1
|
Pan Q, Lu M. Numerical simulation study of power-frequency exposure to driving windings of electromagnetic suspension high-speed maglev trains. RADIATION PROTECTION DOSIMETRY 2025:ncaf017. [PMID: 40381211 DOI: 10.1093/rpd/ncaf017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/06/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
As a potential mode of future passenger transport, the electromagnetic environment inside maglev trains is directly related to the safety and health of passengers. To study the electromagnetic exposure risk within the maglev train compartment, numerical models were established in this paper for the maglev track's long stator three-phase drive windings (serving as radiation sources), as well as for the train body and simplistic human body models representing passengers. The exposure levels of 50 Hz three-phase symmetrical current electromagnetic fields (EMFs) were numerically calculated for passengers positioned within the carriage. The numerical simulations focused on passengers' electromagnetic exposure resulting from the leakage of 50 Hz EMFs within the carriage and compared the results with established electromagnetic exposure limit guidelines. The findings indicated that the long stator three-phase drive windings generate electromagnetic leakage within the carriage, especially near the windows. Electromagnetic exposure levels vary, with passengers close to the windows experiencing more pronounced effects. Within the carriage, the maximum values of magnetic flux density (|B|) and induced electric field strength (|E|) for passengers' heads are ⁓0.59 μT and 337 μV/m, respectively. For passengers' torsos, the maximum values are ⁓1.53 μT for |B| and 57.8 μV/m for |E|. Passengers seated near the window exhibit higher values of |E| for their heads and higher values of |B| for their torsos. However, all of these values are well below the electromagnetic exposure limits (50 Hz) set by the International Commission on Non-Ionizing Radiation Protection. These findings provide valuable reference data for studying extremely low-frequency EMF exposure dosimetry in electromagnetic suspension high-speed maglev train systems.
Collapse
Affiliation(s)
- Qiangqiang Pan
- Key Laboratory of Opto-Electronic Technology and Intelligent Control, Ministry of Education, Lanzhou Jiaotong University, 88 West Anning Road, Anning District, Gansu Province, Lanzhou 730070, China
| | - Mai Lu
- Key Laboratory of Opto-Electronic Technology and Intelligent Control, Ministry of Education, Lanzhou Jiaotong University, 88 West Anning Road, Anning District, Gansu Province, Lanzhou 730070, China
| |
Collapse
|
2
|
Hadzibegovic S, Nicole O, Andelkovic V, de Gannes FP, Hurtier A, Lagroye I, Bontempi B. Examining the effects of extremely low-frequency magnetic fields on cognitive functions and functional brain markers in aged mice. Sci Rep 2025; 15:8365. [PMID: 40069380 PMCID: PMC11897315 DOI: 10.1038/s41598-025-93230-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 03/05/2025] [Indexed: 03/15/2025] Open
Abstract
Extremely low-frequency magnetic fields (ELF-MFs) are ubiquitously present in various environments of everyday life. While surveys from the World Health Organization (WHO) have not demonstrated the existence of ELF-MF-induced harmful consequences in healthy subjects, whether older adults are more vulnerable to the effects of residential and occupational ELF-MF exposure, and therefore may be at risk, remains unsettled. Here, we explored this potential health issue by investigating, in aged mice, the effects of chronic exposure to ELF-MFs (50 Hz ELF-MF at 1 mT for 8 h/day, 5 days/week for 12 consecutive weeks) on cognitive functions and expression profile of brain markers typically associated with aggravated aging or the development of Alzheimer`s disease (AD). Sham-exposed mice showed a significant age-related decline in spatial memory functions compared to young adult mice. However, this expected pattern was neither exacerbated nor counteracted by chronic exposure to ELF-MFs. No difference in hippocampal expression of APP-695, Aβ(1-42), S100b and GFAP proteins or in the pTau/Tau ratio was observed between sham- and ELF-MF-exposed aged mice, suggesting that chronic exposure to ELF-MFs does not aggravate aging and associated neuroinflammation, or promote pathological pathways involved in the initiation of AD. Because care should be taken in extrapolating these results to older adults with various comorbidities, applying current exposure limits to existing or new sensitive ELF-MF locations is recommended.
Collapse
Affiliation(s)
- Senka Hadzibegovic
- Neurocentre Magendie, INSERM U1215, Université de Bordeaux, 33000, Bordeaux, France.
| | - Olivier Nicole
- Institut Interdisciplinaire de Neurosciences, CNRS UMR 5297, Université de Bordeaux, 33000, Bordeaux, France
| | - Vojislav Andelkovic
- Laboratoire de l'Intégration du Matériau au Système, CNRS UMR 5218, Université de Bordeaux, 33405, Talence, France and Ecole Pratique des Hautes Etudes-PSL, 75014, Paris, France
| | - Florence Poulletier de Gannes
- Laboratoire de l'Intégration du Matériau au Système, CNRS UMR 5218, Université de Bordeaux, 33405, Talence, France and Ecole Pratique des Hautes Etudes-PSL, 75014, Paris, France
| | - Annabelle Hurtier
- Laboratoire de l'Intégration du Matériau au Système, CNRS UMR 5218, Université de Bordeaux, 33405, Talence, France and Ecole Pratique des Hautes Etudes-PSL, 75014, Paris, France
| | - Isabelle Lagroye
- Laboratoire de l'Intégration du Matériau au Système, CNRS UMR 5218, Université de Bordeaux, 33405, Talence, France and Ecole Pratique des Hautes Etudes-PSL, 75014, Paris, France
| | - Bruno Bontempi
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, CNRS UMR 5287, Université de Bordeaux and Ecole Pratique des Hautes Etudes, 33000, Bordeaux, France.
| |
Collapse
|
3
|
Zhao J, Ma J, Wang X, Zhang B. Effects of electromagnetic field emitted by a 90 kHz WPT system on the cognitive functions and neuronal excitation of mice. Electromagn Biol Med 2025; 44:1-16. [PMID: 39663857 DOI: 10.1080/15368378.2024.2438607] [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: 08/05/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024]
Abstract
The advantages of Magnetic Coupling Resonant Wireless Power Transfer (MCR-WPT) technology include long transmission distance, high efficiency, and high power. Therefore, it shows great potential in the field of smart home. This study aims to explore the specific impacts on the cognitive functions and neuronal excitation of mice exposed to the electromagnetic fields (EMF) emitted by the MCR-WPT platform, thereby providing biological solid experimental evidence for developing Wireless Power Transfer (WPT) technology. The research employed a frequency of 90 kHz, which is suitable for wireless charging of household appliances. Mice were exposed to EMF emitted by the WPT biosafety experimental platform for various durations. And they were divided into four groups (control group, 2-week exposure group, 4-week exposure group, and 8-week exposure group). Upon completion of the exposure period, the study employed the Novel Object Recognition (NOR) test to evaluate the learning and memory capabilities of the animals. Following this, whole-cell patch-clamp experiments were conducted to record the action potentials (AP) and potassium currents. It was revealed by our observations that, in comparison to mice without electromagnetic exposure, long-term exposure to WPT-emitted EMF resulted in accelerated release of action potentials, inhibited the activation of Voltage-Gated Potassium Channels (VGKCs) current, accelerated the deactivation of K+ channel current, and thus significantly improved the excitability of neurons in the dentate gyrus (DG) of the hippocampus of mice, but did not significantly affect cognitive function.
Collapse
Affiliation(s)
- Jun Zhao
- School of Health Sciences & Biomedical Engineering, Hebei University of Technology, Tianjin, China
- Hebei Key Laboratory of Bioelectromagnetics and Neural Engineering, Hebei University of Technology, Tianjin, China
- Tianjin Key Laboratory of Bioelectricity and Intelligent Health, Hebei University of Technology, Tianjin, China
| | - Jing Ma
- School of Health Sciences & Biomedical Engineering, Hebei University of Technology, Tianjin, China
- Hebei Key Laboratory of Bioelectromagnetics and Neural Engineering, Hebei University of Technology, Tianjin, China
- Tianjin Key Laboratory of Bioelectricity and Intelligent Health, Hebei University of Technology, Tianjin, China
| | - Xiaoxuan Wang
- School of Health Sciences & Biomedical Engineering, Hebei University of Technology, Tianjin, China
- Hebei Key Laboratory of Bioelectromagnetics and Neural Engineering, Hebei University of Technology, Tianjin, China
- Tianjin Key Laboratory of Bioelectricity and Intelligent Health, Hebei University of Technology, Tianjin, China
| | - Bingqian Zhang
- School of Health Sciences & Biomedical Engineering, Hebei University of Technology, Tianjin, China
- Hebei Key Laboratory of Bioelectromagnetics and Neural Engineering, Hebei University of Technology, Tianjin, China
- Tianjin Key Laboratory of Bioelectricity and Intelligent Health, Hebei University of Technology, Tianjin, China
| |
Collapse
|
4
|
Gokce Y, Seker U, Ozoner MP. Safety analysis of different ıntensities of elf-pemf in terms of apoptotic, inflammatory, and transcription factor NF-Κb expression levels in rat liver. HEPATOLOGY FORUM 2024; 5:178-183. [PMID: 39524317 PMCID: PMC11440225 DOI: 10.14744/hf.2024.2024.0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 11/16/2024]
Abstract
Background and Aim The purpose of this research was to ascertain how exposure to extremely low-frequency pulsed electromagnetic fields (ELF-PEMFs) at varying intensities affects apoptosis-related protein expression levels and liver morphology in rats. Materials and Methods In this experimental study, 40 Wistar albino rats were randomly divided into 4 groups, with 10 animals in each group: Control, Sham, 1 milli Tesla (1mT), and 5 mT groups. The control group did not expose any application during the experiment. Animals in the sham group were placed into the closed ELF-PEMF exposure environment, but the device was kept closed. The rats in the 1mT and 5mT groups were placed into a closed ELF-PEMF exposure environment, and the magnetic field application was applied 5 days a week for 4 hours a day for 8 weeks. At the end of the study, the animals were sacrificed, and their liver tissues were examined morphologically, and the expression levels of proteins related to apoptosis and inflammation in these tissues were analyzed. Results Our results indicated that ELF-PEMFs did not lead to any exact morphological alterations in the groups. Tissue apoptotic Bax and Caspase 3 expression levels in the 1mT and 5mT groups were similar (p>0.05) to the control group. Additionally, pro-inflammatory TNF-α and transcription factor NF-κB in the 1mT and 5mT groups were similar (p>0.05) to each other and the control group. Conclusion It is feasible to conclude that neither the administration nor the exposure design of this study is changing the immunoexpression of apoptosis-regulating protein expression levels or liver morphology exposed to ELF-PEMF in rats.
Collapse
Affiliation(s)
- Yasin Gokce
- Department of Biophysics, Harran University School of Medicine, Sanliurfa, Turkiye
| | - Ugur Seker
- Department of Histology and Embryology, Mardin Artuklu University School of Medicine, Mardin, Turkiye
| | - Merve Pekince Ozoner
- Department of Veterinary Histology and Embryology, School of Veterinary Medicine, Siirt University, Siirt, Turkiye
| |
Collapse
|
5
|
Zhang Y, Zhang Y, Chen Z, Ren P, Fu Y. Continuous high-frequency repetitive transcranial magnetic stimulation at extremely low intensity affects exploratory behavior and spatial cognition in mice. Behav Brain Res 2024; 458:114739. [PMID: 37926334 DOI: 10.1016/j.bbr.2023.114739] [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: 07/20/2023] [Revised: 10/13/2023] [Accepted: 11/01/2023] [Indexed: 11/07/2023]
Abstract
High-frequency repetitive transcranial magnetic stimulation (HF-rTMS) has been shown to be effective for cognitive intervention. However, whether HF-rTMS with extremely low intensity could influence cognitive functions is still under investigation. The present study systematically investigated the effects of continuous 40 Hz and 10 Hz rTMS on cognition in young adult mice at extremely low intensity (10 mT and 1 mT) for 11 days (30 min/day). Cognitive functions were assessed using diverse behavioral tasks, including the open field, Y-maze, and Barnes maze paradigms. We found that 40 Hz rTMS significantly impaired exploratory behavior and spatial memory in both 10 mT and 1 mT conditions. In addition, 40 Hz rTMS induced remarkably different effects on exploratory behavior between 10 mT and 1mT, compared to 10 Hz stimulation. Our results indicate that extremely low intensity rTMS can significantly alter cognitive performance depending on intensity and frequency, shedding light on the understanding of the mechanism of rTMS effects.
Collapse
Affiliation(s)
- Yunfan Zhang
- Medical School, Kunming University of Science & Technology, Kunming, Yunnan 650500, China
| | - Yunbin Zhang
- Medical School, Kunming University of Science & Technology, Kunming, Yunnan 650500, China
| | - Zhuangfei Chen
- Medical School, Kunming University of Science & Technology, Kunming, Yunnan 650500, China
| | - Ping Ren
- Department of Geriatric Psychiatry, Shenzhen Mental Health Center / Shenzhen Kangning Hospital, Shenzhen, Guangdong 518020, China.
| | - Yu Fu
- Medical School, Kunming University of Science & Technology, Kunming, Yunnan 650500, China.
| |
Collapse
|
6
|
Zhu J, Li Y, Zhong C, Zhu M, Zheng Y, Xiong A, Meng P, Shan L, Li Y, Huang J. Neuritin affects the activity of neuralized-like 1 by promoting degradation and weakening its affinity for substrate. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1650-1658. [PMID: 37249336 PMCID: PMC10577452 DOI: 10.3724/abbs.2023098] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 05/04/2023] [Indexed: 05/31/2023] Open
Abstract
Neuritin plays a key role in neural development and regeneration by promoting neurite outgrowth and synapse maturation. Our previous research revealed the mechanism by which neuritin inhibits Notch signaling through interaction with neuralized-like 1 (Neurl1) to promote neurite growth. However, how neuritin regulates Notch signaling through Neurl1 has not been elucidated. Here, we first confirm that neuritin is an upstream regulator of Neurl1 and inhibits Notch signaling through Neurl1. Neurl1 is an E3 ubiquitin ligase that can promote ubiquitination and endocytosis of the Notch1 ligand Jagged1. Therefore, we observe the effect of neuritin on the ligase activity of Neurl1. The results indicate that neuritin inhibits Neurl1 activity by reducing the ubiquitination level and endocytosis of the target protein Jagged1. Moreover, we find that decreased activity of Neurl1 results in reduced expression of Notch receptor Notch intracellular domain (NICD) and downstream target gene hairy and enhancer of split-1 ( HES1). Furthermore, we investigate how neuritin affects Neurl1 enzyme activity. The results show that neuritin not only weakens the affinity between Neurl1 and Jagged1 but also promotes the degradation of Neurl1 by the 26S proteasome pathway. Taken together, our results suggest that neuritin negatively regulates Notch signaling by inhibiting the activity of Neurl1, promoting the degradation of Neurl1 and weakening the affinity of Neurl1 for Jagged1. Our study clarifies the molecular mechanisms of neuritin in regulating the Notch signaling pathway and provides new clues about how neuritin mediates neural regeneration and plasticity.
Collapse
Affiliation(s)
- Jingling Zhu
- Department of Biochemistry and Molecular BiologyTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- the Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of BiochemistryShihezi University School of MedicineShihezi832002China
| | - Yu Li
- the Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of BiochemistryShihezi University School of MedicineShihezi832002China
| | - Chen Zhong
- the First Affiliated Hospital of Shihezi University School of MedicineShihezi832000China
| | - Meiyi Zhu
- the First Affiliated Hospital of Shihezi University School of MedicineShihezi832000China
| | - Yan Zheng
- the Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of BiochemistryShihezi University School of MedicineShihezi832002China
| | - Anying Xiong
- the Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of BiochemistryShihezi University School of MedicineShihezi832002China
| | - Pingping Meng
- the Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of BiochemistryShihezi University School of MedicineShihezi832002China
| | - Liya Shan
- the Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of BiochemistryShihezi University School of MedicineShihezi832002China
| | - Yang Li
- the Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of BiochemistryShihezi University School of MedicineShihezi832002China
| | - Jin Huang
- Department of Biochemistry and Molecular BiologyTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- the Key Laboratory of Xinjiang Endemic & Ethnic Diseases and Department of BiochemistryShihezi University School of MedicineShihezi832002China
| |
Collapse
|
7
|
Guo Y, Fu Y, Sun W. 50 Hz Magnetic Field Exposure Inhibited Spontaneous Movement of Zebrafish Larvae through ROS-Mediated syn2a Expression. Int J Mol Sci 2023; 24:ijms24087576. [PMID: 37108734 PMCID: PMC10144198 DOI: 10.3390/ijms24087576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/21/2023] [Accepted: 04/15/2023] [Indexed: 04/29/2023] Open
Abstract
Extremely low frequency electromagnetic field (ELF-EMF) exists widely in public and occupational environments. However, its potential adverse effects and the underlying mechanism on nervous system, especially behavior are still poorly understood. In this study, zebrafish embryos (including a transfected synapsin IIa (syn2a) overexpression plasmid) at 3 h post-fertilization (hpf) were exposed to a 50-Hz magnetic field (MF) with a series of intensities (100, 200, 400 and 800 μT, respectively) for 1 h or 24 h every day for 5 days. Results showed that, although MF exposure did not affect the basic development parameters including hatching rate, mortality and malformation rate, yet MF at 200 μT could significantly induce spontaneous movement (SM) hypoactivity in zebrafish larvae. Histological examination presented morphological abnormalities of the brain such as condensed cell nucleus and cytoplasm, increased intercellular space. Moreover, exposure to MF at 200 μT inhibited syn2a transcription and expression, and increased reactive oxygen species (ROS) level as well. Overexpression of syn2a could effectively rescue MF-induced SM hypoactivity in zebrafish. Pretreatment with N-acetyl-L-cysteine (NAC) could not only recover syn2a protein expression which was weakened by MF exposure, but also abolish MF-induced SM hypoactivity. However, syn2a overexpression did not affect MF-increased ROS. Taken together, the findings suggested that exposure to a 50-Hz MF inhibited spontaneous movement of zebrafish larvae via ROS-mediated syn2a expression in a nonlinear manner.
Collapse
Affiliation(s)
- Yixin Guo
- Bioelectromagnetics Key Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yiti Fu
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wenjun Sun
- Bioelectromagnetics Key Laboratory, Zhejiang University School of Medicine, Hangzhou 310058, China
- Institute of Environmental Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
8
|
Tao Q, Yang D, Qin K, Liu L, Jin M, Zhang F, Zhu J, Wang J, Luo Q, Du J, Yu L, Shen J, Chu D. Studies on the mechanism of Toxoplasma gondii Chinese 1 genotype Wh6 strain causing mice abnormal cognitive behavior. Parasit Vectors 2023; 16:30. [PMID: 36698166 PMCID: PMC9875435 DOI: 10.1186/s13071-022-05618-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/14/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Alzheimer's disease presents an abnormal cognitive behavior. TgCtwh6 is one of the predominant T. gondii strains prevalent in China. Although T. gondii type II strain infection can cause host cognitive behavioral abnormalities, we do not know whether TgCtwh6 could also cause host cognitive behavioral changes. So, in this study, we will focus on the effect of TgCtwh6 on mouse cognitive behavior and try in vivo and in vitro to explore the underlying mechanism by which TgCtwh6 give rise to mice cognitive behavior changes at the cellular and molecular level. METHODS C57BL/6 mice were infected orally with TgCtwh6 cysts. From day 90 post-infection on, all mice were conducted through the open field test and then Morris water maze test to evaluate cognitive behavior. The morphology and number of cells in hippocampus were examined with hematoxylin-eosin (H&E) and Nissl staining; moreover, Aβ protein in hippocampus was determined with immunohistochemistry and thioflavin S plaque staining. Synaptotagmin 1, apoptosis-related proteins, BACE1 and APP proteins and genes from hippocampus were assessed by western blotting or qRT-PCR. Hippocampal neuronal cell line or mouse microglial cell line was challenged with TgCtwh6 tachyzoites and then separately cultured in a well or co-cultured in a transwell device. The target proteins and genes were analyzed by immunofluorescence staining, western blotting and qRT-PCR. In addition, mouse microglial cell line polarization state and hippocampal neuronal cell line apoptosis were estimated using flow cytometry assay. RESULTS The OFT and MWMT indicated that infected mice had cognitive behavioral impairments. The hippocampal tissue assay showed abnormal neuron morphology and a decreased number in infected mice. Moreover, pro-apoptotic proteins, as well as BACE1, APP and Aβ proteins, increased in the infected mouse hippocampus. The experiments in vitro showed that pro-apoptotic proteins and p-NF-κBp65, NF-κBp65, BACE1, APP and Aβ proteins or genes were significantly increased in the infected HT22. In addition, CD80, pro-inflammatory factors, notch, hes1 proteins and genes were enhanced in the infected BV2. Interestingly, not only the APP and pro-apoptotic proteins in HT22, but also the apoptosis rate of HT22 increased after the infected BV2 were co-cultured with the HT22 in a transwell device. CONCLUSIONS Neuron apoptosis, Aβ deposition and neuroinflammatory response involved with microglia polarization are the molecular and cellular mechanisms by which TgCtwh6 causes mouse cognitive behavioral abnormalities.
Collapse
Affiliation(s)
- Qing Tao
- grid.186775.a0000 0000 9490 772XDepartment of Pathogen Biology, Anhui Province Key Laboratory of Microbiology & Parasitology, Anhui Provincial Laboratory of Zoonoses of High Institutions, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Di Yang
- grid.186775.a0000 0000 9490 772XDepartment of Pathogen Biology, Anhui Province Key Laboratory of Microbiology & Parasitology, Anhui Provincial Laboratory of Zoonoses of High Institutions, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Kunpeng Qin
- grid.412679.f0000 0004 1771 3402Department of Orthopaedics, the First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Lei Liu
- grid.59053.3a0000000121679639Department of Blood Transfusion, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Mengmeng Jin
- grid.186775.a0000 0000 9490 772XMaternity and Child Health Hospital of Anhui Province, The Affiliated Maternity and Child Health Hospital of Anhui Medical University, Hefei, China
| | - Famin Zhang
- grid.186775.a0000 0000 9490 772XDepartment of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jinjin Zhu
- grid.186775.a0000 0000 9490 772XDepartment of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jie Wang
- grid.186775.a0000 0000 9490 772XDepartment of Microbiology and Parasitology, Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Provincial Laboratory of Zoonoses of High Institutions, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Qingli Luo
- grid.186775.a0000 0000 9490 772XDepartment of Pathogen Biology, Anhui Province Key Laboratory of Microbiology & Parasitology, Anhui Provincial Laboratory of Zoonoses of High Institutions, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Jian Du
- grid.186775.a0000 0000 9490 772XDepartment of Microbiology and Parasitology, Anhui Provincial Laboratory of Microbiology and Parasitology, Anhui Provincial Laboratory of Zoonoses of High Institutions, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Li Yu
- grid.186775.a0000 0000 9490 772XDepartment of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Jilong Shen
- grid.186775.a0000 0000 9490 772XDepartment of Pathogen Biology, Anhui Province Key Laboratory of Microbiology & Parasitology, Anhui Provincial Laboratory of Zoonoses of High Institutions, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Deyong Chu
- grid.186775.a0000 0000 9490 772XDepartment of Pathogen Biology, Anhui Province Key Laboratory of Microbiology & Parasitology, Anhui Provincial Laboratory of Zoonoses of High Institutions, School of Basic Medicine, Anhui Medical University, Hefei, China
| |
Collapse
|
9
|
Xu Y, Wu J, Dong L, Di G. The role of Nrf2 in protection against electrostatic field-induced oxidative stress and learning and memory decline in mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:7005-7017. [PMID: 36018411 DOI: 10.1007/s11356-022-22702-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 08/20/2022] [Indexed: 05/21/2023]
Abstract
The intensity of static electric field (SEF) in the surrounding environment of transmission lines has been greatly increased with the rapid development of ultra-high-voltage direct-current transmission. Therefore, the potential health effects of SEF have stimulated great public attention. It has been proven that SEF exposure can cause reversible damage to the nervous system through oxidative stress; however, the mechanism of its recovery is unclear. This study focused on nuclear factor erythroid 2-related factor 2 (Nrf2), a vital regulator of oxidative stress, and has been identified to notably impact the protection of organisms against many external stimuli. Herein, it was found that 56.3 kV/m SEF exposure for 7 days and 14 days significantly improved the expression levels of Nrf2 protein in the cytoplasm and nucleus of mice' hippocampus, as well as antioxidant genes, superoxide dismutase 2, and glutathione peroxidase 1. No significant difference in the expression level of the Nrf2 gene was found. The results indicated that the body could activate the Nrf2 signalling under SEF exposure by means other than up-regulation of Nrf2 gene expression. Inhibiting Nrf2 signalling by isoniazid could block SEF-induced gene transcription and protein expression, resulting in a decrease in antioxidant capacity, an increase in the level of lipid peroxide product, and irretrievability of learning and memory damage. These results demonstrated that the Nrf2 signalling pathway exhibited a protective role in SEF-induced oxidative damage and decline in learning and memory ability, which provides a potential strategy for preventing and treating SEF-related neurotoxicity.
Collapse
Affiliation(s)
- Yaqian Xu
- College of Science and Technology, Ningbo University, Ningbo, 315300, People's Republic of China
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Jiahong Wu
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Li Dong
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Guoqing Di
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China.
| |
Collapse
|
10
|
Zadeh-Haghighi H, Simon C. Magnetic field effects in biology from the perspective of the radical pair mechanism. J R Soc Interface 2022; 19:20220325. [PMID: 35919980 PMCID: PMC9346374 DOI: 10.1098/rsif.2022.0325] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 07/14/2022] [Indexed: 04/07/2023] Open
Abstract
Hundreds of studies have found that weak magnetic fields can significantly influence various biological systems. However, the underlying mechanisms behind these phenomena remain elusive. Remarkably, the magnetic energies implicated in these effects are much smaller than thermal energies. Here, we review these observations, and we suggest an explanation based on the radical pair mechanism, which involves the quantum dynamics of the electron and nuclear spins of transient radical molecules. While the radical pair mechanism has been studied in detail in the context of avian magnetoreception, the studies reviewed here show that magnetosensitivity is widespread throughout biology. We review magnetic field effects on various physiological functions, discussing static, hypomagnetic and oscillating magnetic fields, as well as isotope effects. We then review the radical pair mechanism as a potential unifying model for the described magnetic field effects, and we discuss plausible candidate molecules for the radical pairs. We review recent studies proposing that the radical pair mechanism provides explanations for isotope effects in xenon anaesthesia and lithium treatment of hyperactivity, magnetic field effects on the circadian clock, and hypomagnetic field effects on neurogenesis and microtubule assembly. We conclude by discussing future lines of investigation in this exciting new area of quantum biology.
Collapse
Affiliation(s)
- Hadi Zadeh-Haghighi
- Department of Physics and Astronomy, University of Calgary, Calgary, Alberta, Canada T2N 1N4
- Institute for Quantum Science and Technology, University of Calgary, Calgary, Alberta, Canada T2N 1N4
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada T2N 1N4
| | - Christoph Simon
- Department of Physics and Astronomy, University of Calgary, Calgary, Alberta, Canada T2N 1N4
- Institute for Quantum Science and Technology, University of Calgary, Calgary, Alberta, Canada T2N 1N4
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada T2N 1N4
| |
Collapse
|
11
|
Lai H. Neurological effects of static and extremely-low frequency electromagnetic fields. Electromagn Biol Med 2022; 41:201-221. [DOI: 10.1080/15368378.2022.2064489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Henry Lai
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| |
Collapse
|
12
|
YU JINGTONG, ZHAO CHUNXIA, GU XIAOYU, DI GUOQING, XU YAQIAN. IMPACTS OF STATIC ELECTRIC FIELD PRODUCED BY ULTRA-HIGH-VOLTAGE DIRECT-CURRENT TRANSMISSION LINES ON HIPPOCAMPAL PROTEIN EXPRESSION AND MORPHOLOGICAL STRUCTURE IN MICE. J MECH MED BIOL 2021. [DOI: 10.1142/s0219519421400716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Static electric field (SEF) from ultra-high-voltage direct-current (UHVDC) transmission lines has the potential to produce neurobiological effects. To explore these effects and elucidate their potential mechanisms, protein expression levels and morphological structure in the hippocampi of mice were investigated after SEF exposure. Mice from the Institute of Cancer Research were exposed to an environmental SEF induced by UHVDC transmission lines with the strength of 9.20–21.85[Formula: see text]kV/m for 35 days. Mouse body weight was measured weekly during the exposure. After the exposure, hippocampal Ca[Formula: see text]/calmodulin-dependent protein kinase II (CaMKII) and calcineurin (CaN) expression levels were assayed by Western blot. Hippocampal pathologic morphology and ultrastructure were observed using light microscopy and transmission electron microscopy, respectively. No significant differences in body weight, CaMKII and CaN expression levels, and hippocampal pathologic morphology were observed between mice in the exposed and the control groups. However, cytoplasmic vacuolization of the hippocampal neurons was observed in the exposed group. Thus, hippocampal neuron ultrastructure damage may be a mechanism of SEF-exposure-induced memory decline in mice.
Collapse
Affiliation(s)
- JINGTONG YU
- College of Science and Technology, Ningbo University, Ningbo 315212, P. R. China
| | - CHUNXIA ZHAO
- Ningbo Research Institute of Ecological and Environmental Sciences, Ningbo 315000, P. R. China
| | - XIAOYU GU
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - GUOQING DI
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - YAQIAN XU
- College of Science and Technology, Ningbo University, Ningbo 315212, P. R. China
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| |
Collapse
|
13
|
Liu Y, Yao J, Song Z, Guo W, Sun B, Wei J, Estillore JP, Back TG, Chen SRW. Limiting RyR2 open time prevents Alzheimer's disease-related deficits in the 3xTG-AD mouse model. J Neurosci Res 2021; 99:2906-2921. [PMID: 34352124 DOI: 10.1002/jnr.24936] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/11/2021] [Accepted: 07/14/2021] [Indexed: 12/24/2022]
Abstract
Increasing evidence suggests that Alzheimer's disease (AD) progression is driven by a vicious cycle of soluble β-amyloid (Aβ)-induced neuronal hyperactivity. Thus, breaking this vicious cycle by suppressing neuronal hyperactivity may represent a logical approach to stopping AD progression. In support of this, we have recently shown that genetically and pharmacologically limiting ryanodine receptor 2 (RyR2) open time prevented neuronal hyperactivity, memory impairment, dendritic spine loss, and neuronal cell death in a rapid, early onset AD mouse model (5xFAD). Here, we assessed the impact of limiting RyR2 open time on AD-related deficits in a relatively late occurring, slow developing AD mouse model (3xTG-AD) that bears more resemblance (compared to 5xFAD) to that of human AD. Using behavioral tests, long-term potentiation recordings, and Golgi and Nissl staining, we found that the RyR2-E4872Q mutation, which markedly shortens the open duration of the RyR2 channel, prevented learning and memory impairment, defective long-term potentiation, dendritic spine loss, and neuronal cell death in the 3xTG-AD mice. Furthermore, pharmacologically shortening the RyR2 open time with R-carvedilol rescued these AD-related deficits in 3xTG mice. Therefore, limiting RyR2 open time may offer a promising, neuronal hyperactivity-targeted anti-AD strategy.
Collapse
Affiliation(s)
- Yajing Liu
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada.,Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Jinjing Yao
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Zhenpeng Song
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Wenting Guo
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Bo Sun
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada.,Medical School, Kunming University of Science and Technology, Kunming, China
| | - Jinhong Wei
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - John Paul Estillore
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Thomas G Back
- Department of Chemistry, University of Calgary, Calgary, AB, Canada
| | - S R Wayne Chen
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
14
|
Lu J, Li Z, Zhao Q, Liu D, Mei YA. Neuritin improves the neurological functional recovery after experimental intracerebral hemorrhage in mice. Neurobiol Dis 2021; 156:105407. [PMID: 34058347 DOI: 10.1016/j.nbd.2021.105407] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/06/2021] [Accepted: 05/26/2021] [Indexed: 11/16/2022] Open
Abstract
Stroke is one of the leading causes of death worldwide, with intracerebral hemorrhage (ICH) being the most lethal subtype. Neuritin (Nrn) is a neurotropic factor that has been reported to have neuroprotective effects in acute brain and spinal cord injury. However, whether Nrn has a protective role in ICH has not been investigated. In this study, ICH was induced in C57BL/6 J mice by injection of collagenase VII, while the overexpression of Nrn in the striatum was induced by an adeno-associated virus serotype 9 (AAV9) vector. We found that compared with GFP-ICH mice, Nrn-ICH mice showed improved performance in the corner, cylinder and forelimb tests after ICH, and showed less weight loss and more rapid weight recovery. Overexpression of Nrn reduced brain lesions, edema, neuronal death and white matter and synaptic integrity dysfunction caused by ICH. Western blot results showed that phosphorylated PERK and ATF4 were significantly inhibited, while phosphorylation of Akt/mammalian target of rapamycin was increased in the Nrn-ICH group, compared with the GFP-ICH group. Whole cell recording from motor neurons indicated that overexpression of Nrn reversed the decrease of spontaneous excitatory postsynaptic currents (sEPSCs) and action potential frequencies induced by ICH. These data show that Nrn improves neurological deficits in mice with ICH by reducing brain lesions and edema, inhibiting neuronal death, and possibly by increasing neuronal connections.
Collapse
Affiliation(s)
- Junmei Lu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Zhaoyang Li
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Qianru Zhao
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Dongdong Liu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yan-Ai Mei
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai 200438, China.
| |
Collapse
|
15
|
Yao J, Sun B, Institoris A, Zhan X, Guo W, Song Z, Liu Y, Hiess F, Boyce AKJ, Ni M, Wang R, Ter Keurs H, Back TG, Fill M, Thompson RJ, Turner RW, Gordon GR, Chen SRW. Limiting RyR2 Open Time Prevents Alzheimer's Disease-Related Neuronal Hyperactivity and Memory Loss but Not β-Amyloid Accumulation. Cell Rep 2021; 32:108169. [PMID: 32966798 PMCID: PMC7532726 DOI: 10.1016/j.celrep.2020.108169] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 07/23/2020] [Accepted: 08/27/2020] [Indexed: 12/31/2022] Open
Abstract
Neuronal hyperactivity is an early primary dysfunction in Alzheimer’s disease (AD) in humans and animal models, but effective neuronal hyperactivity-directed anti-AD therapeutic agents are lacking. Here we define a previously unknown mode of ryanodine receptor 2 (RyR2) control of neuronal hyperactivity and AD progression. We show that a single RyR2 point mutation, E4872Q, which reduces RyR2 open time, prevents hyperexcitability, hyperactivity, memory impairment, neuronal cell death, and dendritic spine loss in a severe early-onset AD mouse model (5xFAD). The RyR2-E4872Q mutation upregulates hippocampal CA1-pyramidal cell A-type K+ current, a well-known neuronal excitability control that is downregulated in AD. Pharmacologically limiting RyR2 open time with the R-carvedilol enantiomer (but not racemic carvedilol) prevents and rescues neuronal hyperactivity, memory impairment, and neuron loss even in late stages of AD. These AD-related deficits are prevented even with continued β-amyloid accumulation. Thus, limiting RyR2 open time may be a hyperactivity-directed, non-β-amyloid-targeted anti-AD strategy. Yao et al. show that genetically or pharmacologically limiting the open duration of ryanodine receptor 2 upregulates the A-type potassium current and prevents neuronal hyperexcitability and hyperactivity, memory impairment, neuronal cell death, and dendritic spine loss in a severe early-onset Alzheimer’s disease mouse model, even with continued accumulation of β-amyloid.
Collapse
Affiliation(s)
- Jinjing Yao
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Bo Sun
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; Medical School, Kunming University of Science and Technology, Kunming 650504, China
| | - Adam Institoris
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Xiaoqin Zhan
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Wenting Guo
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Zhenpeng Song
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Yajing Liu
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Florian Hiess
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Andrew K J Boyce
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Mingke Ni
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Ruiwu Wang
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Henk Ter Keurs
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Thomas G Back
- Department of Chemistry, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Michael Fill
- Department of Physiology & Biophysics, Rush University Medical Center, Chicago, IL 60612, USA
| | - Roger J Thompson
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Ray W Turner
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Grant R Gordon
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - S R Wayne Chen
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; Department of Physiology & Biophysics, Rush University Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
16
|
Gonzalez-Figueroa P, Roco JA, Papa I, Núñez Villacís L, Stanley M, Linterman MA, Dent A, Canete PF, Vinuesa CG. Follicular regulatory T cells produce neuritin to regulate B cells. Cell 2021; 184:1775-1789.e19. [PMID: 33711260 DOI: 10.1016/j.cell.2021.02.027] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/25/2021] [Accepted: 02/08/2021] [Indexed: 10/21/2022]
Abstract
Regulatory T cells prevent the emergence of autoantibodies and excessive IgE, but the precise mechanisms are unclear. Here, we show that BCL6-expressing Tregs, known as follicular regulatory T (Tfr) cells, produce abundant neuritin protein that targets B cells. Mice lacking Tfr cells or neuritin in Foxp3-expressing cells accumulated early plasma cells in germinal centers (GCs) and developed autoantibodies against histones and tissue-specific self-antigens. Upon immunization, these mice also produced increased plasma IgE and IgG1. We show that neuritin is taken up by B cells, causes phosphorylation of numerous proteins, and dampens IgE class switching. Neuritin reduced differentiation of mouse and human GC B cells into plasma cells, downregulated BLIMP-1, and upregulated BCL6. Administration of neuritin to Tfr-deficient mice prevented the accumulation of early plasma cells in GCs. Production of neuritin by Tfr cells emerges as a central mechanism to suppress B cell-driven autoimmunity and IgE-mediated allergies.
Collapse
Affiliation(s)
- Paula Gonzalez-Figueroa
- Dept of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Jonathan A Roco
- Dept of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Ilenia Papa
- Dept of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Lorena Núñez Villacís
- Dept of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Maurice Stanley
- Dept of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Michelle A Linterman
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
| | - Alexander Dent
- Dept of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Pablo F Canete
- Dept of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Carola G Vinuesa
- Dept of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia.
| |
Collapse
|
17
|
Klimek A, Rogalska J. Extremely Low-Frequency Magnetic Field as a Stress Factor-Really Detrimental?-Insight into Literature from the Last Decade. Brain Sci 2021; 11:174. [PMID: 33572550 PMCID: PMC7912337 DOI: 10.3390/brainsci11020174] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/23/2021] [Accepted: 01/27/2021] [Indexed: 12/13/2022] Open
Abstract
Biological effects of extremely low-frequency magnetic field (ELF-MF) and its consequences on human health have become the subject of important and recurrent public debate. ELF-MF evokes cell/organism responses that are characteristic to a general stress reaction, thus it can be regarded as a stress factor. Exposure to ELF-MF "turns on" different intracellular mechanisms into both directions: compensatory or deleterious ones. ELF-MF can provoke morphological and physiological changes in stress-related systems, mainly nervous, hormonal, and immunological ones. This review summarizes the ELF-MF-mediated changes at various levels of the organism organization. Special attention is placed on the review of literature from the last decade. Most studies on ELF-MF effects concentrate on its negative influence, e.g., impairment of behavior towards depressive and anxiety disorders; however, in the last decade there was an increase in the number of research studies showing stimulating impact of ELF-MF on neuroplasticity and neurorehabilitation. In the face of numerous studies on the ELF-MF action, it is necessary to systematize the knowledge for a better understanding of the phenomenon, in order to reduce the risk associated with the exposure to this factor and to recognize the possibility of using it as a therapeutic agent.
Collapse
Affiliation(s)
| | - Justyna Rogalska
- Department of Animal Physiology and Neurobiology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Torun, Poland;
| |
Collapse
|
18
|
Neuritin-overexpressing transgenic mice demonstrate enhanced neuroregeneration capacity and improved spatial learning and memory recovery after ischemia-reperfusion injury. Aging (Albany NY) 2020; 13:2681-2699. [PMID: 33323541 PMCID: PMC7880330 DOI: 10.18632/aging.202318] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 10/01/2020] [Indexed: 02/01/2023]
Abstract
Acute ischemia-reperfusion (IR)-induced brain injury is further exacerbated by a series of slower secondary pathogenic events, including delayed apoptosis due to neurotrophic factor deficiency. Neuritin, a neurotrophic factor regulating nervous system development and plasticity, is a potential therapeutic target for treatment of IR injury. In this study, Neuritin-overexpressing transgenic (Tg) mice were produced by pronuclear injection and offspring with high overexpression used to generate a line with stable inheritance for testing the neuroprotective capacity of Neuritin against transient global ischemia (TGI). Compared to wild-type mice, transgenic mice demonstrated reduced degradation of the DNA repair factor poly [ADP-ribose] polymerase 1 (PARP 1) in the hippocampus, indicating decreased hippocampal apoptosis rate, and a greater number of surviving hippocampal neurons during the first week post-TGI. In addition, Tg mice showed increased expression of the regeneration markers NF-200, synaptophysin, and GAP-43, and improved recovery of spatial learning and memory. Our findings exhibited that the window of opportunity of neural recovery in Neuritin transgenic mice group had a tendency to move ahead after TGI, which indicated that Neuritin can be used as a potential new therapeutic strategy for improving the outcome of cerebral ischemia injury.
Collapse
|
19
|
Song D, Li G, Hong Y, Zhang P, Zhu J, Yang L, Huang J. miR‑199a decreases Neuritin expression involved in the development of Alzheimer's disease in APP/PS1 mice. Int J Mol Med 2020; 46:384-396. [PMID: 32626916 PMCID: PMC7255456 DOI: 10.3892/ijmm.2020.4602] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 04/23/2020] [Indexed: 01/16/2023] Open
Abstract
Neuritin plays an important role in neural development and plasticity. A recent study demonstrated that increasing Neuritin levels attenuated synaptic damage in mice with Alzheimer's disease (AD), which exhibit a decreased Neuritin expression. However, it remains unclear as to whether Neuritin expression is regulated by microRNAs (miRNAs or miRs) in AD. In the present study, it was found that miR-199a decreased Neuritin expression and was therefore involved in the development of AD. Subsequently, differentially expressed miRNAs in AD from datasets and the literature were recruited, and those that could bind Neuritin were predicted using bioinformatics analysis. The present study then focused on the candidate miRNAs that were highly associated with Neuritin and were upregulated in AD. The expression patterns of the candidate miRNAs and Neuritin in the hippocampus and cortex of APP/PS1 (AD model) mice at different stages were then detected and analyzed. It was found that miR-199a expression was significantly increased in the early stages of AD and was negatively associated with Neuritin expression. Furthermore, it was revealed that the decreased Neuritin expression was due to the direct targeting of the Neuritin 3′-UTR by miR-199a. Finally, the association between the spatial memory capacity of APP/PS1 mice and the changes in miR-199a and Neuritin expression protein was investigated. On the whole, the data of the present study suggest that miR-199a is involved in the development of AD by regulating Neuritin expression.
Collapse
Affiliation(s)
- Dandan Song
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, P.R. China
| | - Guoxiang Li
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, P.R. China
| | - Yu Hong
- Department of Prevention Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 310036, P.R. China
| | - Pan Zhang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, P.R. China
| | - Jingling Zhu
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, P.R. China
| | - Lei Yang
- Department of Prevention Medicine, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 310036, P.R. China
| | - Jin Huang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases and Department of Biochemistry, Shihezi University School of Medicine, Shihezi, Xinjiang 832002, P.R. China
| |
Collapse
|
20
|
Zheng Y, Ma XX, Dong L, Gao Y, Tian L. Effects of single- and hybrid-frequency extremely low-frequency electromagnetic field stimulations on long-term potentiation in the hippocampal Schaffer collateral pathway. Int J Radiat Biol 2019; 95:1319-1325. [PMID: 31140893 DOI: 10.1080/09553002.2019.1625463] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Purpose: To study the different effects of single- and hybrid-frequency magnetic fields on long-term potentiation (LTP) in synaptic plasticity. Materials and methods: Based on the online electromagnetic field stimulation system and field excitatory postsynaptic potentials (fEPSPs) recording system, we applied four different single- and hybrid-frequency magnetic fields with an intensity of 1 mT to the Schaffer collateral (CA1) pathway of rat hippocampal slices in vitro. Results: The amplitude of fEPSPs decreased significantly under both single- and hybrid-frequency magnetic stimulation. Lower single-frequency magnetic stimulation on LTP had a greater regulating effect, while the regulating effect among four different hybrid-frequency extremely low-frequency electromagnetic fields (ELF-EMFs) stimulations on LTP showed no significant differences. Conclusion: Single-frequency magnetic stimulation produces more significant regulatory effects, and the lower the frequency, the more significant the regulatory effect. The effect of hybrid-frequency magnetic stimulation in each group was similar, and there was no significant difference between each group. The 15-Hz single-frequency magnetic stimulation group showed the most significant regulatory effect, but once it was mixed with other higher frequency magnetic stimulation, its regulation effect was significantly weakened.
Collapse
Affiliation(s)
- Yu Zheng
- School of Electronics and Information Engineering, Tianjin Polytechnic University , Tianjin , China
| | - Xiao-Xu Ma
- School of Electronics and Information Engineering, Tianjin Polytechnic University , Tianjin , China
| | - Lei Dong
- State Key Laboratory of Precision Measurement Technology and Instruments, Tianjin University , Tianjin , China
| | - Yang Gao
- The School of Information Technology and Electrical Engineering, The University of Queensland , Brisbane , Australia
| | - Lei Tian
- School of Electronics and Information Engineering, Tianjin Polytechnic University , Tianjin , China
| |
Collapse
|
21
|
Di G, Kim H, Xu Y, Kim J, Gu X. A comparative study on influences of static electric field and power frequency electric field on cognition in mice. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2019; 66:91-95. [PMID: 30639900 DOI: 10.1016/j.etap.2019.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 12/30/2018] [Accepted: 01/03/2019] [Indexed: 06/09/2023]
Abstract
Recently, electromagnetic fields around ultra-high voltage transmission lines have received considerable attentions for their potential biological effects. This study aimed to investigate the effects of static electric field (SEF) and power frequency electric field (PFEF) on cognition. Mice were exposed to SEF and PFEF with the same strength (35 kV/m) for 49 days, respectively. Behaviors in Morris water maze test and amino acid neurotransmitter levels in hippocampus were examined during exposure. Results indicated that the exposure of 35 kV/m SEF would not cause significant influences on learning and memory ability in mice, while the exposure of 35 kV/m PFEF would cause significant positive effects on learning and memory ability in mice on day 33. This difference in effects from SEF and PFEF on cognition was possibly induced by the difference in the degree of molecular polarization and ion migration in organisms under exposure of two kinds of electric fields with different frequency.
Collapse
Affiliation(s)
- Guoqing Di
- Institute of Environmental Process, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, PR China.
| | - Hakbong Kim
- Institute of Environmental Process, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, PR China; Kim Chaek University of Technology, Pyongyang, Democratic People's Republic of Korea
| | - Yaqian Xu
- Institute of Environmental Process, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Jonghyok Kim
- Institute of Environmental Process, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Xiaoyu Gu
- Institute of Environmental Process, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, PR China
| |
Collapse
|
22
|
Yao JJ, Zhao QR, Lu JM, Mei YA. Functions and the related signaling pathways of the neurotrophic factor neuritin. Acta Pharmacol Sin 2018; 39:1414-1420. [PMID: 29595190 PMCID: PMC6289377 DOI: 10.1038/aps.2017.197] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/08/2017] [Indexed: 12/29/2022]
Abstract
Neuritin is a member of the neurotrophic factor family, which is activated by neural activity and neurotrophins, and promotes neurite growth and branching. It has shown to play an important role in neuronal plasticity and regeneration. It is also involved in other biological processes such as angiogenesis, tumorigenesis and immunomodulation. Thus far, however, the primary mechanisms of neuritin, including whether or not it acts through a receptor or which downstream signals might be activated following binding, are not fully understood. Recent evidence suggests that neuritin may be a potential therapeutic target in several neurodegenerative diseases. This review focuses on the recent advances in studies regarding the newly identified functions of neuritin and the signaling pathways related to these functions. We also discuss current hot topics and difficulties in neuritin research.
Collapse
Affiliation(s)
- Jin-Jing Yao
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Qian-Ru Zhao
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Jun-Mei Lu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Yan-Ai Mei
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
23
|
Zhao QR, Lu JM, Li ZY, Mei YA. Neuritin promotes neurite and spine growth in rat cerebellar granule cells via L-type calcium channel-mediated calcium influx. J Neurochem 2018; 147:40-57. [PMID: 29920676 PMCID: PMC6220818 DOI: 10.1111/jnc.14535] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/21/2018] [Accepted: 06/13/2018] [Indexed: 01/15/2023]
Abstract
Neuritin is a neurotrophic factor that is activated by neural activity and neurotrophins. Its major function is to promote neurite growth and branching; however, the underlying mechanisms are not fully understood. To address this issue, this study investigated the effects of neuritin on neurite and spine growth and intracellular Ca2+ concentration in rat cerebellar granule neurons (CGNs). Incubation of CGNs for 24 h with neuritin increased neurite length and spine density; this effect was mimicked by insulin and abolished by inhibiting insulin receptor (IR) or mitogen‐activated protein kinase kinase/extracellular signal‐regulated kinase (ERK) activity. Calcium imaging and western blot analysis revealed that neuritin enhanced the increase in intracellular Ca2+ level induced by high K+, and stimulated the cell surface expression of CaV1.2 and CaV1.3 α subunits of the L‐type calcium channel, which was suppressed by inhibition of IR or mitogen‐activated protein kinase kinase/ERK. Treatment with inhibitors of L‐type calcium channels, calmodulin, and calcineurin (CaN) abrogated the effects of neuritin on neurite length and spine density. A similar result was obtained by silencing nuclear factor of activated T cells c4, which is known to be activated by neuritin in CGNs. These results indicate that IR and ERK signaling as well as the Ca2+/CaN/nuclear factor of activated T cells c4 axis mediate the effects of neuritin on neurite and spine growth in CGNs. Open Practices
Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/ ![]()
Cover Image for this issue: doi: 10.1111/jnc.14195.
Collapse
Affiliation(s)
- Qian-Ru Zhao
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai, China
| | - Jun-Mei Lu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai, China
| | - Zhao-Yang Li
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai, China
| | - Yan-Ai Mei
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
24
|
Xu Y, Gu X, Di G. Duration-dependent effect of exposure to static electric field on learning and memory ability in mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:23864-23874. [PMID: 29881961 DOI: 10.1007/s11356-018-2458-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/30/2018] [Indexed: 06/08/2023]
Abstract
With the rapid development of ultra-high-voltage direct-current (UHVDC) transmission, the strength of environmental static electric field (SEF) around UHVDC transmission lines increased substantially, which has aroused widely public attention on the potential health effects of SEF. In this study, the effect of SEF exposure on learning and memory ability was investigated. Institute of Cancer Research mice were exposed to 56.3 kV/m SEF for a short term (7 days) or long term (49 days). Behaviors in the Morris water maze (MWM) test, hippocampal neurotransmitter contents, and oxidative stress indicators were examined. Results showed that short-term SEF exposure significantly prolonged escape latency and decreased the number of platform-site crossovers, as well as decreased the time spent in the target quadrant in the MWM test. Meanwhile, serotonin level and the ratio of glutamate level to γ-aminobutyric acid level changed significantly. Besides, malondialdehyde content and glutathione peroxidase activity increased significantly, while superoxide dismutase activity decreased significantly. After long-term SEF exposure, all indices above showed no significant differences between the SEF and sham exposure groups. These data indicated that short-term exposure to 56.3 kV/m SEF could cause abnormal neurotransmitter levels and oxidative stress in the hippocampus, which led to the decline in learning and memory ability. Under the condition of long-term exposure, the SEF-induced disturbances in neurotransmitter contents and redox balance were offset by the compensatory responses of mice, and thus, the learning and memory ability returned to normal level. The temporary and reversible decline in learning and memory ability was only a common biological effect of SEF rather than a health hazard.
Collapse
Affiliation(s)
- Yaqian Xu
- Department of Environmental Science, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Xiaoyu Gu
- Department of Environmental Science, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Guoqing Di
- Department of Environmental Science, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China.
| |
Collapse
|
25
|
Zhu BY, Yang ZD, Chen XR, Zhou J, Gao YH, Xian CJ, Chen KM. Exposure Duration Is a Determinant of the Effect of Sinusoidal Electromagnetic Fields on Peak Bone Mass of Young Rats. Calcif Tissue Int 2018; 103:95-106. [PMID: 29362823 DOI: 10.1007/s00223-018-0396-2] [Citation(s) in RCA: 5] [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: 11/22/2017] [Accepted: 01/17/2018] [Indexed: 11/26/2022]
Abstract
We proposed a three-step strategy to obtain the optimal therapeutic parameters, which is composed of large-scale screening at cellular level, verification in animal experiments, and confirmation by a clinical trial. The objective of the current study was to test the feasibility of our strategy. Newborn rat calvarial osteoblasts were treated by 50 Hz 1.8 mT sinusoidal electromagnetic fields (SEMFs) with 0.5, 1.0, 1.5, 2.0, 2.5, and 3.0 h/days, respectively. The osteogenic differentiation and maturation of the osteoblast were assayed and compared to obtain the optimal duration. One-month-old growing rats were then treated by the same SEMFs with 0.5, 1.5, and 2.5 h/days, respectively, and the peak bone mass was analyzed after 2 months. It was found that the optimal exposure duration to promote the osteogenic differentiation and maturation of osteoblasts was 1.5 h/days, judging by the increasing degrees of ALP activity, calcified nodules formed, the gene and protein expression levels of Runx-2, BMP-2, and Col-I, as well as the expression levels of signaling proteins of the BMP-2/Smad1/5/8 pathway. The highest increase of peak bone mass after 2 months was also obtained by 1.5 h/days, judging by the results of X-ray dual-energy absorptiometry, mechanical property analysis, micro-CT scanning, and serum bone turnover marker examinations. The above results indicated that exposure duration is a determinant for the therapeutic effect of EMFs, and the optimal therapeutic effects only can be obtained by the optimal exposure duration.
Collapse
Affiliation(s)
- B Y Zhu
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, 730050, People's Republic of China
- Institute of Orthopaedics, Lanzhou General Hospital of CPLA, Lanzhou, 730050, People's Republic of China
| | - Z D Yang
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, 730050, People's Republic of China.
| | - X R Chen
- College of Life Sciences, Northwest A & F University, Yanglin, 712100, People's Republic of China
| | - J Zhou
- Institute of Orthopaedics, Lanzhou General Hospital of CPLA, Lanzhou, 730050, People's Republic of China
| | - Y H Gao
- Institute of Orthopaedics, Lanzhou General Hospital of CPLA, Lanzhou, 730050, People's Republic of China
| | - C J Xian
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, 5001, Australia
| | - K M Chen
- Institute of Orthopaedics, Lanzhou General Hospital of CPLA, Lanzhou, 730050, People's Republic of China.
| |
Collapse
|
26
|
Kumari K, Koivisto H, Viluksela M, Paldanius KMA, Marttinen M, Hiltunen M, Naarala J, Tanila H, Juutilainen J. Behavioral testing of mice exposed to intermediate frequency magnetic fields indicates mild memory impairment. PLoS One 2017; 12:e0188880. [PMID: 29206232 PMCID: PMC5714647 DOI: 10.1371/journal.pone.0188880] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/14/2017] [Indexed: 12/27/2022] Open
Abstract
Human exposure to intermediate frequency magnetic fields (MF) is increasing due to applications like electronic article surveillance systems and induction heating cooking hobs. However, limited data is available on their possible health effects. The present study assessed behavioral and histopathological consequences of exposing mice to 7.5 kHz MF at 12 or 120 μT for 5 weeks. No effects were observed on body weight, spontaneous activity, motor coordination, level of anxiety or aggression. In the Morris swim task, mice in the 120 μT group showed less steep learning curve than the other groups, but did not differ from controls in their search bias in the probe test. The passive avoidance task indicated a clear impairment of memory over 48 h in the 120 μT group. No effects on astroglial activation or neurogenesis were observed in the hippocampus. The mRNA expression of brain-derived neurotrophic factor did not change but expression of the proinflammatory cytokine tumor necrosis factor alpha mRNA was significantly increased in the 120 μT group. These findings suggest that 7.5 kHz MF exposure may lead to mild learning and memory impairment, possibly through an inflammatory reaction in the hippocampus.
Collapse
Affiliation(s)
- Kajal Kumari
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
- * E-mail:
| | | | - Matti Viluksela
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
- National Institute for Health and Welfare, Environmental Health Unit, Kuopio, Finland
| | | | - Mikael Marttinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Jonne Naarala
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Heikki Tanila
- A. I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland
| | - Jukka Juutilainen
- Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
27
|
Zhang P, Luo X, Guo Z, Xiong A, Dong H, Zhang Q, Liu C, Zhu J, Wang H, Yu N, Zhang J, Hong Y, Yang L, Huang J. Neuritin Inhibits Notch Signaling through Interacted with Neuralized to Promote the Neurite Growth. Front Mol Neurosci 2017. [PMID: 28642682 PMCID: PMC5462965 DOI: 10.3389/fnmol.2017.00179] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Neuritin plays a key role in neural development and regeneration by promoting neurite outgrowth and synapse maturation. However, the mechanism of neuritin in modulating neurite growth has not been elucidated. Here, using yeast two-hybrid we screened and discovered the interaction of neuritin and neuralized (NEURL1), which is an important regulator that can activate Notch signaling through promoting endocytosis of Notch ligand. And then we identified the interaction of neuritin and neuralized by co-immunoprecipitation (IP) assays, and clarified that neuritin and NEURL1 were co-localized on the cell membrane of SH-SY5Y cells. Moreover, neuritin significantly suppressed Notch ligand Jagged1 (JAG1) endocytosis promoted by NEURL1, and then inhibited the activation of Notch receptor Notch intracellular domain (NICD) and decreased the expression of downstream gene hairy and enhancer of split-1 (HES1). Importantly, the effect of neuritin on inhibiting Notch signaling was rescued by NEURL1, which indicated that neuritin is an upstream and negative regulator of NEURL1 to inhibit Notch signaling through interaction with NEURL1. Notably, recombinant neuritin restored the retraction of neurites caused by activation of Notch, and neurite growth stimulated by neuritin was partially blocked by NEURL1. These findings establish neuritin as an upstream and negative regulator of NEURL1 that inhibits Notch signaling to promote neurite growth. This mechanism connects neuritin with Notch signaling, and provides a valuable foundation for further investigation of neuritin's role in neurodevelopment and neural plasticity.
Collapse
Affiliation(s)
- Pan Zhang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Xing Luo
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Zheng Guo
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Anying Xiong
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Hongchang Dong
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Qiao Zhang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Chunyan Liu
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Jingling Zhu
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Haiyan Wang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Na Yu
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Jinli Zhang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| | - Yu Hong
- School of Medicine, Hangzhou Normal UniversityHangzhou, China
| | - Lei Yang
- School of Medicine, Hangzhou Normal UniversityHangzhou, China
| | - Jin Huang
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Department of Biochemistry, Shihezi University School of MedicineShihezi, China
| |
Collapse
|
28
|
Lu JM, Liu DD, Li ZY, Ling C, Mei YA. Neuritin Enhances Synaptic Transmission in Medial Prefrontal Cortex in Mice by Increasing CaV3.3 Surface Expression. Cereb Cortex 2017; 27:3842-3855. [DOI: 10.1093/cercor/bhx082] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 03/23/2017] [Indexed: 02/02/2023] Open
Affiliation(s)
- Jun-Mei Lu
- State Key Laboratory of Medical Neurobiology and School of Life Sciences, Institutes of Brain Science, Fudan University, Shanghai 200433, China
| | - Dong-Dong Liu
- State Key Laboratory of Medical Neurobiology and School of Life Sciences, Institutes of Brain Science, Fudan University, Shanghai 200433, China
| | - Zhao-Yang Li
- State Key Laboratory of Medical Neurobiology and School of Life Sciences, Institutes of Brain Science, Fudan University, Shanghai 200433, China
| | - Chen Ling
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yan-Ai Mei
- State Key Laboratory of Medical Neurobiology and School of Life Sciences, Institutes of Brain Science, Fudan University, Shanghai 200433, China
| |
Collapse
|
29
|
Effect of 1.8 GHz radiofrequency electromagnetic radiation on novel object associative recognition memory in mice. Sci Rep 2017; 7:44521. [PMID: 28303965 PMCID: PMC5355939 DOI: 10.1038/srep44521] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 02/10/2017] [Indexed: 02/06/2023] Open
Abstract
Mounting evidence suggests that exposure to radiofrequency electromagnetic radiation (RF-EMR) can influence learning and memory in rodents. In this study, we examined the effects of single exposure to 1.8 GHz RF-EMR for 30 min on subsequent recognition memory in mice, using the novel object recognition task (NORT). RF-EMR exposure at an intensity of >2.2 W/kg specific absorption rate (SAR) power density induced a significant density-dependent increase in NORT index with no corresponding changes in spontaneous locomotor activity. RF-EMR exposure increased dendritic-spine density and length in hippocampal and prefrontal cortical neurons, as shown by Golgi staining. Whole-cell recordings in acute hippocampal and medial prefrontal cortical slices showed that RF-EMR exposure significantly altered the resting membrane potential and action potential frequency, and reduced the action potential half-width, threshold, and onset delay in pyramidal neurons. These results demonstrate that exposure to 1.8 GHz RF-EMR for 30 min can significantly increase recognition memory in mice, and can change dendritic-spine morphology and neuronal excitability in the hippocampus and prefrontal cortex. The SAR in this study (3.3 W/kg) was outside the range encountered in normal daily life, and its relevance as a potential therapeutic approach for disorders associated with recognition memory deficits remains to be clarified.
Collapse
|
30
|
Pulsed or continuous electromagnetic field induce p53/p21-mediated apoptotic signaling pathway in mouse spermatogenic cells in vitro and thus may affect male fertility. Toxicology 2017; 382:84-92. [PMID: 28323003 DOI: 10.1016/j.tox.2017.03.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 02/24/2017] [Accepted: 03/14/2017] [Indexed: 11/22/2022]
Abstract
The impact of electromagnetic field (EMF) on the human health and surrounding environment is a common topic investigated over the years. A significant increase in the electromagnetic field concentration arouses public concern about the long-term effects of EMF on living organisms associated with many aspects. In the present study, we investigated the effects of pulsed and continuous electromagnetic field (PEMF/CEMF) on mouse spermatogenic cell lines (GC-1 spg and GC-2 spd) in terms of cellular and biochemical features in vitro. We evaluated the effect of EMF on mitochondrial metabolism, morphology, proliferation rate, viability, cell cycle progression, oxidative stress balance and regulatory proteins. Our results strongly suggest that EMF induces oxidative and nitrosative stress-mediated DNA damage, resulting in p53/p21-dependent cell cycle arrest and apoptosis. Therefore, spermatogenic cells due to the lack of antioxidant enzymes undergo oxidative and nitrosative stress-mediated cytotoxic and genotoxic events, which contribute to infertility by reduction in healthy sperm cells pool. In conclusion, electromagnetic field present in surrounding environment impairs male fertility by inducing p53/p21-mediated cell cycle arrest and apoptosis.
Collapse
|
31
|
Xu Y, Wu S, Di G, Ling P, Jiang J, Bao H. Influence of static electric field on cognition in mice. Bioengineered 2017; 7:241-5. [PMID: 27282242 DOI: 10.1080/21655979.2016.1197632] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
With the rapid development of high voltage direct current transmission, the possibility of health effects associated with static electric field (SEF) has caused wide public concern. To examine the effects of long-lasting, full-body exposure to SEF on cognition, Institute of Cancer Research mice were exposed to SEF for 35 d. The intensities of SEF in experimental group I (EG-I), experimental group II (EG-II) and control group (CG) were 2.30∼15.40 kV/m, 9.20∼21.85 kV/m and 0 kV/m, respectively. The performance in learning and memory of mice were tested by Morris water maze (MWM) on days 2∼6, 16∼20 and 30∼34 during the exposure period. The concentrations of hippocampal amino acid neurotransmitters were evaluated on days 7, 21 and 35. Results showed that the latency in the MWM test had no significant difference among the EG-I, EG-II and CG (P > 0.05) during the exposure period. The percentage of time spent in the target quadrant was significantly decreased in the EG-II on day 34 during the exposure period (P < 0.05), whereas the percentage of time spent in the opposite quadrant increased markedly (P < 0.01). The glutamate and gamma-aminobutyric acid concentrations showed no significant differences among the EG-I, EG-II and CG (P > 0.05) during the exposure period. These results indicated that long-lasting, full-body exposure to SEF with certain intensity would not cause significant influence on learning ability, but it might associate with memory impairment of receptors. Meanwhile, this effect of memory impairment was dose-dependent and not causally linked to the glutamate and gamma-aminobutyric acid levels in the hippocampus.
Collapse
Affiliation(s)
- Yaqian Xu
- a Institute of Environmental Pollution & Control Technology, Zhejiang University , Hangzhou , China
| | - Sixia Wu
- a Institute of Environmental Pollution & Control Technology, Zhejiang University , Hangzhou , China
| | - Guoqing Di
- a Institute of Environmental Pollution & Control Technology, Zhejiang University , Hangzhou , China
| | - Ping Ling
- b State Grid Shanghai Municipal Electric Power Company , Shanghai , China
| | - Jianhua Jiang
- b State Grid Shanghai Municipal Electric Power Company , Shanghai , China
| | - Hailong Bao
- b State Grid Shanghai Municipal Electric Power Company , Shanghai , China
| |
Collapse
|
32
|
Yao JJ, Zhao QR, Liu DD, Chow CW, Mei YA. Neuritin Up-regulates Kv4.2 α-Subunit of Potassium Channel Expression and Affects Neuronal Excitability by Regulating the Calcium-Calcineurin-NFATc4 Signaling Pathway. J Biol Chem 2016; 291:17369-81. [PMID: 27307045 PMCID: PMC5016134 DOI: 10.1074/jbc.m115.708883] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Indexed: 11/06/2022] Open
Abstract
Neuritin is an important neurotrophin that regulates neural development, synaptic plasticity, and neuronal survival. Elucidating the downstream molecular signaling is important for potential therapeutic applications of neuritin in neuronal dysfunctions. We previously showed that neuritin up-regulates transient potassium outward current (IA) subunit Kv4.2 expression and increases IA densities, in part by activating the insulin receptor signaling pathway. Molecular mechanisms of neuritin-induced Kv4.2 expression remain elusive. Here, we report that the Ca(2+)/calcineurin (CaN)/nuclear factor of activated T-cells (NFAT) c4 axis is required for neuritin-induced Kv4.2 transcriptional expression and potentiation of IA densities in cerebellum granule neurons. We found that neuritin elevates intracellular Ca(2+) and increases Kv4.2 expression and IA densities; this effect was sensitive to CaN inhibition and was eliminated in Nfatc4(-/-) mice but not in Nfatc2(-/-) mice. Stimulation with neuritin significantly increased nuclear accumulation of NFATc4 in cerebellum granule cells and HeLa cells, which expressed IR. Furthermore, NFATc4 was recruited to the Kv4.2 gene promoter loci detected by luciferase reporter and chromatin immunoprecipitation assays. More importantly, data obtained from cortical neurons following adeno-associated virus-mediated overexpression of neuritin indicated that reduced neuronal excitability and increased formation of dendritic spines were abrogated in the Nfatc4(-/-) mice. Together, these data demonstrate an indispensable role for the CaN/NFATc4 signaling pathway in neuritin-regulated neuronal functions.
Collapse
Affiliation(s)
- Jin-Jing Yao
- From the Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China and
| | - Qian-Ru Zhao
- From the Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China and
| | - Dong-Dong Liu
- From the Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China and
| | - Chi-Wing Chow
- the Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, New York 10461
| | - Yan-Ai Mei
- From the Institutes of Brain Science, School of Life Sciences and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200433, China and
| |
Collapse
|
33
|
Zhang Y, Lai J, Ruan G, Chen C, Wang DW. Meta-analysis of extremely low frequency electromagnetic fields and cancer risk: a pooled analysis of epidemiologic studies. ENVIRONMENT INTERNATIONAL 2016; 88:36-43. [PMID: 26703095 DOI: 10.1016/j.envint.2015.12.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 11/23/2015] [Accepted: 12/10/2015] [Indexed: 06/05/2023]
Abstract
Studies have suggested that extremely low frequency electromagnetic fields (ELF-EMF) may affect physiological functions in animal models. However, epidemiologic studies investigating the association of ELF-EMF with the susceptibility to cancer yield contradictory results. In this comprehensive analysis, we conducted a search for case-control surveys regarding the associations of ELF-EMF and cancer susceptibility in electronic databases. A total of 42 studies involving 13,259 cases and 100,882 controls were retrieved. Overall, increased susceptibility to cancer was identified in the ELF-EMF exposed population (OR=1.08, 95% CI: 1.01, 1.15, P=0.02). In the stratified analyses, increased risk was found in North America (OR=1.10; 95% CI: 1.02, 1.20, P=0.02), especially the United States (OR=1.10; 95% CI: 1.01, 1.20, P=0.03). However, studies from Europe contradict these results. Moreover, a higher risk was found to be statistically significantly associated with the residential exposed population (OR=1.18; 95% CI: 1.02, 1.37, P=0.03). Furthermore, an increased cancer risk was found in interview-based surveys (OR=1.16; 95% CI: 1.00, 1.35, P=0.04). In device measurement-based studies, a slight increased risk was found only in premenopausal breast cancer (OR=1.23; 95% CI: 1.01, 1.49, P=0.04). Our meta-analysis suggests that ELF-EMFs are associated with cancer risk, mainly in the United States and in residential exposed populations. Methodological challenges might explain the differences among studies.
Collapse
Affiliation(s)
- Yemao Zhang
- High Voltage Research Institute, China Electric Power Research Institute, Wuhan, People's Republic of China
| | - Jinsheng Lai
- Department of Internal Medicine and the Institute of Hypertension, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Guoran Ruan
- Department of Internal Medicine and the Institute of Hypertension, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Chen Chen
- Department of Internal Medicine and the Institute of Hypertension, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, People's Republic of China.
| | - Dao Wen Wang
- Department of Internal Medicine and the Institute of Hypertension, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|