1
|
Wang L, Zhang J, Zhao Y, Li J, Lu X, Song J, Zhang L, Niu Q. Nuclear factor kappa B (NF-κB) participates in the aluminum-induced down-regulation of miR29a/b1. J Trace Elem Med Biol 2023; 80:127309. [PMID: 37801786 DOI: 10.1016/j.jtemb.2023.127309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 08/11/2023] [Accepted: 09/20/2023] [Indexed: 10/08/2023]
Abstract
BACKGROUND Studies have shown that aluminum (Al) is one of the environmental risk factors leading to Alzheimer's disease (AD), and Al exposure can cause elevated levels of BACE1mRNA, β-secretase (BACE1), and amyloid beta (Aβ) in vivo and in vitro. Previous studies by our research group have shown that this is partly caused by the negative regulation of BACE1 by miRNA29a/b1 (miR29a/b1). Despite the observed the role of nuclear factor kappa B (NF-κB) on many miRNAs, the upstream regulation of NF-κB protein on miR29 remains poorly understood. The purpose of this study was to better define the relationship between NF-κB and miR29a/b1 and the potentially relevant signaling pathways. METHODS On the one hand, we constructed the animal model of Al exposure by the intraperitoneal injection of aluminum-maltolate (Al(mal)3) in rats. Conversely, NF- κB inhibitors were added to adrenal phaeochromocytoma (PC12) cells exposed to Al(mal)3. RESULTS We verified that NF-κB shows an increasing trend with Al accumulation in the brain of rats, which is accompanied by a downward trend of miR29a/b1. Notably, the suppression of NF-κB significantly increased miR29a/b1 and affected the expression of BACE1mRNA and downstream proteins. CONCLUSION Al-induced NF-κB can negatively regulate the expression of miR29a/b1, which then significantly enhances the expression of BACE1 and Aβ plaques.
Collapse
Affiliation(s)
- Linping Wang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; NHC Key Laboratory of Pneumoconiosis (Shanxi), Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Jingqi Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; NHC Key Laboratory of Pneumoconiosis (Shanxi), Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Yue Zhao
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; NHC Key Laboratory of Pneumoconiosis (Shanxi), Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Juan Li
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; NHC Key Laboratory of Pneumoconiosis (Shanxi), Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Xiaoting Lu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; NHC Key Laboratory of Pneumoconiosis (Shanxi), Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Jing Song
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; NHC Key Laboratory of Pneumoconiosis (Shanxi), Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Ling Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; NHC Key Laboratory of Pneumoconiosis (Shanxi), Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; NHC Key Laboratory of Pneumoconiosis (Shanxi), Medical University, Taiyuan, 030001 Shanxi Province, China.
| |
Collapse
|
2
|
Aydin N, Turkez H, Tozlu OO, Arslan ME, Yavuz M, Sonmez E, Ozpolat OF, Cacciatore I, Di Stefano A, Mardinoglu A. Ameliorative Effects by Hexagonal Boron Nitride Nanoparticles against Beta Amyloid Induced Neurotoxicity. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12152690. [PMID: 35957121 PMCID: PMC9370266 DOI: 10.3390/nano12152690] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/30/2022] [Accepted: 08/02/2022] [Indexed: 05/28/2023]
Abstract
Alzheimer’s disease (AD) is considered as the most common neurodegenerative disease. Extracellular amyloid beta (Aβ) deposition is a hallmark of AD. The options based on degradation and clearance of Aβ are preferred as promising therapeutic strategies for AD. Interestingly, recent findings indicate that boron nanoparticles not only act as a carrier but also play key roles in mediating biological effects. In the present study, the aim was to investigate the effects of different concentrations (0−500 mg/L) of hexagonal boron nitride nanoparticles (hBN-NPs) against neurotoxicity by beta amyloid (Aβ1-42) in differentiated human SH-SY5Y neuroblastoma cell cultures for the first time. The synthesized hBN-NPs were characterized by X-ray diffraction (XRD) measurements, scanning electron microscopy (SEM) and transmission electron microscopy (TEM). Aβ1-42-induced neurotoxicity and therapeutic potential by hBN-NPs were assessed on differentiated SH-SY5Y cells using MTT and LDH release assays. Levels of total antioxidant capacity (TAC) and total oxidant status (TOS), expression levels of genes associated with AD and cellular morphologies were examined. The exposure to Aβ1-42 significantly decreased the rates of viable cells which was accompanied by elevated TOS level. Aβ1-42 induced both apoptotic and necrotic cell death. Aβ exposure led to significant increases in expression levels of APOE, BACE 1, EGFR, NCTSN and TNF-α genes and significant decreases in expression levels of ADAM 10, APH1A, BDNF, PSEN1 and PSENEN genes (p < 0.05). All the Aβ1-42-induced neurotoxic insults were inhibited by the applications with hBN-NPs. hBN-NPs also suppressed the remarkable elevation in the signal for Aβ following exposure to Aβ1-42 for 48 h. Our results indicated that hBN-NPs could significantly prevent the neurotoxic damages by Aβ. Thus, hBN-NPs could be a novel and promising anti-AD agent for effective drug development, bio-nano imaging or drug delivery strategies.
Collapse
Affiliation(s)
- Nursah Aydin
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum 25050, Turkey
| | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum 25240, Turkey
- East Anatolia High Technology Application and Research Center (DAYTAM), Ataturk University, Erzurum 25240, Turkey
| | - Ozlem Ozdemir Tozlu
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum 25050, Turkey
| | - Mehmet Enes Arslan
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum 25050, Turkey
| | - Mehmet Yavuz
- REEM Neuropsychiatry Clinics, İstanbul 34245, Turkey
| | - Erdal Sonmez
- Department of Nanoscience and Nanoengineering, Graduate School of Natural and Applied Sciences, Ataturk University, Erzurum 25240, Turkey
- Department of Physics, Kazım Karabekir Education Faculty, Atatürk University, Erzurum 25240, Turkey
| | - Ozgur Fırat Ozpolat
- Computer Sciences Research and Application Center, Atatürk University, Erzurum 25240, Turkey
| | - Ivana Cacciatore
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti Scalo, CH, Italy
| | - Antonio Di Stefano
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini, 31, 66100 Chieti Scalo, CH, Italy
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH-Royal Institute of Technology, SE-17121 Stockholm, Sweden
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK
| |
Collapse
|
3
|
Makhdoomi S, Mahboobian MM, Haddadi R, Komaki A, Mohammadi M. Silibinin-loaded nanostructured lipid carriers (NLCs) ameliorated cognitive deficits and oxidative damages in aluminum chloride-induced neurotoxicity in male mice. Toxicology 2022; 477:153260. [PMID: 35850386 DOI: 10.1016/j.tox.2022.153260] [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] [Received: 05/13/2022] [Revised: 07/09/2022] [Accepted: 07/14/2022] [Indexed: 10/17/2022]
Abstract
Aluminum chloride (AlCl3) and its accumulation in the brain are associated with neurodegenerative disease. Recent investigations have illustrated that silibinin is known to have neuroprotective properties. The present study investigates the neuroprotective effects of silibinin-loaded nanostructured lipid carriers (Sili-NLCs) against AlCl3-induced neurotoxicity in male mice. Sili-NLCs were prepared using the emulsification-solvent evaporation method and subjected to particle size, zeta potential, and entrapment efficiency (% EE) analysis. Mice were treated with AlCl3 (100 mg/kg/day, p.o.) and with the same concentration of silibinin and Sili-NLCs (50,100, and 200 mg/kg/day, p.o.) for 30 days in different groups. After treating animals, behavioral studies were assessed. Also, the brain tissue samples were collected from all mice to evaluate oxidative damage and histological changes. The particle size, polydispersity index, zeta potential, and entrapment efficiency (% EE) of prepared Sili-NLCs found 239.7 ± 4.04 nm, 0.082 ± 0.003, - 16.33 ± 0.15 mV, and 72.65 ± 2.03 %, respectively. Brain uptake studies showed that Sili-NLCs had a 5.7-fold greater uptake in the mice brain than the free drug. The AlCl3 caused significant cognitive impairment and increased the level of lipid peroxidation accompanied by decreasing antioxidant enzyme activity in the brain tissue. These findings correlated well with the histopathological experiments. Furthermore, treatment with Sili-NLCs significantly improved the AlCl3-induced cognitive impairment, neurochemical anomalies, and histopathological changes. Given these results, silibinin, when delivered using NLCs, is potentially more effective than free silibinin in decreasing AlCl3- induced neurotoxicity.
Collapse
Affiliation(s)
- Sajjad Makhdoomi
- Department of Pharmacology & Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Mehdi Mahboobian
- Department of Pharmaceutics, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rasool Haddadi
- Department of Pharmacology and Toxicology, School of Pharmacy, Herbal Medicine and natural product Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Komaki
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mojdeh Mohammadi
- Department of Pharmacology & Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
4
|
Abdel-Aal RA, Hussein OA, Elsaady RG, Abdelzaher LA. Naproxen as a potential candidate for promoting rivastigmine anti-Alzheimer activity against aluminum chloride-prompted Alzheimer's-like disease in rats; neurogenesis and apoptosis modulation as a possible underlying mechanism. Eur J Pharmacol 2022; 915:174695. [PMID: 34914971 DOI: 10.1016/j.ejphar.2021.174695] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/29/2021] [Accepted: 12/07/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND AND AIM Alzheimer's disease (AD) is one of the leading causes of dependence and disability among the elderly worldwide. The traditional anti-Alzheimer medication, rivastigmine, one of the cholinesterase inhibitors (ChEIs), fails to achieve a definitive cure. We tested the hypothesis that naproxen administration to the rivastigmine-treated aluminum chloride (AlCl3) Alzheimer's rat model could provide an additive neuroprotective effect compared to rivastigmine alone. MATERIALS AND METHODS The studied groups were control (Cont), AlCl3 treated (Al), rivastigmine treated (RIVA), naproxen treated (Napro), and combined rivastigmine and naproxen treated (RIVA + Napro). Rats' memory, spatial learning, and cognitive behavior were assessed followed by evaluation of hippocampal acetylcholinesterase (AChE) activity. Hippocampal and cerebellar histopathology were thoroughly examined. Activated caspase-3 and the neuroepithelial stem cells marker; nestin expressions were immunohistochemically assayed. RESULTS AD rats displayed significantly impaired memory and cognitive function, augmented hippocampal AChE activity; massive neurodegeneration associated with enhanced astrogliosis, apoptosis, and impaired neurogenesis. Except for the enhancement of neurogenesis and suppression of apoptosis, the combination therapy had no additional neuroprotective benefit over rivastigmine-only therapy. CONCLUSION Naproxen's efficacy was established by its ability to function at the cellular level, improved neurogenesis, and decreased, apoptosis without having an additional mitigating impact on cognitive impairment in rivastigmine-treated AD rats.
Collapse
Affiliation(s)
- Raafat A Abdel-Aal
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ola A Hussein
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Reham G Elsaady
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Lobna A Abdelzaher
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt.
| |
Collapse
|
5
|
Song J, Yuan C, Li W, Gao T, Lu X, Wang L. APP palmitoylation is involved in the increase in Aβ 1-42 induced by aluminum. Brain Res 2022; 1774:147709. [PMID: 34758347 DOI: 10.1016/j.brainres.2021.147709] [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: 08/06/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 11/29/2022]
Abstract
The increase in Aβ1-42 is a neurotoxic effect induced by aluminum which can lead to impairment of learning and memory, but its mechanism has yet to be fully elucidated. Studies have shown that APP palmitoylation is appears to be involved in the production process of Aβ1-42. Here, we investigated whether APP palmitoylation is related to the increase in Aβ caused by aluminum and its specific mechanism of action. In this study, APP palmitoylation was studied in the setting of aluminum-induced increases in Aβ1-42 from two perspectives: whole animal experiments and in vitro cell experiments. First, the learning and memory of rats were impaired and the number of rat cortical neurons was decreased after staining with aluminum. Second, the expression of palmitoyl APP, APP in lipid rafts and palmitoyl acyltransferase zDHHC7 both in rat cerebral cortex and PC12 cells increased with the production of Aβ1-42 induced by aluminum in a dose-dependent manner. Finally, the intervention with the palmitoylation inhibitors 2-BP and siRNA zDHHC7 in PC12 cells reduced levels of palmitoyl APP, the expression of APP in lipid rafts and the content of Aβ1-42 induced by aluminum to a certain extent. Our results indicate that increased APP palmitoylation levels may be related to the increase in Aβ1-42 caused by aluminum, and the mechanism may involve APP palmitoylation promoting the accumulation of APP protein on lipid rafts and the cleavage of APP by BACE1 in amyloidogenic pathway. The increase in expression of zDHHC7 may be one of the reasons for the increase in levels of APP palmitoylation caused by aluminum.
Collapse
Affiliation(s)
- Jing Song
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, China.
| | - Chunman Yuan
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China
| | - Wenjing Li
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China
| | - Ting Gao
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China
| | - Xiaoting Lu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, China
| | - Linping Wang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, China
| |
Collapse
|
6
|
Santos J, Quimque MT, Liman RA, Agbay JC, Macabeo APG, Corpuz MJA, Wang YM, Lu TT, Lin CH, Villaflores OB. Computational and Experimental Assessments of Magnolol as a Neuroprotective Agent and Utilization of UiO-66(Zr) as Its Drug Delivery System. ACS OMEGA 2021; 6:24382-24396. [PMID: 34604621 PMCID: PMC8482410 DOI: 10.1021/acsomega.1c02555] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Indexed: 05/26/2023]
Abstract
The phenolic natural product magnolol exhibits neuroprotective properties through β-amyloid toxicity in PC-12 cells and ameliorative effects against cognitive deficits in a TgCRND8 transgenic mice model. Its bioavailability and blood-brain barrier crossing ability have been significantly improved using the metal-organic framework (MOF) UiO-66(Zr) as a drug delivery system (DDS). To investigate the neuroprotective effects of the Zr-based DDS, magnolol and magnolol-loaded-UiO-66(Zr) (Mag@UiO-66(Zr)) were evaluated for inhibitory activity against β-secretase and AlCl3-induced neurotoxicity. Due to the moderate inhibition observed for magnolol in vitro, in silico binding studies were explored against β-secretase along with 11 enzymes known to affect Alzheimer's disease (AD). Favorable binding energies against CDK2, CKD5, MARK, and phosphodiesterase 3B (PDE3B) and dynamically stable complexes were noted through molecular docking and molecular dynamic simulation experiments, respectively. The magnolol-loaded DDS UiO-66(Zr) also showed enhanced neuroprotective activity against two pathological indices, namely, neutrophil infiltration and apoptotic neurons, in addition to damage reversal compared to magnolol. Thus, MOFs are promising drug delivery platforms for poorly bioavailable drugs.
Collapse
Affiliation(s)
- Joshua Santos
- The
Graduate School, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
- Phytochemistry
Laboratory, Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
| | - Mark Tristan Quimque
- The
Graduate School, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
- Laboratory
of Organic Reactivity, Discovery, and Synthesis (LORDS), Research
Center for Natural and Applied Sciences, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
- Department
of Chemistry, College of Science and Mathematics, Mindanao State University-Iligan Institute of Technology, Tibanga, 9200 Iligan City, Philippines
| | - Rhenz Alfred Liman
- The
Graduate School, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
- Phytochemistry
Laboratory, Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
| | - Jay Carl Agbay
- Department
of Chemistry, College of Science and Mathematics, Mindanao State University-Iligan Institute of Technology, Tibanga, 9200 Iligan City, Philippines
- Philippine
Science High School-Central Mindanao Campus, 9217 Balo-i, Lanao del Norte, Philippines
| | - Allan Patrick G. Macabeo
- Laboratory
of Organic Reactivity, Discovery, and Synthesis (LORDS), Research
Center for Natural and Applied Sciences, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
| | - Mary Jho-Anne Corpuz
- The
Graduate School, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
- Pharmacology
Laboratory, Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
- Department
of Pharmacy, Faculty of Pharmacy, University
of Santo Tomas, España
Blvd., 1015 Manila, Philippines
| | - Yun-Ming Wang
- Department
of Biological Science and Technology, Institute of Molecular Medicine
and Bioengineering, Center for Intelligent Drug Systems and Smart
Bio-devices (IDS2B), National Chiao Tung
University, 30010 Hsinchu, Taiwan
| | - Tsai-Te Lu
- Institute
of Biomedical Engineering, National Tsing
Hua University, 30013 Hsinchu, Taiwan
| | - Chia-Her Lin
- College
of Science, Chung Yuan Christian University, Zhongli District, 320 Taoyuan City, Taiwan
| | - Oliver B. Villaflores
- The
Graduate School, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
- Phytochemistry
Laboratory, Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
- Department of Biochemistry, Faculty of Pharmacy, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
| |
Collapse
|
7
|
Li H, Liu Q, Xue X, Lu X, Song J, He C, Hao Y, Nie J, Zhang Q, Zhao Y, Pan B, Wang L, Niu Q. miR-29a/b1 Regulates BACE1 in Aluminum-Induced Aβ Deposition in Vitro. ACS Chem Neurosci 2021; 12:3250-3265. [PMID: 34415727 DOI: 10.1021/acschemneuro.1c00444] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Aluminum is an environmental neurotoxin that comes extensively in contact with human beings. Animal and human studies demonstrated that aluminum exposure increases the deposition of beta amyloid proteins in the brain as it was observed in Alzheimer's disease. The purpose of this study was to investigate whether miR-29a/b1 affected the expression of beta-secrete enzymes (BACE1) in the process of amyloid β-protein (Aβ) deposition caused by aluminum exposure. The study was performed using two different cell lines. Our results showed that after rat primary cortical neurons were exposed to aluminum, BACE1 gene and protein levels increased to different degrees, and the expression level of Aβ1-42 increased. In aluminum-exposed groups, the expression of miR-29a and miR-29b1 decreased, while the expression of amyloid protein Aβ1-42 and BACE1 increased. In miRs transfection groups, the expression of amyloid protein Aβ1-42 and BACE1 decreased. Aluminum may affect the expression of BACE1 by lowering miR-29a and miR-29b1. AEK293 cells were utilized in this research since they present elevated levels of miR-29a and miR-29b1. After HEK293 cells were exposed to aluminum alone, BACE1 mRNA and BACE1 protein expression levels increased with the increase of aluminum exposure dose (p < 0.05), and the level of Aβ1-42 also increased (p < 0.05). Compared with the group exposed to aluminum alone at the same doses, the expression levels of BACE1 mRNA and BACE1 protein in the miRs transfected plus aluminum-exposed groups significantly decreased (p < 0.05), and the level of Aβ1-42 also decreased (p < 0.05). This result is consistent with the investigation in rat primary neurons. The results of two types of cells showed that aluminum may cause abnormal down-regulation of the expressions of miR-29a and miR-29b1, thus negatively regulating the increase of BACE1 expression and finally leading to the increase of Aβ.
Collapse
Affiliation(s)
- Huan Li
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
- Department of Occupational Health, School of Public Health, Jining Medical University, Jining 272000, China
| | - Qun Liu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Xingli Xue
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Xiaoting Lu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
- Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan 030001, China
- Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, Taiyuan 030001, China
| | - Jing Song
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
- Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan 030001, China
- Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, Taiyuan 030001, China
| | - Chanting He
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Yanxia Hao
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Jisheng Nie
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
- Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan 030001, China
- Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, Taiyuan 030001, China
| | - Qinli Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
- Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan 030001, China
- Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, Taiyuan 030001, China
| | - Yue Zhao
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Baolong Pan
- Sixth Hospital of Shanxi Medical University (General hospital of Tisco), Taiyuan 030001, China
| | - Linping Wang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
- Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan 030001, China
- Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, Taiyuan 030001, China
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
- Department of Occupational Health, School of Public Health, Xuzhou Medical University, Xuzhou 221000, China
- Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, Taiyuan 030001, China
- Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
8
|
Maugeri G, Bucolo C, Drago F, Rossi S, Di Rosa M, Imbesi R, D'Agata V, Giunta S. Attenuation of High Glucose-Induced Damage in RPE Cells through p38 MAPK Signaling Pathway Inhibition. Front Pharmacol 2021; 12:684680. [PMID: 34025440 PMCID: PMC8138305 DOI: 10.3389/fphar.2021.684680] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/26/2021] [Indexed: 12/18/2022] Open
Abstract
This study aimed to investigate the high glucose damage on human retinal pigment epithelial (RPE) cells, the role of p38 MAPK signaling pathway and how dimethyl fumarate can regulate that. We carried out in vitro studies on ARPE-19 cells exposed to physiological and high glucose (HG) conditions, to evaluate the effects of DMF on cell viability, apoptosis, and expression of inflammatory and angiogenic biomarkers such as COX-2, iNOS, IL-1β, and VEGF. Our data have demonstrated that DMF treatment attenuated HG-induced apoptosis, as confirmed by reduction of BAX/Bcl-2 ratio. Furthermore, in RPE cells exposed to HG we observed a significant increase of iNOS, COX-2, and IL-1β expression, that was reverted by DMF treatment. Moreover, DMF reduced the VEGF levels elicited by HG, inhibiting p38 MAPK signaling pathway. The present study demonstrated that DMF provides a remarkable protection against high glucose-induced damage in RPE cells through p38 MAPK inhibition and the subsequent down-regulation of VEGF levels, suggesting that DMF is a small molecule that represents a good candidate for diabetic retinopathy treatment and warrants further in vivo and clinical evaluation.
Collapse
Affiliation(s)
- Grazia Maugeri
- Section of Anatomy, Histology and Movement Sciences, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Claudio Bucolo
- Pharmacology Section, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Center for Research in Ocular Pharmacology (CERFO), University of Catania, Catania, Italy
| | - Filippo Drago
- Pharmacology Section, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Center for Research in Ocular Pharmacology (CERFO), University of Catania, Catania, Italy
| | - Settimio Rossi
- Eye Clinic, Multidisciplinary Department of Medical, Surgical and Dental Sciences, University of Campania "Luigi Vanvitelli", Napoli, Italy
| | - Michelino Di Rosa
- Section of Anatomy, Histology and Movement Sciences, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rosa Imbesi
- Section of Anatomy, Histology and Movement Sciences, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Velia D'Agata
- Section of Anatomy, Histology and Movement Sciences, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Salvatore Giunta
- Section of Anatomy, Histology and Movement Sciences, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
9
|
Molecular mechanisms of aluminum neurotoxicity: Update on adverse effects and therapeutic strategies. ADVANCES IN NEUROTOXICOLOGY 2021; 5:1-34. [PMID: 34263089 DOI: 10.1016/bs.ant.2020.12.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
10
|
Syeda T, Cannon JR. Environmental exposures and the etiopathogenesis of Alzheimer's disease: The potential role of BACE1 as a critical neurotoxic target. J Biochem Mol Toxicol 2021; 35:e22694. [PMID: 33393683 DOI: 10.1002/jbt.22694] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a major public health crisis due to devastating cognitive symptoms, a lack of curative treatments, and increasing prevalence. Most cases are sporadic (>95% of cases) after the age of 65 years, implicating an important role of environmental factors in disease pathogenesis. Environmental neurotoxicants have been implicated in neurodegenerative disorders including Parkinson's Disease and AD. Animal models of AD and in vitro studies have shed light on potential neuropathological mechanisms, yet the biochemical and molecular underpinnings of AD-relevant environmental neurotoxicity remain poorly understood. Beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) is a potentially critical pathogenic target of environmentally induced neurotoxicity. BACE1 clearly has a critical role in AD pathophysiology: It is required for amyloid beta production and expression and activity of BACE1 are increased in the AD brain. Though the literature on BACE1 in response to environmental insults is limited, current studies, along with extensive AD neurobiology literature suggest that BACE1 deserves attention as an important neurotoxic target. Here, we critically review research on environmental neurotoxicants such as metals, pesticides, herbicides, fungicides, polyfluoroalkyl substances, heterocyclic aromatic amines, advanced glycation end products, and acrolein that modulate BACE1 and potential mechanisms of action. Though more research is needed to clearly understand whether BACE1 is a critical mediator of AD-relevant neurotoxicity, available reports provide convincing evidence that BACE1 is altered by environmental risk factors associated with AD pathology, implying that BACE1 inhibition and its use as a biomarker should be considered in AD management and research.
Collapse
Affiliation(s)
- Tauqeerunnisa Syeda
- School of Health Sciences, Purdue University, West Lafayette, Indiana, USA.,Purdue Institute for Integrative Neurosciences, Purdue University, West Lafayette, Indiana, USA
| | - Jason R Cannon
- School of Health Sciences, Purdue University, West Lafayette, Indiana, USA.,Purdue Institute for Integrative Neurosciences, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
11
|
Aluminum affects neural phenotype determination of embryonic neural progenitor cells. Arch Toxicol 2019; 93:2515-2524. [PMID: 31363819 DOI: 10.1007/s00204-019-02522-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 07/18/2019] [Indexed: 10/26/2022]
Abstract
Aluminum (Al) is a neurotoxin and is associated with the etiology of neurodegenerative diseases, such as Alzheimer's disease (AD). The Al-free ion (Al3+) is the biologically reactive and toxic form. However, the underlying mechanisms of Al toxicity in the brain remain unclear. Here, we evaluated the effects of Al3+ (in the chloride form-AlCl3) at different concentrations (0.1-100 µM) on the morphology, proliferation, apoptosis, migration and differentiation of neural progenitor cells (NPCs) isolated from embryonic telencephalons, cultured as neurospheres. Our results reveal that Al3+ at 100 µM reduced the number and diameter of neurospheres. Cell cycle analysis showed that Al3+ had a decisive function in proliferation inhibition of NPCs during neural differentiation and induced apoptosis on neurospheres. In addition, 1 µM Al3+ resulted in deleterious effects on neural phenotype determination. Flow cytometry and immunocytochemistry analysis showed that Al3+ promoted a decrease in immature neuronal marker β3-tubulin expression and an increase in co-expression of the NPC marker nestin and glial fibrillary acidic protein. Thus, our findings indicate that Al3+ caused cellular damage and reduced proliferation and migration, resulting in global inhibition of NPC differentiation and neurogenesis.
Collapse
|
12
|
Yang MH, Chen SC, Lin YF, Lee YC, Huang MY, Chen KC, Wu HY, Lin PC, Gozes I, Tyan YC. Reduction of aluminum ion neurotoxicity through a small peptide application - NAP treatment of Alzheimer's disease. J Food Drug Anal 2019; 27:551-564. [PMID: 30987727 PMCID: PMC9296191 DOI: 10.1016/j.jfda.2018.11.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 11/16/2018] [Accepted: 11/20/2018] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in late life. It is difficult to precisely diagnose AD at early stages, making biomarker search essential for further developments. The objective of this study was to identify protein biomarkers associated with aluminum ions toxicity (AD-like toxicity) in a human neuroblastoma cell model, SH-SY5Y and assess potential prevention by NAP (NAPVSIPQ). Complete proteomic techniques were implemented. Four proteins were identified as up-regulated with aluminum ion treatment, CBP80/20-dependent translation initiation factor (CTIF), Early endosome antigen 1 (EEA1), Leucine-rich repeat neuronal protein 4 (LRRN4) and Phosphatidylinositol 3-kinase regulatory subunit beta (PI3KR2). Of these four proteins, EEA1 and PI3KR2 were down-regulated after NAP-induced neuroprotective activity in neuroblastoma cells. Thus, aluminum ions may increase the risk for neurotoxicity in AD, and the use of NAP is suggested as a treatment to provide additional protection against the effects of aluminum ions, via EEA1 and PI3KR2, associated with sorting and processing of the AD amyloid precursor protein (APP) through the endosomal system.
Collapse
Affiliation(s)
- Ming-Hui Yang
- Institute of Biological Chemistry, Academia Sinica, Taipei, 115, Taiwan; Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Shih-Cheng Chen
- Office of Research and Development, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yu-Fen Lin
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yi-Chia Lee
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ming-Yii Huang
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; Department of Radiation Oncology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ko-Chin Chen
- Department of Pathology, Changhua Christian Hospital, Changhua 500, Taiwan
| | - Hsin-Yi Wu
- Instrumentation Center, National Taiwan University, Taipei 106, Taiwan
| | - Po-Chiao Lin
- Department of Chemistry, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Illana Gozes
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Adams Super Center for Brain Studies and Sagol School for Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel.
| | - Yu-Chang Tyan
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 804, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| |
Collapse
|
13
|
Bilgic Y, Demir EA, Bilgic N, Dogan H, Tutuk O, Tumer C. Detrimental effects of chia (Salvia hispanica L.) seeds on learning and memory in aluminum chloride-induced experimental Alzheimer’s disease. Acta Neurobiol Exp (Wars) 2018. [DOI: 10.21307/ane-2018-031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
14
|
Liang R. Cross Talk Between Aluminum and Genetic Susceptibility and Epigenetic Modification in Alzheimer’s Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1091:173-191. [DOI: 10.1007/978-981-13-1370-7_10] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
15
|
Saba K, Rajnala N, Veeraiah P, Tiwari V, Rana RK, Lakhotia SC, Patel AB. Energetics of Excitatory and Inhibitory Neurotransmission in Aluminum Chloride Model of Alzheimer's Disease: Reversal of Behavioral and Metabolic Deficits by Rasa Sindoor. Front Mol Neurosci 2017; 10:323. [PMID: 29089867 PMCID: PMC5651029 DOI: 10.3389/fnmol.2017.00323] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 09/25/2017] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) is an age-related neurodegenerative disorder, characterized by progressive loss of cognitive functions and memory. Excessive intake of aluminum chloride in drinking water is associated with amyloid plaques and neurofibrillary tangles in the brain, which are the hallmark of AD. We have evaluated brain energy metabolism in aluminum chloride (AlCl3) mouse model of AD. In addition, effectiveness of Rasa Sindoor (RS), a formulation used in Indian Ayurvedic medicine, for alleviation of symptoms of AD was evaluated. Mice were administered AlCl3 (40 mg/kg) intraperitoneally once a day for 60 days. The memory of mice was measured using Morris Water Maze test. The 13C labeling of brain amino acids was measured ex vivo in tissue extracts using 1H-[13C]-NMR spectroscopy with timed infusion of [1,6-13C2]glucose. The 13C turnover of brain amino acids was analyzed using a three-compartment metabolic model to derive the neurotransmitter cycling and TCA cycle rates associated with glutamatergic and GABAergic pathways. Exposure of AlCl3 led to reduction in memory of mice. The glutamatergic and GABAergic neurotransmitter cycling and glucose oxidation were found to be reduced in the cerebral cortex, hippocampus, and striatum following chronic AlCl3 treatment. The perturbation in metabolic rates was highest in the cerebral cortex. However, reduction in metabolic fluxes was higher in hippocampus and striatum following one month post AlCl3 treatment. Most interestingly, oral administration of RS (2 g/kg) restored memory as well as the energetics of neurotransmission in mice exposed to AlCl3. These data suggest therapeutic potential of RS to manage cognitive functions and memory in preclinical AD.
Collapse
Affiliation(s)
- Kamal Saba
- NMR Microimaging and Spectroscopy, CSIR-Centre for Cellular and Molecular Biology, Habsiguda, India
| | - Niharika Rajnala
- NMR Microimaging and Spectroscopy, CSIR-Centre for Cellular and Molecular Biology, Habsiguda, India
| | - Pandichelvam Veeraiah
- NMR Microimaging and Spectroscopy, CSIR-Centre for Cellular and Molecular Biology, Habsiguda, India
| | - Vivek Tiwari
- NMR Microimaging and Spectroscopy, CSIR-Centre for Cellular and Molecular Biology, Habsiguda, India
| | - Rohit K Rana
- CSIR-Indian Institute of Chemical Technology, Tarnaka, India
| | - Subhash C Lakhotia
- Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, India
| | - Anant B Patel
- NMR Microimaging and Spectroscopy, CSIR-Centre for Cellular and Molecular Biology, Habsiguda, India
| |
Collapse
|
16
|
La Cognata V, Maugeri G, D'Amico AG, Saccone S, Federico C, Cavallaro S, D'Agata V. Differential expression of PARK2 splice isoforms in an in vitro model of dopaminergic-like neurons exposed to toxic insults mimicking Parkinson's disease. J Cell Biochem 2017; 119:1062-1073. [PMID: 28688199 DOI: 10.1002/jcb.26274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 07/07/2017] [Indexed: 12/16/2022]
Abstract
Mutations in PARK2 (or parkin) are responsible for 50% of cases of autosomal-recessive juvenile-onset Parkinson's disease (PD). To date, 21 alternative splice variants of the human gene have been cloned. Yet most studies have focused on the full-length protein, whereas the spectrum of the parkin isoforms expressed in PD has never been investigated. In this study, the role of parkin proteins in PD neurodegeneration was explored for the first time by analyzing their expression profile in an in vitro model of PD. To do so, undifferentiated and all-trans-retinoic-acid (RA)-differentiated SH-SY5Y cells (which thereby acquire a PD-like phenotype) were exposed to PD-mimicking neurotoxins: 1-methyl-4-phenylpyridinium (MPP+ ) and 6-hydroxydopamine (6-OHDA) are widely used in PD models, whereas carbonyl cyanide m-chlorophenyl hydrazone (CCCP) and carbobenzoxy-Leu-Leu-leucinal (MG132) interfere, respectively, with mitochondrial mitophagy and proteasomal degradation. Following treatment with each neurotoxin H1, the first parkin isoform to be cloned, was down-regulated compared to the respective controls both in undifferentiated and RA-differentiated cells. In contrast, the expression pattern of the minor splice isoforms varied as a function of the compound used: it was largely unchanged in both cell cultures (eg, H21-H6, H12, XP isoform) or it showed virtually opposite alterations in undifferentiated and RA-differentiated cells (eg, H20 and H3 isoform). This complex picture suggests that up- or down-regulation may be a direct effect of toxin exposure, and that the different isoforms may exert different actions in neurodegeneration via modulation of different molecular pathways.
Collapse
Affiliation(s)
- Valentina La Cognata
- Section of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Institute of Neurological Sciences, National Research Council, Catania, Italy
| | - Grazia Maugeri
- Section of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Agata Grazia D'Amico
- Section of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Department of Human Science and Promotion of Quality of Life, San Raffaele Open University of Rome, Rome, Italy
| | - Salvatore Saccone
- Section of Animal Biology, Department of Biological, Geological, and Environmental Sciences, University of Catania, Catania, Italy
| | - Concetta Federico
- Section of Animal Biology, Department of Biological, Geological, and Environmental Sciences, University of Catania, Catania, Italy
| | | | - Velia D'Agata
- Section of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
17
|
Wang F, Guo RX, Li WX, Yu BF, Han B, Liu LX, Han DW. The role of intestinal endotoxemia in a rat model of aluminum neurotoxicity. Mol Med Rep 2017. [PMID: 28627692 PMCID: PMC5562103 DOI: 10.3892/mmr.2017.6780] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the effects of intestinal endotoxemia (IETM) in a rat model of aluminum neurotoxicity established by D-galactose and aluminum trichloride (AlCl3). Adult Wistar rats were administered D‑galactose and AlCl3 to create the aluminum neurotoxicity model. The learning and memory abilities of the rats were subsequently observed using a Morris water maze test and the serum levels of lipopolysaccharide (LPS), tumor necrosis factor (TNF)‑α, interleukin (IL)‑1, diamine oxidase (DAO), glutamine (Gln) and glutaminase were measured. The expression of S‑100β in the serum was detected using an enzyme‑linked immunosorbent assay. The expression levels of the amyloid β‑protein (Aβ) precursor (APP), presenilin 1 (PS1), β‑site APP‑cleaving enzyme (BACE), zona occludens protein (ZO)‑1 and Aβ 1‑40 in the brain of rats were detected via reverse‑transcription polymerase chain reaction, western blotting and immunohistochemistry. The levels of LPS, TNF‑α, IL‑1, DAO, Gln and S‑100β in serum and the mRNA and protein expression levels of APP, PS1, BACE and Aβ1‑40 in the brain were markedly increased in the model rats compared with controls. The level of glutaminase in the serum and the expression of ZO‑1 in the brain were decreased in the model rats compared with controls. IETM was present in the rat model of aluminum neurotoxicity established by D‑galactose and AlCl3 and may be important in the development of this neurotoxicity.
Collapse
Affiliation(s)
- Feng Wang
- Department of Pathophysiology, Key Laboratory of Cell Physiology of Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Rui-Xia Guo
- Department of Pathophysiology, Key Laboratory of Cell Physiology of Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Wen-Xing Li
- Department of Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030013, P.R. China
| | - Bao-Feng Yu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Cell Physiology of Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Bai Han
- Department of Psychiatry, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Li-Xin Liu
- The Experimental Center of Science and Research, The First Hospital, Institute of Hepatopathy, Key Laboratory of Cell Physiology of Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - De-Wu Han
- Department of Pathophysiology, Key Laboratory of Cell Physiology of Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
18
|
Maugeri G, D'Amico AG, Reitano R, Magro G, Cavallaro S, Salomone S, D'Agata V. PACAP and VIP Inhibit the Invasiveness of Glioblastoma Cells Exposed to Hypoxia through the Regulation of HIFs and EGFR Expression. Front Pharmacol 2016; 7:139. [PMID: 27303300 PMCID: PMC4885839 DOI: 10.3389/fphar.2016.00139] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/12/2016] [Indexed: 11/30/2022] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal
peptide (VIP) through the binding of vasoactive intestinal peptide receptors (VIPRs),
perform a wide variety of effects in human cancers, including glioblastoma multiforme
(GBM). This tumor is characterized by extensive areas of hypoxia, which triggers the
expression of hypoxia-inducible factors (HIFs). HIFs not only mediate angiogenesis
but also tumor cell migration and invasion. Furthermore, HIFs activation is linked to
epidermal growth factor receptor (EGFR) overexpression. Previous studies have shown
that VIP interferes with the invasive nature of gliomas by regulating cell migration.
However, the role of VIP family members in GBM infiltration under low oxygen tension
has not been clarified yet. Therefore, in the present study we have investigated, for
the first time, the molecular mechanisms involved in the anti-invasive effect of
PACAP or VIP in U87MG glioblastoma cells exposed to hypoxia induced by treatment with
desferrioxamine (DFX). The results suggest that either PACAP or VIP exert an
anti-infiltrative effect under low oxygen tension by modulating HIFs and EGFR
expression, key elements involved in cell migration and angiogenesis. These peptides
act through the inhibition of PI3K/Akt and MAPK/ERK signaling pathways, which are
known to have a crucial role in HIFs regulation.
Collapse
Affiliation(s)
- Grazia Maugeri
- Sections of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| | - Agata Grazia D'Amico
- Sections of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of CataniaCatania, Italy; San Raffaele Open University of RomeRome, Italy
| | - Rita Reitano
- Sections of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| | - Gaetano Magro
- Section of Anatomic Pathology, Department of Medical and Surgical Sciences and Advanced Technologies, G.F. Ingrassia, Azienda Ospedaliero-Universitaria "Policlinico-Vittorio Emanuele", University of Catania Catania, Italy
| | | | - Salvatore Salomone
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| | - Velia D'Agata
- Sections of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania Catania, Italy
| |
Collapse
|
19
|
Maugeri G, D'Amico AG, Rasà DM, Reitano R, Saccone S, Federico C, Parenti R, Magro G, D'Agata V. Expression profile of Wilms Tumor 1 (WT1) isoforms in undifferentiated and all-trans retinoic acid differentiated neuroblastoma cells. Genes Cancer 2016; 7:47-58. [PMID: 27014421 PMCID: PMC4773705 DOI: 10.18632/genesandcancer.94] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Wilms tumor 1 gene (WT1) is a tumor suppressor gene originally identified in nephroblastoma. It is also expressed in neuroblastoma which represents the most aggressive extracranial pediatric tumor. Many evidences have shown that neuroblastoma may undergo maturation, by transforming itself in a more differentiated tumors such as ganglioneuroblastoma and ganglioneuroma, or progressing into a highly aggressive metastatic malignancy. To date, 13 WT1 mRNA alternative splice variants have been identified. However, most of the studies have focused their attention only on isoform of ∼49 kDa. In the present study, it has been investigated the expression pattern of WT1 isoforms in an in vitro model of neuroblastoma consisting in undifferentiated or all-trans retinoic acid (RA) differentiated cells. These latter representing the less malignant phenotype of this tumor. Results have demonstrated that WT1.1-WT1.5, WT1.6-WT1.9, WT1.10 WT1.11-WT1.12 and WT1.13 isoforms are expressed in both groups of cells, but their levels are significantly increased after RA treatment. These data have also been confirmed by immunofluorescence analysis. Moreover, the inhibition of PI3K/Akt and MAPK/ERK, that represent two signalling pathway specifically involved in NB differentiation, induces an overexpression of WT1 isoforms. These data suggest that WT1 isoforms might be involved in differentiation of neuroblastic into mature ganglion cells.
Collapse
Affiliation(s)
- Grazia Maugeri
- Sections of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Agata Grazia D'Amico
- Sections of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy; San Raffaele Telematic University of Rome, Rome, Italy
| | - Daniela Maria Rasà
- Sections of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rita Reitano
- Sections of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Salvatore Saccone
- Section of Animal Biology, Department of Biological, Geological and Environmental Sciences, University of Catania, Catania, Italy
| | - Concetta Federico
- Section of Animal Biology, Department of Biological, Geological and Environmental Sciences, University of Catania, Catania, Italy
| | - Rosalba Parenti
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Gaetano Magro
- Section of Anatomic Pathology, Department of Medical and Surgical Sciences and Advanced Technologies, G.F. Ingrassia, Azienda Ospedaliero-Universitaria "Policlinico-Vittorio Emanuele", University of Catania, Catania, Italy
| | - Velia D'Agata
- Sections of Human Anatomy and Histology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
20
|
Marzagalli R, Castorina A. The seeming paradox of adenosine receptors as targets for the treatment of Alzheimer's disease: agonists or antagonists? Neural Regen Res 2015; 10:205-7. [PMID: 25883615 PMCID: PMC4392664 DOI: 10.4103/1673-5374.152370] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2014] [Indexed: 11/25/2022] Open
Affiliation(s)
- Rubina Marzagalli
- Department of Biomedical Sciences and Biotechnologies, Section of Human Anatomy and Histology, University of Catania, Italy
| | - Alessandro Castorina
- Department of Biomedical Sciences and Biotechnologies, Section of Human Anatomy and Histology, University of Catania, Italy
| |
Collapse
|
21
|
Kaur P, Sodhi R. Memory recuperative potential of rifampicin in aluminum chloride-induced dementia: Role of pregnane X receptors. Neuroscience 2015; 288:24-36. [DOI: 10.1016/j.neuroscience.2014.12.033] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 12/12/2014] [Accepted: 12/13/2014] [Indexed: 11/26/2022]
|
22
|
Wang H, Ye M, Yu L, Wang J, Guo Y, Lei W, Yang J. Hippocampal neuronal cyclooxygenase-2 downstream signaling imbalance in a rat model of chronic aluminium gluconate administration. Behav Brain Funct 2015; 11:8. [PMID: 25888969 PMCID: PMC4336726 DOI: 10.1186/s12993-015-0054-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 01/21/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Acute and chronic brain damages including neurodegenerative diseases are a group of neuroinflammation-associated diseases characterized by cognitive function defect and progressive neuron loss. The pathophysiological procession of brain damages involves the overexpression of cyclooxygenase (COX)-2. Owing to the limited benefit to chronic brain damage and the late adverse effect of COX-2 inhibitors, the COX downstream signaling pathway has become a focus in neurological research. In order to explore the mechanism of aluminum neurotoxicity and the importance of COX2 downstream signaling pathways to chronic brain damage, the present study was designed to simultaneously observe the prostaglandin (PG) contents, and the expressions of PG synthases and PG receptors of hippocampus in a rat model induced by chronic administration of aluminium gluconate. METHODS A rat model of chronic brain damage was established by chronic intragastric administration of aluminium gluconate (Al3+ 200 mg/kg per day, 5d a week for 20 weeks). PG contents, the expressions of PG synthases, and the expressions of PG receptors in rats were measured by ELISA, RT-PCR and Western blotting, respectively. RESULTS Chronic aluminium gluconate administration resulted in hippocampal neuron injury and learning and memory disorders in rats. Aluminium gluconate administration also resulted in increased levels of PGE2, PGD2, TXA2, PGI2, and PGF2α in rat hippocampus. The DP1, EP2, IP, mPGES-1, EP4, PGIS and TXAS mRNA expressions, and the DP1, EP2 and IP protein expressions significantly increased in the Al-treated hippocampus, while the EP3 and FP mRNA and protein expressions and the TP mRNA expression decreased. CONCLUSIONS The PGS/PGs/PG receptors signaling pathway in chronic aluminium gluconate-overloaded rat hippocampus is disturbed, which may be involved in the mechanism of aluminium neurotoxicity.
Collapse
Affiliation(s)
- Hong Wang
- Department of Pharmacology, Chongqing Medical University, Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, 400016, China.
| | - Mengliang Ye
- Department of Biostatistics, School of Public Health, Chongqing Medical University, Chongqing, Chongqing, 400016, China.
| | - Lijuan Yu
- Department of Pharmacology, Chongqing Medical University, Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, 400016, China.
| | - Jianfeng Wang
- Department of Pharmacology, Chongqing Medical University, Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, 400016, China.
| | - Yuanxin Guo
- Department of Pharmacology, Chongqing Medical University, Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, 400016, China.
| | - Wenjuan Lei
- Department of Pharmacology, Chongqing Medical University, Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, 400016, China.
| | - Junqing Yang
- Department of Pharmacology, Chongqing Medical University, Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, 400016, China.
| |
Collapse
|
23
|
Willhite CC, Karyakina NA, Yokel RA, Yenugadhati N, Wisniewski TM, Arnold IMF, Momoli F, Krewski D. Systematic review of potential health risks posed by pharmaceutical, occupational and consumer exposures to metallic and nanoscale aluminum, aluminum oxides, aluminum hydroxide and its soluble salts. Crit Rev Toxicol 2014; 44 Suppl 4:1-80. [PMID: 25233067 PMCID: PMC4997813 DOI: 10.3109/10408444.2014.934439] [Citation(s) in RCA: 244] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Abstract Aluminum (Al) is a ubiquitous substance encountered both naturally (as the third most abundant element) and intentionally (used in water, foods, pharmaceuticals, and vaccines); it is also present in ambient and occupational airborne particulates. Existing data underscore the importance of Al physical and chemical forms in relation to its uptake, accumulation, and systemic bioavailability. The present review represents a systematic examination of the peer-reviewed literature on the adverse health effects of Al materials published since a previous critical evaluation compiled by Krewski et al. (2007) . Challenges encountered in carrying out the present review reflected the experimental use of different physical and chemical Al forms, different routes of administration, and different target organs in relation to the magnitude, frequency, and duration of exposure. Wide variations in diet can result in Al intakes that are often higher than the World Health Organization provisional tolerable weekly intake (PTWI), which is based on studies with Al citrate. Comparing daily dietary Al exposures on the basis of "total Al"assumes that gastrointestinal bioavailability for all dietary Al forms is equivalent to that for Al citrate, an approach that requires validation. Current occupational exposure limits (OELs) for identical Al substances vary as much as 15-fold. The toxicity of different Al forms depends in large measure on their physical behavior and relative solubility in water. The toxicity of soluble Al forms depends upon the delivered dose of Al(+3) to target tissues. Trivalent Al reacts with water to produce bidentate superoxide coordination spheres [Al(O2)(H2O4)(+2) and Al(H2O)6 (+3)] that after complexation with O2(•-), generate Al superoxides [Al(O2(•))](H2O5)](+2). Semireduced AlO2(•) radicals deplete mitochondrial Fe and promote generation of H2O2, O2 (•-) and OH(•). Thus, it is the Al(+3)-induced formation of oxygen radicals that accounts for the oxidative damage that leads to intrinsic apoptosis. In contrast, the toxicity of the insoluble Al oxides depends primarily on their behavior as particulates. Aluminum has been held responsible for human morbidity and mortality, but there is no consistent and convincing evidence to associate the Al found in food and drinking water at the doses and chemical forms presently consumed by people living in North America and Western Europe with increased risk for Alzheimer's disease (AD). Neither is there clear evidence to show use of Al-containing underarm antiperspirants or cosmetics increases the risk of AD or breast cancer. Metallic Al, its oxides, and common Al salts have not been shown to be either genotoxic or carcinogenic. Aluminum exposures during neonatal and pediatric parenteral nutrition (PN) can impair bone mineralization and delay neurological development. Adverse effects to vaccines with Al adjuvants have occurred; however, recent controlled trials found that the immunologic response to certain vaccines with Al adjuvants was no greater, and in some cases less than, that after identical vaccination without Al adjuvants. The scientific literature on the adverse health effects of Al is extensive. Health risk assessments for Al must take into account individual co-factors (e.g., age, renal function, diet, gastric pH). Conclusions from the current review point to the need for refinement of the PTWI, reduction of Al contamination in PN solutions, justification for routine addition of Al to vaccines, and harmonization of OELs for Al substances.
Collapse
Affiliation(s)
- Calvin C. Willhite
- Risk Sciences International, Ottawa, ON, Canada
- McLaughlin Centre for Population Health Risk Assessment, Ottawa, ON, Canada
| | | | - Robert A. Yokel
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky, USA
| | | | - Thomas M. Wisniewski
- Departments of Neurology, Psychiatry and Pathology, New York University School of Medicine, New York City, New York, USA
| | - Ian M. F. Arnold
- Occupational Health Program, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Franco Momoli
- Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Epidemiology and Community Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - Daniel Krewski
- Risk Sciences International, Ottawa, ON, Canada
- McLaughlin Centre for Population Health Risk Assessment, Ottawa, ON, Canada
- Department of Epidemiology and Community Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
24
|
Giunta S, Andriolo V, Castorina A. Dual blockade of the A1 and A2A adenosine receptor prevents amyloid beta toxicity in neuroblastoma cells exposed to aluminum chloride. Int J Biochem Cell Biol 2014; 54:122-36. [PMID: 25058312 DOI: 10.1016/j.biocel.2014.07.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 06/25/2014] [Accepted: 07/11/2014] [Indexed: 02/06/2023]
Abstract
In a previous work we have shown that exposure to aluminum (Al) chloride (AlCl3) enhanced the neurotoxicity of the amyloid beta(25-35) fragment (Abeta(25-35)) in neuroblastoma cells and affected the expression of Alzheimer's disease (AD)-related genes. Caffein, a compound endowed with beneficial effects against AD, exerts neuroprotection primarily through its antagonist activity on A2A adenosine receptors (A2AR), although it also inhibits A1Rs with similar potency. Still, studies on the specific involvement of these receptors in neuroprotection in a model of combined neurotoxicity (Abeta(25-35)+AlCl3) are missing. To address this issue, cultured SH-SY5Y cells exposed to Abeta(25-35)+AlCl3 were assessed for cell viability, morphology, intracellular ROS activity and expression of apoptosis-, stress- and AD-related proteins. To define the role of A1R and A2ARs, pretreatment with caffein, specific receptor antagonists (DPCPX or SCH58261) or siRNA-mediated gene knockdown were delivered. Results indicate that AlCl3 treatment exacerbated Abeta(25-35) toxicity, increased ROS production, lipid peroxidation, β-secretase-1 (BACE1) and amyloid precursor protein (APP). Interestingly, SCH58261 successfully prevented toxicity associated to Abeta(25-35) only, whereas pretreatment with both DPCPX and SCH58261 was required to fully avert Abeta(25-35)+AlCl3-induced damage, suggesting that A1Rs might also be critically involved in protection during combined toxicity. The effects of caffein were mimicked by both N-acetyl cysteine, an antioxidant, and desferrioxamine, likely acting through distinct mechanisms. Altogether, our data establish a novel protective function associated with A1R inhibition in the setting of combined Abeta(25-35)+AlCl3 neurotoxicity, and expand our current knowledge on the potential beneficial role of caffein to prevent AD progression in subjects environmentally exposed to aluminum.
Collapse
Affiliation(s)
- Salvatore Giunta
- Department of Bio-Medical Sciences, Section of Anatomy and Histology, University of Catania, Italy
| | - Violetta Andriolo
- Department of Pediatrics and Public Health Sciences, University of Turin, Italy
| | - Alessandro Castorina
- Department of Bio-Medical Sciences, Section of Anatomy and Histology, University of Catania, Italy.
| |
Collapse
|
25
|
Wang L, Hu J, Zhao Y, Lu X, Zhang Q, Niu Q. Effects of aluminium on β-amyloid (1-42) and secretases (APP-cleaving enzymes) in rat brain. Neurochem Res 2014; 39:1338-45. [PMID: 24792732 DOI: 10.1007/s11064-014-1317-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Revised: 04/16/2014] [Accepted: 04/21/2014] [Indexed: 01/22/2023]
Abstract
Chronic administration of aluminium has been proposed as an environmental factor that may affect some pathological changes related to neurotoxicity and Alzheimer's disease (AD). The abnormal generation and deposition of β-amyloid (Aβ) in senile plaques are hallmark features in the brains of AD patients. Furthermore, Aβ is generated by the sequential cleavage of the amyloid precursor protein (APP) via the APP cleaving enzyme (α-secretase, or β-secretase) and γ-secretase. In the present study, we investigated the modulation of Aβ deposition and neurotoxicity in aluminium-maltolate-treated (0, 15, 30, 45 mmol/kg body weight via intraperitoneal injection) in experimental rats. We measured Aβ1-40 and Aβ1-42 in the cortex and hippocampus in rat brains using ELISA. Subtypes of α-secretase, β-secretase, and γ-secretase, including ADAM9, ADAM10, ADAM17 (TACE), BACE1, presenilin 1 (PS1) and nicastrin (NCT), were determined using western blotting analyses. These results indicated that aluminium-maltolate induced an AD-like behavioural deficit in rats at 30 and 45 mmol/kg body weight. Moreover, the Aβ1-42 content increased significantly, both in the cortex and hippocampus, although no changes were observed in Aβ1-40. Furthermore, ADAM9, ADAM10, and ADAM17 decreased significantly; in contrast, BACE1, PS1, and NCT showed significant increase. Taken together, these results suggest that the changes in secretases may correlate to the abnormal deposition of Aβ by aluminium in rat brains.
Collapse
Affiliation(s)
- Linping Wang
- School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | | | | | | | | | | |
Collapse
|
26
|
Nam SM, Kim JW, Yoo DY, Kim W, Jung HY, Hwang IK, Seong JK, Yoon YS. Additive or synergistic effects of aluminum on the reduction of neural stem cells, cell proliferation, and neuroblast differentiation in the dentate gyrus of high-fat diet-fed mice. Biol Trace Elem Res 2014; 157:51-9. [PMID: 24265032 DOI: 10.1007/s12011-013-9861-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 11/04/2013] [Indexed: 01/20/2023]
Abstract
Aluminum is the most plentiful metal on the Earth's crust, and its usage in cooking utensils, cosmetics, drinking containers, food additives, pharmaceutical products, and building materials provides many opportunities for potential aluminum consumption. However, its toxicity is low and harmful effects only develop with large-scale deposition of aluminum. In this study, we investigated the effects of subchronic exposure to aluminum (40 mg/kg/day) on neural stem cells, cell proliferation, neuroblast differentiation, and mature neurons in the dentate gyrus of the hippocampus. These experiments were performed in both high-fat diet and low-fat diet-fed C57BL/6J mice via immunohistochemistry using the relevant marker for each cell type, including nestin, Ki67, doublecortin, and NeuN. Subchronic exposure to aluminum in both low-fat and high-fat diet-fed mice reduced neural stem cells, cell proliferation, and neuroblast differentiation without any changes in mature neurons. Furthermore, this reduction effect was exacerbated in high-fat diet-fed mice. These results suggest that aluminum accelerates the reduction of neural stem cells, cell proliferation, and neuroblast differentiation additively or synergistically in high-fat diet-fed mice without any harmful changes in mature neurons.
Collapse
Affiliation(s)
- Sung Min Nam
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 151-742, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Chen SM, Fan CC, Chiue MS, Chou C, Chen JH, Hseu RS. Hemodynamic and neuropathological analysis in rats with aluminum trichloride-induced Alzheimer's disease. PLoS One 2013; 8:e82561. [PMID: 24376545 PMCID: PMC3869709 DOI: 10.1371/journal.pone.0082561] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 10/25/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND AND AIMS Hemodynamic normality is crucial to maintaining the integrity of cerebral vessels and, therefore, preserving the cognitive functions of Alzheimer's disease patients. This study investigates the implications of the hemodynamic changes and the neuropathological diversifications of AlCl3-induced AD. METHODS The experimental animals were 8- to 12-wk-old male Wistar rats. The rats were randomly divided into 2 groups: a control group and a (+)control group. Food intake, water intake, and weight changes were recorded daily for 22 wk. Synchronously, the regional cerebral blood flow (rCBF) of the rats with AlCl3-induced AD were measured using magnetic resonance imaging (MRI). The hemorheological parameters were analyzed using a computerized auto-rotational rheometer. The brain tissue of the subjects was analyzed using immunohistological chemical (IHC) staining to determine the beta-amyloid (Aβ) levels. RESULTS The results of hemodynamic analysis revealed that the whole blood viscosity (WBV), fibrinogen, plasma viscosity and RBC aggregation index (RAI) in (+)control were significantly higher than that of control group, while erythrocyte electrophoresis (EI) of whole blood in (+)control were significantly lower than that of control group. The results of acetylcholinesterase-RBC (AChE-RBC)in the (+)control group was significantly higher than that of the control group. The results also show that the reduction of rCBF in rats with AlCl3-induced AD was approximately 50% to 60% that of normal rats. IHC stain results show that significantly more Aβ plaques accumulated in the hippocampus and cortex of the (+)control than in the control group. CONCLUSION The results accentuate the importance of hemorheology and reinforce the specific association between hemodynamic and neuropathological changes in rats with AlCl3-induced AD. Hemorheological parameters, such as WBV and fibrinogen, and AChE-RBC were ultimately proven to be useful biomarkers of the severity and progression of AD patients. In addition, the parameters can be substituted for invasive inspection in therapeutic intervention.
Collapse
Affiliation(s)
- Szu-Ming Chen
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University, Hsinchu, Taiwan
| | - Chi-Chen Fan
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University, Hsinchu, Taiwan
- Department of Physiology, Mackay Memorial Hospital, Taipei, Taiwan
| | - Ming-Shiuan Chiue
- Interdisciplinary MRI/MRS Lab, Department of Electrical Engineering and Molecular Imaging Center, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Chi Chou
- Department of Physiology, Mackay Memorial Hospital, Taipei, Taiwan
| | - Jyh-Horng Chen
- Interdisciplinary MRI/MRS Lab, Department of Electrical Engineering and Molecular Imaging Center, National Taiwan University, Taipei, Taiwan
| | - Ruey-Shyang Hseu
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
28
|
Ismail MF, Elmeshad AN, Salem NAH. Potential therapeutic effect of nanobased formulation of rivastigmine on rat model of Alzheimer's disease. Int J Nanomedicine 2013; 8:393-406. [PMID: 23378761 PMCID: PMC3558309 DOI: 10.2147/ijn.s39232] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Background To sustain the effect of rivastigmine, a hydrophilic cholinesterase inhibitor, nanobased formulations were prepared. The efficacy of the prepared rivastigmine liposomes (RLs) in comparison to rivastigmine solution (RS) was assessed in an aluminium chloride (AlCl3)-induced Alzheimer’s model. Methods Liposomes were prepared by lipid hydration (F1) and heating (F2) methods. Rats were treated with either RS or RLs (1 mg/kg/day) concomitantly with AlCl3 (50 mg/kg/day). Results The study showed that the F1 method produced smaller liposomes (67.51 ± 14.2 nm) than F2 (528.7 ± 15.5 nm), but both entrapped the same amount of the drug (92.1% ± 1.4%). After 6 hours, 74.2% ± 1.5% and 60.8% ± 2.3% of rivastigmine were released from F1 and F2, respectively. Both RLs and RS improved the deterioration of spatial memory induced by AlCl3, with RLs having a superior effect. Further biochemical measurements proved that RS and RLs were able to lower plasma C-reactive protein, homocysteine and asymmetric dimethy-larginine levels. RS significantly attenuated acetylcholinesterase (AChE) activity, whereas Na+/K+-adenosine triphosphatase (ATPase) activity was enhanced compared to the AlCl3-treated animals; however, RLs succeeded in normalization of AChE and Na+/K+ ATPase activities. Gene-expression profile showed that cotreatment with RS to AlCl3-treated rats succeeded in exerting significant decreases in BACE1, AChE, and IL1B gene expression. Normalization of the expression of the aforementioned genes was achieved by coadministration of RLs to AlCl3-treated rats. The profound therapeutic effect of RLs over RS was evidenced by nearly preventing amyloid plaque formation, as shown in the histopathological examination of rat brain. Conclusion RLs could be a potential drug-delivery system for ameliorating Alzheimer’s disease.
Collapse
|
29
|
Li XB, Zhang ZY, Yin LH, Schluesener HJ. The profile of β-amyloid precursor protein expression of rats induced by aluminum. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2012; 33:135-140. [PMID: 22209725 DOI: 10.1016/j.etap.2011.12.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 11/11/2011] [Accepted: 12/04/2011] [Indexed: 05/31/2023]
Abstract
The environmental agent aluminum has been extensively investigated for a potential relationship with amyloid precursor protein (APP) expression. Despite many investigations, there is at present no definite proof from which to draw a conclusion. Since APP is an integral membrane protein expressed in different tissues and capable of fluxes across the blood-brain barrier (BBB), which may ultimately affect APP level in brain, it is necessary to assess the expression profile among vital body organs. The present study compared aluminum oxide and aluminum chloride injected rats with control rats (saline treated) to observe if aluminum affected APP expression patterns in different organs by immunohistochemistry (IHC). The expression of APP was observed in the brain of aluminum chloride treated rats and in the liver of aluminum oxide injected group. Results of double IHC staining showed that it is Kupffer cells, which are located in liver sinus and expressed APP after aluminum oxide treatment. Oxidative stress is suggested as the potential pathway that aluminum chloride exert effects in brain. These results suggest that different aluminum compounds may impact the expression of APP in brain and liver tissues. The mechanism that aluminum induced liver APP expression still needs further investigation.
Collapse
Affiliation(s)
- Xiao-Bo Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| | | | | | | |
Collapse
|
30
|
Zhang Q, Jia L, Jiao X, Guo W, Ji J, Yang H, Niu Q. APP/PS1 Transgenic Mice Treated with Aluminum: An Update of Alzheimer's Disease Model. Int J Immunopathol Pharmacol 2012; 25:49-58. [DOI: 10.1177/039463201202500107] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
There is still no animal model available that can mimic all the cognitive, behavioral, biochemical, and histopathological abnormalities observed in patients with Alzheimer's disease (AD). We undertook to consider the interaction between genetic factors, including amyloid precursor protein (APP) and presenilin-1 (PS1), and environmental factors, such as Aluminum (Al) in determining susceptibility outcomes when studying the pathogenesis of AD. In this article, we provide an AD model in APP/PS1 transgenic mice triggered by Al. The animal model was established via intracerebral ventricular microinjection of aluminum chloride once a day for 5 days in APP/PS1 transgenic mice. Twenty wild type (WT) mice and 20 APP/PS1 transgenic (TG) mice were separately divided into 2 groups (control and Al group), and a stainless steel injector with stopper was used for microinjection into the left-lateral cerebral ventricle of each mouse. The Morris water maze task was used to evaluate behavioral function of learning and memory ability on the 20th day after the last injection. This AD model's brain was analyzed by: (1) amyloid β immunohistochemical staining; (2) Tunnel staining; (3) apoptotic rates; (4) caspase-3 gene expression. Here, decrease of cognitive ability and neural cells loss were shown in APP/PS1 transgenic mice exposed to Al, which were more extensive than those in APP/PS1 TG alone and WT mice exposed to Al alone. These findings indicate that there is a close relationship between over-expression of APP and PS1 genes and Al overload. It is also suggested that APP/PS1 TG mice exposed to Al have potential value for improving AD models.
Collapse
Affiliation(s)
- Q.L. Zhang
- Ministry of Education Key Laboratory, Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - L. Jia
- Ministry of Education Key Laboratory, Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - X. Jiao
- Ministry of Education Key Laboratory, Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - W.L. Guo
- Ministry of Education Key Laboratory, Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - J.W. Ji
- Ministry of Education Key Laboratory, Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - H.L. Yang
- The First Affiliated Hospital of Shanxi Medical University, Taiyuan, China
| | - Q. Niu
- Ministry of Education Key Laboratory, Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
31
|
Kawahara M, Kato-Negishi M. Link between Aluminum and the Pathogenesis of Alzheimer's Disease: The Integration of the Aluminum and Amyloid Cascade Hypotheses. Int J Alzheimers Dis 2011; 2011:276393. [PMID: 21423554 PMCID: PMC3056430 DOI: 10.4061/2011/276393] [Citation(s) in RCA: 153] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Revised: 11/29/2010] [Accepted: 01/05/2011] [Indexed: 12/21/2022] Open
Abstract
Whilst being environmentally abundant, aluminum is not essential for life. On the contrary, aluminum is a widely recognized neurotoxin that inhibits more than 200 biologically important functions and causes various adverse effects in plants, animals, and humans. The relationship between aluminum exposure and neurodegenerative diseases, including dialysis encephalopathy, amyotrophic lateral sclerosis and Parkinsonism dementia in the Kii Peninsula and Guam, and Alzheimer's disease (AD) has been suggested. In particular, the link between aluminum and Alzheimer's disease has been the subject of scientific debate for several decades. However, the complex characteristics of aluminum bioavailability make it difficult to evaluate its toxicity and therefore, the relationship remains to be established. Mounting evidence has suggested that significance of oligomerization of β-amyloid protein and neurotoxicity in the molecular mechanism of AD pathogenesis. Aluminum may play crucial roles as a cross-linker in β-amyloid oligomerization. Here, we review the detailed characteristics of aluminum neurotoxicity based on our own studies and the recent literatures. Our aim is to revisit the link between aluminum and AD and to integrate aluminum and amyloid cascade hypotheses in the context of β-amyloid oligomerization and the interactions with other metals.
Collapse
Affiliation(s)
- Masahiro Kawahara
- Department of Analytical Chemistry, School of Pharmaceutical Sciences, Kyushu University of Health and Welfare, 1714-1 Yoshino-cho, Nobeoka-shi, Miyazaki 882-8508, Japan
| | | |
Collapse
|
32
|
Thinnes FP. Amyloid Aß , cut from APP by ß-secretase BACE1 and γ-secretase, induces apoptosis via opening type-1 porin/VDAC in cell membranes of hypometabolic cells-A basic model for the induction of apoptosis!? Mol Genet Metab 2010; 101:301-3. [PMID: 20675165 DOI: 10.1016/j.ymgme.2010.07.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2010] [Revised: 07/08/2010] [Accepted: 07/08/2010] [Indexed: 12/27/2022]
|