the level of acetyl cholinesterase activity, inflammatory cytokines such as TNF-α, IL-6 and IL-1β, and oxidative stress biomarkers in rat brain in this model. The neurobehavioral parameters were also assessed in animals by using spontaneous locomotor activity, passive avoidance, rotarod test and novel object recognition test to evaluate alteration in learning, memory and muscle co-ordination. We found that chronic oral exposure of aluminum chloride causes a significant increase in structural hallmarks such as Aβ1-42 and p231-tau levels along with proinflammatory cytokines (TNF-α and IL-6), oxidative stress, and a decrease in antioxidant markers such as GSH and catalase. in the brain tissue. These biomarkers significantly affected neurobehavioral parameters in animals. This study provides a mechanistic understanding of chronic aluminum-induced neuronal toxicity in brain with relevance to Alzheimer's disease.
Collapse
Affiliation(s)
- Mangaldeep Dey
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli. Transit campus, Bijnour-sisendi road, Sarojini nagar, Lucknow-226002, Uttar Pradesh, India
| | - Rakesh Kumar Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli. Transit campus, Bijnour-sisendi road, Sarojini nagar, Lucknow-226002, Uttar Pradesh, India
| |
Collapse
|
10
|
Ding M, Shu Q, Zhang N, Yan C, Niu H, Li X, Guan P, Hu X. Electrochemical Immunosensor for the Sensitive Detection of Alzheimer's Biomarker Amyloid‐β (1–42) Using the Heme‐amyloid‐β (1–42) Complex as the Signal Source. ELECTROANAL 2021. [DOI: 10.1002/elan.202100392] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Minling Ding
- School of Chemistry and Chemical Engineering Northwestern Polytechnical University Xi'an 710072 P. R. China
| | - Qi Shu
- School of Chemistry and Chemical Engineering Northwestern Polytechnical University Xi'an 710072 P. R. China
| | - Nan Zhang
- School of Chemistry and Chemical Engineering Northwestern Polytechnical University Xi'an 710072 P. R. China
| | - Chaoren Yan
- School of Chemistry and Chemical Engineering Northwestern Polytechnical University Xi'an 710072 P. R. China
| | - Huizhe Niu
- School of Chemistry and Chemical Engineering Northwestern Polytechnical University Xi'an 710072 P. R. China
| | - Xiaoqian Li
- School of Chemistry and Chemical Engineering Northwestern Polytechnical University Xi'an 710072 P. R. China
| | - Ping Guan
- School of Chemistry and Chemical Engineering Northwestern Polytechnical University Xi'an 710072 P. R. China
| | - Xiaoling Hu
- School of Chemistry and Chemical Engineering Northwestern Polytechnical University Xi'an 710072 P. R. China
| |
Collapse
|
11
|
Kong L, Wu Y, Hu W, Liu L, Xue Y, Liang G. Mechanisms underlying reproductive toxicity induced by nickel nanoparticles identified by comprehensive gene expression analysis in GC-1 spg cells. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 275:116556. [PMID: 33588191 DOI: 10.1016/j.envpol.2021.116556] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 01/04/2021] [Accepted: 01/17/2021] [Indexed: 06/12/2023]
Abstract
The public around the world is increasingly concerned about male reproductive health. The impact of nickel nanoparticles (Ni NPs) on male reproductive toxicity including sperm production, motility and fertilizing capacity has been confirmed by our previous researches. In the current study of Ni NPs-inducing toxicity, the expression profiles of piRNAs and their predicted target genes associated with male infertility, were obtained. The results showed that piR-mmu-32362259 was the highest differential expression multiples in both the testis tissues of male mice and GC-1 cells similarly. Notably, piR-mmu-32362259 target gene was significantly enriched in the PI3K-AKT signaling pathway. All these results suggest that piR-mmu-32362259 may affect the occurrence and development of injury in the mouse spermatogenesis process by regulating the PI3K-AKT signaling pathway. In order to verify the result, piR-mmu-32362259 low-expression lentivirus was used to transfect GC-1 cells to establish a stable transfected cell model. The effects of piR-mmu-32362259 on the viability, cycle and apoptosis as well as related protein expression levels of GC-1 cells induced by Ni NPs were detected using CCK8, flow cytometry and western blot assay, respectively. The results showed that low expression of piR-mmu-32362259 could not only alleviate the decrease of GC-1 cell viability, affect the cell cycle and reduce the apoptosis rate, but also significantly affect the expression levels of key proteins and their downstream molecules of PI3K/AKT/mTOR signaling pathway. Collectively, our current results provide a theoretical basis for further exploring the molecular regulatory mechanism of male reproductive toxicity induced by Ni NPs.
Collapse
Affiliation(s)
- Lu Kong
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, PR China.
| | - Yongya Wu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, PR China.
| | - Wangcheng Hu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, PR China.
| | - Lin Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, PR China.
| | - Yuying Xue
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, PR China.
| | - Geyu Liang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu, 210009, PR China.
| |
Collapse
|
12
|
Chen H, Chen F, Zhang M, Chen Y, Cui L, Liang C. A Review of APOE Genotype-Dependent Autophagic Flux Regulation in Alzheimer's Disease. J Alzheimers Dis 2021; 84:535-555. [PMID: 34569952 DOI: 10.3233/jad-210602] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Autophagy is a basic physiological process maintaining cell renewal, the degradation of dysfunctional organelles, and the clearance of abnormal proteins and has recently been identified as a main mechanism underlying the onset and progression of Alzheimer's disease (AD). The APOE ɛ4 genotype is the strongest genetic determinant of AD pathogenesis and initiates autophagic flux at different times. This review synthesizes the current knowledge about the potential pathogenic effects of ApoE4 on autophagy and describes its associations with the biological hallmarks of autophagy and AD from a novel perspective. Via a remarkable variety of widely accepted signaling pathway markers, such as mTOR, TFEB, SIRT1, LC3, p62, LAMP1, LAMP2, CTSD, Rabs, and V-ATPase, ApoE isoforms differentially modulate autophagy initiation; membrane expansion, recruitment, and enclosure; autophagosome and lysosome fusion; and lysosomal degradation. Although the precise pathogenic mechanism varies for different genes and proteins, the dysregulation of autophagic flux is a key mechanism on which multiple pathogenic processes converge.
Collapse
Affiliation(s)
- Huiyi Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Yuebei People's Hospital, Affiliated Hospital of Shantou University Medical College, Shaoguan, China
| | - Feng Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Miaoping Zhang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yanting Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lili Cui
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Chunmei Liang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
13
|
Kawahara M, Kato-Negishi M, Tanaka KI. Amyloids: Regulators of Metal Homeostasis in the Synapse. Molecules 2020; 25:molecules25061441. [PMID: 32210005 PMCID: PMC7145306 DOI: 10.3390/molecules25061441] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/17/2020] [Accepted: 03/20/2020] [Indexed: 12/11/2022] Open
Abstract
Conformational changes in amyloidogenic proteins, such as β-amyloid protein, prion proteins, and α-synuclein, play a critical role in the pathogenesis of numerous neurodegenerative diseases, including Alzheimer’s disease, prion disease, and Lewy body disease. The disease-associated proteins possess several common characteristics, including the ability to form amyloid oligomers with β-pleated sheet structure, as well as cytotoxicity, although they differ in amino acid sequence. Interestingly, these amyloidogenic proteins all possess the ability to bind trace metals, can regulate metal homeostasis, and are co-localized at the synapse, where metals are abundantly present. In this review, we discuss the physiological roles of these amyloidogenic proteins in metal homeostasis, and we propose hypothetical models of their pathogenetic role in the neurodegenerative process as the loss of normal metal regulatory functions of amyloidogenic proteins. Notably, these amyloidogenic proteins have the capacity to form Ca2+-permeable pores in membranes, suggestive of a toxic gain of function. Therefore, we focus on their potential role in the disruption of Ca2+ homeostasis in amyloid-associated neurodegenerative diseases.
Collapse
|