1
|
Kajikawa H, Matsuura K, Ii Y, Tabei K, Nakamura N, Ishikawa H, Nishiguchi Y, Matsuda K, Kagawa K, Ichikawa N, Araki T, Shindo A. Effects of Goreisan in the Perioperative Period of Subthalamic Deep Brain Stimulation in Parkinson's Disease. Brain Behav 2024; 14:e70069. [PMID: 39467102 PMCID: PMC11516107 DOI: 10.1002/brb3.70069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/23/2024] [Accepted: 09/08/2024] [Indexed: 10/30/2024] Open
Abstract
INTRODUCTION Patients with Parkinson's disease (PD) may benefit from deep brain stimulation (DBS). Perifocal brain edema sometimes occurs after DBS surgery, but it is transient and does not affect the final prognosis. Transient deterioration of cognitive function has been reported in patients with frontal edema in the first postoperative week. This study aimed to investigate the effect of Goreisan in preventing edematous changes after DBS and determine the influence of edema on cognition. METHODS We included 29 patients with PD who underwent bilateral subthalamic nucleus (STN) DBS and who were divided into 2 groups: those using (11 patients) and those not using Goreisan (18 patients). At 1 week postoperatively, all patients underwent magnetic resonance imaging. We measured the volume of edema either in the frontal white matter or STN on fluid-attenuated inversion recovery (FLAIR) images. Finally, brain edema, motor function, and cognitive function were compared between the groups with and without Goreisan. RESULTS In the FLAIR image 1 week postoperatively, the average postoperative frontal subcortical edema (FE) volume of the group with Goreisan was significantly lower than that without Goreisan (2249 ± 2186 mm3, 6261 ± 7213 mm3, respectively, p = 0.023). Multivariate analysis with age, preoperative Mini-Mental State Examination (MMSE) score, FE, and peri-STN edema (SE) as factors, and MMSE at 1 week postoperatively as the dependent variable showed that preoperative MMSE score and SE were significant as associated factors. CONCLUSIONS FE after DBS surgery may be alleviated using Goreisan. SE and preoperative MMSE scores were associated with MMSE scores 1 week postoperatively. TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
- Hiroyuki Kajikawa
- Department of Neurology, Graduate School of MedicineMie UniversityMieJapan
- Department of NeurorolgySuzukakaisei HospitalMieJapan
| | - Keita Matsuura
- Department of Neurology, Graduate School of MedicineMie UniversityMieJapan
- Department of NeurorolgySuzukakaisei HospitalMieJapan
| | - Yuichiro Ii
- Department of Neuroimaging and PathophysiologyMie University School of MedicineMieJapan
| | - Ken‐ichi Tabei
- School of Industrial Technology, Advanced Institute of Industrial TechnologyTokyo Metropolitan Public University CorporationTokyoJapan
| | - Naoko Nakamura
- Department of Neurology, Graduate School of MedicineMie UniversityMieJapan
| | - Hidehiro Ishikawa
- Department of Neurology, Graduate School of MedicineMie UniversityMieJapan
| | - Yamato Nishiguchi
- Department of Neurology, Graduate School of MedicineMie UniversityMieJapan
| | - Kana Matsuda
- Department of Dementia Prevention and TherapeuticsMie University Graduate School of MedicineMieJapan
| | - Ken Kagawa
- Department of NeurorolgySuzukakaisei HospitalMieJapan
| | | | | | - Akihiro Shindo
- Department of Neurology, Graduate School of MedicineMie UniversityMieJapan
| |
Collapse
|
2
|
Zheng H, Xiao X, Han Y, Wang P, Zang L, Wang L, Zhao Y, Shi P, Yang P, Guo C, Xue J, Zhao X. Research progress of propofol in alleviating cerebral ischemia/reperfusion injury. Pharmacol Rep 2024; 76:962-980. [PMID: 38954373 DOI: 10.1007/s43440-024-00620-6] [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: 01/30/2024] [Revised: 06/21/2024] [Accepted: 06/23/2024] [Indexed: 07/04/2024]
Abstract
Ischemic stroke is a leading cause of adult disability and death worldwide. The primary treatment for cerebral ischemia patients is to restore blood supply to the ischemic region as quickly as possible. However, in most cases, more severe tissue damage occurs, which is known as cerebral ischemia/reperfusion (I/R) injury. The pathological mechanisms of brain I/R injury include mitochondrial dysfunction, oxidative stress, excitotoxicity, calcium overload, neuroinflammation, programmed cell death and others. Propofol (2,6-diisopropylphenol), a short-acting intravenous anesthetic, possesses not only sedative and hypnotic effects but also immunomodulatory and neuroprotective effects. Numerous studies have reported the protective properties of propofol during brain I/R injury. In this review, we summarize the potential protective mechanisms of propofol to provide insights for its better clinical application in alleviating cerebral I/R injury.
Collapse
Affiliation(s)
- Haijing Zheng
- Basic Medical College, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
- Zhengzhou Central Hospital, Zhengzhou, China
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
| | - Xian Xiao
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
| | - Yiming Han
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
| | - Pengwei Wang
- Department of Pharmacy, the First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Weihui, Henan, 453100, China
| | - Lili Zang
- Department of Surgery, the First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Weihui, China
| | - Lilin Wang
- Department of Pediatric Surgery, the First Affiliated Hospital of Xinxiang Medical University, No. 88 Jiankang Road, Weihui, China
| | - Yinuo Zhao
- Basic Medical College, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
| | - Peijie Shi
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China
| | - Pengfei Yang
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China.
| | - Chao Guo
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China.
| | - Jintao Xue
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China.
| | - Xinghua Zhao
- Basic Medical College, Xinxiang Medical University, 601, Jin Sui Avenue, Xinxiang, Henan, China.
| |
Collapse
|
3
|
Martínez-Torres AM, Morán J. Aquaporin 4 and the endocannabinoid system: a potential therapeutic target in brain injury. Exp Brain Res 2024; 242:2041-2058. [PMID: 39043897 PMCID: PMC11306651 DOI: 10.1007/s00221-024-06896-7] [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: 02/20/2024] [Accepted: 07/14/2024] [Indexed: 07/25/2024]
Abstract
Brain edema is a critical complication arising from stroke and traumatic brain injury (TBI) with an important impact on patient recovery and can lead to long-term consequences. Therapeutic options to reduce edema progression are limited with variable patient outcomes. Aquaporin 4 (AQP4) is a water channel that allows bidirectional water diffusion across the astrocyte membrane and participates in the distinct phases of cerebral edema. The absence or inhibition of this channel has been demonstrated to ameliorate edema and brain damage. The endocannabinoid system (ECS) is a neuromodulator system with a wide expression in the brain and its activation has shown neuroprotective properties in diverse models of neuronal damage. This review describes and discusses the major features of ECS and AQP4 and their role during brain damage, observing that ECS stimulation reduces edema and injury size in diverse models of brain damage, however, the relationship between AQP4 expression and dynamics and ECS activation remains unclear. The research on these topics holds promising therapeutic implications for the treatment of brain edema following stroke and TBI.
Collapse
Affiliation(s)
- Ari Misael Martínez-Torres
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Coyoacán, Apartado Postal 70-253, 04510, Ciudad de Mexico, México
| | - Julio Morán
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Coyoacán, Apartado Postal 70-253, 04510, Ciudad de Mexico, México.
| |
Collapse
|
4
|
Mo S, Yang C, Zheng X, Lv H, Mao S, Liu N, Yang Q, Liao B, Yang M, Lu Z, Tang L, Huang X, Jian C, Li X, Shang J. Neuroprotective Effects of AER-271 in a tMCAO Mouse Model: Modulation of Autophagy, Apoptosis, and Inflammation. Inflammation 2024:10.1007/s10753-024-02082-7. [PMID: 39117789 DOI: 10.1007/s10753-024-02082-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 08/10/2024]
Abstract
Following ischemic stroke, aquaporin 4 (AQP4) expression modifications have been associated with increased inflammation. However, the underlying mechanisms are not fully understood. This study aims to elucidate the mechanistic basis of post-cerebral ischemia-reperfusion (I/R) inflammation by employing the AQP4-specific inhibitor, AER-271. The middle cerebral artery occlusion (MCAO) model was used to induce ischemic stroke in mice. C57BL/6 mice were randomly allocated into four groups: sham operation, I/R, AER-271, and 2-(nicotinamide)-1,3,4-thiadiazole (TGN-020) treatment, with observations recorded at 1 day, 3 days, and 7 days post-tMCAO. Each group consisted of 15 mice. Procedures included histological examination through HE staining, neurological scoring, Western blot analysis, and immunofluorescence staining. AER-271 treatment yielded significant improvements in post-stroke weight recovery and neurological scores, accompanied by a reduction in cerebral infarction volume. Moreover, AER-271 exhibited a noticeable influence on autophagic and apoptotic pathways, affecting the activation of both pro-inflammatory and anti-inflammatory cytokines. Alterations in the levels of inflammatory biomarkers MCP-1, NLRP3, and caspase 1 were also detected. Finally, a comparative assessment of the effects of AER-271 and TGN-020 in mitigating apoptosis and microglial polarization in ischemic mice revealed neuroprotective effects with no significant difference in efficacy. This study provides essential insights into the neuroprotective mechanisms of AER-271 in cerebral ischemia-reperfusion injury, offering potential clinical applications in the treatment of ischemic cerebrovascular disorders.
Collapse
Affiliation(s)
- Shenglong Mo
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
- Biological Molecule Laboratory, Guangxi University Key Laboratory of High Incidence Prevention and Control Research in Western Guangxi, Baise, 53300, Guangxi, China
- Graduate School of Youjiang, Medical University for Nationalities, Baise, Guangxi, China
| | - Chengmin Yang
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
- Biological Molecule Laboratory, Guangxi University Key Laboratory of High Incidence Prevention and Control Research in Western Guangxi, Baise, 53300, Guangxi, China
| | - Xingwu Zheng
- Department of Geriatrics, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Hui Lv
- Modern Industrial College of Biomedicine and Great Health, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Sanyin Mao
- Department of Neurology, The First People's Hospital of Jiande, Hangzhou, China
| | - Ning Liu
- School of Basic Medical Sciences, Beihua University, Jilin, China
| | - Qin Yang
- Department of Neurology, BAISE PEOPLE'S HOSPITAL, Baise, Guangxi, China
| | - Bao Liao
- Department of Neurology, BAISE PEOPLE'S HOSPITAL, Baise, Guangxi, China
| | - Meiling Yang
- Graduate School of Youjiang, Medical University for Nationalities, Baise, Guangxi, China
| | - Zhicheng Lu
- Graduate School of Youjiang, Medical University for Nationalities, Baise, Guangxi, China
| | - Lina Tang
- Graduate School of Youjiang, Medical University for Nationalities, Baise, Guangxi, China
| | - Xiaorui Huang
- Department of Psychiatry and Psychology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Chongdong Jian
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
- Biological Molecule Laboratory, Guangxi University Key Laboratory of High Incidence Prevention and Control Research in Western Guangxi, Baise, 53300, Guangxi, China.
| | - Xuebin Li
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
- Biological Molecule Laboratory, Guangxi University Key Laboratory of High Incidence Prevention and Control Research in Western Guangxi, Baise, 53300, Guangxi, China.
| | - Jingwei Shang
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
- Biological Molecule Laboratory, Guangxi University Key Laboratory of High Incidence Prevention and Control Research in Western Guangxi, Baise, 53300, Guangxi, China.
| |
Collapse
|
5
|
Chen YY, Gong ZC, Zhang MM, Huang ZH. Brain-Targeting Emodin Mitigates Ischemic Stroke via Inhibiting AQP4-Mediated Swelling and Neuroinflammation. Transl Stroke Res 2024; 15:818-830. [PMID: 37380800 DOI: 10.1007/s12975-023-01170-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/26/2023] [Accepted: 06/20/2023] [Indexed: 06/30/2023]
Abstract
Failure to achieve target-specific delivery to ischemic brain sites has hampered the clinical efficacy of newly developed therapies for ischemic stroke. Emodin, an active ingredient isolated from traditional Chinese medicine, has been indicated to alleviate ischemic stroke; however, the underlying mechanism remains unclear. In this study, we aimed to achieve brain-targeted delivery of emodin to maximize its therapeutic efficacy and elucidate the mechanisms by which emodin alleviates ischemic stroke. A polyethylene glycol (PEG)/cyclic Arg-Gly-Asp (cRGD)-modified liposome was used to encapsulate emodin. TTC, HE, Nissl staining, and immunofluorescence staining were employed to evaluate the therapeutic efficacy of brain-targeting emodin in MCAO and OGD/R models. Inflammatory cytokine levels were determined using ELISA. Immunoprecipitation, immunoblotting, and RT-qPCR were utilized for clarifying the changes in key downstream signaling. Lentivirus-mediated gene restoration was employed to verify the core effector of emodin for relieving ischemic stroke. Encapsulating emodin in a PEG/cRGD-modified liposome enhanced its accumulation in the infarct region and substantially raised its therapeutic efficacy. Furthermore, we demonstrated that AQP4, the most abundant water transporter subunit expressed in astrocytes, plays a crucial role in mediating the mechanisms by which emodin inhibits astrocyte swelling, neuroinflammatory blood-brain barrier (BBB) breakdown in vivo and in vitro, and brain edema in general. Our study unveiled the critical target of emodin responsible for alleviating ischemic stroke and a localizable drug delivery vehicle in the therapeutic strategy for ischemic stroke and other brain injuries.
Collapse
Affiliation(s)
- Yan-Yan Chen
- Wuxi Cancer Institute, and Wuxi Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China.
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi, China.
| | - Zhi-Cheng Gong
- Wuxi Cancer Institute, and Wuxi Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China
| | - Mei-Mei Zhang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi, China
| | - Zhao-Hui Huang
- Wuxi Cancer Institute, and Wuxi Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu, China.
| |
Collapse
|
6
|
Czyżewski W, Litak J, Sobstyl J, Mandat T, Torres K, Staśkiewicz G. Aquaporins: Gatekeepers of Fluid Dynamics in Traumatic Brain Injury. Int J Mol Sci 2024; 25:6553. [PMID: 38928258 PMCID: PMC11204105 DOI: 10.3390/ijms25126553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Aquaporins (AQPs), particularly AQP4, play a crucial role in regulating fluid dynamics in the brain, impacting the development and resolution of edema following traumatic brain injury (TBI). This review examines the alterations in AQP expression and localization post-injury, exploring their effects on brain edema and overall injury outcomes. We discuss the underlying molecular mechanisms regulating AQP expression, highlighting potential therapeutic strategies to modulate AQP function. These insights provide a comprehensive understanding of AQPs in TBI and suggest novel approaches for improving clinical outcomes through targeted interventions.
Collapse
Affiliation(s)
- Wojciech Czyżewski
- Department of Neurosurgery, Maria Sklodowska-Curie National Research Institute of Oncology, ul. W.K. Roentgena 5, 02-781 Warsaw, Poland;
- Department of Didactics and Medical Simulation, Medical University of Lublin, 20-954 Lublin, Poland
| | - Jakub Litak
- Department of Clinical Immunology, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Jan Sobstyl
- Department of Interventional Radiology and Neuroradiology, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Tomasz Mandat
- Department of Neurosurgery, Maria Sklodowska-Curie National Research Institute of Oncology, ul. W.K. Roentgena 5, 02-781 Warsaw, Poland;
| | - Kamil Torres
- Department of Plastic, Reconstructive Surgery with Microsurgery, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Grzegorz Staśkiewicz
- Department of Human, Clinical and Radiological Anatomy, Medical University, 20-954 Lublin, Poland;
| |
Collapse
|
7
|
Sun M, Baker TL, Wilson CT, Brady RD, Yamakawa GR, Wright DK, Mychasiuk R, Vo A, Wilson T, Allen J, McDonald SJ, Shultz SR. Treatment with the vascular endothelial growth factor-A antibody, bevacizumab, has sex-specific effects in a rat model of mild traumatic brain injury. J Cereb Blood Flow Metab 2024; 44:542-555. [PMID: 37933736 PMCID: PMC10981407 DOI: 10.1177/0271678x231212377] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 11/08/2023]
Abstract
Mild traumatic brain injury (mTBI) involves damage to the cerebrovascular system. Vascular endothelial growth factor-A (VEGF-A) is an important modulator of vascular health and VEGF-A promotes the brain's ability to recover after more severe forms of brain injury; however, the role of VEGF-A in mTBI remains poorly understood. Bevacizumab (BEV) is a monoclonal antibody that binds to VEGF-A and neutralises its actions. To better understand the role of VEGF-A in mTBI recovery, this study examined how BEV treatment affected outcomes in rats given a mTBI. Adult Sprague-Dawley rats were assigned to sham-injury + vehicle treatment (VEH), sham-injury + BEV treatment, mTBI + VEH treatment, mTBI + BEV treatment groups. Treatment was administered intracerebroventricularly via a cannula beginning at the time of injury and continuing until the end of the study. Rats underwent behavioral testing after injury and were euthanized on day 11. In both females and males, BEV had a negative impact on cognitive function. mTBI and BEV treatment increased the expression of inflammatory markers in females. In males, BEV treatment altered markers related to hypoxia and vascular health. These novel findings of sex-specific responses to BEV and mTBI provide important insights into the role of VEGF-A in mTBI.
Collapse
Affiliation(s)
- Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Tamara L Baker
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Campbell T Wilson
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Rhys D Brady
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Anh Vo
- Monash Health Translation Precinct, Monash University, Melbourne, VIC, Australia
| | - Trevor Wilson
- Monash Health Translation Precinct, Monash University, Melbourne, VIC, Australia
| | - Josh Allen
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Health Sciences, Vancouver Island University, Nanaimo, BC, Canada
| |
Collapse
|
8
|
Das N, Dhamija R, Sarkar S. The role of astrocytes in the glymphatic network: a narrative review. Metab Brain Dis 2024; 39:453-465. [PMID: 38008886 DOI: 10.1007/s11011-023-01327-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 11/17/2023] [Indexed: 11/28/2023]
Abstract
To date, treatment of Central Nervous System (CNS) pathology has largely focused on neuronal structure and function. Yet, revived attention towards fluid circulation within the CNS has exposed the need to further explore the role of glial cells in maintaining homeostasis within neural networks. In the past decade, discovery of the neural glymphatic network has revolutionized traditional understanding of fluid dynamics within the CNS. Advancements in neuroimaging have revealed alternative pathways of cerebrospinal fluid (CSF) generation and efflux. Here, we discuss emerging perspectives on the role of astrocytes in CSF hydrodynamics, with particular focus on the contribution of aquaporin-4 channels to the glymphatic network. Astrocytic structural features and expression patterns are detailed in relation to their function in maintaining integrity of the Blood Brain Barrier (BBB) as part of the neurovascular unit (NVU). This narrative also highlights the potential role of glial dysfunction in pathogenesis of neurodegenerative disease, hydrocephalus, intracranial hemorrhage, ischemic stroke, and traumatic brain injury. The purpose of this literature summary is to provide an update on the changing landscape of scientific theory surrounding production, flow, and absorption of cerebrospinal fluid. The overarching aim of this narrative review is to advance the conception of basic, translational, and clinical research endeavors investigating glia as therapeutic targets for neurological disease.
Collapse
Affiliation(s)
- Nikita Das
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Ravi Dhamija
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Sumit Sarkar
- Division of Neurotoxicology, HFT-132, National Center for Toxicological Research, U.S. Food & Drug Administration, Jefferson, AR, 72079, USA.
| |
Collapse
|
9
|
Boland R, Kokiko-Cochran ON. Deplete and repeat: microglial CSF1R inhibition and traumatic brain injury. Front Cell Neurosci 2024; 18:1352790. [PMID: 38450286 PMCID: PMC10915023 DOI: 10.3389/fncel.2024.1352790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/25/2024] [Indexed: 03/08/2024] Open
Abstract
Traumatic brain injury (TBI) is a public health burden affecting millions of people. Sustained neuroinflammation after TBI is often associated with poor outcome. As a result, increased attention has been placed on the role of immune cells in post-injury recovery. Microglia are highly dynamic after TBI and play a key role in the post-injury neuroinflammatory response. Therefore, microglia represent a malleable post-injury target that could substantially influence long-term outcome after TBI. This review highlights the cell specific role of microglia in TBI pathophysiology. Microglia have been manipulated via genetic deletion, drug inhibition, and pharmacological depletion in various pre-clinical TBI models. Notably, colony stimulating factor 1 (CSF1) and its receptor (CSF1R) have gained much traction in recent years as a pharmacological target on microglia. CSF1R is a transmembrane tyrosine kinase receptor that is essential for microglia proliferation, differentiation, and survival. Small molecule inhibitors targeting CSF1R result in a swift and effective depletion of microglia in rodents. Moreover, discontinuation of the inhibitors is sufficient for microglia repopulation. Attention is placed on summarizing studies that incorporate CSF1R inhibition of microglia. Indeed, microglia depletion affects multiple aspects of TBI pathophysiology, including neuroinflammation, oxidative stress, and functional recovery with measurable influence on astrocytes, peripheral immune cells, and neurons. Taken together, the data highlight an important role for microglia in sustaining neuroinflammation and increasing risk of oxidative stress, which lends to neuronal damage and behavioral deficits chronically after TBI. Ultimately, the insights gained from CSF1R depletion of microglia are critical for understanding the temporospatial role that microglia develop in mediating TBI pathophysiology and recovery.
Collapse
Affiliation(s)
- Rebecca Boland
- Department of Neuroscience, College of Medicine, Chronic Brain Injury Program, Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| | - Olga N Kokiko-Cochran
- Department of Neuroscience, College of Medicine, Chronic Brain Injury Program, Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
10
|
Duan HZ, Zhou X, Hu Q, Liu ML, Wang SH, Zhang J, Jiang XH, Zhang TX, Yu AY. Mannitol inhibits the proliferation of neural stem cell by a p38 mitogen-activated protein kinase-dependent signaling pathway. Chin J Traumatol 2024; 27:42-52. [PMID: 37953130 PMCID: PMC10859289 DOI: 10.1016/j.cjtee.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/12/2023] [Accepted: 10/20/2023] [Indexed: 11/14/2023] Open
Abstract
PURPOSE Mannitol is one of the first-line drugs for reducing cerebral edema through increasing the extracellular osmotic pressure. However, long-term administration of mannitol in the treatment of cerebral edema triggers damage to neurons and astrocytes. Given that neural stem cell (NSC) is a subpopulation of main regenerative cells in the central nervous system after injury, the effect of mannitol on NSC is still elusive. The present study aims to elucidate the role of mannitol in NSC proliferation. METHODS C57 mice were derived from the animal house of Zunyi Medical University. A total of 15 pregnant mice were employed for the purpose of isolating NSCs in this investigation. Initially, mouse primary NSCs were isolated from the embryonic cortex of mice and subsequently identified through immunofluorescence staining. In order to investigate the impact of mannitol on NSC proliferation, both cell counting kit-8 assays and neurospheres formation assays were conducted. The in vitro effects of mannitol were examined at various doses and time points. In order to elucidate the role of Aquaporin 4 (AQP4) in the suppressive effect of mannitol on NSC proliferation, various assays including reverse transcription polymerase chain reaction, western blotting, and immunocytochemistry were conducted on control and mannitol-treated groups. Additionally, the phosphorylated p38 (p-p38) was examined to explore the potential mechanism underlying the inhibitory effect of mannitol on NSC proliferation. Finally, to further confirm the involvement of the p38 mitogen-activated protein kinase-dependent (MAPK) signaling pathway in the observed inhibition of NSC proliferation by mannitol, SB203580 was employed. All data were analyzed using SPSS 20.0 software (SPSS, Inc., Chicago, IL). The statistical analysis among multiple comparisons was performed using one-way analysis of variance (ANOVA), followed by Turkey's post hoc test in case of the data following a normal distribution using a Shapiro-Wilk normality test. Comparisons between 2 groups were determined using Student's t-test, if the data exhibited a normal distribution using a Shapiro-Wilk normality test. Meanwhile, data were shown as median and interquartile range and analyzed using the Mann-Whitney U test, if the data failed the normality test. A p < 0.05 was considered as significant difference. RESULTS Primary NSC were isolated from the mice, and the characteristics were identified using immunostaining analysis. Thereafter, the results indicated that mannitol held the capability of inhibiting NSC proliferation in a dose-dependent and time-dependent manner using cell counting kit-8, neurospheres formation, and immunostaining of Nestin and Ki67 assays. During the process of mannitol suppressing NSC proliferation, the expression of AQP4 mRNA and protein was downregulated, while the gene expression of p-p38 was elevated by reverse transcription polymerase chain reaction, immunostaining, and western blotting assays. Subsequently, the administration of SB203580, one of the p38 MAPK signaling pathway inhibitors, partially abrogated this inhibitory effect resulting from mannitol, supporting the fact that the p38 MAPK signaling pathway participated in curbing NSC proliferation induced by mannitol. CONCLUSIONS Mannitol inhibits NSC proliferation through downregulating AQP4, while upregulating the expression of p-p38 MAPK.
Collapse
Affiliation(s)
- Hai-Zhen Duan
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou province, China
| | - Xin Zhou
- Dazhou Vocational College of Chinese Medicine, Dazhou, 635000, Sichuan province, China; Dachuan District Traditional Chinese Medicine Hospital, Dazhou, 635000, Sichuan province, China
| | - Quan Hu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou province, China
| | - Meng-Long Liu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou province, China
| | - Shu-Hong Wang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou province, China
| | - Ji Zhang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou province, China
| | - Xu-Heng Jiang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou province, China
| | - Tian-Xi Zhang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou province, China
| | - An-Yong Yu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou province, China.
| |
Collapse
|
11
|
Abou-El-Hassan H, Bernstock JD, Chalif JI, Yahya T, Rezende RM, Weiner HL, Izzy S. Elucidating the neuroimmunology of traumatic brain injury: methodological approaches to unravel intercellular communication and function. Front Cell Neurosci 2023; 17:1322325. [PMID: 38162004 PMCID: PMC10756680 DOI: 10.3389/fncel.2023.1322325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/15/2023] [Indexed: 01/03/2024] Open
Abstract
The neuroimmunology of traumatic brain injury (TBI) has recently gained recognition as a crucial element in the secondary pathophysiological consequences that occur following neurotrauma. Both immune cells residing within the central nervous system (CNS) and those migrating from the periphery play significant roles in the development of secondary brain injury. However, the precise mechanisms governing communication between innate and adaptive immune cells remain incompletely understood, partly due to a limited utilization of relevant experimental models and techniques. Therefore, in this discussion, we outline current methodologies that can aid in the exploration of TBI neuroimmunology, with a particular emphasis on the interactions between resident neuroglial cells and recruited lymphocytes. These techniques encompass adoptive cell transfer, intra-CNS injection(s), selective cellular depletion, genetic manipulation, molecular neuroimaging, as well as in vitro co-culture systems and the utilization of organoid models. By incorporating key elements of both innate and adaptive immunity, these methods facilitate the examination of clinically relevant interactions. In addition to these preclinical approaches, we also detail an emerging avenue of research that seeks to leverage human biofluids. This approach enables the investigation of how resident and infiltrating immune cells modulate neuroglial responses after TBI. Considering the growing significance of neuroinflammation in TBI, the introduction and application of advanced methodologies will be pivotal in advancing translational research in this field.
Collapse
Affiliation(s)
- Hadi Abou-El-Hassan
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Joshua D. Bernstock
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Joshua I. Chalif
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Taha Yahya
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Rafael M. Rezende
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Howard L. Weiner
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Saef Izzy
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
12
|
Li C, Chen S, Siedhoff HR, Grant D, Liu P, Balderrama A, Jackson M, Zuckerman A, Greenlief CM, Kobeissy F, Wang KW, DePalma RG, Cernak I, Cui J, Gu Z. Low-intensity open-field blast exposure effects on neurovascular unit ultrastructure in mice. Acta Neuropathol Commun 2023; 11:144. [PMID: 37674234 PMCID: PMC10481586 DOI: 10.1186/s40478-023-01636-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 08/14/2023] [Indexed: 09/08/2023] Open
Abstract
Mild traumatic brain injury (mTBI) induced by low-intensity blast (LIB) is a serious health problem affecting military service members and veterans. Our previous reports using a single open-field LIB mouse model showed the absence of gross microscopic damage or necrosis in the brain, while transmission electron microscopy (TEM) identified ultrastructural abnormalities of myelin sheaths, mitochondria, and synapses. The neurovascular unit (NVU), an anatomical and functional system with multiple components, is vital for the regulation of cerebral blood flow and cellular interactions. In this study, we delineated ultrastructural abnormalities affecting the NVU in mice with LIB exposure quantitatively and qualitatively. Luminal constrictive irregularities were identified at 7 days post-injury (DPI) followed by dilation at 30 DPI along with degeneration of pericytes. Quantitative proteomic analysis identified significantly altered vasomotor-related proteins at 24 h post-injury. Endothelial cell, basement membrane and astrocyte end-foot swellings, as well as vacuole formations, occurred in LIB-exposed mice, indicating cellular edema. Structural abnormalities of tight junctions and astrocyte end-foot detachment from basement membranes were also noted. These ultrastructural findings demonstrate that LIB induces multiple-component NVU damage. Prevention of NVU damage may aid in identifying therapeutic targets to mitigate the effects of primary brain blast injury.
Collapse
Affiliation(s)
- Chao Li
- Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, One Hospital Drive, Medical Science Building, M741, Columbia, MO, 65212, USA
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510630, China
| | - Shanyan Chen
- Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, One Hospital Drive, Medical Science Building, M741, Columbia, MO, 65212, USA
- Truman VA Hospital Research Service, Columbia, MO, 65201, USA
| | - Heather R Siedhoff
- Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, One Hospital Drive, Medical Science Building, M741, Columbia, MO, 65212, USA
- Truman VA Hospital Research Service, Columbia, MO, 65201, USA
| | - DeAna Grant
- Electron Microscopy Core Facility, University of Missouri, Columbia, MO, 65211, USA
| | - Pei Liu
- Charles W. Gehrke Proteomic Center, University of Missouri, Columbia, MO, 65211, USA
| | - Ashley Balderrama
- Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, One Hospital Drive, Medical Science Building, M741, Columbia, MO, 65212, USA
- Truman VA Hospital Research Service, Columbia, MO, 65201, USA
| | - Marcus Jackson
- Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, One Hospital Drive, Medical Science Building, M741, Columbia, MO, 65212, USA
| | - Amitai Zuckerman
- Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, One Hospital Drive, Medical Science Building, M741, Columbia, MO, 65212, USA
- Truman VA Hospital Research Service, Columbia, MO, 65201, USA
| | - C Michael Greenlief
- Charles W. Gehrke Proteomic Center, University of Missouri, Columbia, MO, 65211, USA
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, 30310-1458, USA
- Atlanta VA Medical and Rehab Center, Decatur, GA, 30033, USA
| | - Kevin W Wang
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, 30310-1458, USA
- Atlanta VA Medical and Rehab Center, Decatur, GA, 30033, USA
| | - Ralph G DePalma
- Office of Research and Development, Department of Veterans Affairs, Washington, DC, 20420, USA
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Ibolja Cernak
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA, 31207, USA
| | - Jiankun Cui
- Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, One Hospital Drive, Medical Science Building, M741, Columbia, MO, 65212, USA
- Truman VA Hospital Research Service, Columbia, MO, 65201, USA
| | - Zezong Gu
- Department of Pathology & Anatomical Sciences, University of Missouri School of Medicine, One Hospital Drive, Medical Science Building, M741, Columbia, MO, 65212, USA.
- Truman VA Hospital Research Service, Columbia, MO, 65201, USA.
| |
Collapse
|
13
|
Wang D, Wang S, Zhu Q, Shen Z, Yang G, Chen Y, Luo C, Du Y, Hu Y, Wang W, Yang J. Prospects for Nerve Regeneration and Gene Therapy in the Treatment of Traumatic Brain Injury. J Mol Neurosci 2023; 73:578-586. [PMID: 37458921 DOI: 10.1007/s12031-023-02144-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/12/2023] [Indexed: 09/24/2023]
Abstract
Traumatic brain injury (TBI) is a prevalent neurological disorder and a leading cause of death and disability worldwide. The high mortality rates result in a tremendous burden on society and families in terms of public health and economic costs. Despite advances in biomedical research, treatment options for TBI still remain limited, and there is no effective therapy to restore the structure and function of the injured brain. Regrettably, due to the excessive heterogeneity of TBI and the lack of objective and reliable efficacy evaluation indicators, no proven therapeutic drugs or drugs with clear benefits on functional outcomes have been successfully developed to date. Therefore, it is urgent to explore new therapeutic approaches to protect or regenerate the injured brain from different perspectives. In this review, we first provide a brief overview of the causes and current status of TBI and then summarize the preclinical and clinical research status of cutting-edge treatment methods, including nerve regeneration therapy and gene therapy, with the aim of providing valuable references for effective therapeutic strategies for TBI.
Collapse
Affiliation(s)
- Daliang Wang
- Department of Critical Care Medicine, The First People Hospital of Jiashan, Jiaxing, 314199, Zhejiang, China
| | - Shengguo Wang
- Department of Critical Care Medicine, The First People Hospital of Jiashan, Jiaxing, 314199, Zhejiang, China
| | - Qunchao Zhu
- Department of Critical Care Medicine, The First People Hospital of Jiashan, Jiaxing, 314199, Zhejiang, China
| | - Zhe Shen
- Department of Critical Care Medicine, The First People Hospital of Jiashan, Jiaxing, 314199, Zhejiang, China
| | - Guohuan Yang
- Department of Critical Care Medicine, The First People Hospital of Jiashan, Jiaxing, 314199, Zhejiang, China
| | - Yanfei Chen
- Department of Critical Care Medicine, The First People Hospital of Jiashan, Jiaxing, 314199, Zhejiang, China
| | - Chen Luo
- Department of Critical Care Medicine, The First People Hospital of Jiashan, Jiaxing, 314199, Zhejiang, China
| | - Yanglin Du
- Department of Critical Care Medicine, The First People Hospital of Jiashan, Jiaxing, 314199, Zhejiang, China
| | - Yelang Hu
- Biological Medicine Research and Development Center, Yangtze Delta of Zhejiang, Hangzhou, 314006, Zhejiang, China
| | - Wenmin Wang
- Biological Medicine Research and Development Center, Yangtze Delta of Zhejiang, Hangzhou, 314006, Zhejiang, China
| | - Jie Yang
- Department of Critical Care Medicine, The First People Hospital of Jiashan, Jiaxing, 314199, Zhejiang, China.
| |
Collapse
|
14
|
Ahluwalia M, Mcmichael H, Kumar M, Espinosa MP, Bosomtwi A, Lu Y, Khodadadi H, Jarrahi A, Khan MB, Hess DC, Rahimi SY, Vender JR, Vale FL, Braun M, Baban B, Dhandapani KM, Vaibhav K. Altered endocannabinoid metabolism compromises the brain-CSF barrier and exacerbates chronic deficits after traumatic brain injury in mice. Exp Neurol 2023; 361:114320. [PMID: 36627040 PMCID: PMC9904276 DOI: 10.1016/j.expneurol.2023.114320] [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: 10/02/2022] [Revised: 12/07/2022] [Accepted: 01/06/2023] [Indexed: 01/09/2023]
Abstract
Endocannabinoids [2-arachidonoylglycerol (2-AG) and N-arachidonoylethanolamine (AEA)], endogenously produced arachidonate-based lipids, are anti-inflammatory physiological ligands for two known cannabinoid receptors, CB1 and CB2, yet the molecular and cellular mechanisms underlying their effects after brain injury are poorly defined. In the present study, we hypothesize that traumatic brain injury (TBI)-induced loss of endocannabinoids exaggerates neurovascular injury, compromises brain-cerebrospinal fluid (CSF) barriers (BCB) and causes behavioral dysfunction. Preliminary analysis in human CSF and plasma indicates changes in endocannabinoid levels. This encouraged us to investigate the levels of endocannabinoid-metabolizing enzymes in a mouse model of controlled cortical impact (CCI). Reductions in endocannabinoid (2-AG and AEA) levels in plasma were supported by higher expression of their respective metabolizing enzymes, monoacylglycerol lipase (MAGL), fatty acid amide hydrolase (FAAH), and cyclooxygenase 2 (Cox-2) in the post-TBI mouse brain. Following increased metabolism of endocannabinoids post-TBI, we observed increased expression of CB2, non-cannabinoid receptor Transient receptor potential vanilloid-1 (TRPV1), aquaporin 4 (AQP4), ionized calcium binding adaptor molecule 1 (IBA1), glial fibrillary acidic protein (GFAP), and acute reduction in cerebral blood flow (CBF). The BCB and pericontusional cortex showed altered endocannabinoid expressions and reduction in ventricular volume. Finally, loss of motor functions and induced anxiety behaviors were observed in these TBI mice. Taken together, our findings suggest endocannabinoids and their metabolizing enzymes play an important role in the brain and BCB integrity and highlight the need for more extensive studies on these mechanisms.
Collapse
Affiliation(s)
- Meenakshi Ahluwalia
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Hannah Mcmichael
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Manish Kumar
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Mario P Espinosa
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Asamoah Bosomtwi
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Yujiao Lu
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Hesam Khodadadi
- Department of Oral Biology and Diagnostic Sciences, Center for Excellence in Research, Scholarship and Innovation, Dental College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Abbas Jarrahi
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Mohammad Badruzzaman Khan
- Department of Neurology, Neuroscience Center of Excellence, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - David C Hess
- Department of Neurology, Neuroscience Center of Excellence, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Scott Y Rahimi
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - John R Vender
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Fernando L Vale
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Molly Braun
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America; Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, United States of America; VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA, United States of America
| | - Babak Baban
- Department of Oral Biology and Diagnostic Sciences, Center for Excellence in Research, Scholarship and Innovation, Dental College of Georgia, Augusta University, Augusta, GA, United States of America; Department of Neurology, Neuroscience Center of Excellence, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Krishnan M Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Kumar Vaibhav
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America; Department of Oral Biology and Diagnostic Sciences, Center for Excellence in Research, Scholarship and Innovation, Dental College of Georgia, Augusta University, Augusta, GA, United States of America.
| |
Collapse
|
15
|
Ibeh S, Bakkar NMZ, Ahmad F, Nwaiwu J, Barsa C, Mekhjian S, Reslan MA, Eid AH, Harati H, Nabha S, Mechref Y, El-Yazbi AF, Kobeissy F. High fat diet exacerbates long-term metabolic, neuropathological, and behavioral derangements in an experimental mouse model of traumatic brain injury. Life Sci 2023; 314:121316. [PMID: 36565814 DOI: 10.1016/j.lfs.2022.121316] [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: 10/20/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
AIMS Traumatic brain injury (TBI) constitutes a serious public health concern. Although TBI targets the brain, it can exert several systemic effects which can worsen the complications observed in TBI subjects. Currently, there is no FDA-approved therapy available for its treatment. Thus, there has been an increasing need to understand other factors that could modulate TBI outcomes. Among the factors involved are diet and lifestyle. High-fat diets (HFD), rich in saturated fat, have been associated with adverse effects on brain health. MAIN METHODS To study this phenomenon, an experimental mouse model of open head injury, induced by the controlled cortical impact was used along with high-fat feeding to evaluate the impact of HFD on brain injury outcomes. Mice were fed HFD for a period of two months where several neurological, behavioral, and molecular outcomes were assessed to investigate the impact on chronic consequences of the injury 30 days post-TBI. KEY FINDINGS Two months of HFD feeding, together with TBI, led to a notable metabolic, neurological, and behavioral impairment. HFD was associated with increased blood glucose and fat-to-lean ratio. Spatial learning and memory, as well as motor coordination, were all significantly impaired. Notably, HFD aggravated neuroinflammation, oxidative stress, and neurodegeneration. Also, cell proliferation post-TBI was repressed by HFD, which was accompanied by an increased lesion volume. SIGNIFICANCE Our research indicated that chronic HFD feeding can worsen functional outcomes, predispose to neurodegeneration, and decrease brain recovery post-TBI. This sheds light on the clinical impact of HFD on TBI pathophysiology and rehabilitation as well.
Collapse
Affiliation(s)
- Stanley Ibeh
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nour-Mounira Z Bakkar
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Fatima Ahmad
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Neuroscience Research Center, Lebanese University, Beirut, Lebanon
| | - Judith Nwaiwu
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Deparment of Chemistry, Texas Tech University, Lubbock, TX, USA
| | - Chloe Barsa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Sarine Mekhjian
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mohammad Amine Reslan
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Hayat Harati
- Neuroscience Research Center, Lebanese University, Beirut, Lebanon
| | - Sanaa Nabha
- Neuroscience Research Center, Lebanese University, Beirut, Lebanon
| | - Yehia Mechref
- Deparment of Chemistry, Texas Tech University, Lubbock, TX, USA
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Deparment of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Faculty of Pharmacy, Alamein International University, Al-Alamein, Egypt.
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Morehouse School of Medicine, Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), 720 Westview Dr. SW, Atlanta, GA 30310, USA.
| |
Collapse
|
16
|
Zheng S, Wang C, Lin L, Mu S, Liu H, Hu X, Chen X, Wang S. TNF-α Impairs Pericyte-Mediated Cerebral Microcirculation via the NF-κB/iNOS Axis after Experimental Traumatic Brain Injury. J Neurotrauma 2023; 40:349-364. [PMID: 35972751 DOI: 10.1089/neu.2022.0016] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Secondary structural and functional abnormalities of the neurovascular unit are important pathological mechanisms following traumatic brain injury (TBI). The neurovascular unit maintains blood-brain barrier and vascular integrity through interactions among glial cells, pericytes and endothelial cells. Trauma-induced neuroinflammation and oxidative stress may act as initiating factors for pathological damage after TBI, which in turn impairs cerebral microcirculatory function. Studies have shown that the tumor necrosis factor α (TNF-α)/nuclear factor-κB (NF-κB) pathway regulates inflammation and oxidative damage, but its role in pericyte-mediated cerebral microcirculation are currently unknown. Herein, we assessed TNF-α/NF-κB signaling and inducible nitric oxide synthase (iNOS), and the effects of the TNF-α inhibitor infliximab after TBI. Whether pericyte damage is dependent on the TNF-α/NF-κB/iNOS axis was also evaluated to explore the mechanisms underlying disturbances in the microcirculation after TBI. Microglia are activated after TBI to promote inflammatory factors and free radical release, and upregulate NF-κB and iNOS expression. After lipopolysaccharide treatment, the activity of TNF-α/NF-κB/iNOS in BV2 cells was also upregulated. Inhibition of TNF-α using infliximab reduced NF-κB phosphorylation and nuclear translocation and downregulated iNOS expression, which attenuated the inflammation and oxidative damage. Meanwhile, inhibition of TNF-α reversed pericyte marker loss, and improved pericyte function and microcirculation perfusion after TBI. In conclusion, our study suggests that microglia released TNF-α after TBI, which promoted neuroinflammation and oxidative stress by activating downstream NF-κB/iNOS signals, and this led to pericyte-mediated disturbance of the cerebral microcirculation.
Collapse
Affiliation(s)
- Shaorui Zheng
- Department of Neurosurgery, Fuzong Clinical Medical College, the Second Affiliated Hospital, Fujian Medical University, Fujian Province, China
- Department of Neurosurgery, Affiliated Hospital of Putian University, Fujian Province, China
| | - Cheng Wang
- Department of Neurosurgery, the First Affiliated Hospital of Wannan Medical College, Anhui Province, China
| | - Long Lin
- Department of Neurosurgery, Fuzong Clinical Medical College, the Second Affiliated Hospital, Fujian Medical University, Fujian Province, China
| | - Shuwen Mu
- Department of Neurosurgery, Fuzong Clinical Medical College, the Second Affiliated Hospital, Fujian Medical University, Fujian Province, China
| | - Haibing Liu
- Department of Neurosurgery, the First Affiliated Hospital of Wannan Medical College, Anhui Province, China
| | - Xiaofang Hu
- Department of Neurosurgery, 900th Hospital of PLA, Fujian Province, China
| | - Xiangrong Chen
- Department of Neurosurgery, the Second Affiliated Hospital, Fujian Medical University, Fujian Province, China
| | - Shousen Wang
- Department of Neurosurgery, 900th Hospital of PLA, Fujian Province, China
| |
Collapse
|
17
|
Kocheril PA, Moore SC, Lenz KD, Mukundan H, Lilley LM. Progress Toward a Multiomic Understanding of Traumatic Brain Injury: A Review. Biomark Insights 2022; 17:11772719221105145. [PMID: 35719705 PMCID: PMC9201320 DOI: 10.1177/11772719221105145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/17/2022] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is not a single disease state but describes an array
of conditions associated with insult or injury to the brain. While some
individuals with TBI recover within a few days or months, others present with
persistent symptoms that can cause disability, neuropsychological trauma, and
even death. Understanding, diagnosing, and treating TBI is extremely complex for
many reasons, including the variable biomechanics of head impact, differences in
severity and location of injury, and individual patient characteristics. Because
of these confounding factors, the development of reliable diagnostics and
targeted treatments for brain injury remains elusive. We argue that the
development of effective diagnostic and therapeutic strategies for TBI requires
a deep understanding of human neurophysiology at the molecular level and that
the framework of multiomics may provide some effective solutions for the
diagnosis and treatment of this challenging condition. To this end, we present
here a comprehensive review of TBI biomarker candidates from across the
multiomic disciplines and compare them with known signatures associated with
other neuropsychological conditions, including Alzheimer’s disease and
Parkinson’s disease. We believe that this integrated view will facilitate a
deeper understanding of the pathophysiology of TBI and its potential links to
other neurological diseases.
Collapse
Affiliation(s)
- Philip A Kocheril
- Physical Chemistry and Applied Spectroscopy Group, Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Shepard C Moore
- Physical Chemistry and Applied Spectroscopy Group, Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Kiersten D Lenz
- Physical Chemistry and Applied Spectroscopy Group, Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Harshini Mukundan
- Physical Chemistry and Applied Spectroscopy Group, Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Laura M Lilley
- Physical Chemistry and Applied Spectroscopy Group, Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| |
Collapse
|
18
|
Xiong A, Li J, Xiong R, Xia Y, Jiang X, Cao F, Lu H, Xu J, Shan F. Inhibition of HIF-1α-AQP4 axis ameliorates brain edema and neurological functional deficits in a rat controlled cortical injury (CCI) model. Sci Rep 2022; 12:2701. [PMID: 35177771 PMCID: PMC8854620 DOI: 10.1038/s41598-022-06773-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 01/24/2022] [Indexed: 02/06/2023] Open
Abstract
Traumatic brain injury (TBI) is an important cause of death in young adults and children. Till now, the treatment of TBI in the short- and long-term complications is still a challenge. Our previous evidence implied aquaporin 4 (AQP4) and hypoxia inducible factor-1α (HIF-1α) might be potential targets for TBI. In this study, we explored the roles of AQP4 and HIF-1α on brain edema formation, neuronal damage and neurological functional deficits after TBI using the controlled cortical injury (CCI) model. The adult male Sprague Dawley rats were randomly divided into sham and TBI group, the latter group was further divided into neutralized-AQP4 antibody group, 2-methoxyestradiol (2-ME2) group, and their corresponding control, IgG and isotonic saline groups, respectively. Brain edema was examined by water content. Hippocampal neuronal injury was assessed by neuron loss and neuronal skeleton related protein expressions. Spatial learning and memory deficits were evaluated by Morris water maze test and memory-related proteins were detected by western blot. Our data showed that increased AQP4 protein level was closely correlated with severity of brain edema after TBI. Compared with that in the control group, both blockage of AQP4 with neutralized-AQP4 antibody and inhibition of HIF-1α with 2-ME2 for one-time treatment within 30-60 min post TBI significantly ameliorated brain edema on the 1st day post-TBI, and markedly alleviated hippocampal neuron loss and spatial learning and memory deficits on the 21st day post-TBI. In summary, our preliminary study revealed the short-term and long-term benefits of targeting HIF-1α-AQP4 axis after TBI, which may provide new clues for the selection of potential therapeutic targets for TBI in clinical practice.
Collapse
Affiliation(s)
- Ao Xiong
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Army Occupational Disease, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Department of Orthopaedics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450042, Henan, China
| | - Junxia Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Department of Traumatic Shock and Blood Transfusion, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Renping Xiong
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Army Occupational Disease, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yiming Xia
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Army Occupational Disease, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xu Jiang
- Department of Orthopaedics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450042, Henan, China
| | - Fuyang Cao
- Department of Orthopaedics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450042, Henan, China
| | - Hong Lu
- Department of Radiology, Chongqing No. 7 Hospital of Chongqing University of Technology, Chongqing, 400054, China
| | - Jianzhong Xu
- Department of Orthopaedics, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450042, Henan, China.
| | - Fabo Shan
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Army Occupational Disease, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
19
|
Szczygielski J, Kopańska M, Wysocka A, Oertel J. Cerebral Microcirculation, Perivascular Unit, and Glymphatic System: Role of Aquaporin-4 as the Gatekeeper for Water Homeostasis. Front Neurol 2021; 12:767470. [PMID: 34966347 PMCID: PMC8710539 DOI: 10.3389/fneur.2021.767470] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/12/2021] [Indexed: 12/13/2022] Open
Abstract
In the past, water homeostasis of the brain was understood as a certain quantitative equilibrium of water content between intravascular, interstitial, and intracellular spaces governed mostly by hydrostatic effects i.e., strictly by physical laws. The recent achievements in molecular bioscience have led to substantial changes in this regard. Some new concepts elaborate the idea that all compartments involved in cerebral fluid homeostasis create a functional continuum with an active and precise regulation of fluid exchange between them rather than only serving as separate fluid receptacles with mere passive diffusion mechanisms, based on hydrostatic pressure. According to these concepts, aquaporin-4 (AQP4) plays the central role in cerebral fluid homeostasis, acting as a water channel protein. The AQP4 not only enables water permeability through the blood-brain barrier but also regulates water exchange between perivascular spaces and the rest of the glymphatic system, described as pan-cerebral fluid pathway interlacing macroscopic cerebrospinal fluid (CSF) spaces with the interstitial fluid of brain tissue. With regards to this, AQP4 makes water shift strongly dependent on active processes including changes in cerebral microcirculation and autoregulation of brain vessels capacity. In this paper, the role of the AQP4 as the gatekeeper, regulating the water exchange between intracellular space, glymphatic system (including the so-called neurovascular units), and intravascular compartment is reviewed. In addition, the new concepts of brain edema as a misbalance in water homeostasis are critically appraised based on the newly described role of AQP4 for fluid permeation. Finally, the relevance of these hypotheses for clinical conditions (including brain trauma and stroke) and for both new and old therapy concepts are analyzed.
Collapse
Affiliation(s)
- Jacek Szczygielski
- Department of Neurosurgery, Institute of Medical Sciences, University of Rzeszów, Rzeszów, Poland.,Department of Neurosurgery, Faculty of Medicine and Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Marta Kopańska
- Department of Pathophysiology, Institute of Medical Sciences, University of Rzeszów, Rzeszów, Poland
| | - Anna Wysocka
- Chair of Internal Medicine and Department of Internal Medicine in Nursing, Faculty of Health Sciences, Medical University of Lublin, Lublin, Poland
| | - Joachim Oertel
- Department of Neurosurgery, Faculty of Medicine and Saarland University Medical Center, Saarland University, Homburg, Germany
| |
Collapse
|
20
|
Trillo-Contreras JL, Toledo-Aral JJ, Villadiego J, Echevarría M. Aquaporin-4 Mediates Permanent Brain Alterations in a Mouse Model of Hypoxia-Aged Hydrocephalus. Int J Mol Sci 2021; 22:ijms22189745. [PMID: 34575909 PMCID: PMC8471142 DOI: 10.3390/ijms22189745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 11/29/2022] Open
Abstract
Aquaporin-4 (AQP4) is the principal water channel in the brain being expressed in astrocytes and ependymal cells. AQP4 plays an important role in cerebrospinal fluid (CSF) homeostasis, and alterations in its expression have been associated with hydrocephalus. AQP4 contributes to the development of hydrocephalus by hypoxia in aged mice, reproducing such principal characteristics of the disease. Here, we explore whether these alterations associated with the hydrocephalic state are permanent or can be reverted by reexposure to normoxia. Alterations such as ventriculomegaly, elevated intracranial pressure, and cognitive deficits were reversed, whereas deficits in CSF outflow and ventricular distensibility were not recovered, remaining impaired even one month after reestablishment of normoxia. Interestingly, in AQP4−/− mice, the impairment in CSF drainage and ventricular distensibility was completely reverted by re-normoxia, indicating that AQP4 has a structural role in the chronification of those alterations. Finally, we show that aged mice subjected to two hypoxic episodes experience permanent ventriculomegaly. These data reveal that repetitive hypoxic events in aged cerebral tissue promote the permanent alterations involved in hydrocephalic pathophysiology, which are dependent on AQP4 expression.
Collapse
Affiliation(s)
- José Luis Trillo-Contreras
- Institute of Biomedicine of Seville-IBiS, University Hospital Virgen del Rocío, CSIC, University of Seville, 41013 Seville, Spain; (J.L.T.-C.); (J.J.T.-A.)
- Department of Medical Physiology and Biophysics, University of Seville, 41009 Seville, Spain
| | - Juan José Toledo-Aral
- Institute of Biomedicine of Seville-IBiS, University Hospital Virgen del Rocío, CSIC, University of Seville, 41013 Seville, Spain; (J.L.T.-C.); (J.J.T.-A.)
- Department of Medical Physiology and Biophysics, University of Seville, 41009 Seville, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
| | - Javier Villadiego
- Institute of Biomedicine of Seville-IBiS, University Hospital Virgen del Rocío, CSIC, University of Seville, 41013 Seville, Spain; (J.L.T.-C.); (J.J.T.-A.)
- Department of Medical Physiology and Biophysics, University of Seville, 41009 Seville, Spain
- Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain
- Correspondence: (J.V.); (M.E.); Tel.: +34-955-920-034 (J.V.); +34-955-920-036 (M.E.)
| | - Miriam Echevarría
- Institute of Biomedicine of Seville-IBiS, University Hospital Virgen del Rocío, CSIC, University of Seville, 41013 Seville, Spain; (J.L.T.-C.); (J.J.T.-A.)
- Department of Medical Physiology and Biophysics, University of Seville, 41009 Seville, Spain
- Correspondence: (J.V.); (M.E.); Tel.: +34-955-920-034 (J.V.); +34-955-920-036 (M.E.)
| |
Collapse
|