1
|
Sidhu RK, Maparu K, Singh S, Aran KR. Unveiling the role of Na⁺/K⁺-ATPase pump: neurodegenerative mechanisms and therapeutic horizons. Pharmacol Rep 2025:10.1007/s43440-025-00717-6. [PMID: 40117043 DOI: 10.1007/s43440-025-00717-6] [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: 12/27/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
Sodium and potassium-activated adenosine 5'-triphosphatase (Na+/K+-ATPase) is a pivotal plasma membrane enzyme involved in neuronal activity and cellular homeostasis. The dysregulation of these enzymes has been implicated in a spectrum of neurodegenerative disorders like Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and neurodevelopmental disorders including autism spectrum disorder (ASD), psychiatric disorders such as schizophrenia, and neurological problems like epilepsy. A hallmark of these disorders is the gradual loss of neuronal integrity and function, often exacerbated by protein accumulation within brain cells. This review delves into the multifaceted role of Na+/K+-ATPase dysfunction in driving oxidative stress, excitotoxicity, and neuroinflammation, contributing to synaptic and neuronal damage. Emerging therapeutic strategies, such as gene therapy and developing isoform-specific enzyme modulators, offer promising avenues for targeted interventions. Furthermore, this review highlights innovative research directions, including the role of Na⁺/K⁺-ATPase in synaptic plasticity, the identification of endogenous regulators, and its contribution to neuroinflammatory pathways. Personalized medicine and advanced gene-editing technologies are positioned as transformative tools for crafting safer and more precise therapies tailored to individual patients. This comprehensive exploration underscores the enzyme's therapeutic potential and sets the stage for developing novel targeted strategies to mitigate the burden of Na⁺/K⁺-ATPase-linked neurological disorders.
Collapse
Affiliation(s)
- Ramandeep Kaur Sidhu
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Kousik Maparu
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Shamsher Singh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Khadga Raj Aran
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
2
|
Singh A, Singh L. Acyclic sesquiterpenes nerolidol and farnesol: mechanistic insights into their neuroprotective potential. Pharmacol Rep 2025; 77:31-42. [PMID: 39436564 DOI: 10.1007/s43440-024-00672-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/23/2024]
Abstract
Sesquiterpenes are a class of organic compounds found in plants, fungi, and some insects. They are characterized by the presence of three isoprene units, resulting in a molecular formula that typically contains 15 carbon atoms (C₁₅H₂₄). Nerolidol and farnesol are both sesquiterpene alcohols present in the essential oils of numerous plants. They have drawn attention due to their potential neuroprotective properties. Nerolidol and farnesol are structural isomers, specifically geometric isomers, haring the same molecular formula (C₁₅H₂₄O) but differing in the spatial arrangement of their atoms. This variation in structure may contribute to their distinct biological activities. Scientific evidence suggests that nerolidol and farnesol exhibit antioxidant and anti-inflammatory characteristics which are crucial for neuroprotection. Nerolidol has been specifically noted for its ability to alleviate conditions such as Alzheimer's disease, Parkinson's disease, encephalomyelitis, depression, and anxiety by modulating inflammatory and oxidative stress pathways. Moreover, research indicates that both nerolidol and farnesol may modulate the Nrf-2/HO-1 antioxidant signaling pathway to mitigate oxidative stress-induced neurological damage. Activation of Nrf-2/HO-1 signaling cascade promotes cell survival and enhances the brain's ability to resist various insults. Nerolidol has also been reported to alleviate neuroinflammation by inhibiting the TLR-4/NF-κB and COX-2/NF-κB inflammatory signaling pathway. Besides, this nerolidol also modulates BDNF/TrkB/CREB signaling pathway to improve neuronal health. To date, limited research has delved into the anti-inflammatory properties of farnesol concerning neurodegenerative diseases. Further investigation is warranted to comprehensively elucidate the mechanisms underlying its action and potential therapeutic uses in neuroprotection. Initial observations indicate that farnesol exhibits promising prospects as a natural agent for safeguarding brain functions. Henceforth, drawing upon existing literature elucidating the neuroprotective attributes of nerolidol and farnesol, the current review endeavors to provide a detailed analysis of their mechanistic underpinnings in neuroprotection.
Collapse
Affiliation(s)
- Anish Singh
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, 140413, India
| | - Lovedeep Singh
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, 140413, India.
| |
Collapse
|
3
|
Yuan Y, Yu L, Zhuang X, Wen D, He J, Hong J, Xie J, Ling S, Du X, Chen W, Wang X. Drosophila models used to simulate human ATP1A1 gene mutations that cause Charcot-Marie-Tooth type 2 disease and refractory seizures. Neural Regen Res 2025; 20:265-276. [PMID: 38767491 PMCID: PMC11246156 DOI: 10.4103/1673-5374.391302] [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: 04/25/2023] [Revised: 09/21/2023] [Accepted: 11/06/2023] [Indexed: 05/22/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202501000-00034/figure1/v/2024-05-14T021156Z/r/image-tiff Certain amino acids changes in the human Na+/K+-ATPase pump, ATPase Na+/K+ transporting subunit alpha 1 (ATP1A1), cause Charcot-Marie-Tooth disease type 2 (CMT2) disease and refractory seizures. To develop in vivo models to study the role of Na+/K+-ATPase in these diseases, we modified the Drosophila gene homolog, Atpα, to mimic the human ATP1A1 gene mutations that cause CMT2. Mutations located within the helical linker region of human ATP1A1 (I592T, A597T, P600T, and D601F) were simultaneously introduced into endogenous DrosophilaAtpα by CRISPR/Cas9-mediated genome editing, generating the AtpαTTTF model. In addition, the same strategy was used to generate the corresponding single point mutations in flies (AtpαI571T, AtpαA576T, AtpαP579T, and AtpαD580F). Moreover, a deletion mutation (Atpαmut) that causes premature termination of translation was generated as a positive control. Of these alleles, we found two that could be maintained as homozygotes (AtpαI571T and AtpαP579T). Three alleles (AtpαA576T, AtpαP579 and AtpαD580F) can form heterozygotes with the Atpαmut allele. We found that the Atpα allele carrying these CMT2-associated mutations showed differential phenotypes in Drosophila. Flies heterozygous for AtpαTTTF mutations have motor performance defects, a reduced lifespan, seizures, and an abnormal neuronal morphology. These Drosophila models will provide a new platform for studying the function and regulation of the sodium-potassium pump.
Collapse
Affiliation(s)
- Yao Yuan
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian Province, China
| | - Lingqi Yu
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian Province, China
| | - Xudong Zhuang
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, Fujian Province, China
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Dongjing Wen
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian Province, China
| | - Jin He
- Department of Neurology and Institute of Neurology of The First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jingmei Hong
- Department of Neurology and Institute of Neurology of The First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jiayu Xie
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian Province, China
| | - Shengan Ling
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian Province, China
| | - Xiaoyue Du
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian Province, China
| | - Wenfeng Chen
- Institute of Life Sciences, College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian Province, China
| | - Xinrui Wang
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-human Primate (Fujian Maternity and Child Health Hospital), Fuzhou, Fujian Province, China
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
4
|
Huang S, Dong W, Lin X, Bian J. Na+/K+-ATPase: ion pump, signal transducer, or cytoprotective protein, and novel biological functions. Neural Regen Res 2024; 19:2684-2697. [PMID: 38595287 PMCID: PMC11168508 DOI: 10.4103/nrr.nrr-d-23-01175] [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: 07/11/2023] [Revised: 10/23/2023] [Accepted: 12/09/2023] [Indexed: 04/11/2024] Open
Abstract
Na+/K+-ATPase is a transmembrane protein that has important roles in the maintenance of electrochemical gradients across cell membranes by transporting three Na+ out of and two K+ into cells. Additionally, Na+/K+-ATPase participates in Ca2+-signaling transduction and neurotransmitter release by coordinating the ion concentration gradient across the cell membrane. Na+/K+-ATPase works synergistically with multiple ion channels in the cell membrane to form a dynamic network of ion homeostatic regulation and affects cellular communication by regulating chemical signals and the ion balance among different types of cells. Therefore, it is not surprising that Na+/K+-ATPase dysfunction has emerged as a risk factor for a variety of neurological diseases. However, published studies have so far only elucidated the important roles of Na+/K+-ATPase dysfunction in disease development, and we are lacking detailed mechanisms to clarify how Na+/K+-ATPase affects cell function. Our recent studies revealed that membrane loss of Na+/K+-ATPase is a key mechanism in many neurological disorders, particularly stroke and Parkinson's disease. Stabilization of plasma membrane Na+/K+-ATPase with an antibody is a novel strategy to treat these diseases. For this reason, Na+/K+-ATPase acts not only as a simple ion pump but also as a sensor/regulator or cytoprotective protein, participating in signal transduction such as neuronal autophagy and apoptosis, and glial cell migration. Thus, the present review attempts to summarize the novel biological functions of Na+/K+-ATPase and Na+/K+-ATPase-related pathogenesis. The potential for novel strategies to treat Na+/K+-ATPase-related brain diseases will also be discussed.
Collapse
Affiliation(s)
- Songqiang Huang
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Wanting Dong
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiaoqian Lin
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Jinsong Bian
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| |
Collapse
|
5
|
Sims SL, Frazier HN, Case SL, Lin RL, Trosper JN, Vekaria HJ, Sullivan PG, Thibault O. Variable bioenergetic sensitivity of neurons and astrocytes to insulin and extracellular glucose. NPJ METABOLIC HEALTH AND DISEASE 2024; 2:33. [PMID: 39524535 PMCID: PMC11549053 DOI: 10.1038/s44324-024-00037-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024]
Abstract
Energy flow within cellular elements of the brain is a well-orchestrated, tightly regulated process, however, details underlying these functions at the single-cell level are still poorly understood. Studying hypometabolism in aging and neurodegenerative diseases may benefit from experimentation on unicellular bioenergetics. Here, we examined energy status in neurons and astrocytes using mixed hippocampal cultures and PercevalHR, an ATP:ADP nanosensor. We assessed exposures of several compounds including KCl, glutamate, FCCP, insulin, and glucose. A mitochondrial stress test was performed, and PercevalHR's fluorescence was corrected for pH using pHrodo. Results demonstrate that PercevalHR can reliably report on the energetic status of two cell types that communicate in a mixed-culture setting. While KCl, glutamate, and FCCP showed clear changes in PercevalHR fluorescence, insulin and glucose responses were found to be more subtle and sensitive to extracellular glucose. These results may highlight mechanisms that mediate insulin sensitivity in the brain.
Collapse
Affiliation(s)
- Sophiya L. Sims
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY USA
| | - Hilaree N. Frazier
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY USA
| | - Sami L. Case
- Department of Biomedical Sciences, College of Veterinary Medicine & Biomedical Sciences, Colorado State University, Fort Collins, CO USA
| | - Ruei-Lung Lin
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY USA
| | - James N. Trosper
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY USA
| | - Hemendra J. Vekaria
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY USA
- Department of Neuroscience, University of Kentucky, Lexington, KY USA
| | - Patrick G. Sullivan
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY USA
- Department of Neuroscience, University of Kentucky, Lexington, KY USA
| | - Olivier Thibault
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY USA
| |
Collapse
|
6
|
da Motta K, Martins CC, da Rocha VME, Soares MP, Mesko MF, Luchese C, Wilhelm EA. Insights into Vincristine-Induced Peripheral Neuropathy in Aged Rats: Wallerian Degeneration, Oxidative Damage, and Alterations in ATPase Enzymes. ACS Chem Neurosci 2024; 15:3954-3969. [PMID: 39207203 PMCID: PMC11587511 DOI: 10.1021/acschemneuro.4c00342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
This study aimed to elucidate vincristine (VCR)-induced peripheral neuropathy in aged rats, a poorly understood neurotoxicity. Both young and old Wistar rats were administered VCR (0.1 mg/kg, intraperitoneally (i.p.)) and compared to age-matched controls (0.9% saline; 10 mg/mL, i.p.). Mechanical (MN) and thermal nociceptive (TN) responses were assessed on days 0, 6, 11, and 17. Locomotor response, cognitive ability, and anxious-like behavior were evaluated on days 14, 15, and 16. Results showed MN and TN responses in both young and old VCR-exposed rats. In old rats, VCR exacerbated MN (on days 6, 11, and 17) and TN (on days 6 and 17) responses. VCR also induced cognitive impairments and anxiety-like behavior. Histological analysis revealed Wallerian degeneration in the spinal cords of VCR-exposed rats accompanied by macrophage migration. Furthermore, VCR increased Ca2+-ATPase activity while inhibiting Na+, K+-ATPase activity in young and old rats. VCR altered the homeostasis of Mg2+-ATPase activity. Lipid peroxidation and nitrite and nitrate levels increased in young and old rats exposed to VCR. This study provides valuable insights into VCR's mechanistic pathways in aged rats, emphasizing the need for further research in this area.
Collapse
Affiliation(s)
- Ketlyn
P. da Motta
- Postgraduate
Program in Biochemistry and Bioprospecting, Research Laboratory in
Biochemical Pharmacology (LaFarBio), Center for Chemical, Pharmaceutical
and Food Sciences, Federal University of
Pelotas, Box 354, CEP, 96010-900 Pelotas, RS, Brazil
| | - Carolina C. Martins
- Postgraduate
Program in Biochemistry and Bioprospecting, Research Laboratory in
Biochemical Pharmacology (LaFarBio), Center for Chemical, Pharmaceutical
and Food Sciences, Federal University of
Pelotas, Box 354, CEP, 96010-900 Pelotas, RS, Brazil
| | - Vanessa M. E. da Rocha
- Postgraduate
Program in Biochemistry and Bioprospecting, Research Laboratory in
Biochemical Pharmacology (LaFarBio), Center for Chemical, Pharmaceutical
and Food Sciences, Federal University of
Pelotas, Box 354, CEP, 96010-900 Pelotas, RS, Brazil
| | - Mauro P. Soares
- Regional
Diagnostic Laboratory Faculty of Veterinary Medicine, Federal University of Pelotas (UFPel), CEP, 96010-900 Pelotas, RS, Brazil
| | - Marcia F. Mesko
- Contaminant
Control Laboratory in Biomaterials (LCCBio), Federal University of Pelotas (UFPel), CEP, 96010-900 Pelotas, RS, Brazil
| | - Cristiane Luchese
- Postgraduate
Program in Biochemistry and Bioprospecting, Research Laboratory in
Biochemical Pharmacology (LaFarBio), Center for Chemical, Pharmaceutical
and Food Sciences, Federal University of
Pelotas, Box 354, CEP, 96010-900 Pelotas, RS, Brazil
| | - Ethel A. Wilhelm
- Postgraduate
Program in Biochemistry and Bioprospecting, Research Laboratory in
Biochemical Pharmacology (LaFarBio), Center for Chemical, Pharmaceutical
and Food Sciences, Federal University of
Pelotas, Box 354, CEP, 96010-900 Pelotas, RS, Brazil
| |
Collapse
|
7
|
Dos Santos GRO, Cararo-Lopes MM, Possebom IR, de Sá Lima L, Scavone C, Kawamoto EM. Sex-dependent changes in AMPAR expression and Na, K-ATPase activity in the cerebellum and hippocampus of α-Klotho-Hypomorphic mice. Neuropharmacology 2024; 258:110097. [PMID: 39094831 DOI: 10.1016/j.neuropharm.2024.110097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024]
Abstract
Aging is characterized by a functional decline in several physiological systems. α-Klotho-hypomorphic mice (Kl-/-) exhibit accelerated aging and cognitive decline. We evaluated whether male and female α-Klotho-hypomorphic mice show changes in the expression of synaptic proteins, N-methyl-d-aspartate receptor (NMDAR) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) subunits, postsynaptic density protein 95 (PSD-95), synaptophysin and synapsin, and the activity of Na+, K+-ATPase (NaK) isoforms in the cerebellum and hippocampus. In this study, we demonstrated that in the cerebellum, Kl-/- male mice have reduced expression of GluA1 (AMPA) compared to wild-type (Kl+/+) males and Kl-/- females. Also, Kl-/- male and female mice show reduced ɑ2/ɑ3-NaK and Mg2+-ATPase activities in the cerebellum, respectively, and sex-based differences in NaK and Mg2+-ATPase activities in both the regions. Our findings suggest that α-Klotho could influence the expression of AMPAR and the activity of NaK isoforms in the cerebellum in a sex-dependent manner, and these changes may contribute, in part, to cognitive decline.
Collapse
Affiliation(s)
| | - Marina Minto Cararo-Lopes
- Faculty of Pharmaceutical Sciences, Food and Nutrition, Federal University of Mato Grosso Do Sul, 79070-900, Campo Grande, MS, Brazil
| | - Isabela Ribeiro Possebom
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-900, São Paulo, SP, Brazil
| | - Larissa de Sá Lima
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-900, São Paulo, SP, Brazil
| | - Cristoforo Scavone
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-900, São Paulo, SP, Brazil
| | - Elisa Mitiko Kawamoto
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, 05508-900, São Paulo, SP, Brazil.
| |
Collapse
|
8
|
Zhang X, Li G, Chen H, Nie XW, Bian JS. Targeting NKAα1 to treat Parkinson's disease through inhibition of mitophagy-dependent ferroptosis. Free Radic Biol Med 2024; 218:190-204. [PMID: 38574977 DOI: 10.1016/j.freeradbiomed.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/04/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024]
Abstract
Dysfunction of the Na+/K+-ATPase (NKA) has been documented in various neurodegenerative diseases, yet the specific role of NKAα1 in Parkinson's disease (PD) remains incompletely understood. In this investigation, we utilized NKAα1 haploinsufficiency (NKAα1+/-) mice to probe the influence of NKAα1 on dopaminergic (DA) neurodegeneration induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Our findings reveal that NKAα1+/- mice displayed a heightened loss of DA neurons and more pronounced motor dysfunction compared to the control group when exposed to MPTP. Intriguingly, this phenomenon coincided with the activation of ferroptosis and impaired mitophagy both in vivo and in vitro. To scrutinize the role and underlying mechanism of NKAα1 in PD, we employed DR-Ab, an antibody targeting the DR-region of the NKA α subunit. Our study demonstrates that the administration of DR-Ab effectively reinstated the membrane abundance of NKAα1, thereby mitigating MPTP-induced DA neuron loss and subsequent improvement in behavioral deficit. Mechanistically, DR-Ab heightened the formation of the surface NKAα1/SLC7A11 complex, inhibiting SLC7A11-dependent ferroptosis. Moreover, DR-Ab disrupted the cytosolic interaction between NKAα1 and Parkin, facilitating the translocation of Parkin to mitochondria and enhancing the process of mitophagy. In conclusion, this study establishes NKAα1 as a key regulator of ferroptosis and mitophagy, identifying its DR-region as a promising therapeutic target for PD.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Guanghong Li
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Hanbin Chen
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Xiao-Wei Nie
- Key Laboratory of Shenzhen Respiratory Disease, Shenzhen Institute of Respiratory Disease, Shenzhen People's Hospital (the First Affiliated Hospital, Southern University of Science and Technology; the Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518055, China.
| | - Jin-Song Bian
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
9
|
Hutton SJ, Siddiqui S, Pedersen EI, Markgraf CY, Segarra A, Hladik ML, Connon RE, Brander SM. Multigenerational, Indirect Exposure to Pyrethroids Demonstrates Potential Compensatory Response and Reduced Toxicity at Higher Salinity in Estuarine Fish. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:2224-2235. [PMID: 38267018 PMCID: PMC10851936 DOI: 10.1021/acs.est.3c06234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/26/2024]
Abstract
Estuarine environments are critical to fish species and serve as nurseries for developing embryos and larvae. They also undergo daily fluctuations in salinity and act as filters for pollutants. Additionally, global climate change (GCC) is altering salinity regimes within estuarine systems through changes in precipitation and sea level rise. GCC is also likely to lead to an increased use of insecticides to prevent pests from damaging agricultural crops as their habitats and mating seasons change from increased temperatures. This underscores the importance of understanding how insecticide toxicity to fish changes under different salinity conditions. In this study, larval Inland Silversides (Menidia beryllina) were exposed to bifenthrin (1.1 ng/L), cyfluthrin (0.9 ng/L), or cyhalothrin (0.7 ng/L) at either 6 or 10 practical salinity units (PSU) for 96 h during hatching, with a subset assessed for end points relevant to neurotoxicity and endocrine disruption by testing behavior, gene expression of a select suite of genes, reproduction, and growth. At both salinities, directly exposed F0 larvae were hypoactive relative to the F0 controls; however, the indirectly exposed F1 larvae were hyperactive relative to the F1 control. This could be evidence of a compensatory response to environmentally relevant concentrations of pyrethroids in fish. Effects on development, gene expression, and growth were also observed. Overall, exposure to pyrethroids at 10 PSU resulted in fewer behavioral and endocrine disruptive effects relative to those observed in organisms at 6 PSU.
Collapse
Affiliation(s)
- Sara J. Hutton
- Department
of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon 97331, United States
| | - Samreen Siddiqui
- Department
of Fisheries, Wildlife, and Conservation Sciences, Coastal Oregon
Marine Experiment Station, Oregon State
University, Newport, Oregon 97365, United States
| | - Emily I. Pedersen
- Department
of Fisheries, Wildlife, and Conservation Sciences, Coastal Oregon
Marine Experiment Station, Oregon State
University, Newport, Oregon 97365, United States
| | - Christopher Y. Markgraf
- Department
of Biochemistry and Biophysics, Oregon State
University, Corvallis, Oregon 97331, United States
| | - Amelie Segarra
- Department
of Anatomy, Physiology, and Cell Biology, University of California, Davis, California 95616, United States
| | - Michelle L. Hladik
- U.S.
Geological Survey, California Water Science
Center, Sacramento, California 95819, United States
| | - Richard E. Connon
- Department
of Anatomy, Physiology, and Cell Biology, University of California, Davis, California 95616, United States
| | - Susanne M. Brander
- Department
of Fisheries, Wildlife, and Conservation Sciences, Coastal Oregon
Marine Experiment Station, Oregon State
University, Newport, Oregon 97365, United States
| |
Collapse
|
10
|
Zeylan M, Senyuz S, Picón-Pagès P, García-Elías A, Tajes M, Muñoz FJ, Oliva B, Garcia-Ojalvo J, Barbu E, Vicente R, Nattel S, Ois A, Puig-Pijoan A, Keskin O, Gursoy A. Shared Proteins and Pathways of Cardiovascular and Cognitive Diseases: Relation to Vascular Cognitive Impairment. J Proteome Res 2024; 23:560-573. [PMID: 38252700 PMCID: PMC10846560 DOI: 10.1021/acs.jproteome.3c00289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/29/2023] [Accepted: 12/06/2023] [Indexed: 01/24/2024]
Abstract
One of the primary goals of systems medicine is the detection of putative proteins and pathways involved in disease progression and pathological phenotypes. Vascular cognitive impairment (VCI) is a heterogeneous condition manifesting as cognitive impairment resulting from vascular factors. The precise mechanisms underlying this relationship remain unclear, which poses challenges for experimental research. Here, we applied computational approaches like systems biology to unveil and select relevant proteins and pathways related to VCI by studying the crosstalk between cardiovascular and cognitive diseases. In addition, we specifically included signals related to oxidative stress, a common etiologic factor tightly linked to aging, a major determinant of VCI. Our results show that pathways associated with oxidative stress are quite relevant, as most of the prioritized vascular cognitive genes and proteins were enriched in these pathways. Our analysis provided a short list of proteins that could be contributing to VCI: DOLK, TSC1, ATP1A1, MAPK14, YWHAZ, CREB3, HSPB1, PRDX6, and LMNA. Moreover, our experimental results suggest a high implication of glycative stress, generating oxidative processes and post-translational protein modifications through advanced glycation end-products (AGEs). We propose that these products interact with their specific receptors (RAGE) and Notch signaling to contribute to the etiology of VCI.
Collapse
Affiliation(s)
- Melisa
E. Zeylan
- Computational
Sciences and Engineering, Graduate School of Science and Engineering, Koç University, Istanbul 34450, Türkiye
| | - Simge Senyuz
- Computational
Sciences and Engineering, Graduate School of Science and Engineering, Koç University, Istanbul 34450, Türkiye
| | - Pol Picón-Pagès
- Laboratory
of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Anna García-Elías
- Laboratory
of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Marta Tajes
- Laboratory
of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Francisco J. Muñoz
- Laboratory
of Molecular Physiology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Baldomero Oliva
- Laboratory
of Structural Bioinformatics (GRIB), Department of Medicine and Life
Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Jordi Garcia-Ojalvo
- Laboratory
of Dynamical Systems Biology, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona 08002, Spain
| | - Eduard Barbu
- Institute
of Computer Science, University of Tartu, Tartu, 50090, Estonia
| | - Raul Vicente
- Institute
of Computer Science, University of Tartu, Tartu, 50090, Estonia
| | - Stanley Nattel
- Department
of Medicine and Research Center, Montreal Heart Institute and Université
de Montréal; Institute of Pharmacology, West German Heart and
Vascular Center, University Duisburg-Essen,
Germany; IHU LIRYC and Fondation Bordeaux Université, Bordeaux 33000, France
| | - Angel Ois
- Department
of Neurology, Hospital Del Mar. Hospital
Del Mar - Medical Research Institute and Universitat Pompeu Fabra, Barcelona 08003, Spain
| | - Albert Puig-Pijoan
- Department
of Neurology, Hospital Del Mar. Hospital
Del Mar - Medical Research Institute and Universitat Pompeu Fabra, Barcelona 08003, Spain
| | - Ozlem Keskin
- Department
of Chemical and Biological Engineering, Koç University, Istanbul 34450, Türkiye
| | - Attila Gursoy
- Department
of Computer Engineering, Koç University, Istanbul 34450, Türkiye
| |
Collapse
|
11
|
Piergiorge RM, da Silva Francisco Junior R, de Vasconcelos ATR, Santos-Rebouças CB. Multi-layered transcriptomic analysis reveals a pivotal role of FMR1 and other developmental genes in Alzheimer's disease-associated brain ceRNA network. Comput Biol Med 2023; 166:107494. [PMID: 37769462 DOI: 10.1016/j.compbiomed.2023.107494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/05/2023] [Accepted: 09/15/2023] [Indexed: 09/30/2023]
Abstract
Alzheimer's disease (AD) is an increasingly neurodegenerative disorder that causes progressive cognitive decline and memory impairment. Despite extensive research, the underlying causes of late-onset AD (LOAD) are still in progress. This study aimed to establish a network of competing regulatory interactions involving circular RNAs (circRNAs), microRNAs (miRNAs), RNA-binding proteins (RBPs), and messenger RNAs (mRNAs) connected to LOAD. A systematic analysis of publicly available expression data was conducted to identify integrated differentially expressed genes (DEGs) from the hippocampus of LOAD patients. Subsequently, gene co-expression analysis identified modules comprising highly expressed DEGs that act cooperatively. The competition between co-expressed DEGs and miRNAs/RBPs and the simultaneous interactions between circRNA and miRNA/RBP revealed a complex ceRNA network responsible for post-transcriptional regulation in LOAD. Hippocampal expression data for miRNAs, circRNAs, and RBPs were used to filter relevant relationships for AD. An integrated topological score was used to identify the highly connected hub gene, from which a brain core ceRNA subnetwork was generated. The Fragile X Messenger Ribonucleoprotein 1 (FMR1) coding for the RBP FMRP emerged as the prominent driver gene in this subnetwork. FMRP has been previously related to AD but not in a ceRNA network context. Also, the substantial number of neurodevelopmental genes in the ceRNA subnetwork and their related biological pathways strengthen that AD shares common pathological mechanisms with developmental conditions. Our results enhance the current knowledge about the convergent ceRNA regulatory pathways underlying AD and provide potential targets for identifying early biomarkers and developing novel therapeutic interventions.
Collapse
Affiliation(s)
- Rafael Mina Piergiorge
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Cíntia Barros Santos-Rebouças
- Department of Genetics, Institute of Biology Roberto Alcantara Gomes, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
12
|
Leite JA, Orellana AM, Andreotti DZ, Matumoto AM, de Souza Ports NM, de Sá Lima L, Kawamoto EM, Munhoz CD, Scavone C. Ouabain Reverts CUS-Induced Disruption of the HPA Axis and Avoids Long-Term Spatial Memory Deficits. Biomedicines 2023; 11:biomedicines11041177. [PMID: 37189795 DOI: 10.3390/biomedicines11041177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 05/17/2023] Open
Abstract
Ouabain (OUA) is a cardiotonic steroid that modulates Na+, K+ -ATPase activity. OUA has been identified as an endogenous substance that is present in human plasma, and it has been shown to be associated with the response to acute stress in both animals and humans. Chronic stress is a major aggravating factor in psychiatric disorders, including depression and anxiety. The present work investigates the effects of the intermittent administration of OUA (1.8 μg/kg) during the chronic unpredictable stress (CUS) protocol in a rat's central nervous system (CNS). The results suggest that the intermittent OUA treatment reversed CUS-induced HPA axis hyperactivity through a reduction in (i) glucocorticoids levels, (ii) CRH-CRHR1 expression, and by decreasing neuroinflammation with a reduction in iNOS activity, without interfering with the expression of antioxidant enzymes. These changes in both the hypothalamus and hippocampus may reflect in the rapid extinction of aversive memory. The present data demonstrate the ability of OUA to modulate the HPA axis, as well as to revert CUS-induced long-term spatial memory deficits.
Collapse
Affiliation(s)
- Jacqueline Alves Leite
- Departament of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Goiás, Goiânia 74690-900, Brazil
| | - Ana Maria Orellana
- Departament of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Diana Zukas Andreotti
- Departament of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Amanda Midori Matumoto
- Departament of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | | | - Larissa de Sá Lima
- Departament of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Elisa Mitiko Kawamoto
- Departament of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Carolina Demarchi Munhoz
- Departament of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | - Cristoforo Scavone
- Departament of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
13
|
Garcia IJP, Kinoshita PF, Valadares JMDM, de Carvalho LED, Cortes VF, Barbosa LA, Scavone C, Santos HDL. Effect of Ouabain on Glutamate Transport in the Hippocampus of Rats with LPS-Induced Neuroinflammation. Biomedicines 2023; 11:biomedicines11030920. [PMID: 36979899 PMCID: PMC10045517 DOI: 10.3390/biomedicines11030920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 03/19/2023] Open
Abstract
A lipopolysaccharide (LPS)-induced neuroinflammation rat model was used to study the effects of ouabain (OUA) at low concentrations, which can interact with the Na,K-ATPase, causing the modulation of intracellular signalling pathways in the Central Nervous System. Our study aimed to analyse the effects of OUA on glutamate transport in the hippocampus of rats with LPS-induced neuroinflammation. Adult male Wistar rats were divided into four groups: OUA (1.8 µg/kg), saline (CTR), LPS (200 µg/kg), and OUA + LPS (OUA 20 min before LPS). The animals were sacrificed after 2 h, and the hippocampus was collected for analysis. After treatment, we determined the activities of Na,K-ATPase and glutamine synthetase (GS). In addition, expression of the α1, α2, and α3 isoforms of Na,K-ATPase and the glutamate transporters, EAAT1 and EAAT2, were also analysed. Treatment with OUA caused a specific increase in the α2 isoform expression (~20%), whereas LPS decreased its expression (~22%), and treatment with OUA before LPS prevented the effects of LPS. Moreover, LPS caused a decrease of approximately 50% in GS activity compared with that in the CTR group; however, OUA pre-treatment attenuated this effect of LPS. Notably, it was found that treatment with OUA caused an increase in the expression of EAAT1 (~30%) and EAAT2 (~25%), whereas LPS caused a decrease in the expression of EAAT1 (~23%) and EAAT2 (~25%) compared with that in the CTR group. When treated with OUA, the effects of LPS were abrogated. In conclusion, the OUA pre-treatment abolished the effect caused by LPS, suggesting that this finding may be related to the restoration of the interaction between FXYD2 and the studied membrane proteins.
Collapse
Affiliation(s)
- Israel José Pereira Garcia
- Cellular Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Membrane and ATPase Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
| | - Paula Fernanda Kinoshita
- Molecular Neuropharmacology Laboratory, Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, Brazil
| | - Jéssica Martins de Moura Valadares
- Cellular Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Membrane and ATPase Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
| | - Luciana Estefani Drumond de Carvalho
- Cellular Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Membrane and ATPase Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
| | - Vanessa Faria Cortes
- Cellular Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Membrane and ATPase Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
| | - Leandro Augusto Barbosa
- Cellular Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Membrane and ATPase Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
| | - Cristoforo Scavone
- Molecular Neuropharmacology Laboratory, Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, Brazil
- Correspondence: (C.S.); (H.d.L.S.)
| | - Hérica de Lima Santos
- Cellular Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Membrane and ATPase Biochemistry Laboratory, Federal University of São João del-Rei, Campus Cento-Oeste, Divinópolis 35501-296, Brazil
- Correspondence: (C.S.); (H.d.L.S.)
| |
Collapse
|
14
|
Zou S, Lan YL, Gong Y, Chen Z, Xu C. The role of ATP1A3 gene in epilepsy: We need to know more. Front Cell Neurosci 2023; 17:1143956. [PMID: 36866063 PMCID: PMC9972585 DOI: 10.3389/fncel.2023.1143956] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 01/23/2023] [Indexed: 02/16/2023] Open
Abstract
The ATP1A3 gene, which encodes the Na+/K+-ATPase α3 catalytic subunit, plays a crucial role in both physiological and pathological conditions in the brain, and mutations in this gene have been associated with a wide variety of neurological diseases by impacting the whole infant development stages. Cumulative clinical evidence suggests that some severe epileptic syndromes have been linked to mutations in ATP1A3, among which inactivating mutation of ATP1A3 has been intriguingly found to be a candidate pathogenesis for complex partial and generalized seizures, proposing ATP1A3 regulators as putative targets for the rational design of antiepileptic therapies. In this review, we introduced the physiological function of ATP1A3 and summarized the findings about ATP1A3 in epileptic conditions from both clinical and laboratory aspects at first. Then, some possible mechanisms of how ATP1A3 mutations result in epilepsy are provided. We think this review timely introduces the potential contribution of ATP1A3 mutations in both the genesis and progression of epilepsy. Taken that both the detailed mechanisms and therapeutic significance of ATP1A3 for epilepsy are not yet fully illustrated, we think that both in-depth mechanisms investigations and systematic intervention experiments targeting ATP1A3 are needed, and by doing so, perhaps a new light can be shed on treating ATP1A3-associated epilepsy.
Collapse
Affiliation(s)
- Shuang Zou
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu-Long Lan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China,*Correspondence: Yu-Long Lan ✉
| | - Yiwei Gong
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Cenglin Xu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China,Cenglin Xu ✉
| |
Collapse
|
15
|
Osman KA, Shaaban MMI, Ahmed NS. Biomarkers of imidacloprid toxicity in Japanese quail, Coturnix coturnix japonica. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:5662-5676. [PMID: 35980528 DOI: 10.1007/s11356-022-22580-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 08/12/2022] [Indexed: 06/15/2023]
Abstract
The in vivo effect of the oral sublethal doses of 3.014 mg kg-1 of IMI (1/25 LD50) for 1, 7, 14, and 28 days every other day on Japanese quail was investigated. The results revealed that certain biomarkers in the selected tissues of the quail such as acetylcholinesterase (AChE), butyrylcholinesterase (BuChE), aminotransaminases (alanine aminotransferase, ALT, and aspartate aminotransaminase, AST), phosphatases (acid phosphatase, ACP, and alkaline phosphatase, ALP), lactate dehydrogenase (LDH), adenosine-triphosphatase (ATPase), glutathione-S-transferase (GST), lipid peroxidation (LPO), and blood glucose showed significant inductions, while significant reductions in the levels of glutathione-reduced (GSH), deoxyribonucleic acid (DNA), and ribonucleic acid (RNA) were noticed. In this study, the molecular mechanisms of the toxic effects of imidacloprid on quails were elucidated regarding neurotoxicity, hepatotoxicity, oxidative stress, lipid peroxidation, antioxidant activity, and genotoxicity. Because IMI induced alterations in the levels of these biomarkers in Japanese quail; therefore, Japanese quail as a wild avian can be used as a suite bioindicator to detect imidacloprid toxicity.
Collapse
Affiliation(s)
- Khaled A Osman
- Department of Pesticide Chemistry and Technology, Faculty of Agriculture, Alexandria University, P.O Box 21545, Alexandria, Egypt.
| | - Mahmoud M I Shaaban
- Department of Pesticide Chemistry and Technology, Faculty of Agriculture, Alexandria University, P.O Box 21545, Alexandria, Egypt
| | - Nabila S Ahmed
- Department of Pesticide Chemistry and Technology, Faculty of Agriculture, Alexandria University, P.O Box 21545, Alexandria, Egypt
| |
Collapse
|
16
|
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
|
17
|
Xiao L, Jiang S, Wang Y, Gao C, Liu C, Huo X, Li W, Guo B, Wang C, Sun Y, Wang A, Feng Y, Wang F, Sun T. Continuous high-frequency deep brain stimulation of the anterior insula modulates autism-like behavior in a valproic acid-induced rat model. J Transl Med 2022; 20:570. [PMID: 36474209 PMCID: PMC9724311 DOI: 10.1186/s12967-022-03787-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Until now, the treatment of patients with autism spectrum disorder (ASD) remain a difficult problem. The insula is involved in empathy and sensorimotor integration, which are often impaired in individuals with ASD. Deep brain stimulation, modulating neuronal activity in specific brain circuits, has recently been considered as a promising intervention for neuropsychiatric disorders. Valproic acid (VPA) is a potential teratogenic agent, and prenatal exposure can cause autism-like symptoms including repetitive behaviors and defective sociability. Herein, we investigated the effects of continuous high-frequency deep brain stimulation in the anterior insula of rats exposed to VPA and explored cognitive functions, behavior, and molecular proteins connected to autism spectrum disorder. METHODS VPA-exposed offspring were bilaterally implanted with electrodes in the anterior insula (Day 0) with a recovery period of 1 week. (Day 0-7). High-frequency deep brain stimulation was applied from days 11 to 29. Three behavioral tests, including three-chamber social interaction test, were performed on days 7, 13, 18, 25 and 36, and several rats were used for analysis of immediate early genes and proteomic after deep brain stimulation intervention. Meanwhile, animals were subjected to a 20 day spatial learning and cognitive rigidity test using IntelliCage on day 11. RESULTS Deep brain stimulation improved the sociability and social novelty preference at day 18 prior to those at day 13, and the improvement has reached the upper limit compared to day 25. As for repetitive/stereotypic-like behavior, self- grooming time were reduced at day 18 and reached the upper limit, and the numbers of burried marbles were reduced at day 13 prior to those at day 18 and day 25. The improvements of sociability and social novelty preference were persistent after the stimulation had ceased. Spatial learning ability and cognitive rigidity were unaffected. We identified 35 proteins in the anterior insula, some of which were intimately linked to autism, and their expression levels were reversed upon administration of deep brain stimulation. CONCLUSIONS Autism-like behavior was ameliorated and autism-related proteins were reversed in the insula by deep brain stimulation intervention, these findings reveal that the insula may be a potential target for DBS in the treatment of autism, which provide a theoretical basis for its clinical application., although future studies are still warranted.
Collapse
Affiliation(s)
- Lifei Xiao
- grid.412194.b0000 0004 1761 9803Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, 750000 China ,grid.413385.80000 0004 1799 1445Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, 750000 China
| | - Shucai Jiang
- grid.416966.a0000 0004 1758 1470Department of Neurosurgery, Weifang People’s Hospital, Weifang, 261000 China
| | - Yangyang Wang
- grid.412194.b0000 0004 1761 9803Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, 750000 China
| | - Caibin Gao
- grid.413385.80000 0004 1799 1445Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, 750000 China
| | - Cuicui Liu
- grid.477991.5Department of Otolaryngology and Head Surgery, The First People’s Hospital of Yinchuan, Yinchuan, 750000 China
| | - Xianhao Huo
- grid.412194.b0000 0004 1761 9803Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, 750000 China ,grid.413385.80000 0004 1799 1445Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, 750000 China
| | - Wenchao Li
- grid.412194.b0000 0004 1761 9803Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, 750000 China
| | - Baorui Guo
- grid.440288.20000 0004 1758 0451Department of Neurosurgery, Shaanxi Provincial People’s Hospital, Xi’an, 710000 China
| | - Chaofan Wang
- grid.412194.b0000 0004 1761 9803Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, 750000 China
| | - Yu Sun
- grid.412194.b0000 0004 1761 9803Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, 750000 China
| | - Anni Wang
- grid.412194.b0000 0004 1761 9803Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, 750000 China
| | - Yan Feng
- grid.412194.b0000 0004 1761 9803Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, 750000 China
| | - Feng Wang
- grid.13402.340000 0004 1759 700XDepartment of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000 China
| | - Tao Sun
- grid.412194.b0000 0004 1761 9803Ningxia Key Laboratory of Cerebrocranial Disease, Incubation Base of National Key Laboratory, Ningxia Medical University, Yinchuan, 750000 China ,grid.413385.80000 0004 1799 1445Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, 750000 China
| |
Collapse
|
18
|
Dos Santos TM, Siebert C, Bobermin LD, Quincozes-Santos A, Wyse ATS. Mild Hyperhomocysteinemia Causes Anxiety-like Behavior and Brain Hyperactivity in Rodents: Are ATPase and Excitotoxicity by NMDA Receptor Overstimulation Involved in this Effect? Cell Mol Neurobiol 2022; 42:2697-2714. [PMID: 34324129 PMCID: PMC11421652 DOI: 10.1007/s10571-021-01132-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 07/18/2021] [Indexed: 10/20/2022]
Abstract
Mild hyperhomocysteinemia is a risk factor for psychiatric and neurodegenerative diseases, whose mechanisms between them are not well-known. In the present study, we evaluated the emotional behavior and neurochemical pathways (ATPases, glutamate homeostasis, and cell viability) in amygdala and prefrontal cortex rats subjected to mild hyperhomocysteinemia (in vivo studies). The ex vivo effect of homocysteine on ATPases and redox status, as well as on NMDAR antagonism by MK-801 in same structures slices were also performed. Wistar male rats received a subcutaneous injection of 0.03 µmol Homocysteine/g of body weight or saline, twice a day from 30 to 60th-67th days of life. Hyperhomocysteinemia increased anxiety-like behavior and tended to alter locomotion/exploration of rats, whereas sucrose preference and forced swimming tests were not altered. Glutamate uptake was not changed, but the activities of glutamine synthetase and ATPases were increased. Cell viability was not altered. Ex vivo studies (slices) showed that homocysteine altered ATPases and redox status and that MK801, an NMDAR antagonist, protected amygdala (partially) and prefrontal cortex (totally) effects. Taken together, data showed that mild hyperhomocysteinemia impairs the emotional behavior, which may be associated with changes in ATPase and glutamate homeostasis, including glutamine synthetase and NMDAR overstimulation that could lead to excitotoxicity. These findings may be associated with the homocysteine risk factor on psychiatric disorders development and neurodegeneration.
Collapse
Affiliation(s)
- Tiago Marcon Dos Santos
- Wyse´s Lab, Institute of Health Basic Sciences, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, CEP 90035-003, Brazil
- Postgraduate Program in Biological Science: Biochemistry, Institute of Health Basic Sciences, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, CEP 90035-003, Brazil
| | - Cassiana Siebert
- Wyse´s Lab, Institute of Health Basic Sciences, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, CEP 90035-003, Brazil
- Postgraduate Program in Biological Science: Biochemistry, Institute of Health Basic Sciences, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, CEP 90035-003, Brazil
| | - Larissa Daniele Bobermin
- Postgraduate Program in Biological Science: Biochemistry, Institute of Health Basic Sciences, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, CEP 90035-003, Brazil
- Department of Biochemistry, Institute of Health Basic Sciences, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, CEP 90035-003, Brazil
| | - André Quincozes-Santos
- Postgraduate Program in Biological Science: Biochemistry, Institute of Health Basic Sciences, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, CEP 90035-003, Brazil
- Department of Biochemistry, Institute of Health Basic Sciences, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, CEP 90035-003, Brazil
| | - Angela T S Wyse
- Wyse´s Lab, Institute of Health Basic Sciences, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, CEP 90035-003, Brazil.
- Postgraduate Program in Biological Science: Biochemistry, Institute of Health Basic Sciences, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, CEP 90035-003, Brazil.
- Department of Biochemistry, Institute of Health Basic Sciences, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, CEP 90035-003, Brazil.
| |
Collapse
|
19
|
Luduvico KP, Spohr L, de Aguiar MSS, Teixeira FC, Bona NP, de Mello JE, Spanevello RM, Stefanello FM. LPS-induced impairment of Na +/K +-ATPase activity: ameliorative effect of tannic acid in mice. Metab Brain Dis 2022; 37:2133-2140. [PMID: 35759073 DOI: 10.1007/s11011-022-01036-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/13/2022] [Indexed: 10/17/2022]
Abstract
Acetylcholine is an excitatory neurotransmitter that modulates synaptic plasticity and communication, and it is essential for learning and memory processes. This neurotransmitter is hydrolyzed by acetylcholinesterase (AChE), which plays other cellular roles in processes such as inflammation and oxidative stress. Ion pumps, such as Na+/K+-ATPase and Ca2+-ATPase, are highly expressed channels that derive energy for their functions from ATP hydrolysis. Impairment of the cholinergic system and ion pumps is associated with neuropsychiatric diseases. Major depressive disorder (MDD) is an example of a complex disease with high morbidity and a heterogenous etiology. Polyphenols have been investigated for their therapeutic effects, and tannic acid (TA) has been reported to show neuroprotective and antidepressant-like activities. Animal models of depression-like behavior, such as lipopolysaccharide (LPS)-induced models of depression, are useful for investigating the pathophysiology of MDD. In this context, effects of TA were evaluated in an LPS-induced mouse model of depression-like behavior. Animals received TA for 7 days, and on the last day of treatment, LPS (830 μg/kg) was administered intraperitoneally. In vitro exposure of healthy brain to TA decreased the AChE activity. Additionally, this enzyme activity was decreased in cerebral cortex of LPS-treated mice. LPS injection increased the activity of Ca2+-ATPase in the cerebral cortex but decreased the enzyme activity in the hippocampus. LPS administration decreased Na+/K+-ATPase activity in the cerebral cortex, hippocampus, and striatum; however, TA administration prevented these changes. In conclusion, tannins may affect Na+/K+-ATPase and Ca2+-ATPase activities, which is interesting in the context of MDD.
Collapse
Affiliation(s)
- Karina Pereira Luduvico
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário s/n, Pelotas, RS, CEP: 96010-900, Brazil.
| | - Luiza Spohr
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário s/n, Pelotas, RS, CEP: 96010-900, Brazil
| | - Mayara Sandrielly Soares de Aguiar
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário s/n, Pelotas, RS, CEP: 96010-900, Brazil
| | - Fernanda Cardoso Teixeira
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário s/n, Pelotas, RS, CEP: 96010-900, Brazil
| | - Natália Pontes Bona
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário s/n, Pelotas, RS, CEP: 96010-900, Brazil
| | - Julia Eisenhardt de Mello
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário s/n, Pelotas, RS, CEP: 96010-900, Brazil
| | - Roselia Maria Spanevello
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário s/n, Pelotas, RS, CEP: 96010-900, Brazil
| | - Francieli Moro Stefanello
- Programa de Pós-Graduação em Bioquímica e Bioprospecção, Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário s/n, Pelotas, RS, CEP: 96010-900, Brazil.
| |
Collapse
|
20
|
Martins CC, Reis AS, Cristiane Luchese KPDM, Wilhelm EA. Mechanistic pathways of fibromyalgia induced by intermittent cold stress in mice is sex-dependently. Brain Res Bull 2022; 187:11-23. [PMID: 35753533 DOI: 10.1016/j.brainresbull.2022.06.005] [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: 01/04/2022] [Revised: 06/03/2022] [Accepted: 06/09/2022] [Indexed: 11/02/2022]
Abstract
Fibromyalgia results from a complex interplay of biochemical and neurobiological elements mediated sensitization of nociceptive pathways. Despite the symptoms of fibromyalgia negatively affect the quality of life of patients, the pathophysiology of this disease remains inconclusive, which difficult the development of an appropriate treatment. The present study investigated the involvement of the serotonergic receptors, the N-methyl-D-aspartate (NMDA)/ nitric oxide (NO)/ cyclic guanosine monophosphate (cGMP) pathway and the oxidative stress in an animal model of fibromyalgia induced by intermittent cold stress (ICS), considering the specificities of male and female Swiss mice. The ICS exposure increased mechanical and thermal sensitivities, and decreased muscle strength in mice of both sexes. Female mice exhibited a longer-lasting mechanical sensitivity than male mice exposed to ICS along with an enhancement of the Na+, K+-ATPase activity in the spinal cord and cerebral cortex. Conversely, an inhibition in the Na+, K+-ATPase and glutathione peroxidase activities accompanied by an increase in the reactive species levels in the cerebral cortex of male mice were observed. The treatment with different serotonergic antagonists (pindolol, ketanserin and ondasetron) reversed the mechanical sensitivity in mice of both sexes, after the ICS exposure. The administration of MK-801, L-arginine and methylene blue also blocked the mechanical sensitivity in female mice exposed to ICS. Except L-arginine, MK-801 and methylene blue also attenuated this nociceptive signal in male mice, after ICS exposure. In conclusion, the modulation of serotonergic receptors, the NMDA/NO/cGMP pathway, and the oxidative stress seems contribute to nociceptive behaviors induced by ICS exposure sex-dependent.
Collapse
Affiliation(s)
- Carolina C Martins
- Laboratório de Pesquisa em Farmacologia Bioquímica - LaFarBio, Programa de Pós-Graduação em Bioquímica e Bioprospecção, CCQFA - Universidade Federal de Pelotas, UFPel, P.O. Box 354 - 96010-900, Pelotas, RS, Brazil
| | - Angélica S Reis
- Laboratório de Pesquisa em Farmacologia Bioquímica - LaFarBio, Programa de Pós-Graduação em Bioquímica e Bioprospecção, CCQFA - Universidade Federal de Pelotas, UFPel, P.O. Box 354 - 96010-900, Pelotas, RS, Brazil
| | - Ketlyn P da Motta Cristiane Luchese
- Laboratório de Pesquisa em Farmacologia Bioquímica - LaFarBio, Programa de Pós-Graduação em Bioquímica e Bioprospecção, CCQFA - Universidade Federal de Pelotas, UFPel, P.O. Box 354 - 96010-900, Pelotas, RS, Brazil.
| | - Ethel A Wilhelm
- Laboratório de Pesquisa em Farmacologia Bioquímica - LaFarBio, Programa de Pós-Graduação em Bioquímica e Bioprospecção, CCQFA - Universidade Federal de Pelotas, UFPel, P.O. Box 354 - 96010-900, Pelotas, RS, Brazil.
| |
Collapse
|
21
|
Handa P, Samkaria A, Sharma S, Arora Y, Mandal PK. Comprehensive Account of Sodium Imaging and Spectroscopy for Brain Research. ACS Chem Neurosci 2022; 13:859-875. [PMID: 35324144 DOI: 10.1021/acschemneuro.2c00027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Sodium (23Na) is a vital component of neuronal cells and plays a key role in various signal transmission processes. Hence, information on sodium distribution in the brain using magnetic resonance imaging (MRI) provides useful information on neuronal health. 23Na MRI and MR spectroscopy (MRS) improve the diagnosis, prognosis, and clinical monitoring of neurological diseases but confront some inherent limitations that lead to low signal-to-noise ratio, longer scan time, and diminished partial volume effects. Recent advancements in multinuclear MR technology have helped in further exploration in this domain. We aim to provide a comprehensive description of 23Na MRI and MRS for brain research including the following aspects: (a) theoretical background for understanding 23Na MRI and MRS fundamentals; (b) technological advancements of 23Na MRI with respect to pulse sequences, RF coils, and sodium compartmentalization; (c) applications of 23Na MRI in the early diagnosis and prognosis of various neurological disorders; (d) structural-chronological evolution of sodium spectroscopy in terms of its numerous applications in human studies; (e) the data-processing tools utilized in the quantitation of sodium in the respective anatomical regions.
Collapse
Affiliation(s)
- Palak Handa
- Neuroimaging and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon 122051, India
| | - Avantika Samkaria
- Neuroimaging and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon 122051, India
| | - Shallu Sharma
- Neuroimaging and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon 122051, India
| | - Yashika Arora
- Neuroimaging and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon 122051, India
| | - Pravat K. Mandal
- Neuroimaging and Neurospectroscopy (NINS) Laboratory, National Brain Research Centre, Gurgaon 122051, India
- Florey Institute of Neuroscience and Mental Health, Melbourne School of Medicine Campus, Melbourne 3010, Australia
| |
Collapse
|
22
|
Lin J, Zhang K, Cao X, Zhao Y, Ullah Khan N, Liu X, Tang X, Chen M, Zhang H, Shen L. iTRAQ-Based Proteomics Analysis of Rat Cerebral Cortex Exposed to Valproic Acid before Delivery. ACS Chem Neurosci 2022; 13:648-663. [PMID: 35138800 DOI: 10.1021/acschemneuro.1c00800] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurological and developmental disorder characterized by social and communication difficulties. Valproic acid (VPA) injection during pregnancy elicits autism-like behavior in the offspring, making it a classic animal model of ASD. However, the mechanisms involved have not yet been determined. In this study, we used iTRAQ (isobaric tags for relative and absolute quantification) proteomics analysis of the cerebral cortex of a VPA rat model (VPA group) and controls (CON group). The results showed that 79 differentially expressed proteins (DEPs) were identified between the VPA group and the CON group. Based on bioinformatics analysis, the DEPs were mainly enriched at synapses, especially glutamatergic synapses and GABAergic synapses. Some DEPs were involved in energy metabolism, thyroid hormone synthesis pathway, and Na+-K+-ATPase. Cytoskeleton and endoplasmic reticulum (ER) stress-related proteins were also involved. Some DEPs matched either the ASD gene database or previous reports on cerebral cortical transcriptome studies in VPA rat models. Dysregulation of these DEPs in the cerebral cortex of VPA rats may be responsible for autism-like behavior in rats. We also found that some DEPs were associated with neuropsychiatric disorders, implying that these diseases share common signaling pathways and mechanisms. Moreover, increased expression of DEPs was associated with energy metabolism in the cerebral cortex of VPA rats, implying that ASD may be a distinct type of mitochondrial dysfunction that requires further investigation.
Collapse
Affiliation(s)
- Jing Lin
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, P. R. China
- Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, P. R. China
| | - Kaoyuan Zhang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, P. R. China
| | - Xueshan Cao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, P. R. China
| | - Yuxi Zhao
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, P. R. China
| | - Naseer Ullah Khan
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, P. R. China
| | - Xukun Liu
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, P. R. China
| | - Xiaoxiao Tang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, P. R. China
- Brain Disease and Big Data Research Institute, Shenzhen University, Shenzhen 518071, P. R. China
| | - Margy Chen
- Department of Psychology, Emory University, Atlanta, Georgia 30322, United States
| | - Huajie Zhang
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, P. R. China
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, Shenzhen 518071, P. R. China
| | - Liming Shen
- College of Life Science and Oceanography, Shenzhen University, Shenzhen 518071, P. R. China
| |
Collapse
|
23
|
Oda I, Danno D, Saigoh K, Wolf J, Kawashita N, Hirano M, Samukawa M, Kitamura S, Kikui S, Takeshima T, Mitsui Y, Kusunoki S, Nagai Y. Hemiplegic migraine type 2 with new mutation of the ATP1A2 gene in Japanese cases. Neurosci Res 2022; 180:83-89. [DOI: 10.1016/j.neures.2022.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/26/2022] [Accepted: 03/02/2022] [Indexed: 11/25/2022]
|
24
|
Interface of Aging and Acute Peripheral Neuropathy Induced by Oxaliplatin in Mice: Target-Directed Approaches for Na +, K +-ATPase, Oxidative Stress, and 7-Chloro-4-(phenylselanyl) quinoline Therapy. Mol Neurobiol 2022; 59:1766-1780. [PMID: 35023057 DOI: 10.1007/s12035-021-02659-5] [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: 08/03/2021] [Accepted: 11/19/2021] [Indexed: 12/17/2022]
Abstract
Almost 90% of patients develop pain immediately after oxaliplatin (OXA) treatment. Here, the impact of aging on OXA-induced acute peripheral neuropathy and the potential of 7-chloro-4-(phenylselanyl) quinoline (4-PSQ) as a new therapeutic strategy were evaluated. In Swiss mice, the oxidative damage and its influence on Mg2+-ATPase and Na+, K+-ATPase activities were investigated. The relationship between the reactive oxygen species (ROS) and nitrate and nitrite (NOx) levels, the activity of glutathione peroxidase (GPx), and superoxide dismutase (SOD) with the development of OXA-induced acute peripheral neuropathy was also studied. In this study, it was evidenced that OXA-induced acute peripheral neuropathy was exacerbated by aging through increased oxidative damage as well as Na+, K+-ATPase, and Mg+2-ATPase inhibition. 4-PSQ reversed hypersensitivity induced by OXA and aging-aggravated by reducing ROS and NOx levels, through modulation of GPx and SOD activities. 4-PSQ partially reestablish Na+, K+-ATPase activity, but not Mg 2+-ATPase activity. Locomotor and exploratory activities were not affected. This study is the first of its kind, providing new insight into the aging impact on mechanisms involved in OXA-induced acute peripheral neuropathy. Also, it provides evidence on promising 4-PSQ effects on this condition, mainly on aging.
Collapse
|
25
|
Osman KA, Ezz El-Din EM, Ahmed NS, El-Seedy AS. Effect of N-acetylcysteine on attenuation of chlropyrifos and its methyl analogue toxicity in male rats. Toxicology 2021; 461:152904. [PMID: 34425170 DOI: 10.1016/j.tox.2021.152904] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 02/01/2023]
Abstract
The attenuating effect of 150 mg/kg of N-acetylcysteine (NAC) against the oral administration of 7.88 and 202.07 mg/kg/day for 14 days of either chlropyrifos-ethyl (CPE-E) or chlropyrifos-methyl (CPF-M), respectively, in male rat was investigated using biochemical and genetic markers. Biomarkers such as acetylcholinesterase (AChE), butyrylcholinesterase (BuChE), paraoxonase (PON), adenosine 5'-triphosphatase (ATP-ase), glutathione-S-transferase (GST), catalase (CAT), glutathione reduced (GSH) in serum showed a significant decline in their levels, while calcium (Ca+2), cytochrome C reduction (CYC-R), lipid peroxidation (LPO), nitric oxide (NO) levels showed a significant increase in serum of treated rats. Regarding the genotoxic parameters, when rats are treated either with CPE-E or CPF-M, liver DNA, chromosomal aberration (CA), and micronucleated polychromatic erythrocytes (MnPCE) significantly increased, while the mitotic index (MI) and polychromatic erythrocytes (PCE)/ normochromatic erythrocytes (NCE) ratio were significantly decreased. However, the administration of NAC following the intoxication of CPF-E or CPF-M attenuated the tested biochemical and genotoxic markers. It can be concluded that NAC can be used to ameliorate the toxicity of certain organophosphorus compounds such as CPF-E and CPF-M.
Collapse
Affiliation(s)
- Khaled A Osman
- Department of Pesticide Chemistry and Technology, Faculty of Agriculture, Alexandria University, Egypt.
| | - Eslam M Ezz El-Din
- Department of Pesticide Chemistry and Technology, Faculty of Agriculture, Alexandria University, Egypt
| | - Nabila S Ahmed
- Department of Pesticide Chemistry and Technology, Faculty of Agriculture, Alexandria University, Egypt
| | - Ayman S El-Seedy
- Laboratory of Cellular and Molecular Genetics, Department of Genetics, Faculty of Agriculture, Alexandria University, P.O Box 21545, Alexandria, Egypt
| |
Collapse
|
26
|
Rodrigues KC, Bortolatto CF, da Motta KP, de Oliveira RL, Paltian JJ, Krüger R, Roman SS, Boeira SP, Alves D, Wilhelm EA, Luchese C. The neurotherapeutic role of a selenium-functionalized quinoline in hypothalamic obese rats. Psychopharmacology (Berl) 2021; 238:1937-1951. [PMID: 33740091 DOI: 10.1007/s00213-021-05821-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 03/08/2021] [Indexed: 12/30/2022]
Abstract
RATIONALE Obesity is considered one of the major global health problems and increases the risk of several medical complications, such as diabetes and mental illnesses. OBJECTIVE The present study investigated the effect of 7-chloro-4-(phenylselanyl) quinoline (4-PSQ) on obesity parameters, behavioral and neurochemical alterations in hypothalamic obese rats. METHODS Male Wistar rats received subcutaneous neonatal injections of monosodium glutamate (MSG, 4g/kg) or saline. After the Lee Index evaluation, rats were divided into groups and treated with 4-PSQ (5 mg/kg, intragastric route) or canola oil once a day (post-natal days (PND) 60→76). Open-field, elevated plus-maze, forced swim task, object recognition/location memory, and stepdown inhibitory avoidance tasks were conducted from PND 66 to 74. On PND 76, rats were euthanized and epididymal fat, blood, cerebral cortex, andhippocampus were removed. Blood biochemical parameters and cortical/hippocampal acetylcholinesterase (AChE) and Na /K -ATPase activities were assessed. RESULTS MSG increased the Lee Index characterizing the chemically induced hypothalamic obesity model. 4-PSQ reversed the increases of epididymal fat, blood glucose, and triglyceride levels caused by MSG exposure. 4-PSQ attenuated anxiety-like and depression-like behaviors induced by neonatal administrations of MSG. Memory deficits found in MSG-obese rats were reversed by treatment with 4-PSQ. Neurochemical alterations produced by MSG evidenced by stimulation ofNa+/K+-ATPase and AChE activities in the cerebral cortex and hippocampus of rats were normalized by 4-PSQ treatment. CONCLUSIONS In brief, 4-PSQ therapy improved hypothalamic obesity-related parameters, as well as psychiatric symptoms, cognitive impairment, and neurochemical alterations found in obese rats.
Collapse
Affiliation(s)
- Karline C Rodrigues
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil
| | - Cristiani F Bortolatto
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Bioquímica e Neurofarmacologia Molecular (LABIONEM), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Pelotas, RS, CEP 96010-900, Brazil
| | - Ketlyn P da Motta
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil
| | - Renata L de Oliveira
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil
| | - Jaini J Paltian
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil
| | - Roberta Krüger
- Programa de Pós-graduação em Química, Laboratório de Síntese Orgânica Limpa - LASOL, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), P.O. Box 354, Pelotas, RS, 96010-900, Brazil
| | - Silvane S Roman
- Universidade Regional Integrada, Campus Erechim, Erechim, RS, CEP 99700-000, Brazil
| | - Silvana P Boeira
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas, LaftamBio Pampa, Universidade Federal do Pampa, Itaqui, RS, CEP 97650-000, Brazil
| | - Diego Alves
- Programa de Pós-graduação em Química, Laboratório de Síntese Orgânica Limpa - LASOL, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), P.O. Box 354, Pelotas, RS, 96010-900, Brazil
| | - Ethel Antunes Wilhelm
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil.
| | - Cristiane Luchese
- Programa de Pós-graduação em Bioquímica e Bioprospecção, Laboratório de Pesquisa em Farmacologia Bioquímica (LaFarBio), Grupo de Pesquisa em Neurobiotecnologia (GPN), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão do Leão, Pelotas, RS, CEP 96010-900, Brazil.
| |
Collapse
|
27
|
Kalia M, Meijer HGE, van Gils SA, van Putten MJAM, Rose CR. Ion dynamics at the energy-deprived tripartite synapse. PLoS Comput Biol 2021; 17:e1009019. [PMID: 34143772 PMCID: PMC8244923 DOI: 10.1371/journal.pcbi.1009019] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 06/30/2021] [Accepted: 04/28/2021] [Indexed: 01/09/2023] Open
Abstract
The anatomical and functional organization of neurons and astrocytes at 'tripartite synapses' is essential for reliable neurotransmission, which critically depends on ATP. In low energy conditions, synaptic transmission fails, accompanied by a breakdown of ion gradients, changes in membrane potentials and cell swelling. The resulting cellular damage and cell death are causal to the often devastating consequences of an ischemic stroke. The severity of ischemic damage depends on the age and the brain region in which a stroke occurs, but the reasons for this differential vulnerability are far from understood. In the present study, we address this question by developing a comprehensive biophysical model of a glutamatergic synapse to identify key determinants of synaptic failure during energy deprivation. Our model is based on fundamental biophysical principles, includes dynamics of the most relevant ions, i.e., Na+, K+, Ca2+, Cl- and glutamate, and is calibrated with experimental data. It confirms the critical role of the Na+/K+-ATPase in maintaining ion gradients, membrane potentials and cell volumes. Our simulations demonstrate that the system exhibits two stable states, one physiological and one pathological. During energy deprivation, the physiological state may disappear, forcing a transit to the pathological state, which can be reverted when blocking voltage-gated Na+ and K+ channels. Our model predicts that the transition to the pathological state is favoured if the extracellular space fraction is small. A reduction in the extracellular space volume fraction, as, e.g. observed with ageing, will thus promote the brain's susceptibility to ischemic damage. Our work provides new insights into the brain's ability to recover from energy deprivation, with translational relevance for diagnosis and treatment of ischemic strokes.
Collapse
Affiliation(s)
- Manu Kalia
- Applied Analysis, Department of Applied Mathematics, University of Twente, Enschede, The Netherlands
- * E-mail:
| | - Hil G. E. Meijer
- Applied Analysis, Department of Applied Mathematics, University of Twente, Enschede, The Netherlands
| | - Stephan A. van Gils
- Applied Analysis, Department of Applied Mathematics, University of Twente, Enschede, The Netherlands
| | | | - Christine R. Rose
- Institute of Neurobiology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
28
|
Chaudhuri T, Chintalapati J, Hosur MV. Identification of 3'-UTR single nucleotide variants and prediction of select protein imbalance in mesial temporal lobe epilepsy patients. PLoS One 2021; 16:e0252475. [PMID: 34086756 PMCID: PMC8177469 DOI: 10.1371/journal.pone.0252475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/16/2021] [Indexed: 11/23/2022] Open
Abstract
The genetic influence in epilepsy, characterized by unprovoked and recurrent seizures, is through variants in genes critical to brain development and function. We have carried out variant calling in Mesial Temporal Lobe Epilepsy (MTLE) patients by mapping the RNA-Seq data available at SRA, NCBI, USA onto human genome assembly hg-19. We have identified 1,75,641 SNVs in patient samples. These SNVs are distributed over 14700 genes of which 655 are already known to be associated with epilepsy. Large number of variants occur in the 3'-UTR, which is one of the regions involved in the regulation of protein translation through binding of miRNAs and RNA-binding proteins (RBP). We have focused on studying the structure-function relationship of the 3'-UTR SNVs that are common to at-least 10 of the 35 patient samples. For the first time we find SNVs exclusively in the 3'-UTR of FGF12, FAR1, NAPB, SLC1A3, SLC12A6, GRIN2A, CACNB4 and FBXO28 genes. Structural modelling reveals that the variant 3'-UTR segments possess altered secondary and tertiary structures which could affect mRNA stability and binding of RBPs to form proper ribonucleoprotein (RNP) complexes. Secondly, these SNVs have either created or destroyed miRNA-binding sites, and molecular modeling reveals that, where binding sites are created, the additional miRNAs bind strongly to 3'-UTR of only variant mRNAs. These two factors affect protein production thereby creating an imbalance in the amounts of select proteins in the cell. We suggest that in the absence of missense and nonsense variants, protein-activity imbalances associated with MTLE patients can be caused through 3'-UTR variants in relevant genes by the mechanisms mentioned above. 3'-UTR SNV has already been identified as causative variant in the neurological disorder, Tourette syndrome. Inhibition of these miRNA-mRNA bindings could be a novel way of treating drug-resistant MTLE patients. We also suggest that joint occurrence of these SNVs could serve as markers for MTLE. We find, in the present study, SNV-mediated destruction of miRNA binding site in the 3'-UTR of the gene encoding glutamate receptor subunit, and, interestingly, overexpression of one of this receptor subunit is also associated with Febrile Seizures.
Collapse
Affiliation(s)
- Tanusree Chaudhuri
- Department of Natural Sciences and Engineering, National Institute of Advanced Studies, IISc campus, Bangalore, India
| | - Janaki Chintalapati
- CDAC-Centre for Development of Advanced Computing, Byappanahalli, Bangalore, India
| | | |
Collapse
|
29
|
Gargas NM, Ethridge VT, Miklasevich MK, Rohan JG. Altered hippocampal function and cytokine levels in a rat model of Gulf War illness. Life Sci 2021; 274:119333. [PMID: 33705732 DOI: 10.1016/j.lfs.2021.119333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 01/09/2023]
Abstract
AIMS Gulf War illness (GWI) is a disorder affecting military personnel deployed in the Gulf War (GW) from 1990 to 1991. Here, we will use a rat model of GWI to evaluate hippocampal function and cytokine levels. MATERIALS AND METHODS Rats were exposed to diethyltoluamide and permethrin via dermal absorption and pyridostigmine bromide via gavage with or without a 5-min restraint for 28 days. Immediate and delayed effects of GW chemical exposure were evaluated using electrophysiology to quantitate hippocampal function, behavioral tests to assess cognitive effects and biochemical assays to measure neurotransmitter and cytokine levels. KEY FINDINGS Behavioral data revealed a statistically significant increase in motor activity at 3 months following completion of exposures, potentially indicating increased excitability, and/or restlessness. Electrophysiology data revealed statistically significant changes in paired pulse facilitation and input-output function of CA1 hippocampal neurons within 24 h and 3 months following completion of exposures. There was also a statistically significant reduction in the frequency of spontaneous firing activity of hippocampal neurons within 24 h following exposures. Naïve hippocampal slices directly incubated in GW chemicals also resulted in similar changes in electrophysiological parameters. Biochemical measurements revealed reduced hippocampal glutamate level at 3 months post-exposure. Furthermore, there was a statistically significant increase in plasma and hippocampal levels of IL-13, as well as decrease in plasma level of IL-1β. SIGNIFICANCE Our data support an effect on glutamate signaling within the hippocampus as indicated by changes in PPF and hippocampal level of glutamate, with some activation of T helper type 2 immune response.
Collapse
Affiliation(s)
- Nathan M Gargas
- Naval Medical Research Unit Dayton, 2728 Q Street, Area B, Building 837, WPAFB, OH 45433, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA; Odyssey Systems Consulting Group, Ltd, 201 Edgewater Drive Suite 270, Wakefield, MA 01880, USA
| | - Victoria T Ethridge
- Naval Medical Research Unit Dayton, 2728 Q Street, Area B, Building 837, WPAFB, OH 45433, USA; Oak Ridge Institute for Science and Education, 1299 Bethel Valley Rd, Oak Ridge, TN 37830, USA
| | - Molly K Miklasevich
- Naval Medical Research Unit Dayton, 2728 Q Street, Area B, Building 837, WPAFB, OH 45433, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817, USA
| | - Joyce G Rohan
- Naval Medical Research Unit Dayton, 2728 Q Street, Area B, Building 837, WPAFB, OH 45433, USA.
| |
Collapse
|
30
|
Moya-Mendez ME, Mueller DM, Pratt M, Bonner M, Elliott C, Hunanyan A, Kucera G, Bock C, Prange L, Jasien J, Keough K, Shashi V, McDonald M, Mikati MA. Early onset severe ATP1A2 epileptic encephalopathy: Clinical characteristics and underlying mutations. Epilepsy Behav 2021; 116:107732. [PMID: 33493807 PMCID: PMC7940561 DOI: 10.1016/j.yebeh.2020.107732] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/09/2020] [Accepted: 12/20/2020] [Indexed: 01/26/2023]
Abstract
BACKGROUND ATP1A2 mutations cause hemiplegic migraine with or without epilepsy or acute reversible encephalopathy. Typical onset is in adulthood or older childhood without subsequent severe long-term developmental impairments. AIM We aimed to describe the manifestations of early onset severe ATP1A2-related epileptic encephalopathy and its underlying mutations in a cohort of seven patients. METHODS A retrospective chart review of a cohort of seven patients was conducted. Response to open-label memantine therapy, used off-label due to its NMDA receptor antagonist effects, was assessed by the Global Rating Scale of Change (GRSC) and Clinical Global Impression Scale of Improvement (CGI-I) methodologies. Molecular modeling was performed using PyMol program. RESULTS Patients (age 2.5-20 years) had symptom onset at an early age (6 days-1 year). Seizures were either focal or generalized. Common features were: drug resistance, recurrent status epilepticus, etc., severe developmental delay with episodes of acute severe encephalopathy often with headaches, dystonias, hemiplegias, seizures, and developmental regression. All had variants predicted to be disease causing (p.Ile293Met, p.Glu1000Lys, c.1017+5G>A, p.Leu809Arg, and 3 patients with p.Met813Lys). Modeling revealed that mutations interfered with ATP1A2 ion binding and translocation sites. Memantine, given to five, was tolerated in all (mean treatment: 2.3 years, range 6 weeks-4.8 years) with some improvements reported in all five. CONCLUSIONS Our observations describe a distinctive clinical profile of seven unrelated probands with early onset severe ATP1A2-related epileptic encephalopathy, provide insights into structure-function relationships of ATP1A2 mutations, and support further studies of NMDAR antagonist therapy in ATP1A2-encephalopathy.
Collapse
Affiliation(s)
- Mary E. Moya-Mendez
- Department of Pediatrics, Division of Pediatric Neurology and Developmental Medicine, Duke University, Durham, NC, United States
| | - David M. Mueller
- Center for Genetic Diseases, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, Chicago, IL, United States
| | - Milton Pratt
- Department of Pediatrics, Division of Pediatric Neurology and Developmental Medicine, Duke University, Durham, NC, United States
| | - Melanie Bonner
- Department of Psychiatry, Duke University, Durham, NC, United States
| | - Courtney Elliott
- Department of Pediatrics, Division of Pediatric Neurology and Developmental Medicine, Duke University, Durham, NC, United States
| | - Arsen Hunanyan
- Department of Pediatrics, Division of Pediatric Neurology and Developmental Medicine, Duke University, Durham, NC, United States
| | - Gary Kucera
- Duke Cancer Institute Rodent Cancer Models Shared Resource, Duke University Medical Center, Durham, NC, United States
| | - Cheryl Bock
- Duke Cancer Institute Rodent Cancer Models Shared Resource, Duke University Medical Center, Durham, NC, United States
| | - Lyndsey Prange
- Department of Pediatrics, Division of Pediatric Neurology and Developmental Medicine, Duke University, Durham, NC, United States
| | - Joan Jasien
- Department of Pediatrics, Division of Pediatric Neurology and Developmental Medicine, Duke University, Durham, NC, United States
| | - Karen Keough
- Dell Medical School at the University of Texas, Austin TX, United States
| | - Vandana Shashi
- Dell Medical School at the University of Texas, Austin TX, United States
| | - Marie McDonald
- Dell Medical School at the University of Texas, Austin TX, United States
| | - Mohamad A. Mikati
- Department of Pediatrics, Division of Pediatric Neurology and Developmental Medicine, Duke University, Durham, NC, United States,Department of Neurobiology, Duke University, Durham, NC, United States,Corresponding Author: Mohamad Mikati, MD, Children Health Center, T913J, Duke University Medical Center, 2301 Erwin Road, Durham, NC 27710, Phone: 919-668-4073, Fax: 919-681-8943,
| |
Collapse
|
31
|
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
|
32
|
García-Hernández JL, Corchete LA, Marcos-Alcalde Í, Gómez-Puertas P, Fons C, Lazo PA. Pathogenic convergence of CNVs in genes functionally associated to a severe neuromotor developmental delay syndrome. Hum Genomics 2021; 15:11. [PMID: 33557955 PMCID: PMC7871650 DOI: 10.1186/s40246-021-00309-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/26/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Complex developmental encephalopathy syndromes might be the consequence of unknown genetic alterations that are likely to contribute to the full neurological phenotype as a consequence of pathogenic gene combinations. METHODS To identify the additional genetic contribution to the neurological phenotype, we studied as a test case a boy, with a KCNQ2 exon-7 partial duplication, by single-nucleotide polymorphism (SNP) microarray to detect copy-number variations (CNVs). RESULTS The proband presented a cerebral palsy like syndrome with a severe motor and developmental encephalopathy. The SNP array analysis detected in the proband several de novo CNVs, nine partial gene losses (LRRC55, PCDH9, NALCN, RYR3, ELAVL2, CDH13, ATP1A2, SLC17A5, ANO3), and two partial gene duplications (PCDH19, EFNA5). The biological functions of these genes are associated with ion channels such as calcium, chloride, sodium, and potassium with several membrane proteins implicated in neural cell-cell interactions, synaptic transmission, and axon guidance. Pathogenically, these functions can be associated to cerebral palsy, seizures, dystonia, epileptic crisis, and motor neuron dysfunction, all present in the patient. CONCLUSIONS Severe motor and developmental encephalopathy syndromes of unknown origin can be the result of a phenotypic convergence by combination of several genetic alterations in genes whose physiological function contributes to the neurological pathogenic mechanism.
Collapse
Affiliation(s)
- Juan L García-Hernández
- Molecular Mechanisms of Cancer Program, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Salamanca, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Departamento de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Luis A Corchete
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Departamento de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain.,Network Center for Biomedical Research in Cancer (CIBERONC), Salamanca, Spain
| | - Íñigo Marcos-Alcalde
- Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain.,Biosciences Research Institute, School of Experimental Sciences, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - Paulino Gómez-Puertas
- Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - Carmen Fons
- Neurology Department, Hospital Sant Joan de Déu, Sant Joan de Déu Research Institute, Esplugues de Llobregat, Barcelona and CIBERER, Instituto de Salud Carlos III, Barcelona, Spain.
| | - Pedro A Lazo
- Molecular Mechanisms of Cancer Program, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Salamanca, Salamanca, Spain. .,Instituto de Investigación Biomédica de Salamanca (IBSAL), Departamento de Hematología, Hospital Universitario de Salamanca, Salamanca, Spain.
| |
Collapse
|
33
|
Decreased content of ascorbic acid (vitamin C) in the brain of knockout mouse models of Na+,K+-ATPase-related neurologic disorders. PLoS One 2021; 16:e0246678. [PMID: 33544780 PMCID: PMC7864419 DOI: 10.1371/journal.pone.0246678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 01/23/2021] [Indexed: 12/21/2022] Open
Abstract
Na+,K+-ATPase is a crucial protein responsible for maintaining the electrochemical gradients across the cell membrane. The Na+,K+-ATPase is comprised of catalytic α, β, and γ subunits. In adult brains, the α3 subunit, encoded by ATP1A3, is predominantly expressed in neurons, whereas the α2 subunit, encoded by ATP1A2, is expressed in glial cells. In foetal brains, the α2 is expressed in neurons as well. Mutations in α subunits cause a variety of neurologic disorders. Notably, the onset of symptoms in ATP1A2- and ATP1A3-related neurologic disorders is usually triggered by physiological or psychological stressors. To gain insight into the distinct roles of the α2 and α3 subunits in the developing foetal brain, whose developmental dysfunction may be a predisposing factor of neurologic disorders, we compared the phenotypes of mouse foetuses with double homozygous knockout of Atp1a2 and Atp1a3 (α2α3-dKO) to those with single knockout. The brain haemorrhage phenotype of α2α3-dKO was similar to that of homozygous knockout of the gene encoding ascorbic acid (ASC or vitamin C) transporter, SVCT2. The α2α3-dKO brain showed significantly decreased level of ASC compared with the wild-type (WT) and single knockout. We found that the ASC content in the basal ganglia and cerebellum was significantly lower in the adult Atp1a3 heterozygous knockout mouse (α3-HT) than in the WT. Interestingly, we observed a significant decrease in the ASC level in the basal ganglia and cerebellum of α3-HT in the peripartum period, during which mice are under physiological stress. These observations indicate that the α2 and α3 subunits independently contribute to the ASC level in the foetal brain and that the α3 subunit contributes to ASC transport in the adult basal ganglia and cerebellum. We propose that decreases in ASC levels may affect neural network development and are linked to the pathophysiology of ATP1A2- and ATP1A3-related neurologic disorders.
Collapse
|
34
|
Lowry CA, Golod ME, Andrew RD, Bennett BM. Expression of Neuronal Na +/K +-ATPase α Subunit Isoforms in the Mouse Brain Following Genetically Programmed or Behaviourally-induced Oxidative Stress. Neuroscience 2020; 442:202-215. [PMID: 32653541 DOI: 10.1016/j.neuroscience.2020.07.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 06/21/2020] [Accepted: 07/04/2020] [Indexed: 12/12/2022]
Abstract
The Na+/K+-ATPase is a transmembrane ion pump that has a critical homeostatic role within every mammalian cell; however, it is vulnerable to the effects of increased oxidative stress. Understanding how expression of this transporter is influenced by oxidative stress may yield insight into its role in the pathophysiology of neurological and neuropsychiatric diseases. In this study we investigated whether increased oxidative stress could influence Na+/K+-ATPase expression in various brain regions of mice. We utilized two different models of oxidative stress: a behavioural chronic unpredictable stress protocol and the Aldh2-/- mouse model of oxidative stress-based and age-related cognitive impairment. We identified distinct regional baseline mRNA and protein expression patterns of the Na+/K+-ATPase α1 and α3 isoforms within the neocortex, hippocampus, and brainstem of wildtype mice. Consistent with previous studies, there was a higher proportion of α3 expression relative to α1 in the brainstem versus neocortex, but a higher proportion of α1 expression relative to α3 in the neocortex versus the brainstem. The hippocampus had similar expression levels of both α1 and α3. Despite increased staining for oxidative stress in higher brain, no differences in α1 or α3 expression were noted in Aldh2-/- mice versus wildtype, or in mice exposed to a 28-day chronic unpredictable stress protocol. In both models of oxidative stress, gene and protein expression of Na+/K+-ATPase α1 and α3 isoforms within the higher and lower brain was remarkably stable. Thus, Na+/K+-ATPase function previously reported as altered by oxidative stress is not through induced changes in the expression of pump isoforms.
Collapse
Affiliation(s)
- Chloe A Lowry
- Centre for Neuroscience Studies, Queen's University, 18 Stuart St., Kingston, Ontario K7L 3N6, Canada.
| | - Michael E Golod
- Centre for Neuroscience Studies, Queen's University, 18 Stuart St., Kingston, Ontario K7L 3N6, Canada.
| | - R David Andrew
- Centre for Neuroscience Studies, Queen's University, 18 Stuart St., Kingston, Ontario K7L 3N6, Canada; Department of Biomedical and Molecular Sciences, Queen's University, 18 Stuart St., Kingston, Ontario K7L 3N6, Canada.
| | - Brian M Bennett
- Centre for Neuroscience Studies, Queen's University, 18 Stuart St., Kingston, Ontario K7L 3N6, Canada; Department of Biomedical and Molecular Sciences, Queen's University, 18 Stuart St., Kingston, Ontario K7L 3N6, Canada.
| |
Collapse
|
35
|
Felix L, Delekate A, Petzold GC, Rose CR. Sodium Fluctuations in Astroglia and Their Potential Impact on Astrocyte Function. Front Physiol 2020; 11:871. [PMID: 32903427 PMCID: PMC7435049 DOI: 10.3389/fphys.2020.00871] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
Astrocytes are the main cell type responsible for the regulation of brain homeostasis, including the maintenance of ion gradients and neurotransmitter clearance. These processes are tightly coupled to changes in the intracellular sodium (Na+) concentration. While activation of the sodium-potassium-ATPase (NKA) in response to an elevation of extracellular K+ may decrease intracellular Na+, the cotransport of transmitters, such as glutamate, together with Na+ results in an increase in astrocytic Na+. This increase in intracellular Na+ can modulate, for instance, metabolic downstream pathways. Thereby, astrocytes are capable to react on a fast time scale to surrounding neuronal activity via intracellular Na+ fluctuations and adjust energy production to the demand of their environment. Beside the well-documented conventional roles of Na+ signaling mainly mediated through changes in its electrochemical gradient, several recent studies have identified more atypical roles for Na+, including protein interactions leading to changes in their biochemical activity or Na+-dependent regulation of gene expression. In this review, we will address both the conventional as well as the atypical functions of astrocytic Na+ signaling, presenting the role of transporters and channels involved and their implications for physiological processes in the central nervous system (CNS). We will also discuss how these important functions are affected under pathological conditions, including stroke and migraine. We postulate that Na+ is an essential player not only in the maintenance of homeostatic processes but also as a messenger for the fast communication between neurons and astrocytes, adjusting the functional properties of various cellular interaction partners to the needs of the surrounding network.
Collapse
Affiliation(s)
- Lisa Felix
- Institute of Neurobiology, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Andrea Delekate
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Division of Vascular Neurology, Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Christine R Rose
- Institute of Neurobiology, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
36
|
Li WX, Li GH, Tong X, Yang PP, Huang JF, Xu L, Dai SX. Systematic metabolic analysis of potential target, therapeutic drug, diagnostic method and animal model applicability in three neurodegenerative diseases. Aging (Albany NY) 2020; 12:9882-9914. [PMID: 32461378 PMCID: PMC7288927 DOI: 10.18632/aging.103253] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/20/2020] [Indexed: 01/10/2023]
Abstract
Considerable evidence suggests that metabolic abnormalities are associated with neurodegenerative diseases. This study aimed to conduct a systematic metabolic analysis of Alzheimer’s disease (AD), Parkinson’s disease (PD) and Huntington’s disease (HD). Human and mouse model microarray datasets were downloaded from the Gene Expression Omnibus database. The metabolic genes and pathways were collected from the Recon 3D human metabolic model. Drug and target information was obtained from the DrugBank database. This study identified ATP1A1, ATP6V1G2, GOT1, HPRT1, MAP2K1, PCMT1 and PLK2 as key metabolic genes that were downregulated in AD, PD and HD. We screened 57 drugs that target these genes, such as digoxin, ouabain and diazoxide. This study constructed multigene diagnostic models for AD, PD and HD by using metabolic gene expression profiles in blood, all models showed high accuracy (AUC > 0.8) both in the experimental and validation sets. Furthermore, analysis of animal models showed that there was almost no consistency among the metabolic changes between mouse models and human diseases. This study systematically revealed the metabolic damage among AD, PD, and HD and uncovered the differences between animal models and human diseases. This information may be helpful for understanding the metabolic mechanisms and drug development for neurodegenerative diseases.
Collapse
Affiliation(s)
- Wen-Xing Li
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, Yunnan, China
| | - Gong-Hua Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China
| | - Xin Tong
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, Yunnan, China
| | - Peng-Peng Yang
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, Yunnan, China
| | - Jing-Fei Huang
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, Yunnan, China
| | - Lin Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, Yunnan, China.,Centre for Excellence in Brain Science and Intelligent Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shao-Xing Dai
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, Yunnan, China
| |
Collapse
|
37
|
Yoneda JS, Sebinelli HG, Itri R, Ciancaglini P. Overview on solubilization and lipid reconstitution of Na,K-ATPase: enzyme kinetic and biophysical characterization. Biophys Rev 2020; 12:49-64. [PMID: 31955383 DOI: 10.1007/s12551-020-00616-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 01/07/2020] [Indexed: 12/11/2022] Open
Abstract
Na,K-ATPase is a membrane protein which plays a vital role. It pumps Na+ and K+ ions across the cellular membranes using energy from ATP hydrolysis, and is responsible for maintaining the osmotic equilibrium and generating the membrane potential. Moreover, Na,K-ATPase has also been involved in cell signaling, interacting with partner proteins. Cardiotonic steroids bind specifically to Na,K-ATPase triggering a number of signaling pathways. Because of its importance, many efforts have been employed to study the structure and function of this protein. Difficulties associated with its removal from natural membranes and the concomitant search for appropriate replacement conditions to keep the protein in solution have presented a challenge that had to be overcome prior to carrying out biophysical and biochemical studies in vitro. In this review, we summarized all of the methods and techniques applied by our group in order to obtain information about Na,K-ATPase in respect to solubilization, reconstitution into mimetic system, influence of lipid composition, stability, oligomerization, and aggregation.
Collapse
Affiliation(s)
- Juliana Sakamoto Yoneda
- Instituto de Fisica, Universidade de Sao Paulo, Rua do Matao, 1371, 05508-090, Sao Paulo, SP, Brazil.
| | - Heitor Gobbi Sebinelli
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto da Universidade de São Paulo (FFCLRP-USP), Ribeirão Preto, SP, 14040-901, Brazil
| | - Rosangela Itri
- Instituto de Fisica, Universidade de Sao Paulo, Rua do Matao, 1371, 05508-090, Sao Paulo, SP, Brazil
| | - Pietro Ciancaglini
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto da Universidade de São Paulo (FFCLRP-USP), Ribeirão Preto, SP, 14040-901, Brazil
| |
Collapse
|
38
|
de Souza Gonçalves B, de Moura Valadares JM, Alves SLG, Silva SC, Rangel LP, Cortes VF, Villar JAFP, Barbosa LA, de Lima Santos H. Evaluation of neuroprotective activity of digoxin and semisynthetic derivatives against partial chemical ischemia. J Cell Biochem 2019; 120:17108-17122. [PMID: 31310381 DOI: 10.1002/jcb.28971] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 03/29/2019] [Accepted: 04/08/2019] [Indexed: 02/06/2023]
Abstract
Recently, cardiotonic steroids (CTS) have been shown to lead to the activation of Na,K-ATPase at low concentrations in brain, promoting neuroprotection against ischemia. We report here the results of the use of digoxin and its semisynthetic derivatives BD-14, BD-15, and BD-16 against partial chemical ischemic induction followed by reperfusion in murine neuroblastoma cells neuro-2a (N2a). For chemical ischemic induction, sodium azide (5 mM) was used for 5 hours, and then reperfusion was induced for 24 hours. Na,K-ATPase activity and protein levels were analyzed in membrane preparation of N2a cells pretreated with the compounds (150 nM), in the controls and in induced chemical ischemia. In the Na,K-ATPase activity and protein levels assays, the steroids digoxin and BD-15 demonstrated a capacity to modulate the activity of the enzyme directly, increasing its levels of expression and activity. Oxidative parameters, such as superoxide dismutase (SOD) activity, lipid peroxidation (thiobarbituric acid reactive substance), glutathione peroxidase (GPx), glutathione (GSH) levels, hydrogen peroxide content, and the amount of free radicals (reactive oxygen species) during induced chemical ischemia were also evaluated. Regarding the redox state, lipid peroxidation, hydrogen peroxide content, and GPx activity, we have observed an increase in the chemical ischemic group, and a reduction in the groups treated with CTS. SOD activity increased in all treated groups when compared to control and GSH levels decreased when treated with sodium azide and did not change with CTS treatments. Regarding the lipid profile, we saw a decrease in the content of phospholipids and cholesterol in the chemical ischemic group, and an increase in the groups treated with CTS. In conclusion, the compounds used in this study demonstrate promising results, since they appear to promote neuroprotection in cells exposed to chemical ischemia.
Collapse
Affiliation(s)
- Bruno de Souza Gonçalves
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Divinópolis, Minas Gerais, Brazil
| | | | - Silmara L G Alves
- Laboratório de Síntese Orgânica e Nanoestruturas, Universidade Federal de São João del Rei, Divinópolis, Minas Gerais, Brazil
| | - Simone C Silva
- Laboratório de Síntese Orgânica e Nanoestruturas, Universidade Federal de São João del Rei, Divinópolis, Minas Gerais, Brazil
| | - Luciana P Rangel
- Laboratório de Bioquímica Tumoral, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vanessa F Cortes
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Divinópolis, Minas Gerais, Brazil
| | - José A F P Villar
- Laboratório de Síntese Orgânica e Nanoestruturas, Universidade Federal de São João del Rei, Divinópolis, Minas Gerais, Brazil
| | - Leandro A Barbosa
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Divinópolis, Minas Gerais, Brazil
| | - Hérica de Lima Santos
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Divinópolis, Minas Gerais, Brazil
| |
Collapse
|
39
|
Schvartz D, González-Ruiz V, Walter N, Antinori P, Jeanneret F, Tonoli D, Boccard J, Zurich MG, Rudaz S, Monnet-Tschudi F, Sandström J, Sanchez JC. Protein pathway analysis to study development-dependent effects of acute and repeated trimethyltin (TMT) treatments in 3D rat brain cell cultures. Toxicol In Vitro 2019; 60:281-292. [PMID: 31176792 DOI: 10.1016/j.tiv.2019.05.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/18/2019] [Accepted: 05/29/2019] [Indexed: 11/25/2022]
Abstract
Trimethyltin is an organometallic compound, described to be neurotoxic and to trigger neuroinflammation and oxidative stress. Previous studies associated TMT with the perturbation of mitochondrial function, or neurotransmission. However, the mechanisms of toxicity may differ depending on the duration of exposure and on the stage of maturation of brain cells. This study aim at elucidating whether the toxicity pathways triggered by a known neurotoxicant (TMT) differs depending on cell maturation stage or duration of exposure. To this end omics profiling of immature and differentiated 3D rat brain cell cultures exposed for 24 h or 10 days (10-d) to 0.5 and 1 μM of TMT was performed to better understand the underlying mechanisms of TMT associated toxicity. Proteomics identified 55 and 17 proteins affected by acute TMT treatment in immature and differentiated cultures respectively, while 10-day treatment altered 96 proteins in immature cultures versus 353 in differentiated. The results suggest different sensitivity to TMT depending on treatment duration and cell maturation. In accordance with known TMT mechanisms oxidative stress and neuroinflammation was observed after 10-d treatment at both maturation stages, whereas the neuroinflammatory process was more prominent in differentiated cultures than in the immature, no development-dependent difference could be detected for oxidative stress or synaptic neurodegeneration. Pathway analysis revealed that both vesicular trafficking and the synaptic machinery were strongly affected by 10-d TMT treatment in both maturation stages, as was GABAergic and glutamatergic neurotransmission. This study shows that omics approaches combined with pathway analysis constitutes an improved tool-set in elucidating toxicity mechanisms.
Collapse
Affiliation(s)
- Domitille Schvartz
- Translational Biomarker Group, Department of Internal Medicine Specialties, University of Geneva, Geneva, Switzerland; Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland
| | - Víctor González-Ruiz
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland; Analytical Sciences, School of Pharmaceutical Sciences, Universities of Geneva and Lausanne, Geneva, Switzerland
| | - Nadia Walter
- Translational Biomarker Group, Department of Internal Medicine Specialties, University of Geneva, Geneva, Switzerland; Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland
| | - Paola Antinori
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland; Neuroproteomics group, Department of Clinical Neurosciences, University of Geneva, Geneva, Switzerland
| | - Fabienne Jeanneret
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland; Analytical Sciences, School of Pharmaceutical Sciences, Universities of Geneva and Lausanne, Geneva, Switzerland
| | - David Tonoli
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland; Analytical Sciences, School of Pharmaceutical Sciences, Universities of Geneva and Lausanne, Geneva, Switzerland
| | - Julien Boccard
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland; Analytical Sciences, School of Pharmaceutical Sciences, Universities of Geneva and Lausanne, Geneva, Switzerland
| | - Marie-Gabrielle Zurich
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland; Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Serge Rudaz
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland; Analytical Sciences, School of Pharmaceutical Sciences, Universities of Geneva and Lausanne, Geneva, Switzerland
| | - Florianne Monnet-Tschudi
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland; Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Jenny Sandström
- Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland; Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Jean-Charles Sanchez
- Translational Biomarker Group, Department of Internal Medicine Specialties, University of Geneva, Geneva, Switzerland; Swiss Centre for Applied Human Toxicology (SCAHT), Switzerland.
| |
Collapse
|
40
|
Waugh DT. Fluoride Exposure Induces Inhibition of Sodium-and Potassium-Activated Adenosine Triphosphatase (Na +, K +-ATPase) Enzyme Activity: Molecular Mechanisms and Implications for Public Health. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:E1427. [PMID: 31010095 PMCID: PMC6518254 DOI: 10.3390/ijerph16081427] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/02/2019] [Accepted: 04/08/2019] [Indexed: 12/24/2022]
Abstract
In this study, several lines of evidence are provided to show that Na + , K + -ATPase activity exerts vital roles in normal brain development and function and that loss of enzyme activity is implicated in neurodevelopmental, neuropsychiatric and neurodegenerative disorders, as well as increased risk of cancer, metabolic, pulmonary and cardiovascular disease. Evidence is presented to show that fluoride (F) inhibits Na + , K + -ATPase activity by altering biological pathways through modifying the expression of genes and the activity of glycolytic enzymes, metalloenzymes, hormones, proteins, neuropeptides and cytokines, as well as biological interface interactions that rely on the bioavailability of chemical elements magnesium and manganese to modulate ATP and Na + , K + -ATPase enzyme activity. Taken together, the findings of this study provide unprecedented insights into the molecular mechanisms and biological pathways by which F inhibits Na + , K + -ATPase activity and contributes to the etiology and pathophysiology of diseases associated with impairment of this essential enzyme. Moreover, the findings of this study further suggest that there are windows of susceptibility over the life course where chronic F exposure in pregnancy and early infancy may impair Na + , K + -ATPase activity with both short- and long-term implications for disease and inequalities in health. These findings would warrant considerable attention and potential intervention, not to mention additional research on the potential effects of F intake in contributing to chronic disease.
Collapse
Affiliation(s)
- Declan Timothy Waugh
- EnviroManagement Services, 11 Riverview, Doherty's Rd, P72 YF10 Bandon, Co. Cork, Ireland.
| |
Collapse
|
41
|
Ferrari F, Viscardi P, Gorini A, Villa RF. Synaptic ATPases system of rat frontal cerebral cortex during aging. Neurosci Lett 2019; 694:74-79. [DOI: 10.1016/j.neulet.2018.11.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 11/01/2018] [Accepted: 11/19/2018] [Indexed: 01/28/2023]
|
42
|
Novaes LS, dos Santos NB, Dragunas G, Perfetto JG, Leza JC, Scavone C, Munhoz CD. Repeated Restraint Stress Decreases Na,K-ATPase Activity via Oxidative and Nitrosative Damage in the Frontal Cortex of Rats. Neuroscience 2018; 393:273-283. [PMID: 30316912 DOI: 10.1016/j.neuroscience.2018.09.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/23/2018] [Accepted: 09/26/2018] [Indexed: 12/23/2022]
|
43
|
Proquin H, Jetten MJ, Jonkhout MCM, Garduño-Balderas LG, Briedé JJ, de Kok TM, van Loveren H, Chirino YI. Transcriptomics analysis reveals new insights in E171-induced molecular alterations in a mouse model of colon cancer. Sci Rep 2018; 8:9738. [PMID: 29950665 PMCID: PMC6021444 DOI: 10.1038/s41598-018-28063-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 06/15/2018] [Indexed: 12/19/2022] Open
Abstract
Titanium dioxide as a food additive (E171) has been demonstrated to facilitate growth of chemically induced colorectal tumours in vivo and induce transcriptomic changes suggestive of an immune system impairment and cancer development. The present study aimed to investigate the molecular mechanisms behind the tumour stimulatory effects of E171 in combination with azoxymethane (AOM)/dextran sodium sulphate (DSS) and compare these results to a recent study performed under the same conditions with E171 only. BALB/c mice underwent exposure to 5 mg/kgbw/day of E171 by gavage for 2, 7, 14, and 21 days. Whole genome mRNA microarray analyses on the distal colon were performed. The results show that E171 induced a downregulation of genes involved in the innate and adaptive immune system, suggesting impairment of this system. In addition, over time, signalling genes involved in colorectal cancer and other types of cancers were modulated. In relation to cancer development, effects potentially associated with oxidative stress were observed through modulation of genes related to antioxidant production. E171 affected genes involved in biotransformation of xenobiotics which can form reactive intermediates resulting in toxicological effects. These transcriptomics data reflect the early biological responses induced by E171 which precede tumour formation in an AOM/DSS mouse model.
Collapse
Affiliation(s)
- Héloïse Proquin
- Department of Toxicogenomics, GROW institute of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.
| | - Marlon J Jetten
- Department of Toxicogenomics, GROW institute of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Marloes C M Jonkhout
- Department of Toxicogenomics, GROW institute of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | | | - Jacob J Briedé
- Department of Toxicogenomics, GROW institute of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Theo M de Kok
- Department of Toxicogenomics, GROW institute of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Henk van Loveren
- Department of Toxicogenomics, GROW institute of Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Yolanda I Chirino
- Laboratorio de Carcinogénesis y Toxicología, Unidad de Biomedicina, FES-Iztacala, UNAM, Estado de México, Mexico.,IUF-Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, DE Düsseldorf, Germany
| |
Collapse
|
44
|
Burjanadze G, Shengelia M, Dachanidze N, Mikadze M, Menabde K, Koshoridze N. Creatine–facilitated protection of stress caused by disrupted circadian rhythm. BIOL RHYTHM RES 2018. [DOI: 10.1080/09291016.2017.1333198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- George Burjanadze
- Faculty of Exact and Natural Sciences, Department of Biology, Ivane Javakhishvili Tbilisi State University, Tbilisi, Georgia
| | - Mariam Shengelia
- Faculty of Exact and Natural Sciences, Department of Biology, Ivane Javakhishvili Tbilisi State University, Tbilisi, Georgia
| | - Natalia Dachanidze
- Faculty of Exact and Natural Sciences, Department of Biology, Ivane Javakhishvili Tbilisi State University, Tbilisi, Georgia
| | - Mariam Mikadze
- US MD Program, Tbilisi State Medical University, Tbilisi, Georgia
| | - Ketevan Menabde
- Faculty of Exact and Natural Sciences, Department of Biology, Ivane Javakhishvili Tbilisi State University, Tbilisi, Georgia
| | - Nana Koshoridze
- Faculty of Exact and Natural Sciences, Department of Biology, Ivane Javakhishvili Tbilisi State University, Tbilisi, Georgia
| |
Collapse
|
45
|
Cerebral Malaria Causes Enduring Behavioral and Molecular Changes in Mice Brain Without Causing Gross Histopathological Damage. Neuroscience 2017; 369:66-75. [PMID: 29113928 DOI: 10.1016/j.neuroscience.2017.10.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/11/2017] [Accepted: 10/30/2017] [Indexed: 01/13/2023]
Abstract
Malaria, parasitic disease considered a major health public problem, is caused by Plasmodium protozoan genus and transmitted by the bite of infected female Anopheles mosquito genus. Cerebral malaria (CM) is the most severe presentation of malaria, caused by P. falciparum and responsible for high mortality and enduring development of cognitive deficits which may persist even after cure and cessation of therapy. In the present study we evaluated selected behavioral, neurochemical and neuropathologic parameters after rescue from experimental cerebral malaria caused by P. berghei ANKA in C57BL/6 mice. Behavioral tests showed impaired nest building activity as well as increased marble burying, indicating that natural behavior of mice remains altered even after cure of infection. Regarding the neurochemical data, we found decreased α2/α3 Na+,K+-ATPase activity and increased immunoreactivity of phosphorylated Na+,K+-ATPase at Ser943 in cerebral cortex after CM. In addition, [3H]-Flunitrazepam binding assays revealed a decrease of benzodiazepine/GABAA receptor binding sites in infected animals. Moreover, in hippocampus, dot blot analysis revealed increased levels of protein carbonyls, suggesting occurrence of oxidative damage to proteins. Interestingly, no changes in the neuropathological markers Fluoro-Jade C, Timm staining or IBA-1 were detected. Altogether, present data indicate that behavioral and neurochemical alterations persist even after parasitemia clearance and CM recovery, which agrees with available clinical findings. Some of the molecular mechanisms reported in the present study may underlie the behavioral changes and increased seizure susceptibility that persist after recovery from CM and may help in the future development of therapeutic strategies for CM sequelae.
Collapse
|
46
|
Mast N, Lin JB, Anderson KW, Bjorkhem I, Pikuleva IA. Transcriptional and post-translational changes in the brain of mice deficient in cholesterol removal mediated by cytochrome P450 46A1 (CYP46A1). PLoS One 2017; 12:e0187168. [PMID: 29073233 PMCID: PMC5658173 DOI: 10.1371/journal.pone.0187168] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 10/13/2017] [Indexed: 01/12/2023] Open
Abstract
Cytochrome P450 46A1 (CYP46A1) converts cholesterol to 24-hydroxycholesterol and thereby controls the major pathways of cholesterol removal from the brain. Cyp46a1-/- mice have a reduction in the rate of cholesterol biosynthesis in the brain and significant impairments to memory and learning. To gain insights into the mechanisms underlying Cyp46a1-/- phenotype, we used Cyp46a1-/- mice and quantified their brain sterol levels and the expression of the genes pertinent to cholesterol homeostasis. We also compared the Cyp46a1-/- and wild type brains for protein phosphorylation and ubiquitination. The data obtained enable the following inferences. First, there seems to be a compensatory upregulation in the Cyp46a1-/- brain of the pathways of cholesterol storage and CYP46A1-independent removal. Second, transcriptional regulation of the brain cholesterol biosynthesis via sterol regulatory element binding transcription factors is not significantly activated in the Cyp46a1-/- brain to explain a compensatory decrease in cholesterol biosynthesis. Third, some of the liver X receptor target genes (Abca1) are paradoxically upregulated in the Cyp46a1-/- brain, possibly due to a reduced activation of the small GTPases RAB8, CDC42, and RAC as a result of a reduced phosphorylation of RAB3IP and PAK1. Fourth, the phosphorylation of many other proteins (a total of 146) is altered in the Cyp46a1-/- brain, including microtubule associated and neurofilament proteins (the MAP and NEF families) along with proteins related to synaptic vesicles and synaptic neurotransmission (e.g., SLCs, SHANKs, and BSN). Fifth, the extent of protein ubiquitination is increased in the Cyp46a1-/- brain, and the affected proteins pertain to ubiquitination (UBE2N), cognition (STX1B and ATP1A2), cytoskeleton function (TUBA1A and YWHAZ), and energy production (ATP1A2 and ALDOA). The present study demonstrates the diverse potential effects of CYP46A1 deficiency on brain functions and identifies important proteins that could be affected by this deficiency.
Collapse
Affiliation(s)
- Natalia Mast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Joseph B. Lin
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Kyle W. Anderson
- Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland, United States of America
- Institute for Bioscience and Biotechnology Research, Rockville, Maryland, United States of America
| | - Ingemar Bjorkhem
- Department of Laboratory Medicine, Division of Clinical Chemistry, Karolinska Institute, Huddinge, Sweden
| | - Irina A. Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
47
|
Hainque E, Caillet S, Leroy S, Flamand-Roze C, Adanyeguh I, Charbonnier-Beaupel F, Retail M, Le Toullec B, Atencio M, Rivaud-Péchoux S, Brochard V, Habarou F, Ottolenghi C, Cormier F, Méneret A, Ruiz M, Doulazmi M, Roubergue A, Corvol JC, Vidailhet M, Mochel F, Roze E. A randomized, controlled, double-blind, crossover trial of triheptanoin in alternating hemiplegia of childhood. Orphanet J Rare Dis 2017; 12:160. [PMID: 28969699 PMCID: PMC5625655 DOI: 10.1186/s13023-017-0713-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 09/25/2017] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Based on the hypothesis of a brain energy deficit, we investigated the safety and efficacy of triheptanoin on paroxysmal episodes in patients with alternating hemiplegia of childhood due to ATP1A3 mutations. METHODS We conducted a randomized, double-blind, placebo-controlled crossover study of triheptanoin, at a target dose corresponding to 30% of daily calorie intake, in ten patients with alternating hemiplegia of childhood due to ATP1A3 mutations. Each treatment period consisted of a 12-week fixed-dose phase, separated by a 4-week washout period. The primary outcome was the total number of paroxysmal events. Secondary outcomes included the number of paroxysmal motor-epileptic events; a composite score taking into account the number, severity and duration of paroxysmal events; interictal neurological manifestations; the clinical global impression-improvement scale (CGI-I); and safety parameters. The paired non-parametric Wilcoxon test was used to analyze treatment effects. RESULTS In an intention-to-treat analysis, triheptanoin failed to reduce the total number of paroxysmal events (p = 0.646), including motor-epileptic events (p = 0.585), or the composite score (p = 0.059). CGI-I score did not differ between triheptanoin and placebo periods. Triheptanoin was well tolerated. CONCLUSIONS Triheptanoin does not prevent paroxysmal events in Alternating hemiplegia of childhood. We show the feasibility of a randomized placebo-controlled trial in this setting. TRIAL REGISTRATION The study has been registered with clinicaltrials.gov ( NCT002408354 ) the 03/24/2015.
Collapse
Affiliation(s)
- Elodie Hainque
- Université de la Sorbonne, UPMC Paris 06, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moëlle, F-75013, Paris, France. .,Département de Neurologie, Groupe Hospitalier Pitié-Salpêtrière, AP-HP, 75013, Paris, France. .,INSERM, Centre d'Investigation Clinique Neurosciences, CIC-1422, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France.
| | - Samantha Caillet
- Service de Diététique, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| | | | - Constance Flamand-Roze
- Centre Hospitalier Sud-Francilien, Université Paris Sud, Corbeil-Essonnes, Service de Neurologie et Unité Neurovasculaire, Corbeil-Essonnes, France.,IFPPC, centre CAMKeys, Paris, France
| | - Isaac Adanyeguh
- Université de la Sorbonne, UPMC Paris 06, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moëlle, F-75013, Paris, France
| | | | - Maryvonne Retail
- INSERM, Centre d'Investigation Clinique Neurosciences, CIC-1422, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| | - Benjamin Le Toullec
- INSERM, Centre d'Investigation Clinique Neurosciences, CIC-1422, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| | - Mariana Atencio
- Université de la Sorbonne, UPMC Paris 06, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moëlle, F-75013, Paris, France
| | - Sophie Rivaud-Péchoux
- Université de la Sorbonne, UPMC Paris 06, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moëlle, F-75013, Paris, France
| | - Vanessa Brochard
- INSERM, Centre d'Investigation Clinique Neurosciences, CIC-1422, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| | - Florence Habarou
- Service de Biochimie Métabolomique et protéomique, Hôpital Necker et Université Paris Descartes, AP-HP, Paris, France
| | - Chris Ottolenghi
- Service de Biochimie Métabolomique et protéomique, Hôpital Necker et Université Paris Descartes, AP-HP, Paris, France
| | - Florence Cormier
- Université de la Sorbonne, UPMC Paris 06, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moëlle, F-75013, Paris, France.,Département de Neurologie, Groupe Hospitalier Pitié-Salpêtrière, AP-HP, 75013, Paris, France.,INSERM, Centre d'Investigation Clinique Neurosciences, CIC-1422, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| | - Aurélie Méneret
- Université de la Sorbonne, UPMC Paris 06, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moëlle, F-75013, Paris, France.,Département de Neurologie, Groupe Hospitalier Pitié-Salpêtrière, AP-HP, 75013, Paris, France
| | - Marta Ruiz
- Université de la Sorbonne, UPMC Paris 06, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moëlle, F-75013, Paris, France.,Département de Neurologie, Groupe Hospitalier Pitié-Salpêtrière, AP-HP, 75013, Paris, France
| | - Mohamed Doulazmi
- Sorbonne Universités, UPMC Paris 06, CNRS UMR8256, Institut de Biologie Paris Seine, Adaptation Biologique et vieillissement, Paris, France
| | - Anne Roubergue
- Département de Neurologie, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Jean-Christophe Corvol
- Université de la Sorbonne, UPMC Paris 06, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moëlle, F-75013, Paris, France.,Département de Neurologie, Groupe Hospitalier Pitié-Salpêtrière, AP-HP, 75013, Paris, France.,INSERM, Centre d'Investigation Clinique Neurosciences, CIC-1422, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| | - Marie Vidailhet
- Université de la Sorbonne, UPMC Paris 06, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moëlle, F-75013, Paris, France.,Département de Neurologie, Groupe Hospitalier Pitié-Salpêtrière, AP-HP, 75013, Paris, France.,INSERM, Centre d'Investigation Clinique Neurosciences, CIC-1422, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| | - Fanny Mochel
- Université de la Sorbonne, UPMC Paris 06, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moëlle, F-75013, Paris, France.,Département de Génétique, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France.,Groupe de Recherche Clinique Neurométabolique, Université Pierre et Marie Curie, Paris, France
| | - Emmanuel Roze
- Université de la Sorbonne, UPMC Paris 06, UMR S 1127, Inserm U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moëlle, F-75013, Paris, France.,Département de Neurologie, Groupe Hospitalier Pitié-Salpêtrière, AP-HP, 75013, Paris, France.,INSERM, Centre d'Investigation Clinique Neurosciences, CIC-1422, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| |
Collapse
|
48
|
Boksha IS, Prokhorova TA, Savushkina OK, Tereshkina EB. Klotho protein: Its role in aging and central nervous system pathology. BIOCHEMISTRY (MOSCOW) 2017; 82:990-1005. [DOI: 10.1134/s0006297917090024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
49
|
Alpha 2 Na +,K +-ATPase silencing induces loss of inflammatory response and ouabain protection in glial cells. Sci Rep 2017; 7:4894. [PMID: 28687727 PMCID: PMC5501845 DOI: 10.1038/s41598-017-05075-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 05/24/2017] [Indexed: 12/12/2022] Open
Abstract
Ouabain (OUA) is a cardiac glycoside that binds to Na+,K+-ATPase (NKA), a conserved membrane protein that controls cell transmembrane ionic concentrations and requires ATP hydrolysis. At nM concentrations, OUA activates signaling pathways that are not related to its typical inhibitory effect on the NKA pump. Activation of these signaling pathways protects against some types of injury of the kidneys and central nervous system. There are 4 isoforms of the alpha subunit of NKA, which are differentially distributed across tissues and may have different physiological roles. Glial cells are important regulators of injury and inflammation in the brain and express the α1 and α2 NKA isoforms. This study investigated the role of α2 NKA in OUA modulation of the neuroinflammatory response induced by lipopolysaccharide (LPS) in mouse primary glial cell cultures. LPS treatment increased lactate dehydrogenase release, while OUA did not decrease cell viability and blocked LPS-induced NF-κB activation. Silencing α2 NKA prevented ERK and NF-κB activation by LPS. α2 NKA also regulates TNF-α and IL-1β levels. The data reported here indicate a significant role of α2 NKA in regulating central LPS effects, with implications in the associated neuroinflammatory processes.
Collapse
|
50
|
The relevance of α-KLOTHO to the central nervous system: Some key questions. Ageing Res Rev 2017; 36:137-148. [PMID: 28323064 DOI: 10.1016/j.arr.2017.03.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 03/10/2017] [Accepted: 03/16/2017] [Indexed: 12/20/2022]
Abstract
α-Klotho is well described as an anti-aging protein, with critical roles in kidney function as a transmembrane co-receptor for FGF23, and as a soluble factor in serum. α-Klotho is also expressed in the choroid plexus, where it is released into the cerebrospinal fluid. Nonetheless, α-Klotho is also expressed in the brain parenchyma. Accumulating evidence indicates that this pool of α-Klotho, which we define as brain α-Klotho, may play important roles as a neuroprotective factor and in promoting myelination, thereby supporting healthy brain aging. Here we summarize what is known about brain α-Klotho before focusing on the outstanding scientific questions related to its function. We believe there is a need for in vitro studies designed to distinguish between brain α-Klotho and other pools of α-Klotho, and for a greater understanding of the basic function of soluble α-Klotho. The mechanism by which the human KL-VS variant affects cognition also requires further elucidation. To help address these questions we suggest some experimental approaches that other laboratories might consider. In short, we hope to stimulate fresh ideas and encourage new research approaches that will allow the importance of α-Klotho for the aging brain to become clear.
Collapse
|