1
|
Ranjbar S, Mohammadi P, Pashaei S, Sadeghi M, Mehrabi M, Shabani S, Ebrahimi A, Brühl AB, Khodarahmi R, Brand S. Effect of Aflatoxin B1 on the Nervous System: A Systematic Review and Network Analysis Highlighting Alzheimer's Disease. BIOLOGY 2025; 14:436. [PMID: 40282301 PMCID: PMC12024953 DOI: 10.3390/biology14040436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/13/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025]
Abstract
Exposure to aflatoxin (AF) triggers the production of inflammatory molecules and free radicals, leading to chronic inflammation, cancer, and neurodegenerative diseases. This systematic review evaluated the effects of AFB1 on the nervous system, particularly focusing on Alzheimer's disease (AD). A comprehensive search was conducted in Scopus, Cochrane Library, PubMed, and Web of Science databases up to 1 June 2024, without restrictions. From 993 records retrieved, 16 articles were included in the systematic review. AFB1 participates in various biochemical processes and pathological conditions. The study highlights that AFB1 contributes to AD by inducing DNA damage, oxidative stress, and endoplasmic reticulum (ER) stress, impairing DNA repair mechanisms. This results in neuronal damage, cognitive decline, and neurodegeneration. AFB1 also affects key signaling pathways, reduces sodium-potassium pump activity, and disrupts cell cycle regulation involving p53, leading to neurotoxicity, inflammation, and the formation of amyloid-beta (Aβ) plaques and neurofibrillary tangles. Additionally, network analysis revealed 309 genes associated with AD, inflammation, angiopathy, and aflatoxin B1 (AFB1). Among these, ESR1 exhibited the highest number of direct connections to other nodes within the network. The gene TP53 played a pivotal role in mediating communication among genes, while the EP300 gene significantly influenced the overall network structure. Additionally, KEGG enrichment analysis demonstrated that these 309 genes are substantially involved in pathways related to cancer, the FoxO signaling pathway, apoptosis, and AD. In summary, the study highlights that AFB1 causes DNA damage and stress, leading to cognitive decline and neurodegeneration. It disrupts signaling pathways, damages neurons, and affects DNA repair, contributing to neurotoxicity and inflammation. PROSPERO registration number: CRD420250651007.
Collapse
Affiliation(s)
- Samira Ranjbar
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran; (S.R.); (P.M.); (S.P.); (M.S.); (M.M.); (S.S.)
| | - Pantea Mohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran; (S.R.); (P.M.); (S.P.); (M.S.); (M.M.); (S.S.)
| | - Somayeh Pashaei
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran; (S.R.); (P.M.); (S.P.); (M.S.); (M.M.); (S.S.)
- Department of Clinical Biochemistry, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran
| | - Masoud Sadeghi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran; (S.R.); (P.M.); (S.P.); (M.S.); (M.M.); (S.S.)
| | - Masomeh Mehrabi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran; (S.R.); (P.M.); (S.P.); (M.S.); (M.M.); (S.S.)
| | - Sasan Shabani
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran; (S.R.); (P.M.); (S.P.); (M.S.); (M.M.); (S.S.)
| | - Ali Ebrahimi
- Dermatology Department, Hajdaie Dermatology Clinic, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran
| | - Annette B. Brühl
- Center for Affective, Stress and Sleep Disorders, Psychiatric Clinics, University of Basel, 4002 Basel, Switzerland;
| | - Reza Khodarahmi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran; (S.R.); (P.M.); (S.P.); (M.S.); (M.M.); (S.S.)
- Department of Pharmacognosy and Biotechnology, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 6714415185, Iran
| | - Serge Brand
- Center for Affective, Stress and Sleep Disorders, Psychiatric Clinics, University of Basel, 4002 Basel, Switzerland;
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
- Substance Abuse Prevention Research Center, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
- Division of Sport Science and Psychosocial Health, Department of Sport, Exercise and Health, University of Basel, 4002 Basel, Switzerland
- School of Medicine, Tehran University of Medical Sciences, Tehran 1416753955, Iran
- Center for Disaster Psychiatry and Disaster Psychology, Center of Competence for Disaster Medicine, Swiss Armed Forces, 4002 Basel, Switzerland
| |
Collapse
|
2
|
Uvarajan D, Gnanarajan R, Karuppusamy PA, Ravichandran N, Govindasamy C, Vellingiri B, Narayanaswamy A, Wang W. Neuroprotective Effects of Berberine Chloride Against the Aluminium Chloride-Induced Alzheimer's Disease in Zebra Fish Larvae. Mol Biotechnol 2025:10.1007/s12033-025-01392-x. [PMID: 40014257 DOI: 10.1007/s12033-025-01392-x] [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: 01/06/2024] [Accepted: 01/30/2025] [Indexed: 02/28/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease distinguished by cognitive and memory deficits. A lack of memory, cognition, and other forms of cognitive dissonance characterizes AD, which affects approximately 50 million people worldwide. This study aimed to identify the neuroprotective effects of berberine chloride (BC) against aluminium chloride (AlCl3)-induced AD in zebrafish larvae by inhibiting oxidative stress and neuroinflammation. BC toxicity was assessed by evaluating survival rates, malformations, and heart rates in zebrafish larvae following treatment with varying concentrations of BC. This study elucidates the mechanisms of BC through an extensive range of biochemical assays, behavioral testing, and molecular docking analysis. The developmental toxicity assessment of BC indicated that doses up to 40 μM did not cause any developmental abnormalities until 96 h post fertilization. The LC50 value of BC in zebrafish larvae was found to be 50.16 μM. The biochemical and behavioral changes induced by AlCl3 in zebrafish larvae were significantly mitigated by BC treatment. Our findings demonstrate that BC can reduce total cholesterol and triglyceride levels in AlCl3-induced AD zebrafish larvae. Our molecular docking results indicated that BC significantly interacted with the ABCA1 protein, suggesting that BC may act as an ABCA1 agonist. Based on our results, it can be concluded that BC may serve as an effective therapeutic agent for mitigating oxidative stress by altering cholesterol metabolism in AlCl3-induced AD.
Collapse
Affiliation(s)
| | - Roselin Gnanarajan
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, India
| | | | - Nandita Ravichandran
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, India
| | - Chandramohan Govindasamy
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, 11433, Riyadh, Saudi Arabia
| | - Balachandhar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, Central University of Punjab, Bathinda, India
| | - Arul Narayanaswamy
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, India
| | - Wei Wang
- The Medical Department of Neurology, Xi'an North Hospital, Xi'an, 710043, China.
| |
Collapse
|
3
|
Chen J, Wang H, Chen Y, Zhu Q, Wan J. Inhibitive effect and mechanism of cinnamaldehyde on growth and OTA production of Aspergillus niger in vitro and in dried red chilies. Food Res Int 2023; 168:112794. [PMID: 37120239 DOI: 10.1016/j.foodres.2023.112794] [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: 12/08/2022] [Revised: 03/23/2023] [Accepted: 04/03/2023] [Indexed: 05/01/2023]
Abstract
Mould and mycotoxin contamination is an ongoing issue in agriculture and food industry. Production by Aspergillus niger DTZ-12 in Guizhou dried red chilies was found, leading to significant economic losses. In this study, the inhibitive efficacy (Effective Concentration, EC) of cinnamaldehyde (CIN), eugenol (EUG), carvacrol (CAR), and linalool (LIN) against A. niger DTZ-12 were evaluated. CIN with the best antifungal capacity was then investigated for the comprehensive inhibitory activity against A. niger DTZ-12 including mycelia, spores, and physiological activities. Results showed that CIN can effectively retard mycelial growth, spore germination, and OTA production of A. niger DTZ-12 in vitro and in dried red chilies during storage. At physiological level, CIN can increase cell membrane permeability by reducing the ergosterol, decrease ATP content and ATPase activity, and promote the accumulation of reactive oxygen species (ROS) and malondialdehyde (MDA) in cell. These results suggested that CIN displayed a great potential to be employed as a natural and effective alternative preservative during dried red chili storage.
Collapse
Affiliation(s)
- Jiang Chen
- College of Life Sciences, Guizhou University, Huaxi District, Guiyang 550025, Guizhou Province, China
| | - Hua Wang
- Department of Liquor and Food Engineering, Guizhou University, Huaxi District, Guiyang 550025, Guizhou Province, China
| | - Yuanshan Chen
- Department of Liquor and Food Engineering, Guizhou University, Huaxi District, Guiyang 550025, Guizhou Province, China
| | - Qiujin Zhu
- Department of Liquor and Food Engineering, Guizhou University, Huaxi District, Guiyang 550025, Guizhou Province, China
| | - Jing Wan
- College of Life Sciences, Guizhou University, Huaxi District, Guiyang 550025, Guizhou Province, China; Department of Liquor and Food Engineering, Guizhou University, Huaxi District, Guiyang 550025, Guizhou Province, China; Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences/Institute of Agro-bioengineering, Guizhou University, Huaxi District, Guiyang 550025, Guizhou Province, China.
| |
Collapse
|
4
|
Liu X, Fang Y, Xu J, Yang T, Xu J, He J, Liu W, Yu X, Wen Y, Zhang N, Li C. Oxidative stress, dysfunctional energy metabolism, and destabilizing neurotransmitters altered the cerebral metabolic profile in a rat model of simulated heliox saturation diving to 4.0 MPa. PLoS One 2023; 18:e0282700. [PMID: 36917582 PMCID: PMC10013885 DOI: 10.1371/journal.pone.0282700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 02/21/2023] [Indexed: 03/15/2023] Open
Abstract
The main objective of the present study was to determine metabolic profile changes in the brains of rats after simulated heliox saturated diving (HSD) to 400 meters of sea water compared to the blank controls. Alterations in the polar metabolome in the rat brain due to HSD were investigated in cortex, hippocampus, and striatum tissue samples by applying an NMR-based metabolomic approach coupled with biochemical detection in the cortex. The reduction in glutathione and taurine levels may hypothetically boost antioxidant defenses during saturation diving, which was also proven by the increased malondialdehyde level, the decreased superoxide dismutase, and the decreased glutathione peroxidase in the cortex. The concomitant decrease in aerobic metabolic pathways and anaerobic metabolic pathways comprised downregulated energy metabolism, which was also proven by the biochemical quantification of the metabolic enzymes Na-K ATPase and LDH in cerebral cortex tissue. The significant metabolic abnormalities of amino acid neurotransmitters, such as GABA, glycine, and aspartate, decreased aromatic amino acids, including tyrosine and phenylalanine, both of which are involved in the metabolism of dopamine and noradrenaline, which are downregulated in the cortex. Particularly, a decline in the level of N-acetyl aspartate is associated with neuronal damage. In summary, hyperbaric decompression of a 400 msw HSD affected the brain metabolome in a rat model, potentially including a broad range of disturbing amino acid homeostasis, metabolites related to oxidative stress and energy metabolism, and destabilizing neurotransmitter components. These disturbances may contribute to the neurochemical and neurological phenotypes of HSD.
Collapse
Affiliation(s)
- Xia Liu
- Department of Diving and Hyperbaric Medicine, Navy Medical Center of PLA, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yiqun Fang
- Department of Diving and Hyperbaric Medicine, Navy Medical Center of PLA, Naval Medical University (Second Military Medical University), Shanghai, China
- * E-mail: (YF); (CL)
| | - Jiajun Xu
- Department of Diving and Hyperbaric Medicine, Navy Medical Center of PLA, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Tao Yang
- Department of Diving and Hyperbaric Medicine, Navy Medical Center of PLA, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Ji Xu
- Department of Diving and Hyperbaric Medicine, Navy Medical Center of PLA, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Jia He
- Department of Diving and Hyperbaric Medicine, Navy Medical Center of PLA, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wenwu Liu
- Department of Diving and Hyperbaric Medicine, Navy Medical Center of PLA, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Xuhua Yu
- Department of Diving and Hyperbaric Medicine, Navy Medical Center of PLA, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yukun Wen
- Department of Diving and Hyperbaric Medicine, Navy Medical Center of PLA, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Naixia Zhang
- Department of Analytical Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Ci Li
- Department of Diving and Hyperbaric Medicine, Navy Medical Center of PLA, Naval Medical University (Second Military Medical University), Shanghai, China
- * E-mail: (YF); (CL)
| |
Collapse
|
5
|
Zhang X, Lee W, Bian JS. Recent Advances in the Study of Na +/K +-ATPase in Neurodegenerative Diseases. Cells 2022; 11:cells11244075. [PMID: 36552839 PMCID: PMC9777075 DOI: 10.3390/cells11244075] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Na+/K+-ATPase (NKA), a large transmembrane protein, is expressed in the plasma membrane of most eukaryotic cells. It maintains resting membrane potential, cell volume and secondary transcellular transport of other ions and neurotransmitters. NKA consumes about half of the ATP molecules in the brain, which makes NKA highly sensitive to energy deficiency. Neurodegenerative diseases (NDDs) are a group of diseases characterized by chronic, progressive and irreversible neuronal loss in specific brain areas. The pathogenesis of NDDs is sophisticated, involving protein misfolding and aggregation, mitochondrial dysfunction and oxidative stress. The protective effect of NKA against NDDs has been emerging gradually in the past few decades. Hence, understanding the role of NKA in NDDs is critical for elucidating the underlying pathophysiology of NDDs and identifying new therapeutic targets. The present review focuses on the recent progress involving different aspects of NKA in cellular homeostasis to present in-depth understanding of this unique protein. Moreover, the essential roles of NKA in NDDs are discussed to provide a platform and bright future for the improvement of clinical research in NDDs.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Weithye Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Jin-Song Bian
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
- Correspondence:
| |
Collapse
|
6
|
Chellammal HSJ, Hasan MH, Kshirsagar RP, Musukula VKR, Ramachandran D, Diwan PV. Metformin inhibits cardiometabolic syndrome associated cognitive deficits in high fat diet rats. J Diabetes Metab Disord 2022; 21:1415-1426. [PMID: 36404813 PMCID: PMC9672285 DOI: 10.1007/s40200-022-01074-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 10/17/2022]
Abstract
Objectives Glucose intolerance and insulin resistance are hallmarks of metabolic syndrome and lead to Alzheimer's disease (AD). The purpose of this study is to elucidate the neuroprotective effect of metformin through insulin regulation with cardiometabolic and neurotransmitter metabolic enzyme regulation in high-fat, high-sucrose diet and streptozotocin (HFHS-STZ)-induced rats. Methods Male Wistar rats were treated with metformin (180 mg/kg and 360 mg/kg). STZ (35 mg/kg i.p.) injection was performed on the 14th day of 42 days of HFHS diet treatment. Brain neurotransmitter metabolic enzymes (acetylcholinesterase and monoamine oxidase) were determined along with sodium-potassium ATPase (Na+K+-ATPase). Plasma lipids and homeostasis model assessment of insulin resistance (HOMA-IR) was performed. Mean arterial blood pressure, heart rate and electrocardiogram (QT, QTc and RR intervals) were analysed with PowerLab. Results Metformin treatment significantly (p < 0.001) reduced the HOMA-IR index and decreased neurotransmitter metabolic enzymes such as AChE and MAO (p < 0.01 and p < 0.05). The lipid profile was significantly (p < 0.001) controlled with cardiometabolic functions. Conclusions Our investigation revealed that metformin has a remarkable role in regulating brain insulin, vascular system with monoaminergic metabolic enzymes and enhancing synaptic plasticity. Metformin may be a selective early therapeutic agent in metabolic syndrome associated with cognitive decline.
Collapse
Affiliation(s)
- Hanish Singh Jayasingh Chellammal
- Department of Pharmacology and Pharmaceutical Chemistry, Faculty of Pharmacy, Universiti Teknologi MARA, Puncak Alam, Selangor 42300 Malaysia
| | - Mizaton Hazizul Hasan
- Department of Pharmacology and Pharmaceutical Chemistry, Faculty of Pharmacy, Universiti Teknologi MARA, Puncak Alam, Selangor 42300 Malaysia
| | - Rahul P Kshirsagar
- Department of Pharmacology, Yashodeep Institute of Pharmacy, Sangram Nagar, Aurangabad, 431001 India
- Department of Pharmacology, Anurag Group of Institutions, Venkatapur, Ghatkesar, Hyderabad, Telangana 500088 India
| | | | - Dhani Ramachandran
- Unit of Pathology, International Medical School, Management & Science University, University Drive, Off Persiaran Olahraga, Section 13, Shah Alam, Selangor Darul Ehsan 40100 Malaysia
| | - Prakash V Diwan
- Department of Pharmacology, Anurag Group of Institutions, Venkatapur, Ghatkesar, Hyderabad, Telangana 500088 India
- Central Research Laboratory, Maratha Mandal Group of Institutions, Belgaum, Karnataka 590019 India
| |
Collapse
|
7
|
Gao Y, Silva LND, Hurley JD, Fan X, Pierre SV, Sodhi K, Liu J, Shapiro JI, Tian J. Gene module regulation in dilated cardiomyopathy and the role of Na/K-ATPase. PLoS One 2022; 17:e0272117. [PMID: 35901050 PMCID: PMC9333241 DOI: 10.1371/journal.pone.0272117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/12/2022] [Indexed: 01/25/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a major cause of cardiac death and heart transplantation. It has been known that black people have a higher incidence of heart failure and related diseases compared to white people. To identify the relationship between gene expression and cardiac function in DCM patients, we performed pathway analysis and weighted gene co-expression network analysis (WGCNA) using RNA-sequencing data (GSE141910) from the NCBI Gene Expression Omnibus (GEO) database and identified several gene modules that were significantly associated with the left ventricle ejection fraction (LVEF) and DCM phenotype. Genes included in these modules are enriched in three major categories of signaling pathways: fibrosis-related, small molecule transporting-related, and immune response-related. Through consensus analysis, we found that gene modules associated with LVEF in African Americans are almost identical as in Caucasians, suggesting that the two groups may have more common rather than disparate genetic regulations in the etiology of DCM. In addition to the identified modules, we found that the gene expression level of Na/K-ATPase, an important membrane ion transporter, has a strong correlation with the LVEF. These clinical results are consistent with our previous findings and suggest the clinical significance of Na/K-ATPase regulation in DCM.
Collapse
Affiliation(s)
- Yingnyu Gao
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV, United States of America
| | - Lilian N. D. Silva
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV, United States of America
| | - John D. Hurley
- Department of Biomedical Sciences, Marshall University Joan C. Edwards Medical School, Huntington, WV, United States of America
| | - Xiaoming Fan
- Department of Medicine, University of Toledo, Toledo, OH, United States of America
| | - Sandrine V. Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV, United States of America
| | - Komal Sodhi
- Department of Biomedical Sciences, Marshall University Joan C. Edwards Medical School, Huntington, WV, United States of America
| | - Jiang Liu
- Department of Biomedical Sciences, Marshall University Joan C. Edwards Medical School, Huntington, WV, United States of America
| | - Joseph I. Shapiro
- Department of Biomedical Sciences, Marshall University Joan C. Edwards Medical School, Huntington, WV, United States of America
| | - Jiang Tian
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, WV, United States of America
- Department of Biomedical Sciences, Marshall University Joan C. Edwards Medical School, Huntington, WV, United States of America
| |
Collapse
|
8
|
Kalocayova B, Snurikova D, Vlkovicova J, Navarova-Stara V, Michalikova D, Ujhazy E, Gasparova Z, Vrbjar N. Effect of handling on ATP utilization of cerebral Na,K-ATPase in rats with trimethyltin-induced neurodegeneration. Mol Cell Biochem 2021; 476:4323-4330. [PMID: 34427815 DOI: 10.1007/s11010-021-04239-6] [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: 03/31/2021] [Accepted: 07/29/2021] [Indexed: 10/20/2022]
Abstract
Previously it was shown that for reduction of anxiety and stress of experimental animals, preventive handling seems to be one of the most effective methods. The present study was oriented on Na,K-ATPase, a key enzyme for maintaining proper concentrations of intracellular sodium and potassium ions. Malfunction of this enzyme has an essential role in the development of neurodegenerative diseases. It is known that this enzyme requires approximately 50% of the energy available to the brain. Therefore in the present study utilization of the energy source ATP by Na,K-ATPase in the frontal cerebral cortex, using the method of enzyme kinetics was investigated. As a model of neurodegeneration treatment with trimethyltin (TMT) was applied. Daily handling (10 min/day) of healthy rats and rats suffering neurodegeneration induced by administration of TMT in a dose of (7.5 mg/kg), at postnatal days 60-102 altered the expression of catalytic subunits of Na,K-ATPase as well as kinetic properties of this enzyme in the frontal cerebral cortex of adult male Wistar rats. In addition to the previously published beneficial effect on spatial memory, daily treatment of rats was accompanied by improved maintenance of sodium homeostasis in the frontal cortex. The key system responsible for this process, Na,K-ATPase, was able to utilize better the energy substrate ATP. In rats, manipulation of TMT-induced neurodegeneration promoted the expression of the α2 isoform of the enzyme, which is typical for glial cells. In healthy rats, manipulation was followed by increased expression of the α3 subunit, which is typical of neurons.
Collapse
Affiliation(s)
- Barbora Kalocayova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Denisa Snurikova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Jana Vlkovicova
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Veronika Navarova-Stara
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Dominika Michalikova
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Eduard Ujhazy
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Zdenka Gasparova
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Norbert Vrbjar
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| |
Collapse
|
9
|
Influence of Nitric Oxide-Cyclic GMP and Oxidative STRESS on Amyloid-β Peptide Induced Decrease of Na,K-ATPase Activity in Rat Hippocampal Slices. J Membr Biol 2021; 254:463-473. [PMID: 34327545 DOI: 10.1007/s00232-021-00196-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 07/19/2021] [Indexed: 12/11/2022]
Abstract
Amyloid-β peptide (Aβ) has been shown to cause synaptic dysfunction and can render neurons vulnerable to excitotoxicity and oxidative stress. Na,K-ATPase plays an important role to maintain cell ionic equilibrium and it can be modulated by N-methyl-D-aspartate (NMDA)-nitric oxide (NO)-cyclic GMP pathway. Disruption of NO synthase (NOS) activity and reactive oxygen species (ROS) production could lead to changes in Na,K-ATPase isoforms' activities that may be detrimental to the cells. Our aim was to evaluate the signaling pathways of Aβ in relation to NMDA-NOS-cyclic GMP versus oxidative stress on α1-/α2,3-Na,K-ATPase activities in rat hippocampal slices. Aβ1-40 induced a concentration-dependent increase of NOS activity and increased cyclic guanosine monophosphate (cGMP), TBARS (thiobarbituric acid reactive substances), and 3-Nitrotyrosine (3-NT)-modified protein levels in rat hippocampal slices. The increase in NOS activity and cyclic GMP levels induced by Aβ1-40 was completely blocked by MK-801 (inhibitor of NMDA receptor) and L-NAME (inhibitor of NOS) pre-treatment but changes in TBARS levels were only partially blocked by both compounds. The Aβ treatment also decreased Na,K-ATPase activity which was reverted by N-nitro-L-arginine methyl ester hydrochloride (L-NAME) but not by MK-801 pre-treatment. The decrease in enzyme activity induced by Aβ was isoform-specific since only α1-Na,K-ATPase was affected. These findings suggest that the activation of NMDA-NOS signaling cascade linked to α2,3-Na,K-ATPase activity may mediate an adaptive, neuroprotective response to Aβ in rat hippocampus.
Collapse
|
10
|
Antibacterial Effect of Dihydromyricetin on Specific Spoilage Organisms of Hybrid Grouper. J FOOD QUALITY 2021. [DOI: 10.1155/2021/5569298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
This study aimed to investigate the mechanism of antibacterial activity level inhibition of dihydromyricetin (DMY) against specific spoilage bacteria of grouper. Firstly, the specific spoilage bacteria of grouper in the cold storage process are Pseudomonas antarctica (P. antarctica), which are selected by calculating the spoilage metabolite yield factor. It was determined that the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of DMY against grouper spoilage bacteria were 2.0 mg/mL and 6.4 mg/mL, respectively. DMY was added to the matrix of chitosan and sodium alginate, and DMY emulsions of different concentrations (0 MIC, 1 MIC, 2 MIC, 4 MIC) were prepared and characterized by differential calorimetry methods. Through analyzing cell permeability, enzyme activity, and images of the confocal laser scanning microscope (CLSM), we further studied the antibacterial mechanism of DMY emulsion on specific spoilage bacteria. The results showed that, with the increase of DMY concentration in the treatment group, the leakage of nucleic acid and protein increased significantly, the activity of ATPase and three critical enzymes in the Embden-Meyerhof-Parnas (EMP) pathway decreased significantly, and the activity of AKPase did not decrease significantly, . The metabolic activity and viability are reduced considerably. Analysis of the above results shows that DMY inhibits the growth and reproduction of P. antarctica by interfering with the metabolic activity of bacteria and destroying the function of bacterial cell membranes but has no inhibitory effect on the activity of AKPase. This study proves that DMY could be an effective and natural antibacterial agent against specific spoilage bacteria in aquatic products.
Collapse
|
11
|
Kinoshita PF, Orellana AMM, Nakao VW, de Souza Port's NM, Quintas LEM, Kawamoto EM, Scavone C. The Janus face of ouabain in Na + /K + -ATPase and calcium signalling in neurons. Br J Pharmacol 2021; 179:1512-1524. [PMID: 33644859 DOI: 10.1111/bph.15419] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/03/2021] [Accepted: 02/14/2021] [Indexed: 12/20/2022] Open
Abstract
Na+ /K+ -ATPase, a transmembrane protein essential for maintaining the electrochemical gradient across the plasma membrane, acts as a receptor for cardiotonic steroids such as ouabain. Cardiotonic steroids binding to Na+ /K+ -ATPase triggers signalling pathways or inhibits Na+ /K+ -ATPas activity in a concentration-dependent manner, resulting in a modulation of Ca2+ levels, which are essential for homeostasis in neurons. However, most of the pharmacological strategies for avoiding neuronal death do not target Na+ /K+ -ATPase activity due to its complexity and the poor understanding of the mechanisms involved in Na+ /K+ -ATPase modulation. The present review aims to discuss two points regarding the interplay between Na+ /K+ -ATPase and Ca2+ signalling in the brain. One, Na+ /K+ -ATPase impairment causing illness and neuronal death due to Ca2+ signalling and two, benefits to the brain by modulating Na+ /K+ -ATPase activity. These interactions play an essential role in neuronal cell fate determination and are relevant to find new targets for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Paula Fernanda Kinoshita
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ana Maria Marques Orellana
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Vinicius Watanabe Nakao
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Natacha Medeiros de Souza Port's
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Luis Eduardo Menezes Quintas
- Laboratory of Biochemical and Molecular Pharmacology, Institute of Biomedical Sciences, Health Sciences Centre Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Elisa Mitiko Kawamoto
- Laboratory of Molecular and Functional Neurobiology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Cristoforo Scavone
- Laboratory of Molecular Neuropharmacology, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
12
|
Cao L, Xiong S, Wu Z, Ding L, Zhou Y, Sun H, Zhu M, Lee WT, Nie X, Bian JS. Anti-Na +/K +-ATPase immunotherapy ameliorates α-synuclein pathology through activation of Na +/K +-ATPase α1-dependent autophagy. SCIENCE ADVANCES 2021; 7:7/5/eabc5062. [PMID: 33571110 PMCID: PMC7840131 DOI: 10.1126/sciadv.abc5062] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 12/09/2020] [Indexed: 05/06/2023]
Abstract
Na+/K+-ATPase (NKA) plays important roles in maintaining cellular homeostasis. Conversely, reduced NKA activity has been reported in aging and neurodegenerative diseases. However, little is known about the function of NKA in the pathogenesis of Parkinson's disease (PD). Here, we report that reduction of NKA activity in NKAα1+/- mice aggravates α-synuclein-induced pathology, including a reduction in tyrosine hydroxylase (TH) and deficits in behavioral tests for memory, learning, and motor function. To reverse this effect, we generated an NKA-stabilizing monoclonal antibody, DR5-12D, against the DR region (897DVEDSYGQQWTYEQR911) of the NKAα1 subunit. We demonstrate that DR5-12D can ameliorate α-synuclein-induced TH loss and behavioral deficits by accelerating α-synuclein degradation in neurons. The underlying mechanism for the beneficial effects of DR5-12D involves activation of NKAα1-dependent autophagy via increased AMPK/mTOR/ULK1 pathway signaling. Cumulatively, this work demonstrates that NKA activity is neuroprotective and that pharmacological activation of this pathway represents a new therapeutic strategy for PD.
Collapse
Affiliation(s)
- Lei Cao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Siping Xiong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Zhiyuan Wu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lei Ding
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yebo Zhou
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Haijian Sun
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mengyuan Zhu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wei Thye Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xiaowei Nie
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Jin-Song Bian
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, PR China
- National University of Singapore (Suzhou) Research Institute, Suzhou 215000, China
| |
Collapse
|
13
|
Cioffi F, Adam RHI, Broersen K. Molecular Mechanisms and Genetics of Oxidative Stress in Alzheimer's Disease. J Alzheimers Dis 2020; 72:981-1017. [PMID: 31744008 PMCID: PMC6971833 DOI: 10.3233/jad-190863] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Alzheimer’s disease is the most common neurodegenerative disorder that can cause dementia in elderly over 60 years of age. One of the disease hallmarks is oxidative stress which interconnects with other processes such as amyloid-β deposition, tau hyperphosphorylation, and tangle formation. This review discusses current thoughts on molecular mechanisms that may relate oxidative stress to Alzheimer’s disease and identifies genetic factors observed from in vitro, in vivo, and clinical studies that may be associated with Alzheimer’s disease-related oxidative stress.
Collapse
Affiliation(s)
- Federica Cioffi
- Nanobiophysics Group, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Rayan Hassan Ibrahim Adam
- Nanobiophysics Group, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Kerensa Broersen
- Applied Stem Cell Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| |
Collapse
|
14
|
Hoshi M. Multi-angle development of therapeutic methods for Alzheimer's disease. Br J Pharmacol 2020; 178:770-783. [PMID: 32592177 DOI: 10.1111/bph.15174] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/07/2020] [Accepted: 06/09/2020] [Indexed: 01/01/2023] Open
Abstract
Recent clinical trial results support the idea that treatment based on the so-called amyloid hypothesis is a promising approach in Alzheimer's disease (AD), but actually, developing effective treatments for AD remains highly challenging. The discovery that neuron-specific sodium pump activity is impaired in AD and other neurodegenerative diseases such as Parkinson's disease has suggested a role for the sodium pump in the pathogenesis of these diseases. This opens up new possibilities for intervention, such as inhibiting the aberrant interaction of the sodium pump with the disease-specific ligand(s) or activating the sodium pump itself or its downstream signalling. In this review article, I would like to discuss possible anti-amyloid therapies, focusing especially on our own research. LINKED ARTICLES: This article is part of a themed issue on Neurochemistry in Japan. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.4/issuetoc.
Collapse
Affiliation(s)
- Minako Hoshi
- Department for Brain and Neurodegenerative Disease Research, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| |
Collapse
|
15
|
Antimicrobial Susceptibility and Antibacterial Mechanism of Limonene against Listeria monocytogenes. Molecules 2019; 25:molecules25010033. [PMID: 31861877 PMCID: PMC6982812 DOI: 10.3390/molecules25010033] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/03/2019] [Accepted: 12/12/2019] [Indexed: 11/24/2022] Open
Abstract
Limonene is a monoterpenoid compound, which is founded in a lot of plants’ essential oils with good antibacterial activity against food-borne pathogens, but it has an ambiguous antimicrobial susceptibility and mechanism against Listeria monocytogenes (L. monocytogenes). In this study, the antimicrobial susceptibility of Limonene to L. monocytogenes was studied, and some new sights regarding its antibacterial mechanism were further explored. Scanning electron microscopy (SEM) verified that limonene caused the destruction of the cell integrity and wall structure of L. monocytogenes. The increase in conductivity and the leakage of intracellular biomacromolecules (nucleic acids and proteins) confirmed that limonene had an obvious effect on cell membrane permeability. The results of Propidium Iodide (PI) fluorescence staining were consistent with the results of the conductivity measurements. This indicated that limonene treatment caused damage to the L. monocytogenes cell membrane. Furthermore, the decrease in ATP content, ATPase (Na+K+-ATPase, Ca2+-ATPase) activity and respiratory chain complex activity indicated that limonene could hinder ATP synthesis by inhibiting the activity of the respiratory complex and ATPase. Finally, differential expression of proteins in the respiratory chain confirmed that limonene affected respiration and energy metabolism by inhibiting the function of the respiratory chain complex.
Collapse
|
16
|
Owen MC, Gnutt D, Gao M, Wärmländer SKTS, Jarvet J, Gräslund A, Winter R, Ebbinghaus S, Strodel B. Effects of in vivo conditions on amyloid aggregation. Chem Soc Rev 2019; 48:3946-3996. [PMID: 31192324 DOI: 10.1039/c8cs00034d] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
One of the grand challenges of biophysical chemistry is to understand the principles that govern protein misfolding and aggregation, which is a highly complex process that is sensitive to initial conditions, operates on a huge range of length- and timescales, and has products that range from protein dimers to macroscopic amyloid fibrils. Aberrant aggregation is associated with more than 25 diseases, which include Alzheimer's, Parkinson's, Huntington's, and type II diabetes. Amyloid aggregation has been extensively studied in the test tube, therefore under conditions that are far from physiological relevance. Hence, there is dire need to extend these investigations to in vivo conditions where amyloid formation is affected by a myriad of biochemical interactions. As a hallmark of neurodegenerative diseases, these interactions need to be understood in detail to develop novel therapeutic interventions, as millions of people globally suffer from neurodegenerative disorders and type II diabetes. The aim of this review is to document the progress in the research on amyloid formation from a physicochemical perspective with a special focus on the physiological factors influencing the aggregation of the amyloid-β peptide, the islet amyloid polypeptide, α-synuclein, and the hungingtin protein.
Collapse
Affiliation(s)
- Michael C Owen
- CEITEC - Central European Institute of Technology, Masaryk University, Kamenice 753/5, Brno 625 00, Czech Republic
| | - David Gnutt
- Institute of Physical and Theoretical Chemistry, TU Braunschweig, Rebenring 56, 38106 Braunschweig, Germany and Lead Discovery Wuppertal, Bayer AG, 42096 Wuppertal, Germany
| | - Mimi Gao
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn Str. 4a, 44227 Dortmund, Germany and Sanofi-Aventis Deutschland GmbH, R&D, Industriepark Höchst, 65926 Frankfurt, Germany
| | - Sebastian K T S Wärmländer
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius väg 16C, 106 91 Stockholm, Sweden
| | - Jüri Jarvet
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius väg 16C, 106 91 Stockholm, Sweden
| | - Astrid Gräslund
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius väg 16C, 106 91 Stockholm, Sweden
| | - Roland Winter
- Faculty of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn Str. 4a, 44227 Dortmund, Germany
| | - Simon Ebbinghaus
- Institute of Physical and Theoretical Chemistry, TU Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
| | - Birgit Strodel
- Institute of Complex Systems: Structural Biochemistry, Forschungszentrum Jülich, 42525 Jülich, Germany. and Institute of Theoretical and Computational Chemistry, Heinrich Heine University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| |
Collapse
|
17
|
Liu S, Cao XL, Liu GQ, Zhou T, Yang XL, Ma BX. Thein silicoandin vivoevaluation of puerarin against Alzheimer's disease. Food Funct 2019; 10:799-813. [DOI: 10.1039/c8fo01696h] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In silicomethods were used to screen the anti-AD effect of puerarin, further mutually verified by anin vivostudy.
Collapse
Affiliation(s)
- Song Liu
- Department of Pharmacy
- School of Medicine
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control
- Wuhan University of Science and Technology
- Wuhan
| | - Xiao-Lu Cao
- Department of Pharmacy
- School of Medicine
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control
- Wuhan University of Science and Technology
- Wuhan
| | - Guang-Qi Liu
- Department of Pharmacy
- School of Medicine
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control
- Wuhan University of Science and Technology
- Wuhan
| | - Tong Zhou
- Department of Pharmacy
- School of Medicine
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control
- Wuhan University of Science and Technology
- Wuhan
| | - Xi-Liang Yang
- Department of Pharmacy
- School of Medicine
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control
- Wuhan University of Science and Technology
- Wuhan
| | - Bing-Xin Ma
- Reproductive Medicine Center
- Tongji Hospital
- Tongji Medical College
- Huazhong University of Science and Technology
- Wuhan, 430030
| |
Collapse
|
18
|
Dinčić M, Krstić DZ, Čolović MB, Nešović Ostojić J, Kovačević S, De Luka SR, Djordjević DM, Ćirković S, Brkić P, Todorović J. Modulation of rat synaptosomal ATPases and acetylcholinesterase activities induced by chronic exposure to the static magnetic field. Int J Radiat Biol 2018; 94:1062-1071. [PMID: 30238840 DOI: 10.1080/09553002.2018.1518611] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
PURPOSE It is considered that exposure to static magnetic fields (SMF) may have both detrimental and therapeutic effect, but the mechanism of SMF influence on the living organisms is not well understood. Since the adenosine triphosphatases (ATPases) and acetylcholinesterase (AChE) are involved in both physiological and pathological processes, the modulation of Na+/K+-ATPase, ecto-ATPases and AChE activities, as well as oxidative stress responses were followed in synaptosomes isolated from rats after chronic exposure toward differently oriented SMF. MATERIAL AND METHODS Wistar albino rats were randomly divided into three experimental groups (six animals per group): Up and Down group - exposed to upward and downward oriented SMF, respectively, and Control group. After 50 days, the rats were sacrificed, and synaptosomes were isolated from the whole rat brain and used for testing the enzyme activities and oxidative stress parameters. RESULTS Chronic exposure to 1 mT SMF significantly increased ATPases, AChE activities, and malondialdehyde (MDA) level in both exposed groups, compared to control values. The significant decrease in synaptosomal catalase activity (1.48 ± 0.17 U/mg protein) induced by exposure to the downward oriented field, compared to those obtained for Control group (2.60 ± 0.29 U/mg protein), and Up group (2.72 ± 0.21 U/mg protein). CONCLUSIONS It could be concluded that chronic exposure to differently oriented SMF increases ATPases and AChE activities in rat synaptosomes. Since brain ATPases and AChE have important roles in the pathogenesis of several neurological diseases, SMF influence on the activity of these enzymes may have potential therapeutic importance.
Collapse
Affiliation(s)
- Marko Dinčić
- a Institute of Pathological Physiology, Faculty of Medicine , University of Belgrade , Belgrade , Serbia
| | - Danijela Z Krstić
- b Institute of Medical Chemistry, Faculty of Medicine , University of Belgrade , Belgrade , Serbia
| | - Mirjana B Čolović
- c Department of Physical Chemistry , Vinča Institute of Nuclear Sciences, University of Belgrade , Belgrade , Serbia
| | - Jelena Nešović Ostojić
- a Institute of Pathological Physiology, Faculty of Medicine , University of Belgrade , Belgrade , Serbia
| | - Sanjin Kovačević
- a Institute of Pathological Physiology, Faculty of Medicine , University of Belgrade , Belgrade , Serbia
| | - Silvio R De Luka
- a Institute of Pathological Physiology, Faculty of Medicine , University of Belgrade , Belgrade , Serbia
| | - Drago M Djordjević
- a Institute of Pathological Physiology, Faculty of Medicine , University of Belgrade , Belgrade , Serbia
| | - Saša Ćirković
- d Institute of Physics , University of Belgrade , Belgrade , Serbia
| | - Predrag Brkić
- e Institute of Physiology, Faculty of Medicine , University of Belgrade , Serbia
| | - Jasna Todorović
- a Institute of Pathological Physiology, Faculty of Medicine , University of Belgrade , Belgrade , Serbia
| |
Collapse
|
19
|
Fan X, Ashraf UM, Drummond CA, Shi H, Zhang X, Kumarasamy S, Tian J. Characterization of a Long Non-Coding RNA, the Antisense RNA of Na/K-ATPase α1 in Human Kidney Cells. Int J Mol Sci 2018; 19:ijms19072123. [PMID: 30037072 PMCID: PMC6073804 DOI: 10.3390/ijms19072123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/17/2018] [Accepted: 07/19/2018] [Indexed: 01/19/2023] Open
Abstract
Non-coding RNAs are important regulators of protein-coding genes. The current study characterized an antisense long non-coding RNA, ATP1A1-AS1, which is located on the opposite strand of the Na/K-ATPase α1 gene. Our results show that four splice variants are expressed in human adult kidney cells (HK2 cells) and embryonic kidney cells (HEK293 cells). These variants can be detected in both cytosol and nuclear fractions. We also found that the inhibition of DNA methylation has a differential effect on the expression of ATP1A1-AS1 and its sense gene. To investigate the physiological role of this antisense gene, we overexpressed the ATP1A1-AS1 transcripts, and examined their effect on Na/K-ATPase expression and related signaling function in human kidney cells. The results showed that overexpression of the ATP1A1-AS1-203 transcript in HK2 cells reduced the Na/K-ATPase α1 (ATP1A1) gene expression by approximately 20% (p < 0.05), while reducing the Na/K-ATPase α1 protein synthesis by approximately 22% (p < 0.05). Importantly, overexpression of the antisense RNA transcript attenuated ouabain-induced Src activation in HK2 cells. It also inhibited the cell proliferation and potentiated ouabain-induced cell death. These results demonstrate that the ATP1A1-AS1 gene is a moderate negative regulator of Na/K-ATPase α1, and can modulate Na/K-ATPase-related signaling pathways in human kidney cells.
Collapse
Affiliation(s)
- Xiaoming Fan
- Department of Medicine, University of Toledo, Toledo, OH 43614, USA.
| | - Usman M Ashraf
- Department of Physiology and Pharmacology, Center for Hypertension and Personalized Medicine, University of Toledo, Toledo, OH 43614, USA.
| | - Christopher A Drummond
- Department of Medicine, University of Toledo, Toledo, OH 43614, USA.
- MPI Research, Mattawan, MI 49071, USA.
| | - Huilin Shi
- Department of Medicine, University of Toledo, Toledo, OH 43614, USA.
| | - Xiaolu Zhang
- Department of Medicine, University of Toledo, Toledo, OH 43614, USA.
| | - Sivarajan Kumarasamy
- Department of Physiology and Pharmacology, Center for Hypertension and Personalized Medicine, University of Toledo, Toledo, OH 43614, USA.
| | - Jiang Tian
- Department of Medicine, University of Toledo, Toledo, OH 43614, USA.
| |
Collapse
|
20
|
Pierozan P, Biasibetti-Brendler H, Schmitz F, Ferreira F, Netto CA, Wyse ATS. Synergistic Toxicity of the Neurometabolites Quinolinic Acid and Homocysteine in Cortical Neurons and Astrocytes: Implications in Alzheimer's Disease. Neurotox Res 2017; 34:147-163. [PMID: 29124681 DOI: 10.1007/s12640-017-9834-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/22/2017] [Accepted: 10/18/2017] [Indexed: 12/19/2022]
Abstract
The brain of patients affected by Alzheimer's disease (AD) develops progressive neurodegeneration linked to the formation of proteins aggregates. However, their single actions cannot explain the extent of brain damage observed in this disorder, and the characterization of co-adjuvant involved in the early toxic processes evoked in AD is essential. In this line, quinolinic acid (QUIN) and homocysteine (Hcy) appear to be involved in the AD neuropathogenesis. Herein, we investigate the effects of QUIN and Hcy on early toxic events in cortical neurons and astrocytes. Exposure of primary cortical cultures to these neurometabolites for 24 h induced concentration-dependent neurotoxicity. In addition, QUIN (25 μM) and Hcy (30 μM) triggered ROS production, lipid peroxidation, diminished of Na+,K+-ATPase activity, and morphologic alterations, culminating in reduced neuronal viability by necrotic cell death. In astrocytes, QUIN (100 μM) and Hcy (30 μM) induced caspase-3-dependent apoptosis and morphologic alterations through oxidative status imbalance. To establish specific mechanisms, we preincubated cell cultures with different protective agents. The combined toxicity of QUIN and Hcy was attenuated by melatonin and Trolox in neurons and by NMDA antagonists and glutathione in astrocytes. Cellular death and morphologic alterations were prevented when co-culture was treated with metabolites, suggesting the activation of protector mechanisms dependent on soluble factors and astrocyte and neuron communication through gap junctions. These findings suggest that early damaging events involved in AD can be magnified by synergistic toxicity of the QUIN and Hcy. Therefore, this study opens new possibilities to elucidate the molecular mechanisms of neuron-astrocyte interactions and their role in neuroprotection against QUIN and Hcy.
Collapse
Affiliation(s)
- Paula Pierozan
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600 anexo, Porto Alegre, RS, 90035-003, Brazil.
| | - Helena Biasibetti-Brendler
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600 anexo, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Felipe Schmitz
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600 anexo, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Fernanda Ferreira
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600 anexo, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carlos Alexandre Netto
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Laboratório de Isquemia Cerebral e Psicobiologia dos Transtornos Mentais, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP, Porto Alegre, RS, 90035-003, Brazil
| | - Angela T S Wyse
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600 anexo, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP, Porto Alegre, RS, 90035-003, Brazil
| |
Collapse
|
21
|
Dorard E, Chasseigneaux S, Gorisse-Hussonnois L, Broussard C, Pillot T, Allinquant B. Soluble Amyloid Precursor Protein Alpha Interacts with alpha3-Na, K-ATPAse to Induce Axonal Outgrowth but Not Neuroprotection: Evidence for Distinct Mechanisms Underlying these Properties. Mol Neurobiol 2017; 55:5594-5610. [PMID: 28983842 DOI: 10.1007/s12035-017-0783-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 09/20/2017] [Indexed: 01/09/2023]
Abstract
Amyloid precursor protein (APP) is cleaved not only to generate the amyloid peptide (Aß), involved in neurodegenerative processes, but can also be metabolized by alpha secretase to produce and release soluble N-terminal APP (sAPPα), which has many properties including the induction of axonal elongation and neuroprotection. The mechanisms underlying the properties of sAPPα are not known. Here, we used proteomic analysis of mouse cortico-hippocampal membranes to identify the neuronal specific alpha3 (α3)-subunit of the plasma membrane enzyme Na, K-ATPase (NKA) as a new binding partner of sAPPα. We showed that sAPPα recruits very rapidly clusters of α3-NKA at neuronal surface, and its binding triggers a cascade of events promoting sAPPα-induced axonal outgrowth. The binding of sAPPα with α3-NKA was not observed for sAPPα-induced Aß1-42 oligomers neuroprotection, neither the downstream events particularly the interaction of sAPPα with APP before endocytosis, ERK signaling, and the translocation of SET from the nucleus to the plasma membrane. These data suggest that the mechanisms of the axonal growth promoting and neuroprotective properties of sAPPα appear to be specific and independent. The signals at the cell surface specific to trigger these mechanisms require further study.
Collapse
Affiliation(s)
- Emilie Dorard
- UMR_S894 INSERM, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, 75014, Paris, France.,SynAging, 54500, Vandoeuvre-les, Nancy, France
| | - Stéphanie Chasseigneaux
- UMR_S894 INSERM, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, 75014, Paris, France.,INSERM U1144, Université Paris Descartes and Université Paris Diderot UMR-S 1144, 75006, Paris, France
| | - Lucie Gorisse-Hussonnois
- UMR_S894 INSERM, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, 75014, Paris, France
| | - Cédric Broussard
- Plate-forme Protéomique, Université Paris Descartes 3P5, Institut Cochin, 75014, Paris, France
| | | | - Bernadette Allinquant
- UMR_S894 INSERM, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, 75014, Paris, France.
| |
Collapse
|
22
|
Boscia F, Begum G, Pignataro G, Sirabella R, Cuomo O, Casamassa A, Sun D, Annunziato L. Glial Na(+) -dependent ion transporters in pathophysiological conditions. Glia 2016; 64:1677-97. [PMID: 27458821 DOI: 10.1002/glia.23030] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/22/2016] [Accepted: 06/29/2016] [Indexed: 12/12/2022]
Abstract
Sodium dynamics are essential for regulating functional processes in glial cells. Indeed, glial Na(+) signaling influences and regulates important glial activities, and plays a role in neuron-glia interaction under physiological conditions or in response to injury of the central nervous system (CNS). Emerging studies indicate that Na(+) pumps and Na(+) -dependent ion transporters in astrocytes, microglia, and oligodendrocytes regulate Na(+) homeostasis and play a fundamental role in modulating glial activities in neurological diseases. In this review, we first briefly introduced the emerging roles of each glial cell type in the pathophysiology of cerebral ischemia, Alzheimer's disease, epilepsy, Parkinson's disease, Amyotrophic Lateral Sclerosis, and myelin diseases. Then, we discussed the current knowledge on the main roles played by the different glial Na(+) -dependent ion transporters, including Na(+) /K(+) ATPase, Na(+) /Ca(2+) exchangers, Na(+) /H(+) exchangers, Na(+) -K(+) -Cl(-) cotransporters, and Na(+) - HCO3- cotransporter in the pathophysiology of the diverse CNS diseases. We highlighted their contributions in cell survival, synaptic pathology, gliotransmission, pH homeostasis, and their role in glial activation, migration, gliosis, inflammation, and tissue repair processes. Therefore, this review summarizes the foundation work for targeting Na(+) -dependent ion transporters in glia as a novel strategy to control important glial activities associated with Na(+) dynamics in different neurological disorders. GLIA 2016;64:1677-1697.
Collapse
Affiliation(s)
- Francesca Boscia
- Division of Pharmacology, Department of Neuroscience, Reproductive, and Odontostomatological Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Gulnaz Begum
- Department of Neurology, University of Pittsburgh Medical School
| | - Giuseppe Pignataro
- Division of Pharmacology, Department of Neuroscience, Reproductive, and Odontostomatological Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Rossana Sirabella
- Division of Pharmacology, Department of Neuroscience, Reproductive, and Odontostomatological Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Ornella Cuomo
- Division of Pharmacology, Department of Neuroscience, Reproductive, and Odontostomatological Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Antonella Casamassa
- Division of Pharmacology, Department of Neuroscience, Reproductive, and Odontostomatological Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh Medical School.,Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational and Clinical Center, Pittsburgh, Pennsylvania, 15213
| | - Lucio Annunziato
- Division of Pharmacology, Department of Neuroscience, Reproductive, and Odontostomatological Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| |
Collapse
|
23
|
Kinoshita PF, Leite JA, Orellana AMM, Vasconcelos AR, Quintas LEM, Kawamoto EM, Scavone C. The Influence of Na(+), K(+)-ATPase on Glutamate Signaling in Neurodegenerative Diseases and Senescence. Front Physiol 2016; 7:195. [PMID: 27313535 PMCID: PMC4890531 DOI: 10.3389/fphys.2016.00195] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/17/2016] [Indexed: 12/17/2022] Open
Abstract
Decreased Na(+), K(+)-ATPase (NKA) activity causes energy deficiency, which is commonly observed in neurodegenerative diseases. The NKA is constituted of three subunits: α, β, and γ, with four distinct isoforms of the catalytic α subunit (α1-4). Genetic mutations in the ATP1A2 gene and ATP1A3 gene, encoding the α2 and α3 subunit isoforms, respectively can cause distinct neurological disorders, concurrent to impaired NKA activity. Within the central nervous system (CNS), the α2 isoform is expressed mostly in glial cells and the α3 isoform is neuron-specific. Mutations in ATP1A2 gene can result in familial hemiplegic migraine (FHM2), while mutations in the ATP1A3 gene can cause Rapid-onset dystonia-Parkinsonism (RDP) and alternating hemiplegia of childhood (AHC), as well as the cerebellar ataxia, areflexia, pescavus, optic atrophy and sensorineural hearing loss (CAPOS) syndrome. Data indicates that the central glutamatergic system is affected by mutations in the α2 isoform, however further investigations are required to establish a connection to mutations in the α3 isoform, especially given the diagnostic confusion and overlap with glutamate transporter disease. The age-related decline in brain α2∕3 activity may arise from changes in the cyclic guanosine monophosphate (cGMP) and cGMP-dependent protein kinase (PKG) pathway. Glutamate, through nitric oxide synthase (NOS), cGMP and PKG, stimulates brain α2∕3 activity, with the glutamatergic N-methyl-D-aspartate (NMDA) receptor cascade able to drive an adaptive, neuroprotective response to inflammatory and challenging stimuli, including amyloid-β. Here we review the NKA, both as an ion pump as well as a receptor that interacts with NMDA, including the role of NKA subunits mutations. Failure of the NKA-associated adaptive response mechanisms may render neurons more susceptible to degeneration over the course of aging.
Collapse
Affiliation(s)
- Paula F. Kinoshita
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| | - Jacqueline A. Leite
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| | - Ana Maria M. Orellana
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| | - Andrea R. Vasconcelos
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| | - Luis E. M. Quintas
- Laboratory of Biochemical and Molecular Pharmacology, Institute of Biomedical Sciences, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Elisa M. Kawamoto
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| | - Cristoforo Scavone
- Department of Pharmacology, Institute of Biomedical Science, University of São PauloSão Paulo, Brazil
| |
Collapse
|
24
|
Tsai SF, Chen PC, Calkins MJ, Wu SY, Kuo YM. Exercise Counteracts Aging-Related Memory Impairment: A Potential Role for the Astrocytic Metabolic Shuttle. Front Aging Neurosci 2016; 8:57. [PMID: 27047373 PMCID: PMC4801859 DOI: 10.3389/fnagi.2016.00057] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/08/2016] [Indexed: 01/19/2023] Open
Abstract
Age-related cognitive impairment has become one of the most common health threats in many countries. The biological substrate of cognition is the interconnection of neurons to form complex information processing networks. Experience-based alterations in the activities of these information processing networks lead to neuroadaptation, which is physically represented at the cellular level as synaptic plasticity. Although synaptic plasticity is known to be affected by aging, the underlying molecular mechanisms are not well described. Astrocytes, a glial cell type that is infrequently investigated in cognitive science, have emerged as energy suppliers which are necessary for meeting the abundant energy demand resulting from glutamatergic synaptic activity. Moreover, the concerted action of an astrocyte-neuron metabolic shuttle is essential for cognitive function; whereas, energetic incoordination between astrocytes and neurons may contribute to cognitive impairment. Whether altered function of the astrocyte-neuron metabolic shuttle links aging to reduced synaptic plasticity is unexplored. However, accumulated evidence documents significant beneficial effects of long-term, regular exercise on cognition and synaptic plasticity. Furthermore, exercise increases the effectiveness of astrocyte-neuron metabolic shuttle by upregulation of astrocytic lactate transporter levels. This review summarizes previous findings related to the neuronal activity-dependent astrocyte-neuron metabolic shuttle. Moreover, we discuss how aging and exercise may shape the astrocyte-neuron metabolic shuttle in cognition-associated brain areas.
Collapse
Affiliation(s)
- Sheng-Feng Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University Tainan, Taiwan
| | - Pei-Chun Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung UniversityTainan, Taiwan; Department of Physiology, College of Medicine, National Cheng Kung UniversityTainan, Taiwan
| | - Marcus J Calkins
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University Tainan, Taiwan
| | - Shih-Ying Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University Tainan, Taiwan
| | - Yu-Min Kuo
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung UniversityTainan, Taiwan; Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung UniversityTainan, Taiwan
| |
Collapse
|
25
|
Ruegsegger C, Maharjan N, Goswami A, Filézac de L'Etang A, Weis J, Troost D, Heller M, Gut H, Saxena S. Aberrant association of misfolded SOD1 with Na(+)/K(+)ATPase-α3 impairs its activity and contributes to motor neuron vulnerability in ALS. Acta Neuropathol 2016; 131:427-51. [PMID: 26619836 DOI: 10.1007/s00401-015-1510-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 11/03/2015] [Accepted: 11/14/2015] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult onset progressive motor neuron disease with no cure. Transgenic mice overexpressing familial ALS associated human mutant SOD1 are a commonly used model for examining disease mechanisms. Presently, it is well accepted that alterations in motor neuron excitability and spinal circuits are pathological hallmarks of ALS, but the underlying molecular mechanisms remain unresolved. Here, we sought to understand whether the expression of mutant SOD1 protein could contribute to altering processes governing motor neuron excitability. We used the conformation specific antibody B8H10 which recognizes a misfolded state of SOD1 (misfSOD1) to longitudinally identify its interactome during early disease stage in SOD1G93A mice. This strategy identified a direct isozyme-specific association of misfSOD1 with Na(+)/K(+)ATPase-α3 leading to the premature impairment of its ATPase activity. Pharmacological inhibition of Na(+)/K(+)ATPase-α3 altered glutamate receptor 2 expression, modified cholinergic inputs and accelerated disease pathology. After mapping the site of direct association of misfSOD1 with Na(+)/K(+)ATPase-α3 onto a 10 amino acid stretch that is unique to Na(+)/K(+)ATPase-α3 but not found in the closely related Na(+)/K(+)ATPase-α1 isozyme, we generated a misfSOD1 binding deficient, but fully functional Na(+)/K(+)ATPase-α3 pump. Adeno associated virus (AAV)-mediated expression of this chimeric Na(+)/K(+)ATPase-α3 restored Na(+)/K(+)ATPase-α3 activity in the spinal cord, delayed pathological alterations and prolonged survival of SOD1G93A mice. Additionally, altered Na(+)/K(+)ATPase-α3 expression was observed in the spinal cord of individuals with sporadic and familial ALS. A fraction of sporadic ALS cases also presented B8H10 positive misfSOD1 immunoreactivity, suggesting that similar mechanism might contribute to the pathology.
Collapse
Affiliation(s)
- Céline Ruegsegger
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Niran Maharjan
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Anand Goswami
- Institute of Neuropathology, Rheinisch-Westfälische Technische Hochschule, Aachen University Hospital, Aachen, Germany
| | - Audrey Filézac de L'Etang
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- Department of Neuroscience, Genentech, Inc., South San Francisco, California, USA
| | - Joachim Weis
- Institute of Neuropathology, Rheinisch-Westfälische Technische Hochschule, Aachen University Hospital, Aachen, Germany
| | - Dirk Troost
- Division of Neuropathology, Department of Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | - Manfred Heller
- Department of Clinical Research, Inselspital, University of Bern, Bern, Switzerland
| | - Heinz Gut
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Smita Saxena
- Institute of Cell Biology, University of Bern, Bern, Switzerland.
| |
Collapse
|
26
|
Abstract
Neurodegeneration correlates with Alzheimer's disease (AD) symptoms, but the molecular identities of pathogenic amyloid β-protein (Aβ) oligomers and their targets, leading to neurodegeneration, remain unclear. Amylospheroids (ASPD) are AD patient-derived 10- to 15-nm spherical Aβ oligomers that cause selective degeneration of mature neurons. Here, we show that the ASPD target is neuron-specific Na(+)/K(+)-ATPase α3 subunit (NAKα3). ASPD-binding to NAKα3 impaired NAKα3-specific activity, activated N-type voltage-gated calcium channels, and caused mitochondrial calcium dyshomeostasis, tau abnormalities, and neurodegeneration. NMR and molecular modeling studies suggested that spherical ASPD contain N-terminal-Aβ-derived "thorns" responsible for target binding, which are distinct from low molecular-weight oligomers and dodecamers. The fourth extracellular loop (Ex4) region of NAKα3 encompassing Asn(879) and Trp(880) is essential for ASPD-NAKα3 interaction, because tetrapeptides mimicking this Ex4 region bound to the ASPD surface and blocked ASPD neurotoxicity. Our findings open up new possibilities for knowledge-based design of peptidomimetics that inhibit neurodegeneration in AD by blocking aberrant ASPD-NAKα3 interaction.
Collapse
|
27
|
Omotayo TI, Akinyemi GS, Omololu PA, Ajayi BO, Akindahunsi AA, Rocha JBT, Kade IJ. Possible involvement of membrane lipids peroxidation and oxidation of catalytically essential thiols of the cerebral transmembrane sodium pump as component mechanisms of iron-mediated oxidative stress-linked dysfunction of the pump's activity. Redox Biol 2014; 4:234-41. [PMID: 25618580 PMCID: PMC4803792 DOI: 10.1016/j.redox.2014.12.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 12/21/2014] [Accepted: 12/22/2014] [Indexed: 11/20/2022] Open
Abstract
The precise molecular events defining the complex role of oxidative stress in the inactivation of the cerebral sodium pump in radical-induced neurodegenerative diseases is yet to be fully clarified and thus still open. Herein we investigated the modulation of the activity of the cerebral transmembrane electrogenic enzyme in Fe2+-mediated in vitro oxidative stress model. The results show that Fe2+ inhibited the transmembrane enzyme in a concentration dependent manner and this effect was accompanied by a biphasic generation of aldehydic product of lipid peroxidation. While dithiothreitol prevented both Fe2+ inhibitory effect on the pump and lipid peroxidation, vitamin E prevented only lipid peroxidation but not inhibition of the pump. Besides, malondialdehyde (MDA) inhibited the pump by a mechanism not related to oxidation of its critical thiols. Apparently, the low activity of the pump in degenerative diseases mediated by Fe2+ may involve complex multi-component mechanisms which may partly involve an initial oxidation of the critical thiols of the enzyme directly mediated by Fe2+ and during severe progression of such diseases; aldehydic products of lipid peroxidation such as MDA may further exacerbate this inhibitory effect by a mechanism that is likely not related to the oxidation of the catalytically essential thiols of the ouabain-sensitive cerebral electrogenic pump. Fe2+ evoked lipid peroxidation (LPO) and inhibition of sodium pump (SP) in rat brain. However, dithiothreitol prevented both Fe2+-mediated LPO and inhibition of SP. Conversely, vitamin E prevented only Fe2+-mediated LPO but not inhibition of SP. Thus Fe2+ mediated inactivation of SP likely by oxidizing the essential thiol on SP. However, malondialdehyde inhibited SP by a mechanism not related to thiol oxidation.
Collapse
Affiliation(s)
- T I Omotayo
- Department of Biochemistry, Federal University of Technology, Akure, Ondo State, Nigeria
| | - G S Akinyemi
- Department of Biochemistry, Federal University of Technology, Akure, Ondo State, Nigeria
| | - P A Omololu
- Department of Biochemistry, Federal University of Technology, Akure, Ondo State, Nigeria
| | - B O Ajayi
- Department of Biochemistry, Federal University of Technology, Akure, Ondo State, Nigeria
| | - A A Akindahunsi
- Department of Biochemistry, Federal University of Technology, Akure, Ondo State, Nigeria
| | - J B T Rocha
- Centro de Ciencias Naturais e Exatas, Programa Posgraduacao em Bioquimica Toxciologica, Universidade Federal de Santa Maria, RS, Brazil
| | - I J Kade
- Department of Biochemistry, Federal University of Technology, Akure, Ondo State, Nigeria.
| |
Collapse
|
28
|
Benkhedah N, Bachert P, Nagel AM. Two-pulse biexponential-weighted 23Na imaging. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2014; 240:67-76. [PMID: 24530955 DOI: 10.1016/j.jmr.2014.01.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 12/23/2013] [Accepted: 01/15/2014] [Indexed: 06/03/2023]
Abstract
A new method is proposed for acquiring 3D biexponential-weighted sodium images with two instead of three RF pulses to allow for shorter repetition time at high magnetic fields (B0≥7 T) and reduced SAR. The second pulse converts single- into triple-quantum coherences in regions containing sodium ions which are restricted in mobility. Since only single-quantum coherences can be detected, an image acquired after the second pulse is intrinsically single-quantum-filtered and can be used to generate a biexponential-weighted sodium image by a weighted subtraction with the spin-density-weighted image acquired between the pulses. The proposed sequence generates biexponential-weighted sodium images of in vivo human brain with 140% higher SNR than triple-quantum-filtered sodium images and 4% higher SNR than a biexponential-weighted sequence with three RF pulses at equal acquisition time and with 1/3 lower SAR. As SAR is reduced, accordingly repetition time can be spared to obtain even higher SNR-time efficiency. In comparison to a difference image generated from two images of a double-readout sequence, the proposed two-pulse sequence yields about 14% higher SNR. Our new two-pulse biexponential-weighted sequence allows for acquisition of full 3D data sets of the human brain in vivo with a nominal resolution of (5 mm)(3) in about 10 min.
Collapse
Affiliation(s)
- Nadia Benkhedah
- German Cancer Research Center (DKFZ), Department of Medical Physics in Radiology, Heidelberg, Germany
| | - Peter Bachert
- German Cancer Research Center (DKFZ), Department of Medical Physics in Radiology, Heidelberg, Germany
| | - Armin M Nagel
- German Cancer Research Center (DKFZ), Department of Medical Physics in Radiology, Heidelberg, Germany.
| |
Collapse
|
29
|
Liu C, Bai Y, Chen Y, Wang Y, Sottejeau Y, Liu L, Li X, Lingrel JB, Malhotra D, Cooper CJ, Shapiro JI, Xie ZJ, Tian J. Reduction of Na/K-ATPase potentiates marinobufagenin-induced cardiac dysfunction and myocyte apoptosis. J Biol Chem 2012; 287:16390-8. [PMID: 22451662 DOI: 10.1074/jbc.m111.304451] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Decreases in cardiac Na/K-ATPase have been documented in patients with heart failure. Reduction of Na/K-ATPase α1 also contributes to the deficiency in cardiac contractility in animal models. Our previous studies demonstrate that reduction of cellular Na/K-ATPase causes cell growth inhibition and cell death in renal proximal tubule cells. To test whether reduction of Na/K-ATPase in combination with increased cardiotonic steroids causes cardiac myocyte death and cardiac dysfunction, we examined heart function in Na/K-ATPase α1 heterozygote knock-out mice (α1(+/-)) in comparison to wild type (WT) littermates after infusion of marinobufagenin (MBG). Adult cardiac myocytes were also isolated from both WT and α1(+/-) mice for in vitro experiments. The results demonstrated that MBG infusion increased myocyte apoptosis and induced significant left ventricle dilation in α1(+/-) mice but not in their WT littermates. Mechanistically, it was found that in WT myocytes MBG activated the Src/Akt/mTOR signaling pathway, which further increased phosphorylation of ribosome S6 kinase (S6K) and BAD (Bcl-2-associated death promoter) and protected cells from apoptosis. In α1(+/-) myocytes, the basal level of phospho-BAD is higher compared with WT myocytes, but MBG failed to induce further activation of the mTOR pathway. Reduction of Na/K-ATPase also caused the activation of caspase 9 but not caspase 8 in these cells. Using cultures of neonatal cardiac myocytes, we demonstrated that inhibition of the mTOR pathway by rapamycin also enabled MBG to activate caspase 9 and induce myocyte apoptosis.
Collapse
Affiliation(s)
- Changxuan Liu
- Department of Physiology and Pharmacology, University of Toledo, Toledo, Ohio 43614, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Hanish Singh JC, Alagarsamy V, Diwan PV, Sathesh Kumar S, Nisha JC, Narsimha Reddy Y. Neuroprotective effect of Alpinia galanga (L.) fractions on Aβ(25-35) induced amnesia in mice. JOURNAL OF ETHNOPHARMACOLOGY 2011; 138:85-91. [PMID: 21911048 DOI: 10.1016/j.jep.2011.08.048] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 08/12/2011] [Accepted: 08/20/2011] [Indexed: 05/31/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The rhizomes of Alpinia galanga (L.) Willd (Zingiberaceae), a ginger substitute for flavouring food was traditionally used as nervine tonic and stimulant. AIM OF THE STUDY This investigation is designed to screen cognitive improvement of Alpinia galanga (AG) fractions in Alzheimer's type of amnesia in mice induced by Aβ((25-35)). MATERIALS AND METHODS Alzheimer's disease induced mice treated with fractions (n-hexane, chloroform and ethyl acetate) of AG in 200 and 400mg/kg. Neurotoxicity was induced by intracerebroventricular injection of Aβ((25-35)) on the 14th day of 21 days drug treatment. Open field and water maze were carried to determine habituation memory and hippocampal memory. Na(+)/K(+)-ATPase, acetylcholinesterase (AChE) and antioxidant enzymes (SOD, GPx, catalase and vitamin C) were determined in brain tissue homogenate to estimate the brain biochemical changes and its anti-amnesic potential with intensity of oxidative stress signaling. Further bioactive (chloroform) fraction was eluted through column chromatography to identify the lead molecules. RESULTS Increased habituation memory and decreased escape latency in behavioral parameter are the indicative of the cognitive enhancement after treatment with Alpinia galanga fractions. Increment in Na(+)/K(+)-ATPase and antioxidant activity depicts brain membrane integrity improvement and free radical scavenging property. AChE level was decreased to improve the cognition by enhancing cholinergic transmission. CONCLUSION Anti-amnesic effect was exerted by various fractions of Alpinia galanga. Among all fractions, preeminent neuroprotection was exerted by chloroform fraction, which has compound, 1'δ-1'-acetoxyeugenol acetate and it may be a potential therapeutic agent for Alzheimer's type of amnesia. These results further motivate us to explore the activity of lead compound's anti-amnesic effect on transgenic mice model of AD.
Collapse
Affiliation(s)
- J C Hanish Singh
- Department of Pharmacology, Lalitha College of Pharmacy, Ghatkesar, Gr. Hyderabad, Andhra Pradesh 501301, India
| | | | | | | | | | | |
Collapse
|
31
|
Omotayo TI, Rocha JBT, Ibukun EO, Kade IJ. Inorganic mercury interacts with thiols at the nucleotide and cationic binding sites of the ouabain-sensitive cerebral electrogenic sodium pump. Neurochem Int 2011; 58:776-84. [PMID: 21397648 DOI: 10.1016/j.neuint.2011.03.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 02/26/2011] [Accepted: 03/04/2011] [Indexed: 11/17/2022]
Abstract
The molecular events leading to neuronal dysfunction often associated with mercury toxicity can be complex and is yet to be fully elucidated. Hence, the present study sought to evaluate the interaction of inorganic mercury (Hg(2+)) with the ouabain-sensitive electrogenic pump in partially purified mammalian brain membrane preparations. The results show that Hg(2+) significantly inhibited the transmembrane enzyme in a concentration dependent manner. In addition, Hg(2+) exerts its inhibitory effect on the activity of the enzyme by interacting with groups at the adenosine triphosphate (ATP), Na(+) and K(+) binding sites. However, preincubation of the enzyme with exogenous monothiols, cysteine, prevented the inhibition of Hg(2+) on the pump's activity suggesting that Hg(2+) may be interacting with the thiols at the nucleotide (ATP) and cationic (Na(+) and K(+)) binding sites. In fact, our data show that Hg(2+) oxidizes sulphydryl groups in cysteine in a time dependent fashion in vitro. Finally, we speculate that the small molecular volume of Hg(2+) in comparison with the substrates (ATP, Na(+) and K(+)) of sodium pump, its possibly high reactivity and strong affinity for thiols may account for its high toxicity towards the membrane bound ouabain-sensitive electrogenic pump.
Collapse
Affiliation(s)
- T I Omotayo
- Department of Biochemistry, Federal University of Technology, Akure, Ondo State, Nigeria
| | | | | | | |
Collapse
|
32
|
Corona JC, Gimenez-Cassina A, Lim F, Díaz-Nido J. Hexokinase II gene transfer protects against neurodegeneration in the rotenone and MPTP mouse models of Parkinson's disease. J Neurosci Res 2010; 88:1943-50. [PMID: 20143419 DOI: 10.1002/jnr.22357] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A typical feature of Parkinson's disease is the progressive loss of dopaminergic neurons in the substantia nigra, in which inhibition of mitochondrial complex I activity may play an important role. Rotenone or 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) inhibit the mitochondrial complex I and they cause the death of substantia nigra dopaminergic neurons, thereby providing acute murine models of Parkinson's disease. We have found that increasing mitochondrial hexokinase II activity can prevent cell death in neuronal cultures treated with rotenone. As a result, we have studied the effects of hexokinase II gene transfer in vivo using a herpes simplex virus type 1 (HSV-1) amplicon vector. The placHK2 amplicon vector was injected into substantia nigra of mice that were subsequently administered rotenone or MPTP. Overexpression of hexokinase II prevented both rotenone and MPTP-induced dopaminergic neuronal cell death, as well as reducing the associated motor defects. Our results provide the first proof-of-principle that hexokinase II protects against dopaminergic neurodegeneration in vivo, emphasizing the role of this enzyme in promoting neuronal survival. Thus, the increase of hexokinase II expression by gene transfer or other means represents a promising approach to treat Parkinson's and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Juan Carlos Corona
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | | |
Collapse
|
33
|
Abstract
Tumor progression involves the acquisition of properties which include growth-factor independent cell proliferation, failure of inhibition by growth-inhibitory signals, ability to invade surrounding tissues, and to evade apoptosis, etc. Characterization of the profile or molecular signature of the tumor may permit the development of rational therapies that target the aberrant pathways. Rapidly growing tumor cells are usually associated with a high rates of glycolysis and in these cells, it may be advantageous to exploit this pathway which most likely is required for optimal synthetic needs. Combinatorial therapeutic agents which target the growth factor signal transduction pathways as well as apoptotic signaling pathways provide an opportunity for maximal therapeutic benefit.
Collapse
Affiliation(s)
- Charles E Wenner
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Buffalo, New York 14263, USA.
| |
Collapse
|
34
|
Jovanova-Nesic K, Jovicic S, Sovilj M, Spector NH. Magnetic brain stimulation upregulates adhesion and prevents Eae: MMP-2, ICAM-1, and VCAM-1 in the choroid plexus as a target. Int J Neurosci 2009; 119:1399-418. [PMID: 19922364 DOI: 10.1080/00207450802324564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Clinical signs appearance and significant increases of ICAM-1 and MMP-2 expressions with the clusters of VCAM-1(+) immunoreactivity in the choroids plexus epithelium to transferred anti-myelin oligodendroglial antibodies into the third brain ventricle, indicate important role of choroids plexus in the induction of acute experimental autoimmune encephalomyelitis (EAE). Magnetic brain stimulation with AKMA micro-magnet flux density of 60 miliTesla, 5 mm in diameter, implanted upon the pineal gland (PG), immediately after antibody injection, significantly decreases the expression of MMP-2 and ICAM-1 in the choroids plexus of the rat brain and abruptly suppresses the induction of acute EAE.
Collapse
Affiliation(s)
- K Jovanova-Nesic
- Immunology Research Center Branislav Jankovic, 45811152 Belgrade, Serbia.
| | | | | | | |
Collapse
|
35
|
Jovanova-Nesic K, Eric-Jovicic M, Spector NH. MAGNETIC STIMULATION OF THE BRAIN INCREASE Na+, K+-ATPase ACTIVITY DECREASED BY INJECTION OF AlCl3INTO NUCLEUS BASALIS MAGNOCELLULARIS OF RATS. Int J Neurosci 2009; 116:681-95. [PMID: 16753895 DOI: 10.1080/00207450600674830] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
This article reports here on the influence of the static magnetic fields (MFs), locally applied to the brain area, on Na, K-ATPase activity in the rat with lesioned nucleus basalis magnocellularis (NBM) by intracerebral injection of 5 microl, 1% AlCl3 into the nucleus. Two AKMA micromagnets (M) flux density of 60 miliTesla, 5 mm in diameter, were bilaterally implanted with "N" polarity facing down to the cranial bones in the vicinity of the pineal gland (PG), immediately after the lesioning of NBM, during the same operation procedure. Ten days after the lesions of NBM, Na, K-ATPase activity on the erythrocyte membranes in the peripheral blood, measured spectrophotometrically, was completely inhibited. Magnetic stimulation (60 mT) of the brain during the 10 days significantly increased Na, K-ATPase activity on the erythrocyte membranes of rats with lesioned NBM. This results suggests that altered by lesions Na, K-ATPase activity in an experimental model of Alzheimer's disease might be ameliorated by magnetic stimulation of the brain.
Collapse
|
36
|
Carageorgiou H, Sideris AC, Messari I, Liakou CI, Tsakiris S. The effects of rivastigmine plus selegiline on brain acetylcholinesterase, (Na, K)-, Mg-ATPase activities, antioxidant status, and learning performance of aged rats. Neuropsychiatr Dis Treat 2008; 4:687-99. [PMID: 19043511 PMCID: PMC2536534 DOI: 10.2147/ndt.s3272] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
UNLABELLED We investigated the effects of rivastigmine (a cholinesterase inhibitor) and selegiline ((-)deprenyl, an irreversible inhibitor of monoamineoxidase-B), alone and in combination, on brain acetylcholinesterase (AChE), (Na(+), K(+))-, Mg(2+)-ATPase activities, total antioxidant status (TAS), and learning performance, after long-term drug administration in aged male rats. The possible relationship between the biochemical and behavioral parameters was evaluated. METHODS Aged rats were treated (for 36 days) with rivastigmine (0.3 mg/kg rat/day ip), selegiline (0.25 mg/kg rat/day im), rivastigmine plus selegiline in the same doses and way of administration as separately. Aged and adult control groups received NaCl 0.9% 0.5 ml ip. RESULTS TAS was lower in aged than in adult rats, rivastigmine alone does not affect TAS, decreases AChE activity, increases (Na(+), K(+))-ATPase and Mg(2+)-ATPase activity of aged rat brain and improves cognitive performance. Selegiline alone decreases free radical production and increases AChE activity and (Na(+), K(+))-ATPase activity, improving cognitive performance as well. In the combination: rivastigmine seems to cancel selegiline action on TAS and AChE activity, while it has additive effect on (Na(+), K(+))-ATPase activity. In the case of Mg(2+)-ATPase selegiline appears to attenuate rivastigmine activity. No statistically significant difference was observed in the cognitive performance. CONCLUSION Reduced TAS, AChE activity and learning performance was observed in old rats. Both rivastigmine and selesiline alone improved performance, although they influenced the biochemical parameters in a different way. The combination of the two drugs did not affect learning performance.
Collapse
Affiliation(s)
- Haris Carageorgiou
- Department of Pharmacology, Medical School, University of Athens Athens, Greece
| | | | | | | | | |
Collapse
|
37
|
Studzinski CM, MacKay WA, Beckett TL, Henderson ST, Murphy MP, Sullivan PG, Burnham WM. Induction of ketosis may improve mitochondrial function and decrease steady-state amyloid-β precursor protein (APP) levels in the aged dog. Brain Res 2008; 1226:209-17. [DOI: 10.1016/j.brainres.2008.06.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Revised: 06/03/2008] [Accepted: 06/03/2008] [Indexed: 01/06/2023]
|
38
|
Sultana R, Butterfield DA. Alterations of some membrane transport proteins in Alzheimer's disease: role of amyloid β-peptide. ACTA ACUST UNITED AC 2008; 4:36-41. [DOI: 10.1039/b715278g] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
39
|
Abstract
Brain cells are highly energy dependent for maintaining ion homeostasis during high metabolic activity. During active periods, full mitochondrial function is essential to generate ATP from electrons that originate with the oxidation of NADH. Decreasing brain metabolism is a significant cause of cognitive abnormalities of Alzheimer disease (AD), but it remains uncertain whether this is the cause of further pathology or whether synaptic loss results in a lower energy demand. Synapses are the first to show pathological symptoms in AD before the onset of clinical symptoms. Because synaptic function has high energy demands, interruption in mitochondrial energy supply could be the major factor in synaptic failure in AD. A newly discovered age-related decline in neuronal NADH and redox ratio may jeopardize this function. Mitochondrial dehydrogenases and several mutations affecting energy transfer are frequently altered in aging and AD. Thus, with the accumulation of genetic defects in mitochondria at the level of energy transfer, the issue of neuronal susceptibility to damage as a function of age and age-related disease becomes important. In an aging rat neuron model, mitochondria are both chronically depolarized and produce more reactive oxygen species with age. These concepts suggest that multiple treatment targets may be needed to reverse this multifactorial disease. This review summarizes new insights based on the interaction of mitoenergetic failure, glutamate excitotoxicity, and amyloid toxicity in the exacerbation of AD.
Collapse
Affiliation(s)
- Mordhwaj S Parihar
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62794-9626, USA
| | | |
Collapse
|
40
|
Mantha AK, Moorthy K, Cowsik SM, Baquer NZ. Membrane Associated Functions of Neurokinin B (NKB) on Aβ (25–35) Induced Toxicity in Aging Rat Brain Synaptosomes. Biogerontology 2006; 7:19-33. [PMID: 16518717 DOI: 10.1007/s10522-005-6044-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2005] [Accepted: 12/14/2005] [Indexed: 11/26/2022]
Abstract
The effect of different concentrations (0.1-5 microM) of neurokinin B (NKB) and Abeta (25-35) on acetylcholine esterase (AChE), Na(+)-K(+) ATPase and membrane fluidity (DPH anisotropy) were investigated in rat brain synaptosomes of 3, 9, 18 and 30 months old. An age dependent decrease was observed for all the three parameters studied. An in vitro incubation of isolated brain synaptosomes with Abeta (25-35) showed toxic effects on all the parameters studied and the peptide had concentration and age dependent effects, while NKB showed stimulating effect on the parameters and the combined NKB+Abeta (25-35) incubations showed a partial reversal effect as compared to the Abeta (25-35) alone. Thus, the results suggest a membrane mediated function for NKB and its role in neuromodulation, neuroprotection and antioxidant property against Abeta (25-35) induced toxicity in aging brain functions.
Collapse
Affiliation(s)
- Anil K Mantha
- Molecular and Structural Biophysics Laboratory, School of Life Sciences, Jawaharlal Nehru University, 110067, New Delhi, India
| | | | | | | |
Collapse
|
41
|
Chen JQ, Contreras RG, Wang R, Fernandez SV, Shoshani L, Russo IH, Cereijido M, Russo J. Sodium/potasium ATPase (Na+, K+-ATPase) and ouabain/related cardiac glycosides: a new paradigm for development of anti- breast cancer drugs? Breast Cancer Res Treat 2005; 96:1-15. [PMID: 16322895 DOI: 10.1007/s10549-005-9053-3] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2005] [Accepted: 08/11/2005] [Indexed: 11/30/2022]
Abstract
Prolonged exposure to 17beta-estradiol (E2) is a key etiological factor for human breast cancer. The biological effects and carcinogenic effects of E2 are mediated via estrogen receptors (ERs), ERalpha and ERbeta. Anti-estrogens, e.g. tamoxifen, and aromatase inhibitors have been used to treat ER-positive breast cancer. While anti-estrogen therapy is initially successful, a major problem is that most tumors develop resistance and the disease ultimately progresses, pointing to the need of developing alternative drugs targeting to other critical targets in breast cancer cells. We have identified that Na+, K+-ATPase, a plasma membrane ion pump, has unique/valuable properties that could be used as a potentially important target for breast cancer treatment: (a) it is a key player of cell adhesion and is involved in cancer progression; (b) it serves as a versatile signal transducer and is a target for a number of hormones including estrogens and (d) its aberrant expression and activity are implicated in the development and progression of breast cancer. There are several lines of evidence indicating that ouabain and related digitalis (the potent inhibitors of Na+, K+-ATPase) possess potent anti-breast cancer activity. While it is not clear how the suggested anti-cancer activity of these drugs work, several observations point to ouabain and digitalis as being potential ER antagonists. We critically reviewed many lines of evidence and postulated a novel concept that Na+, K+-ATPase in combination with ERs could be important targets of anti-breast cancer drugs. Modulators, e.g. ouabain and related digitalis could be useful to develop valuable anti-breast cancer drugs as both Na+, K+-ATPase inhibitors and ER antagonists.
Collapse
Affiliation(s)
- Jin-Qiang Chen
- Breast Cancer Research Laboratory, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Kawamoto EM, Munhoz CD, Glezer I, Bahia VS, Caramelli P, Nitrini R, Gorjão R, Curi R, Scavone C, Marcourakis T. Oxidative state in platelets and erythrocytes in aging and Alzheimer's disease. Neurobiol Aging 2005; 26:857-64. [PMID: 15718044 DOI: 10.1016/j.neurobiolaging.2004.08.011] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2004] [Revised: 07/19/2004] [Accepted: 08/12/2004] [Indexed: 11/29/2022]
Abstract
Several studies have shown involvement of peroxynitrite anion, a potent oxidative agent, in Alzheimer's disease (AD) neuropathology. Herein, we assessed in platelets and erythrocytes of AD patients, age-matched and young adults controls: thiobarbituric acid-reactive substances (TBARS) production; superoxide dismutase (SOD), nitric oxide synthase (NOS) and Na,K-ATPase activities; cyclic GMP (cGMP) content, both basal and after sodium nitroprusside (SNP) stimulation. Aging was associated with an increase in TBARS production and NOS activity, a decrease in basal cGMP content and no change in SOD and Na,K-ATPase activities. AD patients, compared to aged controls, have: increase in TBARS production and in NOS, SOD and Na,K-ATPase activities but no alteration in basal cGMP content. SNP increased cGMP platelets production in all groups. In conclusion, we demonstrated in platelets and erythrocytes a disruption in systemic modulation of oxidative stress in aging and with more intensity in AD.
Collapse
Affiliation(s)
- Elisa Mitiko Kawamoto
- Department of Pharmacology, School of Medicine, University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Dickey CA, Gordon MN, Wilcock DM, Herber DL, Freeman MJ, Morgan D. Dysregulation of Na+/K+ ATPase by amyloid in APP+PS1 transgenic mice. BMC Neurosci 2005; 6:7. [PMID: 15689237 PMCID: PMC549198 DOI: 10.1186/1471-2202-6-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2004] [Accepted: 02/02/2005] [Indexed: 12/05/2022] Open
Abstract
Background The pathology of Alzheimer's disease (AD) is comprised of extracellular amyloid plaques, intracellular tau tangles, dystrophic neurites and neurodegeneration. The mechanisms by which these various pathological features arise are under intense investigation. Here, expanding upon pilot gene expression studies, we have further analyzed the relationship between Na+/K+ ATPase and amyloid using APP+PS1 transgenic mice, a model that develops amyloid plaques and memory deficits in the absence of tangle formation and neuronal or synaptic loss. Results We report that in addition to decreased mRNA expression, there was decreased overall Na+/K+ ATPase enzyme activity in the amyloid-containing hippocampi of the APP+PS1 mice (although not in the amyloid-free cerebellum). In addition, dual immunolabeling revealed an absence of Na+/K+ ATPase staining in a zone surrounding congophilic plaques that was occupied by dystrophic neurites. We also demonstrate that cerebral Na+/K+ ATPase activity can be directly inhibited by high concentrations of soluble Aβ. Conclusions The data suggest that the reductions in Na+/K+ ATPase activity in Alzheimer tissue may not be purely secondary to neuronal loss, but may results from direct effects of amyloid on this enzyme. This disruption of ion homeostasis and osmotic balance may interfere with normal electrotonic properties of dendrites, blocking intraneuronal signal processing, and contribute to neuritic dystrophia. These results suggest that therapies aimed at enhancing Na+/K+ ATPase activity in AD may improve symptoms and/or delay disease progression.
Collapse
Affiliation(s)
- Chad A Dickey
- Alzheimer's Disease Research Laboratory, Department of Pharmacology, University of South Florida, Tampa, USA
| | - Marcia N Gordon
- Alzheimer's Disease Research Laboratory, Department of Pharmacology, University of South Florida, Tampa, USA
| | - Donna M Wilcock
- Alzheimer's Disease Research Laboratory, Department of Pharmacology, University of South Florida, Tampa, USA
| | - Donna L Herber
- Alzheimer's Disease Research Laboratory, Department of Pharmacology, University of South Florida, Tampa, USA
| | - Melissa J Freeman
- Alzheimer's Disease Research Laboratory, Department of Pharmacology, University of South Florida, Tampa, USA
| | - Dave Morgan
- Alzheimer's Disease Research Laboratory, Department of Pharmacology, University of South Florida, Tampa, USA
| |
Collapse
|
44
|
Gu QB, Zhao JX, Fei J, Schwarz W. Modulation of Na(+),K(+) pumping and neurotransmitter uptake by beta-amyloid. Neuroscience 2004; 126:61-7. [PMID: 15145073 DOI: 10.1016/j.neuroscience.2004.03.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2004] [Indexed: 01/19/2023]
Abstract
Micromolar concentrations of beta-amyloid (Abeta), a 40/42-amino-acid-long proteolytic fragment (Abeta(1-40/42)) of the amyloid precursor protein, was shown previously to play a crucial role in pathogenesis of Alzheimer's disease. We used the Xenopus oocyte expression system to investigate specific effects of micromolar concentrations of Abeta(1-42) on the neurotransmitter transporters for gamma-aminobutyric acid (GABA), GAT1, and for the excitatory amino acid glutamate, EAAC1, which are driven by the transmembrane Na(+) gradient that is regulated by the Na(+),K(+)-ATPase. Brief treatment with Abeta(1-42), up to 80 min, leads to a significant inhibition of ion translocation by the Na(+),K(+)-ATPase (30-40%); also glutamate uptake is inhibited (20%) while GABA uptake is not affected. Since reduced glutamate uptake will result in elevated, neurotoxic concentrations of extracellular glutamate, we investigated the effects of Abeta(1-42) and the smaller fragments, Abeta(12-28) and Abeta(25-35), on EAAC1 in more detail. Prolonged incubation in 1 microM Abeta(1-42) leads to further, strong inhibition of glutamate uptake and EAAC1-mediated current (after 4 h inhibition amounts to more than 80%). Abeta(12-28) is less effective with 50% inhibition after 4 h of incubation at 20 microM. Abeta(1-42) and Abeta(12-28) affect EAAC1-mediated current to a similar extent as the rate of glutamate uptake. The effects on EAAC1-mediated current are irreversible if Abeta were applied for longer time periods. Peptides directly microinjected into the oocyte are ineffective suggesting that the observed effect were mediated by extracellular proteins. Abeta(25-35) hardly affects EAAC1-mediated current or glutamate uptake. The results demonstrate that Abeta specifically inhibits the Na(+),K(+) pump and EAAC1. The domain between amino acids 12 and 28 of Abeta seems to play a crucial role for inhibition of EAAC1. The inhibition of EAAC1 by neurotoxic, elevated extracellular glutamate levels may contribute to Alzheimer's pathogenesis.
Collapse
Affiliation(s)
- Q B Gu
- Shanghai Institute of Biochemistry and Cell Biology, 320 YueYang Road, 200031 Shanghai, China
| | | | | | | |
Collapse
|
45
|
Abstract
Impairments of glucose and mitochondrial function are important causes of brain dysfunction and therefore of brain disease. Abnormalities have been found in association with disease of the nervous system in most of the components of glucose/mitochondrial metabolism. In many, molecular genetic abnormalities have been defined. Brain glucose oxidation is abnormal in common diseases of the nervous system, including Alzheimer disease and other dementias, Parkinson disease, delirium, probably schizophrenia and other psychoses, and of course cerebrovascular disease. Defects in a single component and even a single mutation can be associated with different clinical phenotypes. The same clinical phenotype can result from different genotypes. The complex relationship between biological abnormality in brain glucose utilization and clinical disorder is similar to that in other disorders that have been intensively studied at the genetic level. Genes for components of the pathways of brain glucose oxidation are good candidate genes for disease of the brain. Preliminary data support the proposal that treatments to normalize abnormalities in brain glucose oxidation may benefit many patients with common brain diseases.
Collapse
Affiliation(s)
- John P Blass
- Weill Medical College of Cornell University, Burke Medical Research Institute White Plains, New York 10605, USA
| |
Collapse
|
46
|
Kairane C, Roots K, Uusma T, Bogdanovic N, Karelson E, Kõks S, Zilmer M. Regulation of the frontocortical sodium pump by Na+ in Alzheimer's disease: difference from the age-matched control but similarity to the rat model. FEBS Lett 2002; 531:241-4. [PMID: 12417319 DOI: 10.1016/s0014-5793(02)03510-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The Na+ and K+ dependence of the frontocortical Na,K-ATPase in Alzheimer's disease (AD) was compared with that in human control (Co) and rat AD model. In AD, the relationship between the Na/K ratio and the Na,K-ATPase activity showed noticeable left-shift with three-fold increase in the enzyme affinity for Na+ (K(0.5)=10 and 30 mM in AD and Co, respectively). The Na+ dependence of the enzyme in AD showed two different Hill coefficients (n(H)), 1.1 and 0.3, whereas the Co value of n(H) was higher (1.4). The rat AD model generated by ibotenic acid revealed a Na+ dependence similar to AD. The K+ dependence of the Na,K-ATPase showed no significant difference in AD and Co. Compared with Co, AD produced a shift in the break of the Na,K-ATPase Arrhenius plot, suggesting remarkable alterations in the enzyme lipid environment. Our findings support the hypothesis that dysfunction of the Na,K-ATPase in AD is provoked by altered Na+ dependence of the enzyme. An impairment of the pump functionality might serve as an early mechanism of AD that should be interrupted by selective pharmacological agents.
Collapse
Affiliation(s)
- Czeslava Kairane
- Department of Biochemistry, Tartu University, Ravila 19, Tartu, Estonia
| | | | | | | | | | | | | |
Collapse
|
47
|
dos Reis EA, de Oliveira LS, Lamers ML, Netto CA, Wyse ATDS. Arginine administration inhibits hippocampal Na+,K+-ATPase activity and impairs retention of an inhibitory avoidance task in rats. Brain Res 2002; 951:151-7. [PMID: 12270492 DOI: 10.1016/s0006-8993(02)03077-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
In the present study we investigated the effect of acute administration of L-arginine (Arg) on hippocampal Na(+),K(+)-ATPase activity and on retrieval of step-down inhibitory avoidance in adult rats. The action of L-NAME on the effects produced by Arg was also tested. Sixty-day-old rats were treated with a single intraperitoneal injection of saline (group I, control), arginine (0.8 g/kg) (group II), L-NAME (2 mg/kg) (group III) or arginine (0.8 g/kg) plus L-NAME (2 mg/kg) (group IV). Na(+),K(+)-ATPase activity was significantly reduced in arginine-treated rats; this effect was prevented by L-NAME. Retrieval of the avoidance task was also significantly impaired by arginine, whereas the simultaneous injection of L-NAME prevented this effect. Present data strongly indicate that in vivo Arg administration reduces both Na(+),K(+)-ATPase activity and memory modulation in rats probably through NO formation.
Collapse
Affiliation(s)
- Eleonora Araújo dos Reis
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003, RS, Porto Alegre, Brazil
| | | | | | | | | |
Collapse
|
48
|
Koçak H, Oner P, Oztaş B. Comparison of the activities of Na(+),K(+)-ATPase in brains of rats at different ages. Gerontology 2002; 48:279-81. [PMID: 12169791 DOI: 10.1159/000065249] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Na(+),K(+)-ATPase is known to be responsible for the ionic homeostasis in excitable tissues. The energy cost of Na(+),K(+)-ATPase activity is increased in the active brain, so it would be important to ascertain whether defects in brain metabolism in aging are associated with changes in the properties of Na(+),K(+)-ATPase. OBJECTIVE The aim of this study was to investigate the influence of age on the Na(+),K(+)-ATPase activity in developing rat brains from the age of 1 day to 24-28 months. METHODS Crude microsomal preparations were obtained from the brains of newborn (n = 8), 18-day-old (n = 8), 4- to 5-month-old (n = 12), and 24- to 28-month-old (n = 14) rats. Then the ATPase activity was measured and expressed as micromoles of inorganic phosphorus released per milligram of protein per hour. RESULTS The increased tendency in brain Na(+),K(+)-ATPase activity from newborn to 18 days of age suggested that the Na pump is mature soon after birth. No significant differences could be detected in the enzyme activity between newborn and adult rats. In contrast, the Na(+),K(+)-ATPase activity in aged rat brains was found to be significantly lower than in the other age groups of rats tested (p < 0.001). CONCLUSION Our results suggest that aging-induced inhibitions in the brain Na(+),K(+)-ATPase activity may be implicated in the depression of neuronal excitability and in the age-related impairments of cognitive functions.
Collapse
Affiliation(s)
- Hikmet Koçak
- Department of Biochemistry, Istanbul Faculty of Medicine, University of Istanbul, Turkey.
| | | | | |
Collapse
|
49
|
Blass JP, Sheu RK, Gibson GE. Inherent abnormalities in energy metabolism in Alzheimer disease. Interaction with cerebrovascular compromise. Ann N Y Acad Sci 2000; 903:204-21. [PMID: 10818509 DOI: 10.1111/j.1749-6632.2000.tb06370.x] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Alzheimer disease (AD) is a form of the dementia syndrome. AD appears to have a variety of fundamental etiologies that lead to the neuropathological manifestations which define the disease. Patients who are at high risk to develop AD typically show impairments of cerebral metabolic rate in vivo even before they show any evidence of the clinical disease on neuropsychological, electrophysiological, and neuroimaging examinations. Therefore, impairment in energy metabolism in AD can not be attributed to loss of brain substance or to electrophysiological abnormalities. Among the characteristic abnormalities in the AD brain are deficiencies in several enzyme complexes which participate in the mitochondrial oxidation of substrates to yield energy. There include the pyruvate dehydrogenase complex (PDHC), the alpha-ketoglutarate dehydrogenase complex (KGDHC), and Complex IV of the electron transport chain (COX). The deficiency of KGDHC may be due to a mixture of causes including damage by free radicals and perhaps to genetic variation in the DLST gene encoding the core protein of this complex. Inherent impairment of glucose oxidation by the AD brain may reasonably be expected to interact synergistically with an impaired supply of oxygen and glucose to the AD brain, in causing brain damage. These considerations lead to the hypothesis that cerebrovascular compromise and inherent abnormalities in the brain's ability to oxidize substrates can interact to favor the development of AD, in individuals who are genetically predisposed to develop neuritic plaques.
Collapse
Affiliation(s)
- J P Blass
- Dementia Research Service, Burke Medical Research Institute, Weill Medical College of Cornell University, White Plains, New York 10605, USA.
| | | | | |
Collapse
|
50
|
Hattori N, Kitagawa K, Higashida T, Yagyu K, Shimohama S, Wataya T, Perry G, Smith MA, Inagaki C. CI-ATPase and Na+/K(+)-ATPase activities in Alzheimer's disease brains. Neurosci Lett 1998; 254:141-4. [PMID: 10214977 DOI: 10.1016/s0304-3940(98)00654-5] [Citation(s) in RCA: 104] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The enzyme activities and the protein levels of Cl(-)-ATPase and Na+/K(+)-ATPase were examined in Alzheimer's disease (AD) brains. Cl(-)-ATPase and Na+/K(+)-ATPase activities in AD brains (n = 13) were significantly lower than those in age-matched control brains (n = 12). In contrast, there was no significant difference in anion-insensitive Mg2(+)-ATPase activity between the two groups. Western blot analysis revealed that the protein levels of Cl(-)-ATPase, Na+/K(+)-ATPase and neuron specific Na+/K(+)-ATPase alpha3 isoform were also significantly reduced in AD brains, while the amount of protein disulfide isomerase, one of the house keeping membrane proteins, was not different between the two groups. The data first demonstrated that Cl(-)-ATPase and Na+/K(+)-ATPase are selectively impaired in AD brains, which may reduce the gradients of Na(+), K(+) and Cl(-) across the cell membranes to cause excitotoxic cellular response and the resulting neuronal death.
Collapse
Affiliation(s)
- N Hattori
- Department of Pharmacology, Kansai Medical University, Moriguchi-shi, Osaka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|