1
|
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
|
2
|
Li S, Song H, Sun Y, Sun Y, Zhang H, Gao Z. Inhibition of soluble epoxide hydrolase as a therapeutic approach for blood-brain barrier dysfunction. Biochimie 2024; 223:13-22. [PMID: 38531484 DOI: 10.1016/j.biochi.2024.03.015] [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: 01/12/2024] [Revised: 03/20/2024] [Accepted: 03/23/2024] [Indexed: 03/28/2024]
Abstract
The blood-brain barrier (BBB) is a protective semi-permeable structure that regulates the exchange of biomolecules between the peripheral blood and the central nervous system (CNS). Due to its specialized tight junctions and low vesicle trafficking, the BBB strictly limits the paracellular passage and transcellular transport of molecules to maintain the physiological condition of brain tissues. BBB breakdown is associated with many CNS disorders. Soluble epoxide hydrolase (sEH) is a hydrolase enzyme that converts epoxy-fatty acids (EpFAs) to their corresponding diols and is involved in the onset and progression of multiple diseases. EpFAs play a protective role in the central nervous system via preventing neuroinflammation, making sEH a potential therapeutic target for CNS diseases. Recent studies showed that sEH inhibition prevented BBB impairment caused by stroke, hemorrhage, traumatic brain injury, hyperglycemia and sepsis via regulating the expression of tight junctions. In this review, the protective actions of sEH inhibition on BBB and potential mechanisms are summarized, and some important questions that remain to be resolved are also addressed.
Collapse
Affiliation(s)
- Shuo Li
- Hebei Province Key Laboratory of Molecular Chemistry for Drug, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei, China
| | - Huijia Song
- Hebei Province Key Laboratory of Molecular Chemistry for Drug, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei, China
| | - Yanping Sun
- Hebei Province Key Laboratory of Molecular Chemistry for Drug, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei, China
| | - Yongjun Sun
- Hebei Province Key Laboratory of Molecular Chemistry for Drug, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei, China
| | - Huimin Zhang
- Hebei Province Key Laboratory of Molecular Chemistry for Drug, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei, China
| | - Zibin Gao
- Hebei Province Key Laboratory of Molecular Chemistry for Drug, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei, China.
| |
Collapse
|
3
|
Sriram S, Carstens K, Dewing W, Fiacco TA. Astrocyte regulation of extracellular space parameters across the sleep-wake cycle. Front Cell Neurosci 2024; 18:1401698. [PMID: 38988660 PMCID: PMC11233815 DOI: 10.3389/fncel.2024.1401698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/07/2024] [Indexed: 07/12/2024] Open
Abstract
Multiple subfields of neuroscience research are beginning to incorporate astrocytes into current frameworks of understanding overall brain physiology, neuronal circuitry, and disease etiology that underlie sleep and sleep-related disorders. Astrocytes have emerged as a dynamic regulator of neuronal activity through control of extracellular space (ECS) volume and composition, both of which can vary dramatically during different levels of sleep and arousal. Astrocytes are also an attractive target of sleep research due to their prominent role in the glymphatic system, a method by which toxic metabolites generated during wakefulness are cleared away. In this review we assess the literature surrounding glial influences on fluctuations in ECS volume and composition across the sleep-wake cycle. We also examine mechanisms of astrocyte volume regulation in glymphatic solute clearance and their role in sleep and wake states. Overall, findings highlight the importance of astrocytes in sleep and sleep research.
Collapse
Affiliation(s)
- Sandhya Sriram
- Interdepartmental Graduate Program in Neuroscience, University of California, Riverside, Riverside, CA, United States
- Department of Biochemistry and Molecular Biology, University of California, Riverside, Riverside, CA, United States
| | - Kaira Carstens
- Department of Biochemistry and Molecular Biology, University of California, Riverside, Riverside, CA, United States
| | - Wayne Dewing
- Undergraduate Major in Neuroscience, University of California, Riverside, Riverside, CA, United States
| | - Todd A Fiacco
- Interdepartmental Graduate Program in Neuroscience, University of California, Riverside, Riverside, CA, United States
- Department of Biochemistry and Molecular Biology, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
4
|
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
|
5
|
El Baassiri MG, Raouf Z, Badin S, Escobosa A, Sodhi CP, Nasr IW. Dysregulated brain-gut axis in the setting of traumatic brain injury: review of mechanisms and anti-inflammatory pharmacotherapies. J Neuroinflammation 2024; 21:124. [PMID: 38730498 PMCID: PMC11083845 DOI: 10.1186/s12974-024-03118-3] [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: 02/29/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
Traumatic brain injury (TBI) is a chronic and debilitating disease, associated with a high risk of psychiatric and neurodegenerative diseases. Despite significant advancements in improving outcomes, the lack of effective treatments underscore the urgent need for innovative therapeutic strategies. The brain-gut axis has emerged as a crucial bidirectional pathway connecting the brain and the gastrointestinal (GI) system through an intricate network of neuronal, hormonal, and immunological pathways. Four main pathways are primarily implicated in this crosstalk, including the systemic immune system, autonomic and enteric nervous systems, neuroendocrine system, and microbiome. TBI induces profound changes in the gut, initiating an unrestrained vicious cycle that exacerbates brain injury through the brain-gut axis. Alterations in the gut include mucosal damage associated with the malabsorption of nutrients/electrolytes, disintegration of the intestinal barrier, increased infiltration of systemic immune cells, dysmotility, dysbiosis, enteroendocrine cell (EEC) dysfunction and disruption in the enteric nervous system (ENS) and autonomic nervous system (ANS). Collectively, these changes further contribute to brain neuroinflammation and neurodegeneration via the gut-brain axis. In this review article, we elucidate the roles of various anti-inflammatory pharmacotherapies capable of attenuating the dysregulated inflammatory response along the brain-gut axis in TBI. These agents include hormones such as serotonin, ghrelin, and progesterone, ANS regulators such as beta-blockers, lipid-lowering drugs like statins, and intestinal flora modulators such as probiotics and antibiotics. They attenuate neuroinflammation by targeting distinct inflammatory pathways in both the brain and the gut post-TBI. These therapeutic agents exhibit promising potential in mitigating inflammation along the brain-gut axis and enhancing neurocognitive outcomes for TBI patients.
Collapse
Affiliation(s)
- Mahmoud G El Baassiri
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Zachariah Raouf
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Sarah Badin
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Alejandro Escobosa
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Chhinder P Sodhi
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Isam W Nasr
- Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
6
|
Chen DY, Wu PF, Zhu XY, Zhao WB, Shao SF, Xie JR, Yuan DF, Zhang L, Li K, Wang SN, Zhao H. Risk factors and predictive model of cerebral edema after road traffic accidents-related traumatic brain injury. Chin J Traumatol 2024; 27:153-162. [PMID: 38458896 PMCID: PMC11138350 DOI: 10.1016/j.cjtee.2024.02.001] [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: 08/28/2023] [Revised: 11/06/2023] [Accepted: 01/29/2024] [Indexed: 03/10/2024] Open
Abstract
PURPOSE Cerebral edema (CE) is the main secondary injury following traumatic brain injury (TBI) caused by road traffic accidents (RTAs). It is challenging to be predicted timely. In this study, we aimed to develop a prediction model for CE by identifying its risk factors and comparing the timing of edema occurrence in TBI patients with varying levels of injuries. METHODS This case-control study included 218 patients with TBI caused by RTAs. The cohort was divided into CE and non-CE groups, according to CT results within 7 days. Demographic data, imaging data, and clinical data were collected and analyzed. Quantitative variables that follow normal distribution were presented as mean ± standard deviation, those that do not follow normal distribution were presented as median (Q1, Q3). Categorical variables were expressed as percentages. The Chi-square test and logistic regression analysis were used to identify risk factors for CE. Logistic curve fitting was performed to predict the time to secondary CE in TBI patients with different levels of injuries. The efficacy of the model was evaluated using the receiver operator characteristic curve. RESULTS According to the study, almost half (47.3%) of the patients were found to have CE. The risk factors associated with CE were bilateral frontal lobe contusion, unilateral frontal lobe contusion, cerebral contusion, subarachnoid hemorrhage, and abbreviated injury scale (AIS). The odds ratio values for these factors were 7.27 (95% confidence interval (CI): 2.08 - 25.42, p = 0.002), 2.85 (95% CI: 1.11 - 7.31, p = 0.030), 2.62 (95% CI: 1.12 - 6.13, p = 0.027), 2.44 (95% CI: 1.25 - 4.76, p = 0.009), and 1.5 (95% CI: 1.10 - 2.04, p = 0.009), respectively. We also observed that patients with mild/moderate TBI (AIS ≤ 3) had a 50% probability of developing CE 19.7 h after injury (χ2 = 13.82, adjusted R2 = 0.51), while patients with severe TBI (AIS > 3) developed CE after 12.5 h (χ2 = 18.48, adjusted R2 = 0.54). Finally, we conducted a receiver operator characteristic curve analysis of CE time, which showed an area under the curve of 0.744 and 0.672 for severe and mild/moderate TBI, respectively. CONCLUSION Our study found that the onset of CE in individuals with TBI resulting from RTAs was correlated with the severity of the injury. Specifically, those with more severe injuries experienced an earlier onset of CE. These findings suggest that there is a critical time window for clinical intervention in cases of CE secondary to TBI.
Collapse
Affiliation(s)
- Di-You Chen
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China; Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Peng-Fei Wu
- Chongqing Key Laboratory of Traffic Injury and Vehicle Ergonomics, Chongqing, 400042, China
| | - Xi-Yan Zhu
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Wen-Bing Zhao
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Shi-Feng Shao
- Wound Trauma Medical Center, State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jing-Ru Xie
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Dan-Feng Yuan
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Liang Zhang
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Kui Li
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Shu-Nan Wang
- Department of Radiology, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Hui Zhao
- Institute for Traffic Medicine, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
7
|
Mishra S, Grewal J, Wal P, Bhivshet GU, Tripathi AK, Walia V. Therapeutic potential of vasopressin in the treatment of neurological disorders. Peptides 2024; 174:171166. [PMID: 38309582 DOI: 10.1016/j.peptides.2024.171166] [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: 10/19/2023] [Revised: 01/18/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024]
Abstract
Vasopressin (VP) is a nonapeptide made of nine amino acids synthesized by the hypothalamus and released by the pituitary gland. VP acts as a neurohormone, neuropeptide and neuromodulator and plays an important role in the regulation of water balance, osmolarity, blood pressure, body temperature, stress response, emotional challenges, etc. Traditionally VP is known to regulate the osmolarity and tonicity. VP and its receptors are widely expressed in the various region of the brain including cortex, hippocampus, basal forebrain, amygdala, etc. VP has been shown to modulate the behavior, stress response, circadian rhythm, cerebral blood flow, learning and memory, etc. The potential role of VP in the regulation of these neurological functions have suggested the therapeutic importance of VP and its analogues in the management of neurological disorders. Further, different VP analogues have been developed across the world with different pharmacotherapeutic potential. In the present work authors highlighted the therapeutic potential of VP and its analogues in the treatment and management of various neurological disorders.
Collapse
Affiliation(s)
- Shweta Mishra
- SGT College of Pharmacy, SGT University, Gurugram, India
| | - Jyoti Grewal
- Maharisi Markandeshwar University, Sadopur, India
| | - Pranay Wal
- Pranveer Singh Institute of Pharmacy, Kanpur, India
| | | | | | - Vaibhav Walia
- SGT College of Pharmacy, SGT University, Gurugram, India.
| |
Collapse
|
8
|
Wei Z, Yu H, Zhao H, Wei M, Xing H, Pei J, Yang Y, Ren K. Broadening horizons: ferroptosis as a new target for traumatic brain injury. BURNS & TRAUMA 2024; 12:tkad051. [PMID: 38250705 PMCID: PMC10799763 DOI: 10.1093/burnst/tkad051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/24/2023] [Accepted: 10/15/2023] [Indexed: 01/23/2024]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide, with ~50 million people experiencing TBI each year. Ferroptosis, a form of regulated cell death triggered by iron ion-catalyzed and reactive oxygen species-induced lipid peroxidation, has been identified as a potential contributor to traumatic central nervous system conditions, suggesting its involvement in the pathogenesis of TBI. Alterations in iron metabolism play a crucial role in secondary injury following TBI. This study aimed to explore the role of ferroptosis in TBI, focusing on iron metabolism disorders, lipid metabolism disorders and the regulatory axis of system Xc-/glutathione/glutathione peroxidase 4 in TBI. Additionally, we examined the involvement of ferroptosis in the chronic TBI stage. Based on these findings, we discuss potential therapeutic interventions targeting ferroptosis after TBI. In conclusion, this review provides novel insights into the pathology of TBI and proposes potential therapeutic targets.
Collapse
Affiliation(s)
- Ziqing Wei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Erqi District, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Erqi District, Zhengzhou, China
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, No. 1, Longhu Middle Ring Road, Jinshui District, Zhengzhou, China
| | - Haihan Yu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Erqi District, Zhengzhou, China
| | - Huijuan Zhao
- Henan International Joint Laboratory of Thrombosis and Hemostasis, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, No. 1, Longhu Middle Ring Road, Jinshui District, Luoyang, China
| | - Mingze Wei
- The Second Clinical Medical College, Harbin Medical University, No. 263, Kaiyuan Avenue, Luolong District, Harbin, China
| | - Han Xing
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, No. 246, Xuefu Road, Nangang District, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, No. 1, Jianshe East Road, Erqi District, Zhengzhou 450052, China
| | - Jinyan Pei
- Quality Management Department, Henan No.3 Provincial People’s Hospital, No. 198, Funiu Road, Zhongyuan District, Henan province, Zhengzhou 450052, China
| | - Yang Yang
- Clinical Systems Biology Research Laboratories, Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, No. 198, Funiu Road, Zhongyuan District, Zhengzhou 450052, China
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, No. 246, Xuefu Road, Nangang District, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, No. 1, Jianshe East Road, Erqi District, Zhengzhou 450052, China
| |
Collapse
|
9
|
Jazaeri SZ, Taghizadeh G, Babaei JF, Goudarzi S, Saadatmand P, Joghataei MT, Khanahmadi Z. Aquaporin 4 beyond a water channel; participation in motor, sensory, cognitive and psychological performances, a comprehensive review. Physiol Behav 2023; 271:114353. [PMID: 37714320 DOI: 10.1016/j.physbeh.2023.114353] [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: 06/05/2023] [Revised: 08/15/2023] [Accepted: 09/13/2023] [Indexed: 09/17/2023]
Abstract
Aquaporin 4 (AQP4) is a protein highly expressed in the central nervous system (CNS) and peripheral nervous system (PNS) as well as various other organs, whose different sites of action indicate its importance in various functions. AQP4 has a variety of essential roles beyond water homeostasis. In this article, we have for the first time summarized different roles of AQP4 in motor and sensory functions, besides cognitive and psychological performances, and most importantly, possible physiological mechanisms by which AQP4 can exert its effects. Furthermore, we demonstrated that AQP4 participates in pathology of different neurological disorders, various effects depending on the disease type. Since neurological diseases involve a spectrum of dysfunctions and due to the difficulty of obtaining a treatment that can simultaneously affect these deficits, it is therefore suggested that future studies consider the role of this protein in different functional impairments related to neurological disorders simultaneously or separately by targeting AQP4 expression and/or polarity modulation.
Collapse
Affiliation(s)
- Seyede Zohreh Jazaeri
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ghorban Taghizadeh
- Department of Occupational Therapy, School of Rehabilitation Sciences, Iran University of Medical Sciences, Tehran, Iran.
| | - Javad Fahanik Babaei
- Electrophysiology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Goudarzi
- Experimental Medicine Research Center, Tehran University of medical Sciences, Tehran, Iran
| | - Pegah Saadatmand
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghi Joghataei
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Innovation in Medical Education, Faculty of Medicine, Ottawa University, Ottawa, Canada.
| | - Zohreh Khanahmadi
- Department of Occupational Therapy, School of Rehabilitation Services, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
10
|
Freire MAM, Rocha GS, Bittencourt LO, Falcao D, Lima RR, Cavalcanti JRLP. Cellular and Molecular Pathophysiology of Traumatic Brain Injury: What Have We Learned So Far? BIOLOGY 2023; 12:1139. [PMID: 37627023 PMCID: PMC10452099 DOI: 10.3390/biology12081139] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023]
Abstract
Traumatic brain injury (TBI) is one of the leading causes of long-lasting morbidity and mortality worldwide, being a devastating condition related to the impairment of the nervous system after an external traumatic event resulting in transitory or permanent functional disability, with a significant burden to the healthcare system. Harmful events underlying TBI can be classified into two sequential stages, primary and secondary, which are both associated with breakdown of the tissue homeostasis due to impairment of the blood-brain barrier, osmotic imbalance, inflammatory processes, oxidative stress, excitotoxicity, and apoptotic cell death, ultimately resulting in a loss of tissue functionality. The present study provides an updated review concerning the roles of brain edema, inflammation, excitotoxicity, and oxidative stress on brain changes resulting from a TBI. The proper characterization of the phenomena resulting from TBI can contribute to the improvement of care, rehabilitation and quality of life of the affected people.
Collapse
Affiliation(s)
- Marco Aurelio M. Freire
- Graduate Program in Physiological Sciences, University of the State of Rio Grande do Norte, Mossoró 59607-360, RN, Brazil
| | - Gabriel Sousa Rocha
- Graduate Program in Biochemistry and Molecular Biology, University of the State of Rio Grande do Norte, Mossoró 59607-360, RN, Brazil
| | - Leonardo Oliveira Bittencourt
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-900, PA, Brazil
| | - Daniel Falcao
- VCU Health Systems, Virginia Commonwealth University, 23219 Richmond, VA, USA
| | - Rafael Rodrigues Lima
- Laboratory of Functional and Structural Biology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-900, PA, Brazil
| | - Jose Rodolfo Lopes P. Cavalcanti
- Graduate Program in Physiological Sciences, University of the State of Rio Grande do Norte, Mossoró 59607-360, RN, Brazil
- Graduate Program in Biochemistry and Molecular Biology, University of the State of Rio Grande do Norte, Mossoró 59607-360, RN, Brazil
| |
Collapse
|
11
|
Lillian A, Zuo W, Laham L, Hilfiker S, Ye JH. Pathophysiology and Neuroimmune Interactions Underlying Parkinson's Disease and Traumatic Brain Injury. Int J Mol Sci 2023; 24:7186. [PMID: 37108349 PMCID: PMC10138999 DOI: 10.3390/ijms24087186] [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: 02/07/2023] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder clinically defined by motor instability, bradykinesia, and resting tremors. The clinical symptomatology is seen alongside pathologic changes, most notably the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the accumulation of α-synuclein and neuromelanin aggregates throughout numerous neural circuits. Traumatic brain injury (TBI) has been implicated as a risk factor for developing various neurodegenerative diseases, with the most compelling argument for the development of PD. Dopaminergic abnormalities, the accumulation of α-synuclein, and disruptions in neural homeostatic mechanisms, including but not limited to the release of pro-inflammatory mediators and the production of reactive oxygen species (ROS), are all present following TBI and are closely related to the pathologic changes seen in PD. Neuronal iron accumulation is discernable in degenerative and injured brain states, as is aquaporin-4 (APQ4). APQ4 is an essential mediator of synaptic plasticity in PD and regulates edematous states in the brain after TBI. Whether the cellular and parenchymal changes seen post-TBI directly cause neurodegenerative diseases such as PD is a point of considerable interest and debate; this review explores the vast array of neuroimmunological interactions and subsequent analogous changes that occur in TBI and PD. There is significant interest in exploring the validity of the relationship between TBI and PD, which is a focus of this review.
Collapse
Affiliation(s)
- Alyssa Lillian
- New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 08901, USA
| | - Wanhong Zuo
- New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 08901, USA
| | - Linda Laham
- New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 08901, USA
| | - Sabine Hilfiker
- New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 08901, USA
| | - Jiang-Hong Ye
- Department of Anesthesiology, Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, 185 South Orange Avenue, Newark, NJ 08901, USA
| |
Collapse
|
12
|
Mfsd2a attenuated hypoxic-ischemic brain damage via protection of the blood-brain barrier in mfat-1 transgenic mice. Cell Mol Life Sci 2023; 80:71. [PMID: 36820986 PMCID: PMC9950179 DOI: 10.1007/s00018-023-04716-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/10/2023] [Accepted: 02/01/2023] [Indexed: 02/24/2023]
Abstract
Previous studies have shown that mfat-1 transgenic mice have protective effects against some central nervous system (CNS) disorders, owing to the high docosahexaenoic acid (DHA) content enriched in their brains. However, whether this protective effect is connected to the blood-brain barrier (BBB) remains unclear. This study aims to investigate the mechanisms of the protective effect against hypoxic-ischemic brain damage (HIBD) of mfat-1 transgenic mice. mfat-1 mice not only demonstrated a significant amelioration of neurological dysfunction and neuronal damage but also partly maintained the physiological permeability of the BBB after HIBD. We initially showed this was associated with elevated major facilitator superfamily domain-containing 2a (Mfsd2a) expression on the BBB, resulting from more lysophosphatidylcholine (LPC)-DHA entering the brain. Wild-type (WT) mice showed a similar Mfsd2a expression trend after long-term feeding with an LPC-DHA-rich diet. Knockdown of Mfsd2a by siRNA intra-cerebroventricular (ICV) injection neutralized the protective effect against HIBD-induced BBB disruption in mfat-1 mice, further validating the protective function of Mfsd2a on BBB. HIBD-induced BBB high permeability was attenuated by Mfsd2a, primarily through a transcellular pathway to decrease caveolae-like vesicle-mediated transcytosis. Taken together, these findings not only reveal that mfat-1 transgenic mice have higher expression of Mfsd2a on the BBB, which partly sustains BBB permeability via vesicular transcytosis to alleviate the severity of HIBD, but also suggest that dietary intake of LPC-DHA may upregulate Mfsd2a expression as a novel therapeutic strategy for BBB dysfunction and survival in HIBD patients.
Collapse
|
13
|
Fan Y, Huang H, Shao J, Huang W. MicroRNA-mediated regulation of reactive astrocytes in central nervous system diseases. Front Mol Neurosci 2023; 15:1061343. [PMID: 36710937 PMCID: PMC9877358 DOI: 10.3389/fnmol.2022.1061343] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/22/2022] [Indexed: 01/15/2023] Open
Abstract
Astrocytes (AST) are abundant glial cells in the human brain, accounting for approximately 20-50% percent of mammalian central nervous system (CNS) cells. They display essential functions necessary to sustain the physiological processes of the CNS, including maintaining neuronal structure, forming the blood-brain barrier, coordinating neuronal metabolism, maintaining the extracellular environment, regulating cerebral blood flow, stabilizing intercellular communication, participating in neurotransmitter synthesis, and defending against oxidative stress et al. During the pathological development of brain tumors, stroke, spinal cord injury (SCI), neurodegenerative diseases, and other neurological disorders, astrocytes undergo a series of highly heterogeneous changes, which are called reactive astrocytes, and mediate the corresponding pathophysiological process. However, the pathophysiological mechanisms of reactive astrocytes and their therapeutic relevance remain unclear. The microRNAs (miRNAs) are essential for cell differentiation, proliferation, and survival, which play a crucial role in the pathophysiological development of CNS diseases. In this review, we summarize the regulatory mechanism of miRNAs on reactive astrocytes in CNS diseases, which might provide a theoretical basis for the diagnosis and treatment of CNS diseases.
Collapse
|
14
|
Wei T, Zhou M, Gu L, Yang H, Zhou Y, Li M. A Novel Gating Mechanism of Aquaporin-4 Water Channel Mediated by Blast Shockwaves for Brain Edema. J Phys Chem Lett 2022; 13:2486-2492. [PMID: 35271290 DOI: 10.1021/acs.jpclett.2c00321] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
As the principal water channel in the brain, aquaporin-4 (AQP4) plays a vital role in brain edema, but its role in blast brain edema is unclear. On the basis of molecular simulations, we reveal the atomically detailed picture of AQP4 in response to blast shockwaves. The results show that the shockwave alone closes the AQP4 channel; however, shock-induced bubble collapse opens it. The jet from bubble collapse forcefully increases the distance between helices and the tilt angles of six helices relative to the membrane vertical direction in a very short time. The average channel size increases about 2.6 times, and the water flux rate is nearly 20 times higher than for normal states. It is responsible for abnormal water transport and a potential cause of acute blast brain edema. Additionally, the open AQP4 channel quickly returns to its normal state, which is in turn helpful for edema absorption. Thus, a novel gating mechanism for AQP4 related to the secondary structure change has been provided, which is different from the previous residue-mediated gating mechanism.
Collapse
Affiliation(s)
- Tong Wei
- Institute of Chemical Materials, China Academy of Engineering and Physics, Mianyang 621900, China
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei 230026, China
| | - Mi Zhou
- Institute of Chemical Materials, China Academy of Engineering and Physics, Mianyang 621900, China
| | - Lingzhi Gu
- Institute of Chemical Materials, China Academy of Engineering and Physics, Mianyang 621900, China
| | - Hong Yang
- Institute of Chemical Materials, China Academy of Engineering and Physics, Mianyang 621900, China
| | - Yang Zhou
- Institute of Chemical Materials, China Academy of Engineering and Physics, Mianyang 621900, China
| | - Ming Li
- Institute of Chemical Materials, China Academy of Engineering and Physics, Mianyang 621900, China
| |
Collapse
|
15
|
Wichmann TO, Damkier HH, Pedersen M. A Brief Overview of the Cerebrospinal Fluid System and Its Implications for Brain and Spinal Cord Diseases. Front Hum Neurosci 2022; 15:737217. [PMID: 35126070 PMCID: PMC8813779 DOI: 10.3389/fnhum.2021.737217] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 12/29/2021] [Indexed: 12/04/2022] Open
Abstract
A comprehensive understanding of the cerebrospinal fluid (CSF) system is essential for our understanding of health and disease within the central nervous system (CNS). The system of CSF refers to all components involved in CSF production, movement, and absorption. In recent years, extensive research has resulted in vastly improved understanding of the CSF system in health and disease. Yet, several aspects remain to be fully clarified, notably along the spinal cord as the preponderance of research has focused on the brain. This review briefly summarizes the CSF system and its implications for CNS diseases and highlights the knowledge gaps that require further research.
Collapse
Affiliation(s)
- Thea Overgaard Wichmann
- Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark
- *Correspondence: Thea Overgaard Wichmann
| | | | - Michael Pedersen
- Comparative Medicine Lab, Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| |
Collapse
|
16
|
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
|
17
|
Potential roles of vagus nerve stimulation on traumatic brain injury: Evidence from in vivo and clinical studies. Exp Neurol 2021; 347:113887. [PMID: 34624329 DOI: 10.1016/j.expneurol.2021.113887] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/23/2021] [Accepted: 10/02/2021] [Indexed: 01/08/2023]
Abstract
Traumatic Brain Injury (TBI) is a one of the leading causes of death and disability worldwide. The consequences of TBI can be divided into two stages: 1) the immediate neuronal destruction during the initial trauma, resulting in the primary brain injury and pathophysiologic sequelae, and 2) the secondary brain injury, encompassing mitochondrial dysfunction, inflammation, cellular excitotoxicity, oxidative stress, and cortical edema, resulting in increased intracranial pressure (ICP) with exacerbated brain damage. Although the pathophysiology in TBI has been thoroughly investigated, the effectivity of therapeutic approaches for TBI is still lacking. Vagus nerve stimulation (VNS) has been used for treating medical refractory epilepsy and chronic drug-resistant depression. Several previous studies also demonstrated that VNS has beneficial effects for TBI in animal models and patients. The neuroprotective effects of VNS on TBI are possibly explained through several mechanisms, including a noradrenergic mechanism, anti-inflammatory effects, regulation of neurotransmitters, and attenuation of blood brain barrier breakdown, and brain edema. The aims of this review are to summarize and discuss the current evidence pertinent to the effect of VNS on both primary and secondary brain injury following TBI from both in vivo and clinical studies.
Collapse
|
18
|
Mariajoseph-Antony LF, Kannan A, Panneerselvam A, Loganathan C, Shankar EM, Anbarasu K, Prahalathan C. Role of Aquaporins in Inflammation-a Scientific Curation. Inflammation 2021; 43:1599-1610. [PMID: 32435911 DOI: 10.1007/s10753-020-01247-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Inflammation is a universal response mechanism existing as inter-communicator of biological systems. Uncontrolled or dysregulated inflammation addresses chronic low-grade effects eventually resulting in multimorbidity. Active solute transport across the membrane establishes varying osmotic gradients. Aquaporins (AQPs) are a class of critical ubiquitously expressed transmembrane proteins that aid in fluid and small solute transport via facilitated diffusion over established osmotic gradients. Numerous significant data features the biological functions of AQPs rendering them as an appropriate biomarker of health and diseases. Besides their physiological role in well-balanced inflammatory responses, it is worth noting the dysregulation of AQPs during any undesirable inflammatory event. Most literature to date clearly sets out AQPs as potential drug targets instigating AQP-based therapies. In light of this conception, the current review provides a compendious overview on the propitious and portentous out-turns of AQPs under inflammation.
Collapse
Affiliation(s)
- Lezy Flora Mariajoseph-Antony
- Molecular Endocrinology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, 620024, India
| | - Arun Kannan
- Molecular Endocrinology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, 620024, India
| | - Antojenifer Panneerselvam
- Molecular Endocrinology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, 620024, India
| | - Chithra Loganathan
- Molecular Endocrinology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, 620024, India
| | - Esaki M Shankar
- Department of Life Sciences, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur, 610005, India
| | - Kumarasamy Anbarasu
- Department of Marine Biotechnology, Bharathidasan University, Tiruchirappalli, 620024, India
| | - Chidambaram Prahalathan
- Molecular Endocrinology Laboratory, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, 620024, India.
| |
Collapse
|
19
|
Lu H, Zhan Y, Ai L, Chen H, Chen J. AQP4-siRNA alleviates traumatic brain edema by altering post-traumatic AQP4 polarity reversal in TBI rats. J Clin Neurosci 2020; 81:113-119. [PMID: 33222898 DOI: 10.1016/j.jocn.2020.09.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/13/2020] [Accepted: 09/06/2020] [Indexed: 11/18/2022]
Abstract
The spatial and temporal distribution of aquaporin-4 (AQP4) expression in rat brain following brain trauma and AQP4-siRNA treatment, as well as corresponding pathological changes, were studied to explore the mechanism underlying the effect of AQP4-siRNA treatment on traumatic brain injury (TBI). The rats in the sham operation group had normal structure, with AQP4 located in the perivascular end-foot membranes and astrocytic membranes in a polarized pattern. The accelerated polarity reversal was observed in the TBI group in 1-12 h after TBI. During this period, AQP4 abundance on the astrocytic membrane is gradually increased, while AQP4 abundance on the perivascular end-foot membrane declined rapidly. Twelve hours after TBI, AQP4 expression was depolarized, showing a shift from the perivascular end-foot membrane to the astrocytic membrane. Pathological observation showed that vasogenic edema occurred immediately after TBI, at which time the extracellular space was expanded, leading to severe intracellular edema. AQP4-siRNA reduced the polarity reversal index at the early stage of TBI recovery and reduced edema, demonstrating the potential benefit of reduced AQP4 expression during recovery from TBI.
Collapse
Affiliation(s)
- Hong Lu
- Department of Radiology, Chongqing The Seventh Peoplés Hospital, Chongqing, China
| | - Yuefu Zhan
- Department of Radiology, Hainan Women and Children's Medical Center, No. 15, Long Kun Nan road, Haikou, Hainan 572500, China.
| | - Li Ai
- Department of Radiology, Chongqing The Seventh Peoplés Hospital, Chongqing, China
| | - Haixia Chen
- Department of Radiology, Chongqing The Seventh Peoplés Hospital, Chongqing, China
| | - Jianqiang Chen
- Department of Radiology, Xiangya School of Medicine Affiliated Haikou Hospital, Central South University, Haikou, Hainan 570208, China.
| |
Collapse
|
20
|
Liu ZH, Chen NY, Tu PH, Wu CT, Chiu SC, Huang YC, Lim SN, Yip PK. DHA Attenuates Cerebral Edema Following Traumatic Brain Injury via the Reduction in Blood-Brain Barrier Permeability. Int J Mol Sci 2020; 21:ijms21176291. [PMID: 32878052 PMCID: PMC7503959 DOI: 10.3390/ijms21176291] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 08/27/2020] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) could result in edema and cause an increase in intracranial pressure of the brain resulting in mortality and morbidity. Although there is hyperosmolarity therapy available for this pathophysiological event, it remains controversial. Recently, several groups have shown docosahexaenoic acid (DHA) to improve functional and histological outcomes following brain injury based on reduction of neuroinflammation and apoptosis. However, the effect of DHA on blood-brain barrier (BBB) dysfunction after brain injury has not been fully studied. Here, a controlled cortical impact rat model was used to test the effect of a single dose of DHA administered 30 min post injury. Modified neurological severity score (mNSS) and forelimb asymmetry were used to determine the functional outcomes. Neuroimaging and histology were used to characterize the edema and BBB dysfunction. The study showed that DHA-treated TBI rats had better mNSS and forelimb asymmetry score than vehicle-treated TBI rats. Temporal analysis of edema using MRI revealed a significant reduction in edema level with DHA treatment compared to vehicle in TBI rats. Histological analysis using immunoglobulin G (IgG) extravasation showed that there was less extravasation, which corresponded with a reduction in aquaporin 4 and astrocytic metalloprotease 9 expression, and greater endothelial occludin expression in the peri-contusional site of the TBI rat brain treated with DHA in comparison to vehicle treatment. In conclusion, the study shows that DHA can exert its functional improvement by prevention of the edema formation via prevention of BBB dysfunction after TBI.
Collapse
Affiliation(s)
- Zhuo-Hao Liu
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Chang Gung Medical College and University, Taoyuan County 333, Taiwan; (P.-h.T.); (Y.-C.H.)
- Correspondence: (Z.-H.L.); (P.K.Y.)
| | - Nan-Yu Chen
- Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung Medical College and University, Taoyuan County 333, Taiwan;
| | - Po-hsun Tu
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Chang Gung Medical College and University, Taoyuan County 333, Taiwan; (P.-h.T.); (Y.-C.H.)
| | - Chen-Te Wu
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Chang Gung Medical College and University, Taoyuan County 333, Taiwan;
| | - Shao-Chieh Chiu
- Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital at Linkou, Taoyuan County 333, Taiwan;
| | - Ying-Cheng Huang
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Chang Gung Medical College and University, Taoyuan County 333, Taiwan; (P.-h.T.); (Y.-C.H.)
| | - Siew-Na Lim
- Department of Neurology, Chang Gung Memorial Hospital at Linkou, Chang Gung Medical College and University, Taoyuan County 333, Taiwan;
| | - Ping K. Yip
- Queen Mary University of London, Barts and The London School of Medicine and Dentistry, Blizard Institute, Centre for Neuroscience, Surgery & Trauma, London E1 2AT, UK
- Correspondence: (Z.-H.L.); (P.K.Y.)
| |
Collapse
|
21
|
Wang Y, Wang G, Xu D, Jiang B, Ge M, Wu L, Yang C, Mu N, Wang S, Chang C, Chen T, Feng H, Yao J. Terahertz spectroscopic diagnosis of early blast-induced traumatic brain injury in rats. BIOMEDICAL OPTICS EXPRESS 2020; 11:4085-4098. [PMID: 32923030 PMCID: PMC7449730 DOI: 10.1364/boe.395432] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/10/2020] [Accepted: 06/26/2020] [Indexed: 05/23/2023]
Abstract
The early diagnosis of blast-induced traumatic brain injury (bTBI) is of great clinical significance for prognostication and treatment. Here, we report a new strategy for early bTBI diagnosis through serum and cerebrospinal fluid (CSF) based on terahertz time-domain spectroscopy (THz-TDS). The spectral differences of serum and CSF for different degrees of experimental bTBI in rats have been demonstrated in the early period. In addition, the THz spectra of total protein in the hypothalamus and hippocampus were investigated at different time points after blast exposure, which both showed clear differences with time increasing compared with that in the normal brain. This might help to explain the neurological symptoms caused by bTBI. Moreover, based on the THz absorption spectra of serum and CSF, the principal component analysis and machine learning algorithms were performed to automatically identify the degree of bTBI. The highest diagnostic accuracy was up to 95.5%. It is suggested that this method has potential as an alternative method for high-sensitive, rapid, label-free, economical and early diagnosis of bTBI.
Collapse
Affiliation(s)
- Yuye Wang
- Institute of Laser and Optoelectronics, School of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin 300072, China
- Key Laboratory of Optoelectronics Information Technology (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Guoqiang Wang
- Institute of Laser and Optoelectronics, School of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin 300072, China
- Key Laboratory of Optoelectronics Information Technology (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Degang Xu
- Institute of Laser and Optoelectronics, School of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin 300072, China
- Key Laboratory of Optoelectronics Information Technology (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Bozhou Jiang
- Institute of Laser and Optoelectronics, School of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin 300072, China
- Key Laboratory of Optoelectronics Information Technology (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Meilan Ge
- Institute of Laser and Optoelectronics, School of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin 300072, China
- Key Laboratory of Optoelectronics Information Technology (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Limin Wu
- Institute of Laser and Optoelectronics, School of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin 300072, China
- Key Laboratory of Optoelectronics Information Technology (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Chuanyan Yang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ning Mu
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shi Wang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chao Chang
- Innovation Laboratory of Terahertz Biophysics, National Innovation Institute of Defense Technology, Beijing, 100071, China
| | - Tunan Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hua Feng
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jianquan Yao
- Institute of Laser and Optoelectronics, School of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin 300072, China
- Key Laboratory of Optoelectronics Information Technology (Ministry of Education), Tianjin University, Tianjin 300072, China
| |
Collapse
|
22
|
Nasution RA, Islam AA, Hatta M, Prihantono, Kaelan C, Poniman J, Wangi H. Modification of the Marmarou model in developing countries. Ann Med Surg (Lond) 2020; 57:109-113. [PMID: 32742649 PMCID: PMC7385274 DOI: 10.1016/j.amsu.2020.07.029] [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: 06/22/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 01/14/2023] Open
Abstract
Introduction Head injury is an injury or wound of the brain tissue due to external forces; it can cause a decrease or change in the status of consciousness. Many head injury models have used mice as experimental animals; the Marmarou model is the most famous and the most widely-used diffuse brain injury model. In this study, we slightly modified the Marmarou model. The purpose of this study is to help researchers examining head injuries in mice, especially those in developing countries who have limited facilities and infrastructure. Methods This experimental research uses animals models (Rattus novergicus, strain Sprague Dawley) that fit several criteria, including male, aged 10–12 weeks, and body weight of 200–300 g. This study involves a slight modification on the tube used, with a 20 cm-long weight of 20 g. The blood samples for the following assays of ELISA and brain tissue samples were collected at 24 h and 4, 5, 6, and 7 days post-trauma. Results A significant effect on the brain was seen with the Marmarou model modification, at a mass weight of 20 g and height of 20 cm, with 0.04 J energy produced. Changes were also seen in the histological features of brain tissue and the serum levels of AQP-4, F2 IsoPs, MPO, and VEGF from 24 h until 7 days after trauma. Conclusion This report describes the development of an experimental head injury approach modifying the Marmarou model that is able to produce a diffuse brain injury model in mice. Head injury is an injury of the brain tissue due to external forces. The Marmarou model is the most famous and widely-used diffuse brain injury model in mice. A significant effect on the brain was seen with our modified Marmarou trauma model.
Collapse
Affiliation(s)
- Rizha Anshori Nasution
- Department of Neurosurgery, Pelamonia Hospital, Makassar, Indonesia.,Doctoral Program of Biomedical Sciences, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Andi Assadul Islam
- Department of Neurosurgery, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Mochammad Hatta
- Clinical Microbiologist Program, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Prihantono
- Department of Surgery Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Cahyono Kaelan
- Department of Pathological Anatomy, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Jeni Poniman
- Department of Pathological Anatomy, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Harakati Wangi
- Department of Internal Medicine, Pelamonia Hospital, Makassar, Indonesia
| |
Collapse
|
23
|
Zhou Y, Shao A, Yao Y, Tu S, Deng Y, Zhang J. Dual roles of astrocytes in plasticity and reconstruction after traumatic brain injury. Cell Commun Signal 2020; 18:62. [PMID: 32293472 PMCID: PMC7158016 DOI: 10.1186/s12964-020-00549-2] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/06/2020] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the leading causes of fatality and disability worldwide. Despite its high prevalence, effective treatment strategies for TBI are limited. Traumatic brain injury induces structural and functional alterations of astrocytes, the most abundant cell type in the brain. As a way of coping with the trauma, astrocytes respond in diverse mechanisms that result in reactive astrogliosis. Astrocytes are involved in the physiopathologic mechanisms of TBI in an extensive and sophisticated manner. Notably, astrocytes have dual roles in TBI, and some astrocyte-derived factors have double and opposite properties. Thus, the suppression or promotion of reactive astrogliosis does not have a substantial curative effect. In contrast, selective stimulation of the beneficial astrocyte-derived molecules and simultaneous attenuation of the deleterious factors based on the spatiotemporal-environment can provide a promising astrocyte-targeting therapeutic strategy. In the current review, we describe for the first time the specific dual roles of astrocytes in neuronal plasticity and reconstruction, including neurogenesis, synaptogenesis, angiogenesis, repair of the blood-brain barrier, and glial scar formation after TBI. We have also classified astrocyte-derived factors depending on their neuroprotective and neurotoxic roles to design more appropriate targeted therapies. Video Abstract
Collapse
Affiliation(s)
- Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88, Jiefang Road, Zhejiang, 310009, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Province, Zhejiang, 310009, Hangzhou, China.
| | - Yihan Yao
- Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88, Jiefang Road, Zhejiang, 310009, Hangzhou, China
| | - Sheng Tu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, Hangzhou, China
| | - Yongchuan Deng
- Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88, Jiefang Road, Zhejiang, 310009, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Province, Zhejiang, 310009, Hangzhou, China
| |
Collapse
|
24
|
Zusman BE, Kochanek PM, Jha RM. Cerebral Edema in Traumatic Brain Injury: a Historical Framework for Current Therapy. Curr Treat Options Neurol 2020; 22:9. [PMID: 34177248 PMCID: PMC8223756 DOI: 10.1007/s11940-020-0614-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
PURPOSE OF REVIEW The purposes of this narrative review are to (1) summarize a contemporary view of cerebral edema pathophysiology, (2) present a synopsis of current management strategies in the context of their historical roots (many of which date back multiple centuries), and (3) discuss contributions of key molecular pathways to overlapping edema endophenotypes. This may facilitate identification of important therapeutic targets. RECENT FINDINGS Cerebral edema and resultant intracranial hypertension are major contributors to morbidity and mortality following traumatic brain injury. Although Starling forces are physical drivers of edema based on differences in intravascular vs extracellular hydrostatic and oncotic pressures, the molecular pathophysiology underlying cerebral edema is complex and remains incompletely understood. Current management protocols are guided by intracranial pressure measurements, an imperfect proxy for cerebral edema. These include decompressive craniectomy, external ventricular drainage, hyperosmolar therapy, hypothermia, and sedation. Results of contemporary clinical trials assessing these treatments are summarized, with an emphasis on the gap between intermediate measures of edema and meaningful clinical outcomes. This is followed by a brief statement summarizing the most recent guidelines from the Brain Trauma Foundation (4th edition). While many molecular mechanisms and networks contributing to cerebral edema after TBI are still being elucidated, we highlight some promising molecular mechanism-based targets based on recent research including SUR1-TRPM4, NKCC1, AQP4, and AVP1. SUMMARY This review outlines the origins of our understanding of cerebral edema, chronicles the history behind many current treatment approaches, and discusses promising molecular mechanism-based targeted treatments.
Collapse
Affiliation(s)
- Benjamin E. Zusman
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Institute for Clinical Research Education, University of Pittsburgh, Pittsburgh, PA, USA
- Clinical and Translational Science Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Patrick M. Kochanek
- Clinical and Translational Science Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Children’s Hospital of Pittsburgh, UPMC, Pittsburgh, PA, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
| | - Ruchira M. Jha
- Clinical and Translational Science Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Safar Center for Resuscitation Research, John G. Rangos Research Center, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
25
|
Wang X, Tong J, Han X, Qi X, Zhang J, Wu E, Huang JH. Acute effects of human protein S administration after traumatic brain injury in mice. Neural Regen Res 2020; 15:2073-2081. [PMID: 32394965 PMCID: PMC7716047 DOI: 10.4103/1673-5374.282258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Despite years of effort, no effective acute phase treatment has been discovered for traumatic brain injury. One impediment to successful drug development is entangled secondary injury pathways. Here we show that protein S, a natural multifunctional protein that regulates coagulation, inflammation, and apoptosis, is able to reduce the extent of multiple secondary injuries in traumatic brain injury, and therefore improve prognosis. Mice subjected to controlled cortical impact were treated acutely (10–15 minutes post-injury) with a single dose of either protein S (1 mg/kg) or vehicle phosphate buffered saline via intravenous injection. At 24 hours post-injury, compared to the non-treated group, the protein S treated group showed substantial improvement of edema and fine motor coordination, as well as mitigation of progressive tissue loss. Immunohistochemistry and western blot targeting caspase-3, B-cell lymphoma 2 (Bcl-2) along with terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay revealed that apoptosis was suppressed in treated animals. Immunohistochemistry targeting CD11b showed limited leukocyte infiltration in the protein S-treated group. Moreover, protein S treatment increased the ipsilesional expression of aquaporin-4, which may be the underlying mechanism of its function in reducing edema. These results indicate that immediate intravenous protein S treatment after controlled cortical impact is beneficial to traumatic brain injury prognosis. Animal Use Protocols (AUPs) were approved by the University Committee on Animal Resources (UCAR) of University of Rochester Medical Center (approval No. UCAR-2008-102R) on November 12, 2013.
Collapse
Affiliation(s)
- Xiaowei Wang
- Center for Translational Neuromedicine, University of Rochester, Rochester, NY, USA
| | - Jing Tong
- Department of Neurosurgery, 4th Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Xiaodi Han
- Department of Neurosurgery, Tiantan Hospital, Beijing, China
| | - Xiaoming Qi
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX, USA
| | - Jun Zhang
- Department of Neurosurgery, PLA General Hospital, Beijing, China
| | - Erxi Wu
- Department of Neurosurgery, Baylor Scott & White Health; College of Medicine, Texas A&M Health Science Center, Temple, TX, USA
| | - Jason H Huang
- Department of Neurosurgery, Baylor Scott & White Health; College of Medicine, Texas A&M Health Science Center, Temple, TX, USA
| |
Collapse
|
26
|
Abstract
We explored the dynamic features of brain edema after traumatic brain injury (TBI) using healthy adult male Wistar rats. After inducing moderate brain injuries in the rats, we divided them randomly among seven groups on the basis of the time elapsed between TBI and examination: 1, 6, 12, 24, 48, 72, and 168 h. All rats were scanned using diffusion-weighted imaging (DWI) to observe tissue changes in the contusion penumbra (CP) after TBI. Immunoglobulin G expression was also detected. At 1 h after TBI, there was an annular light-colored region in the CP where the intercellular space was enlarged, suggesting vasogenic edema. At 6 h, the cells expanded, their nuclei shrank, and the cytoplasm was replaced by vacuoles, indicating intracellular edema. Vasogenic edema and intracellular edema increased 12 h after TBI, but decreased 24 h after TBI, with vasogenic edema increasing 48 h after TBI. By 72 h after TBI, intracellular edema dominated until resolution of all edema by 168 h after TBI. DWI indicated that the relative apparent diffusion coefficient increased markedly at 1 h after TBI, but was reduced at 6 and 12 h after TBI. At 48 h, relative apparent diffusion coefficient increased gradually and then declined at 72 h. In rats, TBI-related changes include dynamic variations in intracellular and vasogenic edema severity. Routine MRI and DWI examinations do not distinguish between the center of trauma and CP; however, the apparent diffusion coefficient diagram can portray variations in CP edema type and degree at different time-points following TBI.
Collapse
Affiliation(s)
- Huanhuan Ren
- Department of Radiology, Chongqing Seventh People's Hospital, Chongqing, China
| | | |
Collapse
|
27
|
Dinet V, Petry KG, Badaut J. Brain-Immune Interactions and Neuroinflammation After Traumatic Brain Injury. Front Neurosci 2019; 13:1178. [PMID: 31780883 PMCID: PMC6861304 DOI: 10.3389/fnins.2019.01178] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 10/18/2019] [Indexed: 01/04/2023] Open
Abstract
Traumatic brain injury (TBI) is the principal cause of death and disability in children and young adults. Clinical and preclinical research efforts have been carried out to understand the acute, life-threatening pathophysiological events happening after TBI. In the past few years, however, it was recognized that TBI causes significant morbidity weeks, months, or years after the initial injury, thereby contributing substantially to the overall burden of TBI and the decrease of life expectancy in these patients. Long-lasting sequels of TBI include cognitive decline/dementia, sensory-motor dysfunction, and psychiatric disorders, and most important for patients is the need for socio-economic rehabilitation affecting their quality of life. Cerebrovascular alterations have been described during the first week after TBI for direct consequence development of neuroinflammatory process in relation to brain edema. Within the brain-immune interactions, the complement system, which is a family of blood and cell surface proteins, participates in the pathophysiology process. In fact, the complement system is part of the primary defense and clearance component of innate and adaptive immune response. In this review, the complement activation after TBI will be described in relation to the activation of the microglia and astrocytes as well as the blood-brain barrier dysfunction during the first week after the injury. Considering the neuroinflammatory activity as a causal element of neurological handicaps, some major parallel lines of complement activity in multiple sclerosis and Alzheimer pathologies with regard to cognitive impairment will be discussed for chronic TBI. A better understanding of the role of complement activation could facilitate the development of new therapeutic approaches for TBI.
Collapse
Affiliation(s)
- Virginie Dinet
- INSERM U1029, Angiogenesis and Neuroinflammation Group, University of Bordeaux, Bordeaux, France
| | - Klaus G. Petry
- INSERM U1029, Angiogenesis and Neuroinflammation Group, University of Bordeaux, Bordeaux, France
| | - Jerome Badaut
- CNRS UMR 5287, INCIA, Brain molecular Imaging Team, University of Bordeaux, Bordeaux, France
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| |
Collapse
|
28
|
Fraunberger E, Esser MJ. Neuro-Inflammation in Pediatric Traumatic Brain Injury-from Mechanisms to Inflammatory Networks. Brain Sci 2019; 9:E319. [PMID: 31717597 PMCID: PMC6895990 DOI: 10.3390/brainsci9110319] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 12/12/2022] Open
Abstract
Compared to traumatic brain injury (TBI) in the adult population, pediatric TBI has received less research attention, despite its potential long-term impact on the lives of many children around the world. After numerous clinical trials and preclinical research studies examining various secondary mechanisms of injury, no definitive treatment has been found for pediatric TBIs of any severity. With the advent of high-throughput and high-resolution molecular biology and imaging techniques, inflammation has become an appealing target, due to its mixed effects on outcome, depending on the time point examined. In this review, we outline key mechanisms of inflammation, the contribution and interactions of the peripheral and CNS-based immune cells, and highlight knowledge gaps pertaining to inflammation in pediatric TBI. We also introduce the application of network analysis to leverage growing multivariate and non-linear inflammation data sets with the goal to gain a more comprehensive view of inflammation and develop prognostic and treatment tools in pediatric TBI.
Collapse
Affiliation(s)
- Erik Fraunberger
- Alberta Children’s Hospital Research Institute, Calgary, AB T3B 6A8, Canada;
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Michael J. Esser
- Alberta Children’s Hospital Research Institute, Calgary, AB T3B 6A8, Canada;
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Department of Pediatrics, Cumming School Of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
29
|
Kim H, Kim YT, Song ES, Yoon BC, Choi YH, Kim K, Kim DJ. Changes in the gray and white matter of patients with ischemic-edematous insults after traumatic brain injury. J Neurosurg 2019; 131:1243-1253. [PMID: 30485242 DOI: 10.3171/2018.5.jns172711] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 05/10/2018] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Gray matter (GM) and white matter (WM) are vulnerable to ischemic-edematous insults after traumatic brain injury (TBI). The extent of secondary insult after brain injury is quantifiable using quantitative CT analysis. One conventional quantitative CT measure, the gray-white matter ratio (GWR), and a more recently proposed densitometric analysis are used to assess the extent of these insults. However, the prognostic capacity of the GWR in patients with TBI has not yet been validated. This study aims to test the prognostic value of the GWR and evaluate the alternative parameters derived from the densitometric analysis acquired during the acute phase of TBI. In addition, the prognostic ability of the conventional TBI prognostic models (i.e., IMPACT [International Mission for Prognosis and Analysis of Clinical Trials in TBI] and CRASH [Corticosteroid Randomisation After Significant Head Injury] models) were compared to that of the quantitative CT measures. METHODS Three hundred patients with TBI of varying ages (92 pediatric, 94 adult, and 114 geriatric patients) and admitted between 2008 and 2013 were included in this retrospective cohort study. The normality of the density of the deep GM and whole WM was evaluated as the proportion of CT pixels with Hounsfield unit values of 31-35 for GM and 26-30 for WM on CT images of the entire supratentorial brain. The outcome was evaluated using the Glasgow Outcome Scale (GOS) at discharge (GOS score ≤ 3, n = 100). RESULTS Lower proportions of normal densities in the deep GM and whole WM indicated worse outcomes. The proportion of normal WM exhibited a significant prognostic capacity (area under the curve [AUC] = 0.844). The association between the outcome and the normality of the WM density was significant in adult (AUC = 0.792), pediatric (AUC = 0.814), and geriatric (AUC = 0.885) patients. In pediatric patients, the normality of the overall density and the density of the GM were indicative of the outcome (AUC = 0.751). The average GWR was not associated with the outcome (AUC = 0.511). IMPACT and CRASH models showed adequate and reliable performance in the pediatric and geriatric groups but not in the adult group. The highest overall predictive performance was achieved by the densitometry-augmented IMPACT model (AUC = 0.881). CONCLUSIONS Both deep GM and WM are susceptible to ischemic-edematous insults during the early phase of TBI. The extent of the secondary injury was better evaluated by analyzing the normality of the deep GM and WM rather than by calculating the GWR.
Collapse
Affiliation(s)
- Hakseung Kim
- 1Department of Brain and Cognitive Engineering, Korea University, Seongbuk-gu, Seoul, South Korea
| | - Young-Tak Kim
- 1Department of Brain and Cognitive Engineering, Korea University, Seongbuk-gu, Seoul, South Korea
| | - Eun-Suk Song
- 1Department of Brain and Cognitive Engineering, Korea University, Seongbuk-gu, Seoul, South Korea
| | - Byung C Yoon
- 2Department of Radiology, Stanford University School of Medicine, Stanford, California; and
| | | | - Keewon Kim
- 4Rehabilitation, Seoul National University Hospital, College of Medicine, Jongno-gu, Seoul, South Korea
| | - Dong-Joo Kim
- 1Department of Brain and Cognitive Engineering, Korea University, Seongbuk-gu, Seoul, South Korea
| |
Collapse
|
30
|
Szczygielski J, Hubertus V, Kruchten E, Müller A, Albrecht LF, Mautes AE, Schwerdtfeger K, Oertel J. Brain Edema Formation and Functional Outcome After Surgical Decompression in Murine Closed Head Injury Are Modulated by Acetazolamide Administration. Front Neurol 2019; 10:273. [PMID: 30972006 PMCID: PMC6443632 DOI: 10.3389/fneur.2019.00273] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/01/2019] [Indexed: 11/13/2022] Open
Abstract
Acetazolamide (ACZ), carbonic anhydrase inhibitor, has been successfully applied in several neurosurgical conditions for diagnostic or therapeutic purposes. Furthermore, neuroprotective and anti-edematous properties of ACZ have been postulated. However, its use in traumatic brain injury (TBI) is limited, since ACZ-caused vasodilatation according to the Monro-Kellie doctrine may lead to increased intracranial blood volume / raise of intracranial pressure. We hypothesized that these negative effects of ACZ will be reduced or prevented, if the drug is administered after already performed decompression. To test this hypothesis, we used a mouse model of closed head injury (CHI) and decompressive craniectomy (DC). Mice were assigned into following experimental groups: sham, DC, CHI, CHI+ACZ, CHI+DC, and CHI+DC+ACZ (n = 8 each group). 1d and 3d post injury, the neurological function was assessed according to Neurological Severity Score (NSS) and Beam Balance Score (BBS). At the same time points, brain edema was quantified by MRI investigations. Functional impairment and edema volume were compared between groups and over time. Among the animals without skull decompression, the group additionally treated with acetazolamide demonstrated the most severe functional impairment. This pattern was reversed among the mice with decompressive craniectomy: CHI+DC treated but not CHI+DC+ACZ treated animals showed a significant neurological deficit. Accordingly, radiological assessment revealed most severe edema formation in the CHI+DC group while in CHI+DC+ACZ animals, volume of brain edema did not differ from DC-only animals. In our CHI model, the response to acetazolamide treatment varies between animals with decompressive craniectomy and those without surgical treatment. Opening the cranial vault potentially creates an opportunity for acetazolamide to exert its beneficial effects while vasodilatation-related risks are attenuated. Therefore, we recommend further exploration of this potentially beneficial drug in translational research projects.
Collapse
Affiliation(s)
- Jacek Szczygielski
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany.,Institute of Neuropathology, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany.,Faculty of Medicine, University of Rzeszów, Rzeszów, Poland
| | - Vanessa Hubertus
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany.,Department of Neurosurgery, Charité University Medicine, Berlin, Germany
| | - Eduard Kruchten
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany.,Institute of Interventional and Diagnostic Radiology, Karlsruhe Municipal Hospital, Karlsruhe, Germany
| | - Andreas Müller
- Department of Radiology, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| | - Lisa Franziska Albrecht
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| | - Angelika E Mautes
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| | - Karsten Schwerdtfeger
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| | - Joachim Oertel
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg, Germany
| |
Collapse
|
31
|
Zheng Y, Pan C, Chen M, Pei A, Xie L, Zhu S. miR‑29a ameliorates ischemic injury of astrocytes in vitro by targeting the water channel protein aquaporin 4. Oncol Rep 2019; 41:1707-1717. [PMID: 30628716 PMCID: PMC6365700 DOI: 10.3892/or.2019.6961] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 12/13/2018] [Indexed: 01/22/2023] Open
Abstract
Ischemic stroke is the main cause of brain injury and results in a high rate of morbidity, disability and mortality. In the present study, we aimed to determine whether miR-29a played a protective role in oxygen glucose deprivation (OGD) injury via regulation of the water channel protein aquaporin 4 (AQP4). Real-time PCR and western blotting were used to assess miR-29a levels and AQP4 protein levels, respectively. Apoptosis was detected by flow cytometry, and lactate dehydrogenase (LDH) was determined by enzyme-linked immunosorbent assay (ELISA). Overexpression of miR-29a was significantly downregulated in OGD-induced primary astrocytes, and transfection with a miR-29a mimic decreased LDH release and apoptosis, and improved cell health in OGD-induced astrocytes. AQP4 was the target of miR-29a, which suppressed AQP4 expression, and knockdown of AQP4 mitigated OGD-induced astrocyte injury. Furthermore, miR-29a regulated AQP4 expression in OGD-induced astrocytes. AQP4 exacerbated astrocyte injury following ischemic stroke, and knockdown of AQP4 protected OGD/RX-induced primary cultured astrocytes against injury. The effect of miR-29a inhibitor on primary astrocytes was lost following AQP4 knockdown. These findings indicated that miR-29a prevented astrocyte injury in vitro by inhibiting AQP4. Thus, miR-29a may protect primary cultured astrocytes after OGD-induced injury by targeting AQP4, and may be a potential therapeutic target for ischemic injury of astrocytes.
Collapse
Affiliation(s)
- Yueying Zheng
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Caifei Pan
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Manli Chen
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Aijie Pei
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Liwei Xie
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Shengmei Zhu
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
32
|
Chen LH, Zhang HT, Xu RX, Li WD, Zhao H, Yang Y, Sun K. Interaction of aquaporin 4 and N-methyl-D-aspartate NMDA receptor 1 in traumatic brain injury of rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2018; 21:1148-1154. [PMID: 30483388 PMCID: PMC6251393 DOI: 10.22038/ijbms.2018.29135.7037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Objective(s): methyl-D-aspartate NMDA receptor (NMDAR) and aquaporin 4 (AQP4) are involved in the molecular cascade of edema after traumatic brain injury (TBI) and are potential targets of studies in pharmacology and medicine. However, their association and interactions are still unknown. Materials and Methods: We established a rat TBI model in this study. The cellular distribution patterns of AQP4 after inhibition of NMDAR were determined by Western blotting and immunoreactive staining. Furthermore, the regulation of NMDA receptor 1 by AQP4 was studied by injection of a viral vector targeting AQP4 by RNAi into the rat brain before TBI. Results: The results suggest that AQP4 protein expression increased significantly (P<0.05) after TBI and was down-regulated by the NMDAR inhibitor MK801. This decrease could be partly reversed using the NMDAR agonist NMDA. This indicated that AQP4 mRNA levels and protein expression are regulated by the NMDA signaling pathway. By injection of AQP4 RNAi viral vector into the brain of TBI rat models, we found that the mRNA and protein levels of NMDAR decreased significantly (P<0.05). This suggested that NMDAR is also regulated by AQP4. Conclusion: These data suggested that the inhibition of AQP4 down-regulates NMDAR expression, which might be one of the mechanisms involved in edema after TBI.
Collapse
Affiliation(s)
- Li-Hua Chen
- The Affiliated Bayi Brain Hospital, The PLA Army General Hospital, Beijing 100700, China
| | - Hong-Tian Zhang
- The Affiliated Bayi Brain Hospital, The PLA Army General Hospital, Beijing 100700, China
| | - Ru-Xiang Xu
- The Affiliated Bayi Brain Hospital, The PLA Army General Hospital, Beijing 100700, China
| | - Wen-De Li
- The Affiliated Bayi Brain Hospital, The PLA Army General Hospital, Beijing 100700, China
| | - Hao Zhao
- The Affiliated Bayi Brain Hospital, The PLA Army General Hospital, Beijing 100700, China
| | - Yi Yang
- The Affiliated Bayi Brain Hospital, The PLA Army General Hospital, Beijing 100700, China
| | - Kai Sun
- The Affiliated Bayi Brain Hospital, The PLA Army General Hospital, Beijing 100700, China
| |
Collapse
|
33
|
Jha RM, Kochanek PM. A Precision Medicine Approach to Cerebral Edema and Intracranial Hypertension after Severe Traumatic Brain Injury: Quo Vadis? Curr Neurol Neurosci Rep 2018; 18:105. [PMID: 30406315 PMCID: PMC6589108 DOI: 10.1007/s11910-018-0912-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE OF REVIEW Standard clinical protocols for treating cerebral edema and intracranial hypertension after severe TBI have remained remarkably similar over decades. Cerebral edema and intracranial hypertension are treated interchangeably when in fact intracranial pressure (ICP) is a proxy for cerebral edema but also other processes such as extent of mass lesions, hydrocephalus, or cerebral blood volume. A complex interplay of multiple molecular mechanisms results in cerebral edema after severe TBI, and these are not measured or targeted by current clinically available tools. Addressing these underpinnings may be key to preventing or treating cerebral edema and improving outcome after severe TBI. RECENT FINDINGS This review begins by outlining basic principles underlying the relationship between edema and ICP including the Monro-Kellie doctrine and concepts of intracranial compliance/elastance. There is a subsequent brief discussion of current guidelines for ICP monitoring/management. We then focus most of the review on an evolving precision medicine approach towards cerebral edema and intracranial hypertension after TBI. Personalization of invasive neuromonitoring parameters including ICP waveform analysis, pulse amplitude, pressure reactivity, and longitudinal trajectories are presented. This is followed by a discussion of cerebral edema subtypes (continuum of ionic/cytotoxic/vasogenic edema and progressive secondary hemorrhage). Mechanisms of potential molecular contributors to cerebral edema after TBI are reviewed. For each target, we present findings from preclinical models, and evaluate their clinical utility as biomarkers and therapeutic targets for cerebral edema reduction. This selection represents promising candidates with evidence from different research groups, overlap/inter-relatedness with other pathways, and clinical/translational potential. We outline an evolving precision medicine and translational approach towards cerebral edema and intracranial hypertension after severe TBI.
Collapse
Affiliation(s)
- Ruchira M Jha
- Department of Critical Care Medicine, Room 646A, Scaife Hall, 3550 Terrace Street, Pittsburgh, 15261, PA, USA.
- Safar Center for Resuscitation Research John G. Rangos Research Center, 6th Floor; 4401 Penn Avenue, Pittsburgh, PA, 15224, USA.
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Neurological Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Clinical and Translational Science Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Patrick M Kochanek
- Department of Critical Care Medicine, Room 646A, Scaife Hall, 3550 Terrace Street, Pittsburgh, 15261, PA, USA
- Safar Center for Resuscitation Research John G. Rangos Research Center, 6th Floor; 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
- Clinical and Translational Science Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Anesthesiology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- UPMC Children's Hospital of Pittsburgh John G. Rangos Research Center, 6th Floor 4401 Penn Avenue, Pittsburgh, PA, 15224, USA
| |
Collapse
|
34
|
Szczygielski J, Glameanu C, Müller A, Klotz M, Sippl C, Hubertus V, Schäfer KH, Mautes AE, Schwerdtfeger K, Oertel J. Changes in Posttraumatic Brain Edema in Craniectomy-Selective Brain Hypothermia Model Are Associated With Modulation of Aquaporin-4 Level. Front Neurol 2018; 9:799. [PMID: 30333785 PMCID: PMC6176780 DOI: 10.3389/fneur.2018.00799] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 09/04/2018] [Indexed: 12/19/2022] Open
Abstract
Both hypothermia and decompressive craniectomy have been considered as a treatment for traumatic brain injury. In previous experiments we established a murine model of decompressive craniectomy and we presented attenuated edema formation due to focal brain cooling. Since edema development is regulated via function of water channel proteins, our hypothesis was that the effects of decompressive craniectomy and of hypothermia are associated with a change in aquaporin-4 (AQP4) concentration. Male CD-1 mice were assigned into following groups (n = 5): sham, decompressive craniectomy, trauma, trauma followed by decompressive craniectomy and trauma + decompressive craniectomy followed by focal hypothermia. After 24 h, magnetic resonance imaging with volumetric evaluation of edema and contusion were performed, followed by ELISA analysis of AQP4 concentration in brain homogenates. Additional histopathological analysis of AQP4 immunoreactivity has been performed at more remote time point of 28d. Correlation analysis revealed a relationship between AQP4 level and both volume of edema (r2 = 0.45, p < 0.01, **) and contusion (r2 = 0.41, p < 0.01, **) 24 h after injury. Aggregated analysis of AQP4 level (mean ± SEM) presented increased AQP4 concentration in animals subjected to trauma and decompressive craniectomy (52.1 ± 5.2 pg/mL, p = 0.01; *), but not to trauma, decompressive craniectomy and hypothermia (45.3 ± 3.6 pg/mL, p > 0.05; ns) as compared with animals subjected to decompressive craniectomy only (32.8 ± 2.4 pg/mL). However, semiquantitative histopathological analysis at remote time point revealed no significant difference in AQP4 immunoreactivity across the experimental groups. This suggests that AQP4 is involved in early stages of brain edema formation after surgical decompression. The protective effect of selective brain cooling may be related to change in AQP4 response after decompressive craniectomy. The therapeutic potential of this interaction should be further explored.
Collapse
Affiliation(s)
- Jacek Szczygielski
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany.,Institute of Neuropathology, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany.,Faculty of Medicine, University of Rzeszów, Rzeszów, Poland
| | - Cosmin Glameanu
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Andreas Müller
- Department of Radiology, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Markus Klotz
- Working Group Enteric Nervous System (AGENS), University of Applied Sciences Kaiserslautern, Kaiserslautern, Germany
| | - Christoph Sippl
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Vanessa Hubertus
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany.,Department of Neurosurgery, Charité University Medicine, Berlin, Germany
| | - Karl-Herbert Schäfer
- Working Group Enteric Nervous System (AGENS), University of Applied Sciences Kaiserslautern, Kaiserslautern, Germany
| | - Angelika E Mautes
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Karsten Schwerdtfeger
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Joachim Oertel
- Department of Neurosurgery, Faculty of Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| |
Collapse
|
35
|
Teng Z, Wang A, Wang P, Wang R, Wang W, Han H. The Effect of Aquaporin-4 Knockout on Interstitial Fluid Flow and the Structure of the Extracellular Space in the Deep Brain. Aging Dis 2018; 9:808-816. [PMID: 30271658 PMCID: PMC6147590 DOI: 10.14336/ad.2017.1115] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 11/15/2017] [Indexed: 12/22/2022] Open
Abstract
It has been reported that aquaporin-4 (AQP4) deficiency impairs transportation between the cerebrospinal fluid and interstitial fluid (ISF) as well as the clearance of interstitial solutes in the superficial brain. However, the effect of AQP4 on ISF flow in the deep brain remains unclear. This study compared the brain ISF flow in the caudate nucleus and thalamus of normal rats (NO) and AQP4 knockout rats (KO) using tracer-based magnetic resonance imaging. The rate of brain ISF flow slowed to different degrees in the two regions of KO rats’ brains. Compared with NO rats, the half-life of ISF in the thalamus of KO rats was significantly prolonged, with a corresponding decrease in the clearance coefficient. The tortuosity of the brain extracellular space (ECS) was unchanged in the thalamus of KO rats. In the caudate nucleus of KO rats, the volume fraction of the ECS and the diffusion coefficient were increased, with significantly decreased tortuosity; no significant changes in brain ISF flow were demonstrated. Combined with a change in the expression of glial fibrillary acidic protein and AQP4 in two brain regions, we found that the effect of AQP4 knockout on ISF flow and ECS structure in these two regions differed. This difference may be related to the distribution of astrocytes and the extent of AQP4 decline. This study provides evidence for the involvement of AQP4 in ISF transportation in the deep brain and provides a basis for the establishment of a pharmacokinetic model of the brain’s interstitial pathway.
Collapse
Affiliation(s)
- Ze Teng
- 1Department of Radiology, Peking University Third Hospital, Beijing 100191, China.,2Beijing Key Lab. of Magnetic Resonance Imaging Technology, Beijing 100191, China
| | - Aibo Wang
- 1Department of Radiology, Peking University Third Hospital, Beijing 100191, China.,2Beijing Key Lab. of Magnetic Resonance Imaging Technology, Beijing 100191, China
| | - Peng Wang
- 3Department of Orthopedics, Peking University Third Hospital, Beijing 100191, China
| | - Rui Wang
- 2Beijing Key Lab. of Magnetic Resonance Imaging Technology, Beijing 100191, China
| | - Wei Wang
- 2Beijing Key Lab. of Magnetic Resonance Imaging Technology, Beijing 100191, China
| | - Hongbin Han
- 1Department of Radiology, Peking University Third Hospital, Beijing 100191, China.,2Beijing Key Lab. of Magnetic Resonance Imaging Technology, Beijing 100191, China
| |
Collapse
|
36
|
Trillo-Contreras JL, Ramírez-Lorca R, Hiraldo-González L, Sánchez-Gomar I, Galán-Cobo A, Suárez-Luna N, Sánchez de Rojas-de Pedro E, Toledo-Aral JJ, Villadiego J, Echevarría M. Combined effects of aquaporin-4 and hypoxia produce age-related hydrocephalus. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3515-3526. [PMID: 30293570 DOI: 10.1016/j.bbadis.2018.08.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/01/2018] [Accepted: 08/05/2018] [Indexed: 10/28/2022]
Abstract
Aquaporin-4, present in ependymal cells, in glia limiting and abundantly in pericapillary astrocyte foot processes, and aquaporin-1, expressed in choroid plexus epithelial cells, play an important role in cerebrospinal fluid production and may be involved in the pathophysiology of age-dependent hydrocephalus. The finding that brain aquaporins expression is regulated by low oxygen tension led us to investigate how hypoxia and elevated levels of cerebral aquaporins may result in an increase in cerebrospinal fluid production that could be associated with a hydrocephalic condition. Here we have explored, in young and aged mice exposed to hypoxia, whether aquaporin-4 and aquaporin-1 participate in the development of age-related hydrocephalus. Choroid plexus, striatum, cortex and ependymal tissue were analyzed separately both for mRNA and protein levels of aquaporins. Furthermore, parameters such as total ventricular volume, intraventricular pressure, cerebrospinal fluid outflow rate, ventricular compliance and cognitive function were studied in wild type, aquaporin-1 and aquaporin-4 knock-out animals subjected to hypoxia or normoxia. Our data demonstrate that hypoxia is involved in the development of age-related hydrocephalus by a process that depends on aquaporin-4 channels as a main route for cerebrospinal fluid movement. Significant increases in aquaporin-4 expression that occur over the course of animal aging, together with a reduced cerebrospinal fluid outflow rate and ventricular compliance, contribute to produce more severe hydrocephalus related to hypoxic events in aged mice, with a notable impairment in cognitive function. These results indicate that physiological events and/or pathological conditions presenting with cerebral hypoxia/ischemia contribute to the development of chronic adult hydrocephalus.
Collapse
Affiliation(s)
- José Luis Trillo-Contreras
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital. (HUVR)/Spanish National Research Council (CSIC)/University of Seville, Seville 41013, Spain; Department of Physiology and Biophysics, University of Seville, Seville 41009, Spain
| | - Reposo Ramírez-Lorca
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital. (HUVR)/Spanish National Research Council (CSIC)/University of Seville, Seville 41013, Spain; Department of Physiology and Biophysics, University of Seville, Seville 41009, Spain
| | - Laura Hiraldo-González
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital. (HUVR)/Spanish National Research Council (CSIC)/University of Seville, Seville 41013, Spain; Department of Physiology and Biophysics, University of Seville, Seville 41009, Spain
| | - Ismael Sánchez-Gomar
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital. (HUVR)/Spanish National Research Council (CSIC)/University of Seville, Seville 41013, Spain
| | - Ana Galán-Cobo
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital. (HUVR)/Spanish National Research Council (CSIC)/University of Seville, Seville 41013, Spain
| | - Nela Suárez-Luna
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital. (HUVR)/Spanish National Research Council (CSIC)/University of Seville, Seville 41013, Spain; Department of Physiology and Biophysics, University of Seville, Seville 41009, Spain
| | - Eva Sánchez de Rojas-de Pedro
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital. (HUVR)/Spanish National Research Council (CSIC)/University of Seville, Seville 41013, Spain
| | - Juan José Toledo-Aral
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital. (HUVR)/Spanish National Research Council (CSIC)/University of Seville, Seville 41013, Spain; Department of Physiology and Biophysics, University of Seville, Seville 41009, Spain; Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Spain
| | - Javier Villadiego
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital. (HUVR)/Spanish National Research Council (CSIC)/University of Seville, Seville 41013, Spain; Department of Physiology and Biophysics, University of Seville, Seville 41009, Spain; Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Spain.
| | - Miriam Echevarría
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital. (HUVR)/Spanish National Research Council (CSIC)/University of Seville, Seville 41013, Spain; Department of Physiology and Biophysics, University of Seville, Seville 41009, Spain.
| |
Collapse
|
37
|
Pathophysiology and treatment of cerebral edema in traumatic brain injury. Neuropharmacology 2018; 145:230-246. [PMID: 30086289 DOI: 10.1016/j.neuropharm.2018.08.004] [Citation(s) in RCA: 244] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/24/2018] [Accepted: 08/03/2018] [Indexed: 12/30/2022]
Abstract
Cerebral edema (CE) and resultant intracranial hypertension are associated with unfavorable prognosis in traumatic brain injury (TBI). CE is a leading cause of in-hospital mortality, occurring in >60% of patients with mass lesions, and ∼15% of those with normal initial computed tomography scans. After treatment of mass lesions in severe TBI, an important focus of acute neurocritical care is evaluating and managing the secondary injury process of CE and resultant intracranial hypertension. This review focuses on a contemporary understanding of various pathophysiologic pathways contributing to CE, with a subsequent description of potential targeted therapies. There is a discussion of identified cellular/cytotoxic contributors to CE, as well as mechanisms that influence blood-brain-barrier (BBB) disruption/vasogenic edema, with the caveat that this distinction may be somewhat artificial since molecular processes contributing to these pathways are interrelated. While an exhaustive discussion of all pathways with putative contributions to CE is beyond the scope of this review, the roles of some key contributors are highlighted, and references are provided for further details. Potential future molecular targets for treating CE are presented based on pathophysiologic mechanisms. We thus aim to provide a translational synopsis of present and future strategies targeting CE after TBI in the context of a paradigm shift towards precision medicine. This article is part of the Special Issue entitled "Novel Treatments for Traumatic Brain Injury".
Collapse
|
38
|
Zhao H, Wang Y, Chen L, Shi J, Ma K, Tang L, Xu D, Yao J, Feng H, Chen T. High-sensitivity terahertz imaging of traumatic brain injury in a rat model. JOURNAL OF BIOMEDICAL OPTICS 2018; 23:1-7. [PMID: 29595016 DOI: 10.1117/1.jbo.23.3.036015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 03/09/2018] [Indexed: 05/03/2023]
Abstract
We demonstrated that different degrees of experimental traumatic brain injury (TBI) can be differentiated clearly in fresh slices of rat brain tissues using transmission-type terahertz (THz) imaging system. The high absorption region in THz images corresponded well with the injured area in visible images and magnetic resonance imaging results. The THz image and absorption characteristics of dehydrated paraffin-embedded brain slices and the hematoxylin and eosin (H&E)-stained microscopic images were investigated to account for the intrinsic differences in the THz images for the brain tissues suffered from different degrees of TBI and normal tissue aside from water. The THz absorption coefficients of rat brain tissues showed an increase in the aggravation of brain damage, particularly in the high-frequency range, whereas the cell density decreased as the order of mild, moderate, and severe TBI tissues compared with the normal tissue. Our results indicated that the different degrees of TBI were distinguishable owing to the different water contents and probable hematoma components distribution rather than intrinsic cell intensity. These promising results suggest that THz imaging has great potential as an alternative method for the fast diagnosis of TBI.
Collapse
Affiliation(s)
- Hengli Zhao
- Southwest Hospital, Third Military Medical University (Army Medical University), Department of Neuro, China
| | - Yuye Wang
- Southwest Hospital, Third Military Medical University (Army Medical University), Department of Neuro, China
- Tianjin University, Institute of Laser and Optoelectronics, School of Precision Instrument and Optoe, China
- Tianjin University, Key Laboratory of Optoelectronics Information Technology (Ministry of Education), China
| | - Linyu Chen
- Tianjin University, Institute of Laser and Optoelectronics, School of Precision Instrument and Optoe, China
- Tianjin University, Key Laboratory of Optoelectronics Information Technology (Ministry of Education), China
| | - Jia Shi
- Tianjin University, Institute of Laser and Optoelectronics, School of Precision Instrument and Optoe, China
- Tianjin University, Key Laboratory of Optoelectronics Information Technology (Ministry of Education), China
| | - Kang Ma
- Southwest Hospital, Third Military Medical University (Army Medical University), Department of Neuro, China
| | - Longhuang Tang
- Tianjin University, Institute of Laser and Optoelectronics, School of Precision Instrument and Optoe, China
- Tianjin University, Key Laboratory of Optoelectronics Information Technology (Ministry of Education), China
| | - Degang Xu
- Tianjin University, Institute of Laser and Optoelectronics, School of Precision Instrument and Optoe, China
- Tianjin University, Key Laboratory of Optoelectronics Information Technology (Ministry of Education), China
| | - Jianquan Yao
- Tianjin University, Institute of Laser and Optoelectronics, School of Precision Instrument and Optoe, China
- Tianjin University, Key Laboratory of Optoelectronics Information Technology (Ministry of Education), China
| | - Hua Feng
- Southwest Hospital, Third Military Medical University (Army Medical University), Department of Neuro, China
| | - Tunan Chen
- Southwest Hospital, Third Military Medical University (Army Medical University), Department of Neuro, China
| |
Collapse
|
39
|
Wang C, Xu Y, Huang Y, Huang Y. Effects of erythropoietin and methylprednisolone on AQP4 expression in astrocytes. Mol Med Rep 2017; 16:5924-5930. [PMID: 28849166 PMCID: PMC5865770 DOI: 10.3892/mmr.2017.7330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 05/18/2017] [Indexed: 12/28/2022] Open
Abstract
Methylprednisolone sodium succinate (MPSS) has been suggested as a treatment for spinal cord injury (SCI), but its use has been limited due to its adverse effects. Erythropoietin (EPO) has been suggested as a promising candidate for limiting SCI in mammals. The aim of the present study was to investigate the effects of EPO in combination with MPSS on astrocytes following ischemic injury in vitro. Astrocytes were isolated from the cerebral cortex of postnatal day 3 Sprague-Dawley rats and cultured in vitro. Astrocyte ischemic injury was induced by oxygen and glucose deprivation for 4 h, and reperfusion was simulated by subsequent culture under normoxic conditions. The effects of EPO and MPSS on the expression of aquaporin-4 (AQP4) were investigated. Ischemic astrocytes were treated with EPO (10 U/ml), MPSS (10 µg/ml), or EPO (10 U/ml) in combination with MPSS (10 µg/ml) during reperfusion. The cell viability of astrocytes was assessed using an MTT assay. The mRNA and protein expression levels of AQP4 were determined using reverse transcription-quantitative polymerase chain reaction and western blot analysis, respectively. The role of the protein kinase C (PKC) signaling pathway in the molecular mechanisms underlying the effects of EPO and MPSS was also investigated. The present results demonstrated that following treatment with EPO and MPSS, the mRNA expression levels of AQP4 were upregulated and cell viability was enhanced. EPO and MPSS effectively inhibited the oxygen and glucose deprivation-mediated downregulation of AQP4 following reperfusion. In addition, the combined treatment with EPO and MPSS exhibited higher AQP4 expression levels and cell viability compared with each treatment alone. Finally, the effects of EPO and MPSS on AQP4 expression were partially reversed by pretreatment with the PKC inhibitor Ro 31–8220. The present study indicated that EPO and MPSS had a synergistic effect on AQP4 expression following reperfusion, and suggest that they may be combined in the treatment of SCI.
Collapse
Affiliation(s)
- Changchao Wang
- Department of Orthopedics, The Second Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Youjia Xu
- Department of Orthopedics, The Second Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Yadong Huang
- Jiangsu Food and Pharmaceutical Science College, Huai'an, Jiangsu 223003, P.R. China
| | - Yan Huang
- Jiangsu Food and Pharmaceutical Science College, Huai'an, Jiangsu 223003, P.R. China
| |
Collapse
|
40
|
Zheng Y, Wang L, Chen M, Pei A, Xie L, Zhu S. Upregulation of miR-130b protects against cerebral ischemic injury by targeting water channel protein aquaporin 4 (AQP4). Am J Transl Res 2017; 9:3452-3461. [PMID: 28804561 PMCID: PMC5527259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 05/01/2017] [Indexed: 06/07/2023]
Abstract
Altered microRNA regulation has been implicated in the pathogenesis of various disorders, including cerebral ischemia/reperfusion injury (I/RI). However, the regulatory mechanism of miR-130b in cerebral ischemia injury has not been reported. In this study, we explored the role of miR-130b in cerebral ischemia injury and investigated its potential mechanism. Levels of miR-130b were quantified by real-time PCR, and the protein level of AQP4 was detected by Western blotting. Cell apoptosis was detected by flow cytometry. In vitro, miR-130b levels in astrocytes were found significantly downregulated after OGD. Overexpression of miR-130b by miR-130b mimic decreased LDH release and apoptosis, but promoted cell health of astrocytes with OGD, thus playing a protective role in astrocyte I/RI. The level of miR-130b was also downregulated in ischemic tissues in MCAO model compared with the sham group, and the expression of miR-130b was gradually downregulated over time after reperfusion. AQP4 was upregulated both in two models, and as the reperfusion went on, AQP4 expression gradually upregulated. Our results indicated knockdown of AQP4 could ameliorate astrocyte injury induced by OGD. Finally, we found that miR-130b regulated astrocyte expression of AQP4, and rescue experiments further proved the protective role of miR-130b was mediated by AQP4 downregulation. Our study demonstrated that miR-130b might exert a neuroprotective effect following cerebral I/RI by regulating AQP4 expression at the post-transcriptional level. Therefore, miR-130b may be a potential therapeutic target for stroke treatment.
Collapse
Affiliation(s)
- Yueying Zheng
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang Province, People's Republic of China
| | - Liqing Wang
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang Province, People's Republic of China
| | - Manli Chen
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang Province, People's Republic of China
| | - Aijie Pei
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang Province, People's Republic of China
| | - Liwei Xie
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang Province, People's Republic of China
| | - Shengmei Zhu
- Department of Anesthesiology, The First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang Province, People's Republic of China
| |
Collapse
|
41
|
Hubbard JA, Szu JI, Binder DK. The role of aquaporin-4 in synaptic plasticity, memory and disease. Brain Res Bull 2017; 136:118-129. [PMID: 28274814 DOI: 10.1016/j.brainresbull.2017.02.011] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 02/27/2017] [Accepted: 02/28/2017] [Indexed: 12/25/2022]
Abstract
Since the discovery of aquaporins, it has become clear that the various mammalian aquaporins play critical physiological roles in water and ion balance in multiple tissues. Aquaporin-4 (AQP4), the principal aquaporin expressed in the central nervous system (CNS, brain and spinal cord), has been shown to mediate CNS water homeostasis. In this review, we summarize new and exciting studies indicating that AQP4 also plays critical and unanticipated roles in synaptic plasticity and memory formation. Next, we consider the role of AQP4 in Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), multiple sclerosis (MS), neuromyelitis optica (NMO), epilepsy, traumatic brain injury (TBI), and stroke. Each of these conditions involves changes in AQP4 expression and/or distribution that may be functionally relevant to disease physiology. Insofar as AQP4 is exclusively expressed on astrocytes, these data provide new evidence of "astrocytopathy" in the etiology of diverse neurological diseases.
Collapse
Affiliation(s)
- Jacqueline A Hubbard
- Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, School of Medicine, University of California, Riverside, United States
| | - Jenny I Szu
- Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, School of Medicine, University of California, Riverside, United States
| | - Devin K Binder
- Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, School of Medicine, University of California, Riverside, United States.
| |
Collapse
|
42
|
Down-regulated expression of aquaporin-4 in the cerebellum after status epilepticus. Cogn Neurodyn 2016; 11:183-188. [PMID: 28348649 DOI: 10.1007/s11571-016-9420-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 10/15/2016] [Accepted: 11/09/2016] [Indexed: 10/20/2022] Open
Abstract
Status epilepticus (SE) is a common neurological condition associated with high rates of mortality and permanent brain injury. SE usually leads to neuronal death which may be accompanied by edema, epileptogenesis and learning impairment. Aquaporin-4 (AQP4), is a transmembrane water channel protein in the neuropil of the central nervous system that has an important role in water transport in the brain; AQP4 expression is altered in many pathological conditions such as changes in the blood- brain barrier and/or astrocytic activation, including seizures. AQP4 was shown to be downregulated in the piriform cortex and the hippocampus after SE. Although it is normally expressed at a high level in the cerebellum, little is known about AQP4 levels in the cerebellum following SE. We addressed this in the present study in a mouse model of pilocarpine-induced SE. We found that AQP4 expression was reduced from 3 h to 3 days after SE, with the levels recovering on day 7. Moreover, mice in the acute post-SE stages exhibited impaired motor coordination and learning. These results indicate that cerebellar damage following SE involves changes in AQP4 expression.
Collapse
|