1
|
O’Reilly-Fong J, Simpson NJ, Thirouin ZS, Bastone PA, Zaelzer C, Murtaz A, Bourque CW. Acute and Reversible Hypothalamic Symptoms in a Lateral Head Impact Mouse Model of Mild Traumatic Brain Injury. Neurotrauma Rep 2024; 5:749-759. [PMID: 39184177 PMCID: PMC11342051 DOI: 10.1089/neur.2024.0071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024] Open
Abstract
Central autonomic and endocrine dysfunctions following traumatic brain injury (TBI) are believed to involve the hypothalamus; however, underlying mechanisms are unknown. Although chronic deficits might be caused by irreversible tissue damage, various neuroendocrine and autonomic symptoms are only observed transiently, suggesting they might result from a temporary alteration in the activity of hypothalamic neurons. We therefore examined if a mouse model of mild TBI could induce reversible autonomic phenotypes and cause acute changes in c-Fos expression within corresponding regions of the hypothalamus. Adult C57Bl/6 male mice were lightly anesthetized with isoflurane and subjected to TBI by lateral head impact using a Gothenburg impactor. Mice treated the same way, but without the head impact served as controls (shams). We monitored body weight and core body temperature by infrared thermography and performed immunohistochemistry against c-Fos in various regions of the hypothalamus. We determined that a projectile velocity of 9 m/s significantly delayed recovery from the anesthesia without inducing skull fractures and signs of discomfort disappeared within 3 h, as assessed by grimace scale. Compared with shams, TBI mice displayed a rapid decrease in core body temperature which resolved within 48 h. Daily body weight gain was also significantly lower in TBI mice on the day following injury but recovered thereafter. c-Fos analysis revealed a significantly higher density of c-Fos-positive cells in the paraventricular nucleus and a significantly lower density in the median preoptic nucleus and medial preoptic area. We conclude that mild TBI induced by a single lateral head impact in mice at 9 m/s produces acute and reversible symptoms associated with hypothalamic dysfunction accompanied by significant changes in c-Fos expression within relevant areas of the hypothalamus. These findings support the hypothesis that a temporary alteration of neuronal activity may underlie the expression of reversible central autonomic and neuroendocrine symptoms.
Collapse
Affiliation(s)
- Julie O’Reilly-Fong
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | - Nick J. Simpson
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | - Zahra S. Thirouin
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | - Paolo A. Bastone
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | - Cristian Zaelzer
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | - Anzala Murtaz
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | - Charles W. Bourque
- Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Center, Montreal, Canada
| |
Collapse
|
2
|
Suryadi T, Kulsum K. Comparison between premortem histopathology findings in rats with and without traumatic brain injury: prospective application in forensic medicine. F1000Res 2024; 12:1311. [PMID: 39282512 PMCID: PMC11399755 DOI: 10.12688/f1000research.140718.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/14/2024] [Indexed: 09/19/2024] Open
Abstract
Background The aim of this study was to compare pre-mortem histopathology findings in rats with and without traumatic brain injury (TBI) and its prospective application in forensic medicine. Methods This study involved 12 rats with 6 rats for each treatment group. This type of study is a laboratory experimental study with two independent groups design. The first group were rats that did not experience TBI. The second group was a group of rats with TBI. The subjects of this study were Rattus norvegicus rats, adult males, 4-8 weeks old, weighing 150-200 grams. On the 8 th day after the rats experienced traumatic brain injury, the rats were then euthanized using the cervical dislocation method, after euthanasia the rats were given craniotomy and brain tissue was taken for histopathology examination. Results The description of histopathology changes in the brain organs in the group of rat without TBI found that neuron cells looked normal although there were also degeneration (21.16 ± 2.56/FV), necrosis (5.75 ± 0.98/FV), apoptosis (2.91 ± 0.80/FV), congestion ( 0.91 ± 0.49/FV), inflammatory cells (4.58 ± 1.15/FV) and hemorrhage (2.41 ± 1.11/FV). Changes in the rat traumatic brain injury group showed a lot of damage to neuron cells in the form of degeneration (48.41 ± 3.27/FV), necrosis (36.66 ± 2.89/FV), apoptosis (18.91 ± 1.24/FV), congestion (2.50 ±0.31/FV), inflammatory cells (11.41 ± 1.71/FV) and hemorrhage (10.08 ± 2.17/FV). Based on the results of statistical analysis, it can be seen that in all parameters there is a significant difference (p ≤ 0.001). Conclusions The premortem histopathology findings in rats with and without TBI which can be used for the benefit of forensic medicine in determining whether TBI is present or not. It is necessary to look more closely at the histopathology changes in the form of necrosis, apoptosis and hemorrhage.
Collapse
Affiliation(s)
- Taufik Suryadi
- Department of Forensic Medicine and Medicolegal, Dr. Zainoel Abidin Hospital, Banda Aceh, Aceh, 23126, Indonesia
- Department of Forensic Medicine and Medicolegal, Faculty of Medicine, Syiah Kuala University, Banda Aceh, Aceh, 23111, Indonesia
| | - Kulsum Kulsum
- Department of Anesthesiology and Intensive Therapy, Dr. Zainoel Abidin Hospital, Banda Aceh, Aceh, 23126, Indonesia
- Department of Anesthesiology and Intensive Therapy, Faculty of Medicine, Syiah Kuala University, Banda Aceh, Aceh, 23111, Indonesia
| |
Collapse
|
3
|
Feng D, Liu T, Zhang X, Xiang T, Su W, Quan W, Jiang R. Fingolimod improves diffuse brain injury by promoting AQP4 polarization and functional recovery of the glymphatic system. CNS Neurosci Ther 2024; 30:e14669. [PMID: 38459666 PMCID: PMC10924110 DOI: 10.1111/cns.14669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/26/2024] [Accepted: 02/17/2024] [Indexed: 03/10/2024] Open
Abstract
BACKGROUND Diffuse brain injury (DBI) models are characterized by intense global brain inflammation and edema, which characterize the most severe form of TBI. In a previous experiment, we found that fingolimod promoted recovery after controlled cortical impact injury (CCI) by modulating inflammation around brain lesions. However, it remains unclear whether fingolimod can also attenuate DBI because of its different injury mechanisms. Furthermore, whether fingolimod has additional underlying effects on repairing DBI is unknown. METHODS The impact acceleration model of DBI was established in adult Sprague-Dawley rats. Fingolimod (0.5 mg/kg) was administered 0.5, 24, and 48 h after injury for 3 consecutive days. Immunohistochemistry, immunofluorescence analysis, cytokine array, and western blotting were used to evaluate inflammatory cells, inflammatory factors, AQP4 polarization, apoptosis in brain cells, and the accumulation of APP after DBI in rats. To evaluate the function of the glymphatic system (GS), a fluorescent tracer was injected into the cistern. The neural function of rats with DBI was evaluated using various tests, including the modified neurological severity score (mNSS), horizontal ladder-crossing test, beam walking test, and tape sensing and removal test. Brain water content was also measured. RESULTS Fingolimod administration for 3 consecutive days could reduce the levels of inflammatory cytokines, neutrophil recruitment, microglia, and astrocyte activation in the brain following DBI. Moreover, fingolimod reduced apoptotic protein expression, brain cell apoptosis, brain edema, and APP accumulation. Additionally, fingolimod inhibited the loss of AQP4 polarization, improved lymphatic system function, and reduced damage to nervous system function. Notably, inhibiting the GS weakened the therapeutic effect of fingolimod on the neurological function of rats with DBI and increased the accumulation of APP in the brain. CONCLUSIONS In brief, these findings suggest that fingolimod alleviates whole-brain inflammation and GS system damage after DBI and that inhibiting the GS could weaken the positive effect of fingolimod on nerve function in rats with DBI. Thus, inhibiting inflammation and regulating the GS may be critical for the therapeutic effect of fingolimod on DBI.
Collapse
Affiliation(s)
- Dongyi Feng
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| | - Tao Liu
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| | - Xinjie Zhang
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| | - Tangtang Xiang
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| | - Wanqiang Su
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| | - Wei Quan
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| | - Rongcai Jiang
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
- Tianjin Neurological Institute, Key Laboratory of Post Neuro‐injury Neuro‐repair and Regeneration in Central Nervous System, State Key Laboratory of Experimental HematologyMinistry of EducationTianjinChina
| |
Collapse
|
4
|
Vaghebin R, Khalili M, Amiresmaili S, Roghani M, Esmaeili Saber SS, Namdar H. Saphenous vein phlebotomy alleviates neuroinflammatory response and oxidative stress following traumatic brain injury. INTERDISCIPLINARY NEUROSURGERY 2022. [DOI: 10.1016/j.inat.2022.101626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
|
5
|
Allioux C, Achaintre L, Cheataini F, Balança B, Marinesco S. Animal welfare assessment after severe traumatic brain injury in rats. Lab Anim 2022; 56:528-539. [PMID: 35942536 DOI: 10.1177/00236772221107269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Severe traumatic brain injury (TBI) is a multifactorial injury process involving respiratory, cardiovascular and immune functions in addition to the brain. Thus, live animal models are needed to study the molecular, cellular and systemic mechanisms of TBI. The ethical use of laboratory animals requires that the benefits of approaches be carefully weighed against potential harm to animals. Welfare assessments adapted to severe TBI research are lacking. Here, we introduce a scoresheet to describe and monitor potential distress in animals, which includes general welfare (body weight, general appearance and spontaneous behaviour) and TBI-specific indices (respiratory function, pain, locomotor impairment, wound healing). Implementation of this scoresheet in Sprague-Dawley rats subjected to severe lateral fluid percussion TBI revealed a period of suffering limited to four days, followed by a recovery to normal welfare scores within 10-15 days, with females showing a worse impact than males. The scores indicate that animal suffering in this model is transitory compared with TBI consequences in humans. The scoresheet allows for the implementation of refinement measures including (1) analgesia during the initial period following TBI and (2) humane endpoints set (30% weight loss, score ≥90 and/or respiratory problems). This animal scoresheet tailored to TBI research provides a basis for further refinement of animal research paradigms aimed at understanding or treating the sequelae of severe TBI.
Collapse
Affiliation(s)
- Clélia Allioux
- Team TIGER, Lyon Neuroscience Research Centre, Inserm U1028, CNRS UMR 5292, Bron, France.,Claude Bernard Lyon 1 University, Villeurbanne, France
| | - Laëtitia Achaintre
- Neurocampus Animal Housing, Lyon Neuroscience Research Centre, Bron, France
| | - Fatima Cheataini
- Team TIGER, Lyon Neuroscience Research Centre, Inserm U1028, CNRS UMR 5292, Bron, France
| | - Baptiste Balança
- Team TIGER, Lyon Neuroscience Research Centre, Inserm U1028, CNRS UMR 5292, Bron, France.,Department of Neurological Anesthesiology and Intensive Care Medicine, Hospices Civils de Lyon, Hôpital Pierre Wertheimer, Bron, France
| | - Stéphane Marinesco
- Team TIGER, Lyon Neuroscience Research Centre, Inserm U1028, CNRS UMR 5292, Bron, France.,AniRA-BELIV Technological Platform, Lyon Neuroscience Research Centre, Bron, France
| |
Collapse
|
6
|
Zhao Y, Wu J, Li D, Liu J, Chen W, Hou Z, Liu K, Jiang L, Chen X, Wang L, Hu B, Zong F, Wang Y, Wang Y. Human ESC-derived immunity- and matrix- regulatory cells ameliorated white matter damage and vascular cognitive impairment in rats subjected to chronic cerebral hypoperfusion. Cell Prolif 2022; 55:e13223. [PMID: 35437845 PMCID: PMC9136497 DOI: 10.1111/cpr.13223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/24/2022] [Accepted: 03/03/2022] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVES This study investigated the ability of immunity- and matrix- regulatory cells (IMRCs) to improve cognitive function in a rat model of vascular cognitive impairment. MATERIALS AND METHODS A chronic cerebral hypoperfusion (CCH) model was established in rats via permanent bilateral occlusion of the common carotid arteries (two-vessel occlusion, 2VO). The rats then received intravenous injections of IMRCs or saline. A single injection of different doses of IMRCs (1 × 106 cells/rat, 2 × 106 cells/rat, or 4 × 106 cells/rat) was administered via tail vein 72 h after establishment of the model. To evaluate functional recovery, the rats were subjected to behavioural tests after 30 days of CCH. Imaging, western blotting, immunofluorescence staining, and quantitative real-time PCR were used to analyse neuroinflammation and white matter injury after 14 and 40 days of CCH. RNA sequencing (RNA-seq) was used to profile gene expression changes in copine 1 (CPNE1) in response to IMRCs treatment. RESULTS Intravenous injection of 4 × 106 IMRCs alleviated white matter damage and ameliorated cognitive deficits in rats subjected to CCH. Immunofluorescence staining suggested that activation of microglia and astrocytes was reduced, and RNA sequencing showed that CPNE1 expression was significantly elevated following treatment with IMRCs. CONCLUSIONS Intravenous injection of IMRCs protected against CCH-induced white matter injury and cognitive impairment inhibition of microglial activation and regulation of microglia polarization.
Collapse
|
7
|
Kamal SR, Potukutchi S, Gelovani DJ, Bonomi RE, Kallakuri S, Cavanaugh JM, Mangner T, Conti A, Liu RS, Pasqualini R, Arap W, Sidman RL, Perrine SA, Gelovani JG. Spatial and temporal dynamics of HDACs class IIa following mild traumatic brain injury in adult rats. Mol Psychiatry 2022; 27:1683-1693. [PMID: 35027678 DOI: 10.1038/s41380-021-01369-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 09/28/2021] [Accepted: 10/15/2021] [Indexed: 11/09/2022]
Abstract
The fundamental role of epigenetic regulatory mechanisms involved in neuroplasticity and adaptive responses to traumatic brain injury (TBI) is gaining increased recognition. TBI-induced neurodegeneration is associated with several changes in the expression-activity of various epigenetic regulatory enzymes, including histone deacetylases (HDACs). In this study, PET/CT with 6-([18F]trifluoroacetamido)-1- hexanoicanilide ([18F]TFAHA) to image spatial and temporal dynamics of HDACs class IIa expression-activity in brains of adult rats subjected to a weight drop model of diffuse, non-penetrating, mild traumatic brain injury (mTBI). The mTBI model was validated by histopathological and immunohistochemical analyses of brain tissue sections for localization and magnitude of expression of heat-shock protein-70 kDa (HSP70), amyloid precursor protein (APP), cannabinoid receptor-2 (CB2), ionized calcium-binding adapter protein-1 (IBA1), histone deacetylase-4 and -5 (HDAC4 and HDAC5). In comparison to baseline, the expression-activities of HDAC4 and HDAC5 were downregulated in the hippocampus, nucleus accumbens, peri-3rd ventricular part of the thalamus, and substantia nigra at 1-3 days post mTBI, and remained low at 7-8 days post mTBI. Reduced levels of HDAC4 and HDAC5 expression observed in neurons of these brain regions post mTBI were associated with the reduced nuclear and neuropil levels of HDAC4 and HDAC5 with the shift to perinuclear localization of these enzymes. These results support the rationale for the development of therapeutic strategies to upregulate expression-activity of HDACs class IIa post-TBI. PET/CT (MRI) with [18F]TFAHA can facilitate the development and clinical translation of unique therapeutic approaches to upregulate the expression and activity of HDACs class IIa enzymes in the brain after TBI.
Collapse
Affiliation(s)
- Swatabdi R Kamal
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Shreya Potukutchi
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - David J Gelovani
- School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Robin E Bonomi
- School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Srinivasu Kallakuri
- Department of Psychiatry and Behavioral Neurosciences, School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - John M Cavanaugh
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Thomas Mangner
- Cyclotron-Radiochemistry Facility, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Alana Conti
- Research and Development Service, John D. Dingell VA Medical Center, Detroit, MI, 48201, USA
- Departments of Neurosurgery and Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Ren-Shyan Liu
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Department of Nuclear Medicine, Cheng-Hsin General Hospital, Taipei, 112, Taiwan
- Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Renata Pasqualini
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
- Rutgers Cancer Institute of New Jersey, Newark, NJ, 07103, USA
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, NJ, 07103, USA
- Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Richard L Sidman
- Department of Neurology, Harvard Medical School, Boston, MA, 02115, USA
| | - Shane A Perrine
- Department of Psychiatry and Behavioral Neurosciences, School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Juri G Gelovani
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, MI, 48201, USA.
- Molecular Imaging Program, Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA.
- College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE.
| |
Collapse
|
8
|
Xu XJ, Yang MS, Zhang B, Ge QQ, Niu F, Dong JQ, Zhuang Y, Liu BY. Genome-wide interrogation of transfer RNA-derived small RNAs in a mouse model of traumatic brain injury. Neural Regen Res 2022; 17:386-394. [PMID: 34269214 PMCID: PMC8463968 DOI: 10.4103/1673-5374.314315] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Transfer RNA (tRNA)-derived small RNAs (tsRNAs) are a recently established family of regulatory small non-coding RNAs that modulate diverse biological processes. Growing evidence indicates that tsRNAs are involved in neurological disorders and play a role in the pathogenesis of neurodegenerative disease. However, whether tsRNAs are involved in traumatic brain injury-induced secondary injury remains poorly understood. In this study, a mouse controlled cortical impact model of traumatic brain injury was established, and integrated tsRNA and messenger RNA (mRNA) transcriptome sequencing were used. The results revealed that 103 tsRNAs were differentially expressed in the mouse model of traumatic brain injury at 72 hours, of which 56 tsRNAs were upregulated and 47 tsRNAs were downregulated. Based on microRNA-like seed matching and Pearson correlation analysis, 57 differentially expressed tsRNA-mRNA interaction pairs were identified, including 29 tsRNAs and 26 mRNAs. Moreover, Gene Ontology annotation of target genes revealed that the significantly enriched terms were primarily associated with inflammation and synaptic function. Collectively, our findings suggest that tsRNAs may be associated with traumatic brain injury-induced secondary brain injury, and are thus a potential therapeutic target for traumatic brain injury. The study was approved by the Beijing Neurosurgical Institute Animal Care and Use Committee (approval No. 20190411) on April 11, 2019.
Collapse
Affiliation(s)
- Xiao-Jian Xu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Meng-Shi Yang
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bin Zhang
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qian-Qian Ge
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fei Niu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jin-Qian Dong
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuan Zhuang
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Bai-Yun Liu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University; Nerve Injury and Repair Center of Beijing Institute for Brain Disorders; China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
9
|
Jiang H, Li H, Cao Y, Zhang R, Zhou L, Zhou Y, Zeng X, Wu J, Wu D, Wu D, Guo X, Li X, Wu H, Li P. Effects of cannabinoid (CBD) on blood brain barrier permeability after brain injury in rats. Brain Res 2021; 1768:147586. [PMID: 34289379 DOI: 10.1016/j.brainres.2021.147586] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/23/2021] [Accepted: 07/14/2021] [Indexed: 01/30/2023]
Abstract
Cannabidiol is a natural herbal medicine known to protect the brain from traumatic brain injury (TBI). Here, a TBI rat model was established, with cannabidiol administered intraperitoneally at doses of 5, 10, or 20 mg/kg, 30 min before surgery and 6 h after surgery until sacrifice. Brain water content, body weight, and modified neurological severity scores were determined, and enzyme-linked immunosorbent assay, immunofluorescence staining, hematoxylin and eosin staining, Nissl staining, Evans-blue dye extravasation, and western blotting were performed. Results showed that cannabidiol decreased the number of aquaporin-4-positive and glial fibrillary acidic protein-positive cells. Cannabidiol also significantly reduced the protein levels of proinflammatory cytokines (TNF-α and IL-1β) and significantly increased the expression of tight junction proteins (claudin-5 and occludin). Moreover, cannabidiol administration significantly mitigated water content in the brain after TBI and blood-brain barrier disruption and ameliorated the neurological deficit score after TBI. Cannabidiol administration improved the integrity and permeability of the blood-brain barrier and reduced edema in the brain after TBI.
Collapse
Affiliation(s)
- Hongyan Jiang
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China; Department of Pathology, Suining Central Hospital, Suining 629000, China
| | - Hengxi Li
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Yan Cao
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Ruilin Zhang
- Department of Forensic Medicine of Kunming Medical University, Kunming 650500, China
| | - Lei Zhou
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Biomedical Engineering Research Center, Kunming Medical University, Kunming 650500, China
| | - Ying Zhou
- Department of Kunming Medical University Electron Microscope Laboratory, Kunming Medical University, Kunming 650500, China
| | - Xiaofeng Zeng
- Department of Forensic Medicine of Kunming Medical University, Kunming 650500, China
| | - Jia Wu
- Department of Morphology Laboratory, Kunming Medical University, Kunming 650500, China
| | - Douwei Wu
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Deye Wu
- Department of Human Anatomy and Histology/Embryology, Qilu Medical University, Zibo 255213, Shandong, China
| | - Xiaobing Guo
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Xiaowen Li
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Haiying Wu
- Department of Emergency and Intensive Care Unit, First Affiliated Hospital, Kunming Medical University, Kunming 650032, China.
| | - Ping Li
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China.
| |
Collapse
|
10
|
Nong A, Li Q, Huang Z, Xu Y, He K, Jia Y, Cen Z, Liao L, Huang Y. MicroRNA miR-126 attenuates brain injury in septic rats via NF-κB signaling pathway. Bioengineered 2021; 12:2639-2648. [PMID: 34115555 PMCID: PMC8806573 DOI: 10.1080/21655979.2021.1937905] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The purpose of this study was to investigate the impact and mechanism of microRNA miR-126 on brain injury induced by blood-brain barrier (BBB) damage in septic rats. We used cecal ligation and perforation (CLP) to create a rat model of sepsis. The experimental rats were randomly divided into Control group, CLP group, CLP + miR-NC group, CLP + miR-126 group and CLP + miR-126 + NF-κB pathway agonist (PMA) group. MiR-126 expressed in the brain tissue of CLP rats was down-regulated by qRT-PCR. Upregulation of miR-126 in CLP rats could improve brain injury and BBB marker protein level, reduce brain water content, Evans blue extravasation, inflammation, and excessive oxidative stress. This could also result in an inhibition of NF-κB signaling pathway activity. In conclusion, miR-126 overexpression can prevent brain injury caused by BBB damage via the inhibition of NF-κB signaling pathway activity.
Collapse
Affiliation(s)
- Anna Nong
- Graduate School, Youjiang Medical University for Nationalities, Baise, Guangxi China
| | - Qingfeng Li
- Department of Radiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi China
| | - Zhijing Huang
- Department of Pediatric Internal Medicine Ward 1, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi China
| | - Yunan Xu
- Graduate School, Youjiang Medical University for Nationalities, Baise, Guangxi China
| | - Kebin He
- Graduate School, Youjiang Medical University for Nationalities, Baise, Guangxi China
| | - Yuying Jia
- Graduate School, Youjiang Medical University for Nationalities, Baise, Guangxi China
| | - Zhenyi Cen
- Graduate School, Youjiang Medical University for Nationalities, Baise, Guangxi China
| | - Lianghua Liao
- Graduate School, Youjiang Medical University for Nationalities, Baise, Guangxi China
| | - Yueyan Huang
- Department of Radiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi China
| |
Collapse
|
11
|
Yang MS, Xu XJ, Zhang B, Niu F, Liu BY. Comparative transcriptomic analysis of rat versus mouse cerebral cortex after traumatic brain injury. Neural Regen Res 2021; 16:1235-1243. [PMID: 33318400 PMCID: PMC8284282 DOI: 10.4103/1673-5374.301028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The heterogeneity of traumatic brain injury (TBI)-induced secondary injury has greatly hampered the development of effective treatments for TBI patients. Targeting common processes across species may be an innovative strategy to combat debilitating TBI. In the present study, a cross-species transcriptome comparison was performed for the first time to determine the fundamental processes of secondary brain injury in Sprague-Dawley rat and C57/BL6 mouse models of TBI, caused by acute controlled cortical impact. The RNA sequencing data from the mouse model of TBI were downloaded from the Gene Expression Omnibus (ID: GSE79441) at the National Center for Biotechnology Information. For the rat data, peri-injury cerebral cortex samples were collected for transcriptomic analysis 24 hours after TBI. Differentially expressed gene-based functional analysis revealed that common features between the two species were mainly involved in the regulation and activation of the innate immune response, including complement cascades as well as Toll-like and nucleotide oligomerization domain-like receptor pathways. These findings were further corroborated by gene set enrichment analysis. Moreover, transcription factor analysis revealed that the families of signal transducers and activators of transcription (STAT), basic leucine zipper (BZIP), Rel homology domain (RHD), and interferon regulatory factor (IRF) transcription factors play vital regulatory roles in the pathophysiological processes of TBI, and are also largely associated with inflammation. These findings suggest that targeting the common innate immune response might be a promising therapeutic approach for TBI. The animal experimental procedures were approved by the Beijing Neurosurgical Institute Animal Care and Use Committee (approval No. 201802001) on June 6, 2018.
Collapse
Affiliation(s)
- Meng-Shi Yang
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiao-Jian Xu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Bin Zhang
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fei Niu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Bai-Yun Liu
- Beijing Key Laboratory of Central Nervous System Injury, Beijing Neurosurgical Institute; Beijing Key Laboratory of Central Nervous System Injury and Department of Neurosurgery, Beijing Neurosurgical Institute and Beijing Tiantan Hospital, Capital Medical University; Nerve Injury and Repair Center of Beijing Institute for Brain Disorders; China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
12
|
Li D, Zhang Y, Lu L, Zhang L, Ma J, Ji J, Li H, Chen G. Upregulation of Sec22b plays a neuroprotective role in a rat model of traumatic brain injury via inducing protective autophagy. Brain Res Bull 2020; 166:29-36. [PMID: 33186631 DOI: 10.1016/j.brainresbull.2020.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 11/03/2020] [Accepted: 11/06/2020] [Indexed: 11/16/2022]
Abstract
Cortical neuronal cell death following traumatic brain injury (TBI) evoked by the cortical impact is a significant factor that contributes to neurological deficits. In the current study, we harvested the injured area and perilesional area of the injured brain induced by TBI. We explored the functions of Sec22b, an apoptosis-promoting kinase, and a pivotal bridge builder of apoptotic signaling in the etiopathogenesis of an experimental rat model of TBI. We found that Sec22b was expressed in neurons in the injured cortical area, and the expression level significantly decreased after TBI, especially at 24 h. Administration of Sec22b overexpressed plasmid significantly ameliorated TBI-induced apoptosis, neurological deficits, and blood-brain barrier permeability, accompanied by the activation of autophagy. However, the administration of Sec22b knockdown resulted in the opposite eff ;ects. Altogether, these findings indicated that Sec22b plays a neuroprotective role after TBI, suggesting that Sec22b may be a potential therapeutic target for TBI. We speculated that this neuroprotective effect might be achieved by upregulating autophagy levels and required further studies to explore.
Collapse
Affiliation(s)
- Di Li
- Department of Neurosurgery and Translational Medicine Center, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Yan Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lina Lu
- Department of Radiation Oncology, The Affiliated Suzhou Science & Technology Town Hospital of Nanjing Medical University, Suzhou, China
| | - Ling Zhang
- Department of Neurosurgery and Translational Medicine Center, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Jialing Ma
- Department of Anesthesia, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, China
| | - Jiaxuan Ji
- Department of Neurosurgery, Zhangjiagang Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Suzhou, China.
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
13
|
Zhang J, Xiao B, Li CX, Wang Y. Fingolimod (FTY720) improves postoperative cognitive dysfunction in mice subjected to D-galactose-induced aging. Neural Regen Res 2020; 15:1308-1315. [PMID: 31960817 PMCID: PMC7047799 DOI: 10.4103/1673-5374.272617] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 07/18/2019] [Accepted: 08/13/2019] [Indexed: 12/19/2022] Open
Abstract
Neurocognitive dysfunction is a common postoperative complication, especially in older adult patients. Fingolimod (FTY720) is a sphingosine-1-phosphate receptor modulator that has been found to be neuroprotective in several animal models of central nervous system disease. However, few reports have examined whether FTY720 could mitigate postoperative cognitive dysfunction. In this study, we investigated whether FTY720 could prevent postoperative neurocognitive impairment in mice subjected to D-galactose-induced aging. We induced an accelerated model of aging by administering an intraperitoneal injection of D-galactose. Subsequently, we performed a partial hepatolobectomy under sevoflurane anesthesia. FTY720 (1 mg/kg) was administered intraperitoneally 3 hours before and 24 hours after anesthesia and surgery. Our results indicated that anesthesia and surgery significantly impaired spatial memory in the Y-maze test 6 hours after surgery. We also found that problem solving ability and long-term memory in the puzzle box test on postoperative days 2-4 were significantly improved by FTY720 treatment. Immunohistochemical staining and western blot assay demonstrated that FTY720 significantly inhibited microglial activation in the hippocampal CA1 region of mice 6 hours and 3 days after anesthesia, and down-regulated the expression of synaptic-related proteins postsynaptic density protein 95 and GluR2 in the hippocampus. These results indicate that FTY720 improved postoperative neurocognitive dysfunction in mice subjected to D-galactose-induced aging. This study was approved by the Experimental Animal Ethics Committee of the Third Xiangya Hospital of Central South University of China (approval No. LLSC (LA) 2016-025) on September 27, 2016.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Bin Xiao
- Department of Orthopedics, the Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Chen-Xu Li
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Yi Wang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| |
Collapse
|
14
|
Liaudanskaya V, Chung JY, Mizzoni C, Rouleau N, Berk AN, Wu L, Turner JA, Georgakoudi I, Whalen MJ, Nieland TJF, Kaplan DL. Modeling Controlled Cortical Impact Injury in 3D Brain-Like Tissue Cultures. Adv Healthc Mater 2020; 9:e2000122. [PMID: 32406202 PMCID: PMC7395313 DOI: 10.1002/adhm.202000122] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 04/26/2020] [Indexed: 12/13/2022]
Abstract
Traumatic brain injury (TBI) survivors suffer long term from mental illness, neurodegeneration, and neuroinflammation. Studies of 3D tissue models have provided new insights into the pathobiology of many brain diseases. Here, a 3D in vitro contusion model is developed consisting of mouse cortical neurons grown on a silk scaffold embedded in collagen and used outcomes from an in vivo model for benchmarking. Molecular, cellular, and network events are characterized in response to controlled cortical impact (CCI). In this model, CCI induces degradation of neural network structure and function and release of glutamate, which are associated with the expression of programmed necrosis marker phosphorylated Mixed Lineage Kinase Domain Like Pseudokinase (pMLKL). Neurodegeneration is observed first in the directly impacted area and it subsequently spreads over time in 3D space. CCI reduces phosphorylated protein kinase B (pAKT) and Glycogen synthase kinase 3 beta (GSK3β) in neurons in vitro and in vivo, but discordant responses are observed in phosphprylated ribosomal S6 kinase (pS6) and phosphorylated Tau (pTau) expression. In summary, the 3D brain-like culture system mimicked many aspects of in vivo responses to CCI, providing evidence that the model can be used to study the molecular, cellular, and functional sequelae of TBI, opening up new possibilities for discovery of therapeutics.
Collapse
Affiliation(s)
- Volha Liaudanskaya
- Department of Biomedical Engineering, Tufts University, Medford, 02155, MA, USA
| | - Joon Yong Chung
- Neuroscience Center, Massachusetts General Hospital, Charlestown, 02129, MA, USA
| | - Craig Mizzoni
- Department of Biomedical Engineering, Tufts University, Medford, 02155, MA, USA
| | - Nicolas Rouleau
- Department of Biomedical Engineering, Tufts University, Medford, 02155, MA, USA
| | - Alexander N Berk
- Department of Biomedical Engineering, Tufts University, Medford, 02155, MA, USA
| | - Limin Wu
- Neuroscience Center, Massachusetts General Hospital, Charlestown, 02129, MA, USA
| | - Julia A Turner
- Department of Biomedical Engineering, Tufts University, Medford, 02155, MA, USA
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University, Medford, 02155, MA, USA
| | - Michael J Whalen
- Neuroscience Center, Massachusetts General Hospital, Charlestown, 02129, MA, USA
| | - Thomas J F Nieland
- Department of Biomedical Engineering, Tufts University, Medford, 02155, MA, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, 02155, MA, USA
| |
Collapse
|
15
|
Abstract
Translational genomics represents a broad field of study that combines genome and transcriptome-wide studies in humans and model systems to refine our understanding of human biology and ultimately identify new ways to treat and prevent disease. The approaches to translational genomics can be broadly grouped into two methodologies, forward and reverse genomic translation. Traditional (forward) genomic translation begins with model systems and aims at using unbiased genetic associations in these models to derive insight into biological mechanisms that may also be relevant in human disease. Reverse genomic translation begins with observations made through human genomic studies and refines these observations through follow-up studies using model systems. The ultimate goal of these approaches is to clarify intervenable processes as targets for therapeutic development. In this review, we describe some of the approaches being taken to apply translational genomics to the study of diseases commonly encountered in the neurocritical care setting, including hemorrhagic and ischemic stroke, traumatic brain injury, subarachnoid hemorrhage, and status epilepticus, utilizing both forward and reverse genomic translational techniques. Further, we highlight approaches in the field that could be applied in neurocritical care to improve our ability to identify new treatment modalities as well as to provide important information to patients about risk and prognosis.
Collapse
Affiliation(s)
- Pavlos Myserlis
- Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN 6818, Boston, MA, 02114, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Farid Radmanesh
- Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Christopher D Anderson
- Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN 6818, Boston, MA, 02114, USA.
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA.
- Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
16
|
Abstract
Adrenomedullin, a peptide with multiple physiological functions in nervous system injury and disease, has aroused the interest of researchers. This review summarizes the role of adrenomedullin in neuropathological disorders, including pathological pain, brain injury and nerve regeneration, and their treatment. As a newly characterized pronociceptive mediator, adrenomedullin has been shown to act as an upstream factor in the transmission of noxious information for various types of pathological pain including acute and chronic inflammatory pain, cancer pain, neuropathic pain induced by spinal nerve injury and diabetic neuropathy. Initiation of glia-neuron signaling networks in the peripheral and central nervous system by adrenomedullin is involved in the formation and maintenance of morphine tolerance. Adrenomedullin has been shown to exert a facilitated or neuroprotective effect against brain injury including hemorrhagic or ischemic stroke and traumatic brain injury. Additionally, adrenomedullin can serve as a regulator to promote nerve regeneration in pathological conditions. Therefore, adrenomedullin is an important participant in nervous system diseases.
Collapse
Affiliation(s)
- Feng-Jiao Li
- College of Life Sciences, Laboratory of Neuroendocrinology, Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian Province, China
| | - Si-Ru Zheng
- College of Life Sciences, Laboratory of Neuroendocrinology, Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian Province, China
| | - Dong-Mei Wang
- College of Life Sciences, Laboratory of Neuroendocrinology, Provincial Key Laboratory of Developmental Biology and Neuroscience, Fujian Normal University, Fuzhou, Fujian Province, China
| |
Collapse
|