1
|
Hajikarimloo B, Jabbaripour S, Tohidinia AM, Valinejad Qanati A, Fahim F, Javadpour P, Ghasemi R. Insulin potential in preventing brain damage after traumatic brain injury: What we know. J Neuroendocrinol 2025; 37:e13458. [PMID: 39527975 DOI: 10.1111/jne.13458] [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: 11/27/2023] [Revised: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
Traumatic brain injury (TBI) is a major global cause of disability and mortality. TBI results in a spectrum of primary and secondary injuries that impact neural function and overall survival. Insulin, beyond its well-known role in regulating blood glucose levels, plays critical roles in the central nervous system (CNS). These roles include the modulation of synaptic plasticity, neurotransmitter levels, neurogenesis, and neuroprotection. Central insulin resistance, a reduced sensitivity to insulin in the brain, has been observed in TBI patients. This insulin resistance impairs insulin function in the brain and increases the risk of neurodegenerative processes. This review will delve into the central role of insulin resistance in the pathological changes observed after TBI and explore the potential benefits of insulin therapy as a treatment approach for TBI.
Collapse
Affiliation(s)
- Bardia Hajikarimloo
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sama Jabbaripour
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Mohammad Tohidinia
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aysan Valinejad Qanati
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzan Fahim
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pegah Javadpour
- Neuroscience Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rasoul Ghasemi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Neurophysiology Research Center, Institute of Neuroscience and Cognition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Lin K, Hou Y, Li R, Fan F, Hao Y, Wang Y, Huang Y, Li P, Zhu L, Huang X, Zhao YQ. Annexin-A1 tripeptide enhances functional recovery and mitigates brain damage in traumatic brain injury by inhibiting neuroinflammation and preventing ANXA1 nuclear translocation in mice. Metab Brain Dis 2024; 39:1559-1571. [PMID: 39120851 DOI: 10.1007/s11011-024-01404-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
This study explores the role and mechanism of Annexin-A1 Tripeptide (ANXA1sp) in mitigating neuronal damage and promoting functional recovery in a mouse model of traumatic brain injury (TBI). Our goal is to identify ANXA1sp as a potential therapeutic drug candidate for TBI treatment. Adult male C57BL/6J mice were subjected to controlled cortical impact (CCI) to simulate TBI, supplemented by an in vitro model of glutamate-induced TBI in HT22 cells. We assessed neurological deficits using the Modified Neurological Severity Score (mNSS), tested sensorimotor functions with beam balance and rotarod tests, and evaluated cognitive performance via the Morris water maze. Neuronal damage was quantified using Nissl and TUNEL staining, while microglial activation and inflammatory responses were measured through immunostaining, quantitative PCR (qPCR), Western blotting, and ELISA. Additionally, we evaluated cell viability in response to glutamate toxicity using the Cell Counting Kit-8 (CCK-8) assay and lactate dehydrogenase (LDH) release. Intraperitoneal administration of ANXA1sp significantly enhanced neurological outcomes, markedly reducing sensorimotor and cognitive impairments caused by TBI. This treatment resulted in a significant reduction in lesion volume and decreased neuronal cell death in the ipsilateral cortex. Moreover, ANXA1sp effectively diminished microglial activation around the brain lesion and decreased the levels of pro-inflammatory markers such as IL-6, IL-1β, TNF-α, and TGF-β in the cortex, indicating a significant reduction in neuroinflammation post-TBI. ANXA1sp also offered protection against neuronal cell death induced by glutamate toxicity, primarily by inhibiting the nuclear translocation of ANXA1, highlighting its potential as a neuroprotective strategy in TBI management. Administration of ANXA1sp significantly reduced neuroinflammation and neuronal cell death, primarily by blocking the nuclear translocation of ANXA1. This treatment substantially reduced brain damage and improved neurological functional recovery after TBI. Consequently, ANXA1sp stands out as a promising neuroprotective agent for TBI therapy.
Collapse
Affiliation(s)
- Kai Lin
- Department of Clinical Laboratory, Air Force Medical Center, Air Force Medical University, Beijing, 100142, China
| | - Yuejiao Hou
- Department of Cognitive and Stress Medicine, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Ruxin Li
- Department of Cognitive and Stress Medicine, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Fengyan Fan
- Department of Clinical Laboratory, Air Force Medical Center, Air Force Medical University, Beijing, 100142, China
| | - Yinan Hao
- Department of Cognitive and Stress Medicine, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Yuan Wang
- Department of Clinical Laboratory, Air Force Medical Center, Air Force Medical University, Beijing, 100142, China
| | - Yue Huang
- Department of Clinical Laboratory, Air Force Medical Center, Air Force Medical University, Beijing, 100142, China
| | - Peng Li
- Department of Clinical Laboratory, Air Force Medical Center, Air Force Medical University, Beijing, 100142, China
| | - Lingling Zhu
- Department of Cognitive and Stress Medicine, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Xin Huang
- Department of Cognitive and Stress Medicine, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China.
| | - Yong-Qi Zhao
- Department of Cognitive and Stress Medicine, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China.
| |
Collapse
|
3
|
Xiao Y, Zhang Y, Yuan W, Wang C, Ge Y, Huang T, Gao J. Piezo2 Contributes to Traumatic Brain Injury by Activating the RhoA/ROCK1 Pathways. Mol Neurobiol 2024; 61:7419-7430. [PMID: 38388773 PMCID: PMC11415480 DOI: 10.1007/s12035-024-04058-y] [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: 11/09/2023] [Accepted: 02/16/2024] [Indexed: 02/24/2024]
Abstract
Traumatic brain injury (TBI) can lead to short-term and long-term physical and cognitive impairments, which have significant impacts on patients, families, and society. Currently, treatment outcomes for this disease are often unsatisfactory, due at least in part to the fact that the molecular mechanisms underlying the development of TBI are largely unknown. Here, we observed significant upregulation of Piezo2, a key mechanosensitive ion channel protein, in the injured brain tissue of a mouse model of TBI induced by controlled cortical impact. Pharmacological inhibition and genetic knockdown of Piezo2 after TBI attenuated neuronal death, brain edema, brain tissue necrosis, and deficits in neural function and cognitive function. Mechanistically, the increase in Piezo2 expression contributed to TBI-induced neuronal death and subsequent production of TNF-α and IL-1β, likely through activation of the RhoA/ROCK1 pathways in the central nervous system. Our findings suggest that Piezo2 is a key player in and a potential therapeutic target for TBI.
Collapse
Affiliation(s)
- Yinggang Xiao
- Department of Anesthesiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Yangzhou Key Laboratory of Anaesthesiology, Yangzhou, Jiangsu, China
| | - Yang Zhang
- Department of Anesthesiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Yangzhou Key Laboratory of Anaesthesiology, Yangzhou, Jiangsu, China
| | - Wenjuan Yuan
- Department of Anesthesiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Yangzhou Key Laboratory of Anaesthesiology, Yangzhou, Jiangsu, China
| | - Cunjin Wang
- Department of Anesthesiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Yangzhou Key Laboratory of Anaesthesiology, Yangzhou, Jiangsu, China
| | - Yali Ge
- Department of Anesthesiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Yangzhou Key Laboratory of Anaesthesiology, Yangzhou, Jiangsu, China
| | - Tianfeng Huang
- Department of Anesthesiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China.
- Yangzhou Key Laboratory of Anaesthesiology, Yangzhou, Jiangsu, China.
| | - Ju Gao
- Department of Anesthesiology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University/Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, China.
- Yangzhou Key Laboratory of Anaesthesiology, Yangzhou, Jiangsu, China.
| |
Collapse
|
4
|
Wu N, Li W, Chen Q, Chen M, Chen S, Cheng C, Xie Y. Research Advances in Neuroblast Migration in Traumatic Brain Injury. Mol Neurobiol 2024; 61:1-13. [PMID: 38507029 DOI: 10.1007/s12035-024-04117-4] [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: 09/10/2023] [Accepted: 02/17/2024] [Indexed: 03/22/2024]
Abstract
Neuroblasts were first derived from the adult mammalian brains in the 1990s by Reynolds et al. Since then, persistent neurogenesis in the subgranular zone (SGZ) of the hippocampus and subventricular zone (SVZ) has gradually been recognized. To date, reviews on neuroblast migration have largely investigated glial cells and molecular signaling mechanisms, while the relationship between vasculature and cell migration remains a mystery. Thus, this paper underlines the partial biological features of neuroblast migration and unravels the significance and mechanisms of the vasculature in the process to further clarify theoretically the neural repair mechanism after brain injury. Neuroblast migration presents three modes according to the characteristics of cells that act as scaffolds during the migration process: gliophilic migration, neurophilic migration, and vasophilic migration. Many signaling molecules, including brain-derived neurotrophic factor (BDNF), stromal cell-derived factor 1 (SDF-1), vascular endothelial growth factor (VEGF), and angiopoietin-1 (Ang-1), affect vasophilic migration, synergistically regulating the migration of neuroblasts to target areas along blood vessels. However, the precise role of blood vessels in the migration of neuroblasts needs to be further explored. The in-depth study of neuroblast migration will most probably provide theoretical basis and breakthrough for the clinical treatment of brain injury diseases.
Collapse
Affiliation(s)
- Na Wu
- Department of Pediatric Surgery, Chongqing University Three Gorges Hospital, Wanzhou District, No. 165 Xincheng Road, Wanzhou District, Chongqing, 404100, China
| | - Wenlang Li
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, China
| | - Qiang Chen
- Department of Pediatric Surgery, Chongqing University Three Gorges Hospital, Wanzhou District, No. 165 Xincheng Road, Wanzhou District, Chongqing, 404100, China
| | - Meng Chen
- Department of Pediatric Surgery, Chongqing University Three Gorges Hospital, Wanzhou District, No. 165 Xincheng Road, Wanzhou District, Chongqing, 404100, China
| | - Siyuan Chen
- Department of Pediatric Surgery, Chongqing University Three Gorges Hospital, Wanzhou District, No. 165 Xincheng Road, Wanzhou District, Chongqing, 404100, China
| | - Chongjie Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, China
| | - Yimin Xie
- Department of Pediatric Surgery, Chongqing University Three Gorges Hospital, Wanzhou District, No. 165 Xincheng Road, Wanzhou District, Chongqing, 404100, China.
| |
Collapse
|
5
|
Guo R, Yang Q, Zhou X, Li S, Liu Y. Characteristic of clinical trials related to traumatic brain injury registered on ClinicalTrials.gov over the past two decades (2004-2023). Front Med (Lausanne) 2024; 11:1435762. [PMID: 39351009 PMCID: PMC11439763 DOI: 10.3389/fmed.2024.1435762] [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: 05/22/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
Objective The aim of this report is to provide a comprehensive overview of clinical trials and protocols related to traumatic brain injury over the past two decades. Methods We collected information on clinical trials related to traumatic brain injury (TBI) from the ClinicalTrials.gov database, identified key categorical variables, and assessed their characteristics. Results A total of 367 TBI-related trials were identified for analysis. All identified trials were interventional clinical trials. Most trials were small-scale, with 75.2% enrolling 1-100 participants, and only about 20% were funded by industry or the National Institutes of Health (NIH). In most trials, participants were gender-neutral (96.5%), and the primary age group was adults and older adults (56.9%). Of all identified TBI trials, 78.2% were randomized, and 69.4% were blinded. Additionally, the primary purpose of 297 trials (80.9%) was treatment, with drug therapy as the most common intervention. A total of 153 trials (41.7%) were completed; however, only 58 trials submitted results to the registry. Furthermore, 81 trials (22.1%) were discontinued early, primarily due to recruitment problems. Clinical trials started between 2004 and 2013 reported a higher proportion of results compared with those started between 2014 and 2023 (35.1% vs. 11.1%, p < 0.001). In addition, between 2014 and 2023, there was an increase in trials for diagnostic purposes (2.4% vs. 6.5%, p < 0.001). Conclusion Based on the data collected from the ClinicalTrials.gov, our study reveals that most clinical trials related to TBI focus on drug-related treatments, underreporting remains a significant concern, and greater emphasis should be placed on improving the publication and dissemination of clinical trial results.
Collapse
Affiliation(s)
- Ruili Guo
- The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Qingya Yang
- The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Xuan Zhou
- The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Shining Li
- The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yao Liu
- The First Affiliated Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
6
|
Zhang Y, Zheng Z, Sun J, Xu S, Wei Y, Ding X, Ding G. The application of mesenchymal stem cells in the treatment of traumatic brain injury: Mechanisms, results, and problems. Histol Histopathol 2024; 39:1109-1131. [PMID: 38353136 DOI: 10.14670/hh-18-716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells that can be derived from a wide variety of human tissues and organs. They can differentiate into a variety of cell types, including osteoblasts, adipocytes, and chondrocytes, and thus show great potential in regenerative medicine. Traumatic brain injury (TBI) is an organic injury to brain tissue with a high rate of disability and death caused by an external impact or concussive force acting directly or indirectly on the head. The current treatment of TBI mainly includes symptomatic, pharmacological, and rehabilitation treatment. Although some efficacy has been achieved, the definitive recovery effect on neural tissue is still limited. Recent studies have shown that MSC therapies are more effective than traditional treatment strategies due to their strong multi-directional differentiation potential, self-renewal capacity, and low immunogenicity and homing properties, thus MSCs are considered to play an important role and are an ideal cell for the treatment of injurious diseases, including TBI. In this paper, we systematically reviewed the role and mechanisms of MSCs and MSC-derived exosomes in the treatment of TBI, thereby providing new insights into the clinical applications of MSCs and MSC-derived exosomes in the treatment of central nervous system disorders.
Collapse
Affiliation(s)
- Ying Zhang
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Zejun Zheng
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Jinmeng Sun
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Shuangshuang Xu
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Yanan Wei
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Xiaoling Ding
- Clinical Competency Training Center, Shandong Second Medical University, Weifang, Shandong Province, China.
| | - Gang Ding
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong Province, China.
| |
Collapse
|
7
|
Ling Y, Ramalingam M, Lv X, Niu D, Zeng Y, Qiu Y, Si Y, Guo T, Ni Y, Zhang J, Wang Z, Kim HW, Hu J. Human neural stem cell secretome relieves endoplasmic reticulum stress-induced apoptosis and improves neuronal functions after traumatic brain injury in a rat model. J Mol Histol 2024; 55:329-348. [PMID: 38609527 DOI: 10.1007/s10735-024-10192-7] [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: 12/15/2023] [Accepted: 04/01/2024] [Indexed: 04/14/2024]
Abstract
Neural stem cell secretome (NSC-S) plays an important role in neuroprotection and recovery. Studies have shown that endoplasmic reticulum stress (ER stress) is involved in the progression of traumatic brain injury (TBI) and is a crucial cause of secondary damage and neuronal death after brain injury. Whether NSC-S is engaged in ER stress and ER stress-mediated neuronal apoptosis post-TBI has not been investigated. In the study, the Feeney SD male rat model was established. The results showed that NSC-S treatment significantly improved the behavior of rats with TBI. In addition, NSC-S relieved ER stress in TBI rats and was observed by transmission electron microscopy and western blot. The specific mechanism was further elucidated that restoration was achieved by alleviating the PERK-eIF2α pathway and thus protecting neurons from apoptosis. Notably, the discovery of calumenin (CALU) in NSC-S by liquid chromatography-tandem mass spectrometry (LC-MS/MS/MS) may be related to the protective effect of NSC-S on ER stress in neurons. Also, the mechanism by which it functions may be related to ubiquitination. In summary, NSC-S improved prognosis and ER stress in TBI rats and might be a promising treatment for relieving TBI.
Collapse
Affiliation(s)
- Yating Ling
- Institute of Cerebrovascular Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
- Department of Laboratory Medicine, Nanjing Red Cross Blood Center, Nanjing, 210003, Jiangsu, China
| | - Murugan Ramalingam
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea
- School of Basic Medical Sciences, Chengdu University, Chengdu, 610106, China
| | - Xiaorui Lv
- Institute of Cerebrovascular Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Dongdong Niu
- Institute of Cerebrovascular Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Yu Zeng
- Institute of Cerebrovascular Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Yun Qiu
- Institute of Cerebrovascular Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Yu Si
- Institute of Cerebrovascular Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Tao Guo
- Institute of Cerebrovascular Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Yinying Ni
- Institute of Cerebrovascular Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Jingwen Zhang
- Institute of Cerebrovascular Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Ziyu Wang
- Health Clinical Laboratories, Health BioMed Co., Ltd. Ningbo, Zhejiang, 315042, China
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea
- School of Basic Medical Sciences, Chengdu University, Chengdu, 610106, China
| | - Jiabo Hu
- Institute of Cerebrovascular Disease, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
8
|
Maity S, Huang Y, Kilgore MD, Thurmon AN, Vaasjo LO, Galazo MJ, Xu X, Cao J, Wang X, Ning B, Liu N, Fan J. Mapping dynamic molecular changes in hippocampal subregions after traumatic brain injury through spatial proteomics. Clin Proteomics 2024; 21:32. [PMID: 38735925 PMCID: PMC11089002 DOI: 10.1186/s12014-024-09485-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/24/2024] [Indexed: 05/14/2024] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) often results in diverse molecular responses, challenging traditional proteomic studies that measure average changes at tissue levels and fail to capture the complexity and heterogeneity of the affected tissues. Spatial proteomics offers a solution by providing insights into sub-region-specific alterations within tissues. This study focuses on the hippocampal sub-regions, analyzing proteomic expression profiles in mice at the acute (1 day) and subacute (7 days) phases of post-TBI to understand subregion-specific vulnerabilities and long-term consequences. METHODS Three mice brains were collected from each group, including Sham, 1-day post-TBI and 7-day post-TBI. Hippocampal subregions were extracted using Laser Microdissection (LMD) and subsequently analyzed by label-free quantitative proteomics. RESULTS The spatial analysis reveals region-specific protein abundance changes, highlighting the elevation of FN1, LGALS3BP, HP, and MUG-1 in the stratum moleculare (SM), suggesting potential immune cell enrichment post-TBI. Notably, established markers of chronic traumatic encephalopathy, IGHM and B2M, exhibit specific upregulation in the dentate gyrus bottom (DG2) independent of direct mechanical injury. Metabolic pathway analysis identifies disturbances in glucose and lipid metabolism, coupled with activated cholesterol synthesis pathways enriched in SM at 7-Day post-TBI and subsequently in deeper DG1 and DG2 suggesting a role in neurogenesis and the onset of recovery. Coordinated activation of neuroglia and microtubule dynamics in DG2 suggest recovery mechanisms in less affected regions. Cluster analysis revealed spatial variations post-TBI, indicative of dysregulated neuronal plasticity and neurogenesis and further predisposition to neurological disorders. TBI-induced protein upregulation (MUG-1, PZP, GFAP, TJP, STAT-1, and CD44) across hippocampal sub-regions indicates shared molecular responses and links to neurological disorders. Spatial variations were demonstrated by proteins dysregulated in both or either of the time-points exclusively in each subregion (ELAVL2, CLIC1 in PL, CD44 and MUG-1 in SM, and SHOC2, LGALS3 in DG). CONCLUSIONS Utilizing advanced spatial proteomics techniques, the study unveils the dynamic molecular responses in distinct hippocampal subregions post-TBI. It uncovers region-specific vulnerabilities and dysregulated neuronal processes, and potential recovery-related pathways that contribute to our understanding of TBI's neurological consequences and provides valuable insights for biomarker discovery and therapeutic targets.
Collapse
Affiliation(s)
- Sudipa Maity
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Yuanyu Huang
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Mitchell D Kilgore
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Abbigail N Thurmon
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, USA
- Tulane Brain Institute, New Orleans, LA, USA
| | | | - Maria J Galazo
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, USA
- Tulane Brain Institute, New Orleans, LA, USA
| | - Xiaojiang Xu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jing Cao
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaoying Wang
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Bo Ning
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ning Liu
- Clinical Neuroscience Research Center, Department of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA, USA.
- Tulane University Translational Sciences Institute, New Orleans, LA, USA.
| | - Jia Fan
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, LA, USA.
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
9
|
Tan W, Ma J, Fu J, Wu B, Zhu Z, Huang X, Du M, Wu C, Balawi E, Zhou Q, Zhang J, Liao Z. Transcriptomic and bioinformatics analysis of the mechanism by which erythropoietin promotes recovery from traumatic brain injury in mice. Neural Regen Res 2024; 19:171-179. [PMID: 37488864 PMCID: PMC10479836 DOI: 10.4103/1673-5374.374135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 02/04/2023] [Accepted: 03/11/2023] [Indexed: 07/26/2023] Open
Abstract
Recent studies have found that erythropoietin promotes the recovery of neurological function after traumatic brain injury. However, the precise mechanism of action remains unclear. In this study, we induced moderate traumatic brain injury in mice by intraperitoneal injection of erythropoietin for 3 consecutive days. RNA sequencing detected a total of 4065 differentially expressed RNAs, including 1059 mRNAs, 92 microRNAs, 799 long non-coding RNAs, and 2115 circular RNAs. Kyoto Encyclopedia of Genes and Genomes and Gene Ontology analyses revealed that the coding and non-coding RNAs that were differentially expressed after traumatic brain injury and treatment with erythropoietin play roles in the axon guidance pathway, Wnt pathway, and MAPK pathway. Constructing competing endogenous RNA networks showed that regulatory relationship between the differentially expressed non-coding RNAs and mRNAs. Because the axon guidance pathway was repeatedly enriched, the expression of Wnt5a and Ephb6, key factors in the axonal guidance pathway, was assessed. Ephb6 expression decreased and Wnt5a expression increased after traumatic brain injury, and these effects were reversed by treatment with erythropoietin. These findings suggest that erythropoietin can promote recovery of nerve function after traumatic brain injury through the axon guidance pathway.
Collapse
Affiliation(s)
- Weilin Tan
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Ma
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiayuanyuan Fu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Biying Wu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ziyu Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xuekang Huang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mengran Du
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chenrui Wu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ehab Balawi
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiang Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jie Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhengbu Liao
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
10
|
Guo M, Wang L, Yin Z, Chen F, Lei P. Small extracellular vesicles as potential theranostic tools in central nervous system disorders. Biomed Pharmacother 2023; 167:115407. [PMID: 37683594 DOI: 10.1016/j.biopha.2023.115407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/26/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Small extracellular vesicles(sEVs), a subset of extracellular vesicles with a bilateral membrane structure, contain biological cargoes, such as lipids, nucleic acids, and proteins. sEVs are crucial mediators of intercellular communications in the physiological and pathological processes of the central nervous system. Because of the special structure and complex pathogenesis of the brain, central nervous system disorders are characterized by high mortality and morbidity. Increasing evidence has focused on the potential of sEVs in clinical application for central nervous system disorders. sEVs are emerging as a promising diagnostic and therapeutic tool with high sensitivity, low immunogenicity, superior safety profile, and high transfer efficiency. This review highlighted the development of sEVs in central nervous system disorder clinical application. We also outlined the role of sEVs in central nervous system disorders and discussed the limitations of sEVs in clinical translation.
Collapse
Affiliation(s)
- Mengtian Guo
- Department of Internal Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Lu Wang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China; Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhenyu Yin
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China; Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | | | - Ping Lei
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China; Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
11
|
Zhang Y, Zhang Y, Chopp M, Pang H, Chen L, Zhang ZG, Mahmood A, Xiong Y. Therapeutic Role of microRNAs of Small Extracellular Vesicles from Human Mesenchymal Stromal/Stem Cells in Treatment of Experimental Traumatic Brain Injury. J Neurotrauma 2023; 40:758-771. [PMID: 36394949 PMCID: PMC10068676 DOI: 10.1089/neu.2022.0296] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSC)-derived small extracellular vesicles (sEVs) possess therapeutic potential for treatment of traumatic brain injury (TBI). The essential role of micro ribonucleic acids (miRNAs) underlying the beneficial effects of MSC-derived sEVs for treatment of TBI remains elusive. The present study was designed to investigate the role of microRNAs in sEVs from MSCs with Argonaute 2 knockdown (Ago2-KD) in neurological recovery, neuroinflammation, and neurovascular remodeling in TBI rats. Therapeutic effects of sEVs derived from naïve MSCs (naïve-sEV), MSCs transfected with a vector carrying scramble control short hairpin RNA (shRNA; vector-sEV), and MSCs transfected with a lentiviral vector-based shRNA against Ago2 to knock down Ago2 (Ago2-KD-sEV) were determined in adult male rats subjected to a moderate TBI induced by controlled cortical impact (CCI). sEVs (naïve-sEV, vector-sEV, and Ago2-KD-sEV) or vehicle (phosphate-buffered solution [PBS]) were given intravenously 1 day post-injury (PI). Multiple neurological functional tests were performed weekly PI for 5 weeks. The Morris water maze (MWM) test was performed for spatial learning and memory 31-35 days PI. All animals were euthanized 5 weeks PI and the brains were collected for analyses of lesion volume, cell loss, neurovascular remodeling, and neuroinflammation. Ago2-KD reduced global sEV miRNA levels. Compared with the vehicle treatment, both naïve-sEV and vector-sEV treatments significantly improved functional recovery, reduced hippocampal neuronal cell loss, inhibited neuroinflammation, and promoted neurovascular remodeling (angiogenesis and neurogenesis). However, Ago2-KD-sEV treatment had a significantly less therapeutic effect on all the parameters measured above than did naïve-sEV and vector-sEV treatments. The therapeutic effects of Ago2-KD-sEV were comparable to that of vehicle treatment. Our findings demonstrate that attenuation of Ago2 protein in MSCs reduces miRNAs in MSC-derived sEVs and abolishes exosome treatment-induced beneficial effects in TBI recovery, suggesting that miRNAs in MSC-derived sEVs play an essential role in reducing neuronal cell loss, inhibiting neuroinflammation, and augmenting angiogenesis and neurogenesis, as well as improving functional recovery in TBI. The findings underscore the important role of miRNAs in MSC-derived sEVs in the treatment of TBI.
Collapse
Affiliation(s)
- Yanlu Zhang
- Department of Neurosurgery, Henry Ford Hospital, Detroit, Michigan, USA
| | - Yi Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
- Department of Physics, Oakland University, Rochester, Michigan, USA
| | - Haiyan Pang
- Department of Neurosurgery, Henry Ford Hospital, Detroit, Michigan, USA
| | - Liang Chen
- Department of Neurosurgery, Henry Ford Hospital, Detroit, Michigan, USA
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Asim Mahmood
- Department of Neurosurgery, Henry Ford Hospital, Detroit, Michigan, USA
| | - Ye Xiong
- Department of Neurosurgery, Henry Ford Hospital, Detroit, Michigan, USA
| |
Collapse
|
12
|
Acid Sphingomyelinase Inhibitor, Imipramine, Reduces Hippocampal Neuronal Death after Traumatic Brain Injury. Int J Mol Sci 2022; 23:ijms232314749. [PMID: 36499076 PMCID: PMC9740309 DOI: 10.3390/ijms232314749] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Traumatic brain injury (TBI) broadly degrades the normal function of the brain after a bump, blow, or jolt to the head. TBI leads to the aggravation of pre-existing brain dysfunction and promotes neurotoxic cascades that involve processes such as oxidative stress, loss of dendritic arborization, and zinc accumulation. Acid sphingomyelinase (ASMase) is an enzyme that hydrolyzes sphingomyelin to ceramide in cells. Under normal conditions, ceramide plays an important role in various physiological functions, such as differentiation and apoptosis. However, under pathological conditions, excessive ceramide production is toxic and activates the neuronal-death pathway. Therefore, we hypothesized that the inhibition of ASMase activity by imipramine would reduce ceramide formation and thus prevent TBI-induced neuronal death. To test our hypothesis, an ASMase inhibitor, imipramine (10 mg/kg, i.p.), was administrated to rats immediately after TBI. Based on the results of this study, we confirmed that imipramine significantly reduced ceramide formation, dendritic loss, oxidative stress, and neuronal death in the TBI-imipramine group compared with the TBI-vehicle group. Additionally, we validated that imipramine prevented TBI-induced cognitive dysfunction and the modified neurological severity score. Consequently, we suggest that ASMase inhibition may be a promising therapeutic strategy to reduce hippocampal neuronal death after TBI.
Collapse
|
13
|
Nebie O, Buée L, Blum D, Burnouf T. Can the administration of platelet lysates to the brain help treat neurological disorders? Cell Mol Life Sci 2022; 79:379. [PMID: 35750991 PMCID: PMC9243829 DOI: 10.1007/s00018-022-04397-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/09/2022] [Accepted: 05/23/2022] [Indexed: 11/03/2022]
Abstract
Neurodegenerative disorders of the central nervous system (CNS) and brain traumatic insults are characterized by complex overlapping pathophysiological alterations encompassing neuroinflammation, alterations of synaptic functions, oxidative stress, and progressive neurodegeneration that eventually lead to irreversible motor and cognitive dysfunctions. A single pharmacological approach is unlikely to provide a complementary set of molecular therapeutic actions suitable to resolve these complex pathologies. Recent preclinical data are providing evidence-based scientific rationales to support biotherapies based on administering neurotrophic factors and extracellular vesicles present in the lysates of human platelets collected from healthy donors to the brain. Here, we present the most recent findings on the composition of the platelet proteome that can activate complementary signaling pathways in vivo to trigger neuroprotection, synapse protection, anti-inflammation, antioxidation, and neurorestoration. We also report experimental data where the administration of human platelet lysates (HPL) was safe and resulted in beneficial neuroprotective effects in established rodent models of neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, traumatic brain injury, and stroke. Platelet-based biotherapies, prepared from collected platelet concentrates (PC), are emerging as a novel pragmatic and accessible translational therapeutic strategy for treating neurological diseases. Based on this assumption, we further elaborated on various clinical, manufacturing, and regulatory issues that need to be addressed to ensure the ethical supply, quality, and safety of HPL preparations for treating neurodegenerative and traumatic pathologies of the CNS. HPL made from PC may become a unique approach for scientifically based treatments of neurological disorders readily accessible in low-, middle-, and high-income countries.
Collapse
Affiliation(s)
- Ouada Nebie
- College of Biomedical Engineering, Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, 59045, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, LiCEND, 59000, Lille, France
| | - Luc Buée
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, 59045, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, LiCEND, 59000, Lille, France
- NeuroTMULille International Laboratory, Univ. Lille, Lille, France
| | - David Blum
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, 59045, Lille, France.
- Alzheimer and Tauopathies, LabEx DISTALZ, LiCEND, 59000, Lille, France.
- NeuroTMULille International Laboratory, Univ. Lille, Lille, France.
- NeuroTMULille International Laboratory, Taipei Medical University, Taipei, 11031, Taiwan.
| | - Thierry Burnouf
- College of Biomedical Engineering, Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan.
- NeuroTMULille International Laboratory, Taipei Medical University, Taipei, 11031, Taiwan.
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan.
- International PhD Program in Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Brain and Consciousness Research Centre, Taipei Medical University Shuang-Ho Hospital, New Taipei City, 23561, Taiwan.
- Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
14
|
Sarkar C, Lipinski MM. N-acetyl-L-leucine: a promising treatment option for traumatic brain injury. Neural Regen Res 2022; 17:1957-1958. [PMID: 35142673 PMCID: PMC8848596 DOI: 10.4103/1673-5374.335146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Chinmoy Sarkar
- Shock, Trauma and Anesthesiology Research (STAR) Center, Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marta M Lipinski
- Shock, Trauma and Anesthesiology Research (STAR) Center, Department of Anesthesiology; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
15
|
Penning A, Tosoni G, Abiega O, Bielefeld P, Gasperini C, De Pietri Tonelli D, Fitzsimons CP, Salta E. Adult Neural Stem Cell Regulation by Small Non-coding RNAs: Physiological Significance and Pathological Implications. Front Cell Neurosci 2022; 15:781434. [PMID: 35058752 PMCID: PMC8764185 DOI: 10.3389/fncel.2021.781434] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/09/2021] [Indexed: 01/11/2023] Open
Abstract
The adult neurogenic niches are complex multicellular systems, receiving regulatory input from a multitude of intracellular, juxtacrine, and paracrine signals and biological pathways. Within the niches, adult neural stem cells (aNSCs) generate astrocytic and neuronal progeny, with the latter predominating in physiological conditions. The new neurons generated from this neurogenic process are functionally linked to memory, cognition, and mood regulation, while much less is known about the functional contribution of aNSC-derived newborn astrocytes and adult-born oligodendrocytes. Accumulating evidence suggests that the deregulation of aNSCs and their progeny can impact, or can be impacted by, aging and several brain pathologies, including neurodevelopmental and mood disorders, neurodegenerative diseases, and also by insults, such as epileptic seizures, stroke, or traumatic brain injury. Hence, understanding the regulatory underpinnings of aNSC activation, differentiation, and fate commitment could help identify novel therapeutic avenues for a series of pathological conditions. Over the last two decades, small non-coding RNAs (sncRNAs) have emerged as key regulators of NSC fate determination in the adult neurogenic niches. In this review, we synthesize prior knowledge on how sncRNAs, such as microRNAs (miRNAs) and piwi-interacting RNAs (piRNAs), may impact NSC fate determination in the adult brain and we critically assess the functional significance of these events. We discuss the concepts that emerge from these examples and how they could be used to provide a framework for considering aNSC (de)regulation in the pathogenesis and treatment of neurological diseases.
Collapse
Affiliation(s)
- Amber Penning
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Giorgia Tosoni
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Oihane Abiega
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, Netherlands
| | - Pascal Bielefeld
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, Netherlands
| | - Caterina Gasperini
- Neurobiology of miRNAs Lab, Istituto Italiano di Tecnologia, Genova, Italy
| | | | - Carlos P. Fitzsimons
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, Netherlands
| | - Evgenia Salta
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| |
Collapse
|
16
|
Potential of different cells-derived exosomal microRNA cargos for treating spinal cord injury. J Orthop Translat 2021; 31:33-40. [PMID: 34760623 PMCID: PMC8560648 DOI: 10.1016/j.jot.2021.09.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/22/2021] [Accepted: 09/28/2021] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury (SCI) is a disastrous situation that affects many patients worldwide. A profound understanding of the pathology and etiology of SCI is of great importance in inspiring new therapeutic concepts and treatment. In recent years, exosomes, which are complex lipid membrane structures secreted nearly by all kinds of plants and animal cells, can transport their valuable cargoes (e.g., proteins, lipids, RNAs) to the targeted cells and exert their communication and regulation functions, which open up a new field of treatment of SCI. Notably, the exosome's advantage is transporting the carried material to the target cells across the blood-brain barrier and exerting regulatory functions. Among the cargoes of exosomes, microRNAs, through the modulation of their mRNA targets, emerges with great potentiality in the pathological process, diagnosis and treatment of SCI. In this review, we discuss the role of miRNAs transported by different cell-derived exosomes in SCI that are poised to enhance SCI-specific therapeutic capabilities of exosomes.
Collapse
|
17
|
Guo R, Wang X, Fang Y, Chen X, Chen K, Huang W, Chen J, Hu J, Liang F, Du J, Dordoe C, Tian X, Lin L. rhFGF20 promotes angiogenesis and vascular repair following traumatic brain injury by regulating Wnt/β-catenin pathway. Biomed Pharmacother 2021; 143:112200. [PMID: 34649342 DOI: 10.1016/j.biopha.2021.112200] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/13/2021] [Accepted: 09/13/2021] [Indexed: 11/29/2022] Open
Abstract
The pathology of cerebrovascular disorders takes an important role in traumatic brain injury (TBI) by increasing intracranial pressure. Fibroblast growth factor 20 (FGF20) is a brain-derived neurotrophic factor, that has been shown to play an important role in the survival of dopaminergic neurons and the treatment of Parkinson's disease (PD). However, little is known about the role of FGF20 in the treatment of TBI and its underlying mechanism. The purpose of this study was to evaluate the protective effect of recombinant human FGF20 (rhFGF20) on protecting cerebral blood vessels after TBI. In this study, we indicated that rhFGF20 could reduce brain edema, Evans blue penetration and upregulated the expression of blood-brain barrier (BBB)-related tight junction (TJ) proteins, exerting a protective effect on the BBB in vivo after TBI. In the TBI repair phase, rhFGF20 promoted angiogenesis, neurological and cognitive function recovery. In tumor necrosis factor-α (TNF-α)-induced human brain microvascular endothelial cells (hCMEC/D3), an in vitro BBB disruption model, rhFGF20 reversed the impairment in cell migration and tube formation induced by TNF-α. Moreover, in both the TBI mouse model and the in vitro model, rhFGF20 increased the expression of β-catenin and GSK3β, which are the two key regulators in the Wnt/β-catenin signaling pathway. In addition, the Wnt/β-catenin inhibitor IWR-1-endo significantly reversed the effects of rhFGF20. These results indicate that rhFGF20 may prevent vascular repair and angiogenesis through the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Ruili Guo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xue Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yani Fang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiongjian Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Kun Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wenting Huang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 315020, China
| | - Jun Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jian Hu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Fei Liang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jingting Du
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Confidence Dordoe
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xianxi Tian
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 315020, China.
| | - Li Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 315020, China; Research Units of Clinical Translation of Cell Growth Factors and Diseases Research, Chinese Academy of Medical Science, Beijing 100730, China.
| |
Collapse
|
18
|
Antsiferov OV, Cherevatenko RF, Korokin MV, Gureev VV, Gureeva AV, Zatolokina MA, Avdeyeva EV, Zhilinkova LA, Kolesnik IM. A new EPOR/CD131 heteroreceptor agonist EP-11-1: a neuroprotective effect in experimental traumatic brain injury. RESEARCH RESULTS IN PHARMACOLOGY 2021. [DOI: 10.3897/rrpharmacology.7.75301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Introduction: EP-11-1 (UEHLERALNSS) is a short-chain erythropoietin derivative without have erythropoietic activity. It was created by modifying a peptide mimicking the spatial structure of the erythropoietin a-helix B pHBSP. One of the promising directions of its administration is the correction of morphofunctional disorders that occur in traumatic brain injury (TBI).
Materials and methods: The study was performed in 160 male Wistar rats, weighing 180–200 g.TBI was simulated using the drop-weight method. To assess the emerging morphofunctional disorders and a degree of their correction, we used the severity of neurological deficit, indicators of locomotor activity and exploration, a marker of brain injury S100B and morphological examination.
Results and discussion: The combined administration of a new EPOR/CD131 heteroreceptor agonist EP-11-1 with citicoline and trimetazidine led to a more pronounced correction of the neurological deficit when compared not only to the group of the ”untreated” animals, but also to the groups of animals to which these drugs had been administered as monotherapy (p < 0.05). The same tendency was also observed in the study of locomotor activity and exploration. A biochemical study showed that the administration of all three combinations led to a statistically significant (p < 0.05) decrease in the S-100B concentration compared not only to the group of “untreated” animals, but also to the groups of animals to which these drugs had been administered as a monotherapy.
Conclusion: The results of the conducted experiments prove the most pronounced positive dynamics in the combined administration of the new EPOR/CD131 heteroreceptor agonist EP-11-1with citicoline and trimetazidine.
Collapse
|
19
|
Marzano LAS, de Castro FLM, Machado CA, de Barros JLVM, Macedo E Cordeiro T, Simões E Silva AC, Teixeira AL, Silva de Miranda A. Potential Role of Adult Hippocampal Neurogenesis in Traumatic Brain Injury. Curr Med Chem 2021; 29:3392-3419. [PMID: 34561977 DOI: 10.2174/0929867328666210923143713] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/28/2021] [Accepted: 08/18/2021] [Indexed: 11/22/2022]
Abstract
Traumatic brain injury (TBI) is a serious cause of disability and death among young and adult individuals, displaying complex pathophysiology including cellular and molecular mechanisms that are not fully elucidated. Many experimental and clinical studies investigated the potential relationship between TBI and the process by which neurons are formed in the brain, known as neurogenesis. Currently, there are no available treatments for TBI's long-term consequences being the search for novel therapeutic targets, a goal of highest scientific and clinical priority. Some studies evaluated the benefits of treatments aimed at improving neurogenesis in TBI. In this scenario, herein, we reviewed current pre-clinical studies that evaluated different approaches to improving neurogenesis after TBI while achieving better cognitive outcomes, which may consist in interesting approaches for future treatments.
Collapse
Affiliation(s)
- Lucas Alexandre Santos Marzano
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Brazil
| | | | - Caroline Amaral Machado
- Laboratório de Neurobiologia, Departamento de Morfologia, Instituto de Ciências Biológicas, UFMG, Brazil
| | | | - Thiago Macedo E Cordeiro
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Ana Cristina Simões E Silva
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Antônio Lúcio Teixeira
- Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, United States
| | - Aline Silva de Miranda
- Laboratório Interdisciplinar de Investigação Médica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Brazil
| |
Collapse
|
20
|
Traumatic Brain Injury: Mechanistic Insight on Pathophysiology and Potential Therapeutic Targets. J Mol Neurosci 2021; 71:1725-1742. [PMID: 33956297 DOI: 10.1007/s12031-021-01841-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/09/2021] [Indexed: 12/20/2022]
Abstract
Traumatic brain injury (TBI) causes brain damage, which involves primary and secondary injury mechanisms. Primary injury causes local brain damage, while secondary damage begins with inflammatory activity followed by disruption of the blood-brain barrier (BBB), peripheral blood cells infiltration, brain edema, and the discharge of numerous immune mediators including chemotactic factors and interleukins. TBI alters molecular signaling, cell structures, and functions. Besides tissue damage such as axonal damage, contusions, and hemorrhage, TBI in general interrupts brain physiology including cognition, decision-making, memory, attention, and speech capability. Regardless of the deep understanding of the pathophysiology of TBI, the underlying mechanisms still need to be assessed with a desired therapeutic agent to control the consequences of TBI. The current review gives a brief outline of the pathophysiological mechanism of TBI and various biochemical pathways involved in brain injury, pharmacological treatment approaches, and novel targets for therapy.
Collapse
|
21
|
Zhang Y, Zhang Y, Chopp M, Pang H, Zhang ZG, Mahmood A, Xiong Y. MiR-17-92 Cluster-Enriched Exosomes Derived from Human Bone Marrow Mesenchymal Stromal Cells Improve Tissue and Functional Recovery in Rats after Traumatic Brain Injury. J Neurotrauma 2021; 38:1535-1550. [PMID: 33787364 DOI: 10.1089/neu.2020.7575] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Exosomes play an important role in intercellular communication by delivering microribonucleic acids (miRNAs) to recipient cells. Previous studies have demonstrated that multi-potent mesenchymal stromal cell (MSC)-derived exosomes improve functional recovery after experimental traumatic brain injury (TBI). This study was performed to determine efficacy of miR-17-92 cluster-enriched exosomes (Exo-17-92) harvested from human bone marrow MSCs transfected with a miR-17-92 cluster plasmid in enhancing tissue and neurological recovery compared with exosomes derived from MSCs transfected with an empty plasmid vector (Exo-empty) for treatment of TBI. Adult male rats underwent a unilateral moderate cortical contusion. Animals received a single intravenous injection of miR-17-92 cluster-enriched exosomes (100 μg/rat, approximately 3.75x1011 particles, Exo-17-92) or control exosomes (100 μg/rat, Exo-empty) or Vehicle (phosphate-buffered solution) one day after injury. A battery of neurological functional tests was performed weekly after TBI for five weeks. Spatial learning and memory were measured on days 31-35 after TBI using the Morris water maze test. All animals were sacrificed five weeks after injury. Their brains were processed for histopathological and immunohistochemical analyses of lesion volume, cell loss, angiogenesis, neurogenesis, and neuroinflammation. Compared with Vehicle, both Exo-17-92 and Exo-empty treatments significantly improved sensorimotor and cognitive function, reduced neuroinflammation and hippocampal neuronal cell loss, promoted angiogenesis and neurogenesis without altering the lesion volume. Moreover, Exo-17-92 treatment exhibited a significantly more robust therapeutic effect on improvement in functional recovery by reducing neuroinflammation and cell loss, enhancing angiogenesis and neurogenesis than did Exo-empty treatment. Exosomes enriched with miR-17-92 cluster have a significantly better effect on improving functional recovery after TBI compared with Exo-empty, likely by reducing neuroinflammation and enhancing endogenous angiogenesis and neurogenesis. Engineering specific miRNA in exosomes may provide a novel therapeutic strategy for management of unilateral moderate cortical contusion TBI.
Collapse
Affiliation(s)
- Yanlu Zhang
- Department of Neurosurgery and Henry Ford Hospital, Detroit, Michigan, USA
| | - Yi Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physics, Oakland University, Rochester, Michigan, USA
| | - Haiyan Pang
- Department of Neurosurgery and Henry Ford Hospital, Detroit, Michigan, USA
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Asim Mahmood
- Department of Neurosurgery and Henry Ford Hospital, Detroit, Michigan, USA
| | - Ye Xiong
- Department of Neurosurgery and Henry Ford Hospital, Detroit, Michigan, USA
| |
Collapse
|
22
|
Kumar Mishra S, Khushu S, Gangenahalli G. Neuroprotective response and efficacy of intravenous administration of mesenchymal stem cells in traumatic brain injury mice. Eur J Neurosci 2021; 54:4392-4407. [PMID: 33932318 DOI: 10.1111/ejn.15261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 04/06/2021] [Accepted: 04/26/2021] [Indexed: 12/13/2022]
Abstract
Cellular transplantation of stem cells can be a beneficial treatment approach for neurodegenerative diseases such as traumatic brain injury (TBI). In this study, we investigated the proliferation and differentiation potential of infused mesenchymal stem cells (MSCs) after localisation at the injury site. We evaluated the appropriate homing of infused MSCs through immunohistochemistry, followed by Y-chromosome-specific polymerase chain reaction and fluorescent in situ hybridisation analyses. The proliferation and differentiation of infused MSCs were analysed using exogenous cell tracer 5'-bromo-2'-deoxyuridine (BrdU) labelling and neuronal specific markers, respectively. Structural and functional recovery in TBI mice were examined by performing magnetic resonance imaging and different behavioural assessments, respectively. Results demonstrated a significantly high number of BrdU-positive cells in the lesion region in the MSC-infused group compared with control and TBI groups. Infused MSCs were well differentiated into neural-like cells and expressed significantly more neural markers (neuronal nuclear antigen [NeuN], microtubule-associated protein 2 [MAP2] and glial fibrillary acid protein [GFAP]). Improved tissue abnormalities as well as functional behaviours were observed in MSC-infused TBI mice, implying the substantial proliferation and differentiation of infused MSCs. Our findings support the neuroprotective response and efficacy of MSCs after transplantation in TBI mice, and MSCs may serve as potential therapeutic candidates in regenerative medicine.
Collapse
Affiliation(s)
- Sushanta Kumar Mishra
- MRI Research Group, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, India
- Division of Stem Cell and Gene Therapy Research, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, India
| | - Subash Khushu
- MRI Research Group, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, India
| | - Gurudutta Gangenahalli
- Division of Stem Cell and Gene Therapy Research, Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, India
| |
Collapse
|
23
|
Ghiam MK, Patel SD, Hoffer A, Selman WR, Hoffer BJ, Hoffer ME. Drug Repurposing in the Treatment of Traumatic Brain Injury. Front Neurosci 2021; 15:635483. [PMID: 33833663 PMCID: PMC8021906 DOI: 10.3389/fnins.2021.635483] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/19/2021] [Indexed: 01/02/2023] Open
Abstract
Traumatic brain injury (TBI) is the most common cause of morbidity among trauma patients; however, an effective pharmacological treatment has not yet been approved. Individuals with TBI are at greater risk of developing neurological illnesses such as Alzheimer's disease (AD) and Parkinson's disease (PD). The approval process for treatments can be accelerated by repurposing known drugs to treat the growing number of patients with TBI. This review focuses on the repurposing of N-acetyl cysteine (NAC), a drug currently approved to treat hepatotoxic overdose of acetaminophen. NAC also has antioxidant and anti-inflammatory properties that may be suitable for use in therapeutic treatments for TBI. Minocycline (MINO), a tetracycline antibiotic, has been shown to be effective in combination with NAC in preventing oligodendrocyte damage. (-)-phenserine (PHEN), an anti-acetylcholinesterase agent with additional non-cholinergic neuroprotective/neurotrophic properties initially developed to treat AD, has demonstrated efficacy in treating TBI. Recent literature indicates that NAC, MINO, and PHEN may serve as worthwhile repositioned therapeutics in treating TBI.
Collapse
Affiliation(s)
- Michael K. Ghiam
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Shrey D. Patel
- University of Miami Miller School of Medicine, Miami, FL, United States
| | - Alan Hoffer
- Department of Neurological Surgery, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Warren R. Selman
- Department of Neurological Surgery, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Barry J. Hoffer
- Department of Neurological Surgery, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Michael E. Hoffer
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
24
|
Attilio PJ, Snapper DM, Rusnak M, Isaac A, Soltis AR, Wilkerson MD, Dalgard CL, Symes AJ. Transcriptomic Analysis of Mouse Brain After Traumatic Brain Injury Reveals That the Angiotensin Receptor Blocker Candesartan Acts Through Novel Pathways. Front Neurosci 2021; 15:636259. [PMID: 33828448 PMCID: PMC8019829 DOI: 10.3389/fnins.2021.636259] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/19/2021] [Indexed: 12/30/2022] Open
Abstract
Traumatic brain injury (TBI) results in complex pathological reactions, where the initial lesion is followed by secondary inflammation and edema. Our laboratory and others have reported that angiotensin receptor blockers (ARBs) have efficacy in improving recovery from traumatic brain injury in mice. Treatment of mice with a subhypotensive dose of the ARB candesartan results in improved functional recovery, and reduced pathology (lesion volume, inflammation and gliosis). In order to gain a better understanding of the molecular mechanisms through which candesartan improves recovery after controlled cortical impact injury (CCI), we performed transcriptomic profiling on brain regions after injury and drug treatment. We examined RNA expression in the ipsilateral hippocampus, thalamus and hypothalamus at 3 or 29 days post injury (dpi) treated with either candesartan (0.1 mg/kg) or vehicle. RNA was isolated and analyzed by bulk mRNA-seq. Gene expression in injured and/or candesartan treated brain region was compared to that in sham vehicle treated mice in the same brain region to identify genes that were differentially expressed (DEGs) between groups. The most DEGs were expressed in the hippocampus at 3 dpi, and the number of DEGs reduced with distance and time from the lesion. Among pathways that were differentially expressed at 3 dpi after CCI, candesartan treatment altered genes involved in angiogenesis, interferon signaling, extracellular matrix regulation including integrins and chromosome maintenance and DNA replication. At 29 dpi, candesartan treatment reduced the expression of genes involved in the inflammatory response. Some changes in gene expression were confirmed in a separate cohort of animals by qPCR. Fewer DEGs were found in the thalamus, and only one in the hypothalamus at 3 dpi. Additionally, in the hippocampi of sham injured mice, 3 days of candesartan treatment led to the differential expression of 384 genes showing that candesartan in the absence of injury had a powerful impact on gene expression specifically in the hippocampus. Our results suggest that candesartan has broad actions in the brain after injury and affects different processes at acute and chronic times after injury. These data should assist in elucidating the beneficial effect of candesartan on recovery from TBI.
Collapse
Affiliation(s)
- Peter J. Attilio
- Graduate Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Dustin M. Snapper
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Milan Rusnak
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Akira Isaac
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Anthony R. Soltis
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Matthew D. Wilkerson
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Clifton L. Dalgard
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Aviva J. Symes
- Graduate Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
25
|
Lerouet D, Marchand-Leroux C, Besson VC. Neuropharmacology in traumatic brain injury: from preclinical to clinical neuroprotection? Fundam Clin Pharmacol 2021; 35:524-538. [PMID: 33527472 PMCID: PMC9290810 DOI: 10.1111/fcp.12656] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 01/24/2021] [Accepted: 01/26/2021] [Indexed: 12/11/2022]
Abstract
Traumatic brain injury (TBI) constitutes a major health problem worldwide and is a leading cause of death and disability in individuals, contributing to devastating socioeconomic consequences. Despite numerous promising pharmacological strategies reported as neuroprotective in preclinical studies, the translation to clinical trials always failed, albeit the great diversity of therapeutic targets evaluated. In this review, first, we described epidemiologic features, causes, and primary and secondary injuries of TBI. Second, we outlined the current literature on animal models of TBI, and we described their goals, their advantages and disadvantages according to the species used, the type of injury induced, and their clinical relevance. Third, we defined the concept of neuroprotection and discussed its evolution. We also identified the reasons that might explain the failure of clinical translation. Then, we reviewed post‐TBI neuroprotective treatments with a focus on the following pleiotropic drugs, considered “low hanging fruit” with high probability of success: glitazones, glibenclamide, statins, erythropoietin, and progesterone, that were largely tested and demonstrated efficient in preclinical models of TBI. Finally, our review stresses the need to establish a close cooperation between basic researchers and clinicians to ensure the best clinical translation for neuroprotective strategies for TBI.
Collapse
Affiliation(s)
- Dominique Lerouet
- UMR-S1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, Faculté de Pharmacie de Paris, Université de Paris, Paris, France
| | - Catherine Marchand-Leroux
- UMR-S1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, Faculté de Pharmacie de Paris, Université de Paris, Paris, France
| | - Valérie C Besson
- UMR-S1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, Faculté de Pharmacie de Paris, Université de Paris, Paris, France
| |
Collapse
|
26
|
Chavda V, Madhwani K. Coding and non-coding nucleotides': The future of stroke gene therapeutics. Genomics 2021; 113:1291-1307. [PMID: 33677059 DOI: 10.1016/j.ygeno.2021.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/01/2020] [Accepted: 03/02/2021] [Indexed: 01/05/2023]
Abstract
Stroke is the foremost cause of death ranked after heart disease and cancer. It is the fatal life-threatening event that requires immediate medical admissions to overcome following morbidity and mortality. The therapeutic advances in stroke therapy have been manipulated with diverse paths for last 5 years. Recent research and clinical trials have investigated a variety of anti-stroke agents including anti-coagulants, cerebro-protective agents, antiplatelet therapy, stem-cell therapy, and specified gene therapy. In recent advanced studies, genetic therapies including noncoding RNAs (ncRNAs), long non-coding RNAs (LncRNAs), small interfering RNAs (siRNAs), microRNAs (miRNAs), Piwi interacting RNAs (PiWi RNAs) have shown better potential as targeted future therapeutics with a better outcome than conventional stroke therapeutics. The potential of targeted gene therapy is much more advanced in not only the induction of neuroprotection but also safer non-toxic targeted therapeutics. In the current state of the art review, we have focused on the recent advancements made towards the stroke with RNA modifications and targeted gene therapeutics.
Collapse
Affiliation(s)
- Vishal Chavda
- Department of Pharmacology, Nirma University, Ahmadabad, Gujarat, India.
| | - Kajal Madhwani
- Department of Microbiology, Nirma University, Ahmadabad, Gujarat, India
| |
Collapse
|
27
|
de Almeida da Anunciação AR, Favaron PO, de Morais-Pinto L, de Carvalho CMF, Dos Santos Martins D, Conei D, Del Sol M, Vásquez B, Miglino MA. Central nervous system development in rabbits (Oryctolagus cuniculus L. 1758). Anat Rec (Hoboken) 2021; 304:1313-1328. [PMID: 33480146 DOI: 10.1002/ar.24586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 10/27/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023]
Abstract
The present study describes the embryonic and fetal development of the central nervous system in rabbits from the seventh day after conception until the end of the full-term fetal period. A total of 19 embryonic and fetal samples were carefully dissected and microscopically analyzed. Neural tube closure was observed between 7.5 and 8 days of gestation. Primordial encephalic vesicle differentiation and spinal canal delimitation were observed on the 12th day of gestation. Histologically, on the 15th day of gestation, the brain, cerebellum, and brain stem were delimited. On the 18th day of gestation, the cervical and lumbar intumescences of the spinal cord were visible. On the 28th day of gestation, four-cell layers could be distinguished in the cerebral cortex, while the cerebellar cortex was still differentiating. Overall, the morphological aspects of the embryonic and fetal developmental phases in rabbits were highly similar to those in humans. Thus, the present study provides relevant information highlighting rabbits as an excellent candidate animal model for preclinical research on human neurological diseases given the high adaptability of rabbits to bioterium conditions and the similarity of morphological events between rabbits and humans.
Collapse
Affiliation(s)
| | - Phelipe Oliveira Favaron
- General Biology Department, Biological Science Center, Universidade Estadual de Londrina, Londrina, Brazil
| | - Luciano de Morais-Pinto
- Laboratory of Anatomical Design/LabDA, Department of Morphology, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | | | | | - Daniel Conei
- Doctoral Program in Morphological Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
| | - Mariano Del Sol
- Doctoral Program in Morphological Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
| | - Bélgica Vásquez
- Faculty of Health Sciences, Universidad de Tarapacá, Arica, Chile
| | - Maria Angelica Miglino
- Faculty of Veterinary Medicine Animal Sciences, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
28
|
Dave AM, Peeples ES. Cholesterol metabolism and brain injury in neonatal encephalopathy. Pediatr Res 2021; 90:37-44. [PMID: 33106607 PMCID: PMC8511855 DOI: 10.1038/s41390-020-01218-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 01/05/2023]
Abstract
Neonatal encephalopathy (NE) results from impaired cerebral blood flow and oxygen delivery to the brain. The pathophysiology of NE is complex and our understanding of its underlying pathways continues to evolve. There is considerable evidence that cholesterol dysregulation is involved in several adult diseases, including traumatic brain injury, stroke, Huntington's disease, and Parkinson's disease. Although the research is less robust in pediatrics, there is emerging evidence that aberrations in cholesterol metabolism may also be involved in the pathophysiology of neonatal NE. This narrative review provides an overview of cholesterol metabolism in the brain along with several examples from the adult literature where pathologic alterations in cholesterol metabolism have been associated with inflammatory and ischemic brain injury. Using those data as a background, the review then discusses the current preclinical data supporting the involvement of cholesterol in the pathogenesis of NE as well as how brain-specific cholesterol metabolites may serve as serum biomarkers for brain injury. Lastly, we review the potential for using the cholesterol metabolic pathways as therapeutic targets. Further investigation of the shifts in cholesterol synthesis and metabolism after hypoxia-ischemia may prove vital in understanding NE pathophysiology as well as providing opportunities for rapid diagnosis and therapeutic interventions. IMPACT: This review summarizes emerging evidence that aberrations in cholesterol metabolism may be involved in the pathophysiology of NE. Using data from NE as well as analogous adult disease states, this article reviews the potential for using cholesterol pathways as targets for developing novel therapeutic interventions and using cholesterol metabolites as biomarkers for injury. When possible, gaps in the current literature were identified to aid in the development of future studies to further investigate the interactions between cholesterol pathways and NE.
Collapse
Affiliation(s)
- Amanda M Dave
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Eric S Peeples
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
29
|
Hackett EP, Pinho MC, Harrison CE, Reed GD, Liticker J, Raza J, Hall RG, Malloy CR, Barshikar S, Madden CJ, Park JM. Imaging Acute Metabolic Changes in Patients with Mild Traumatic Brain Injury Using Hyperpolarized [1- 13C]Pyruvate. iScience 2020; 23:101885. [PMID: 33344923 PMCID: PMC7736977 DOI: 10.1016/j.isci.2020.101885] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/25/2020] [Accepted: 11/25/2020] [Indexed: 01/13/2023] Open
Abstract
Traumatic brain injury (TBI) involves complex secondary injury processes following the primary injury. The secondary injury is often associated with rapid metabolic shifts and impaired brain function immediately after the initial tissue damage. Magnetic resonance spectroscopic imaging (MRSI) coupled with hyperpolarization of 13C-labeled substrates provides a unique opportunity to map the metabolic changes in the brain after traumatic injury in real-time without invasive procedures. In this report, we investigated two patients with acute mild TBI (Glasgow coma scale 15) but no anatomical brain injury or hemorrhage. Patients were imaged with hyperpolarized [1-13C]pyruvate MRSI 1 or 6 days after head trauma. Both patients showed significantly reduced bicarbonate (HCO3–) production, and one showed hyperintense lactate production at the injured sites. This study reports the feasibility of imaging altered metabolism using hyperpolarized pyruvate in patients with TBI, demonstrating the translatability and sensitivity of the technology to cerebral metabolic changes after mild TBI. Clinical translation of hyperpolarized pyruvate to TBI was demonstrated Patients with mild TBI were imaged with hyperpolarized [1-13C]pyruvate Altered lactate and HCO3– production in the brain nearest the site of trauma
Collapse
Affiliation(s)
- Edward P Hackett
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Marco C Pinho
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Crystal E Harrison
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Galen D Reed
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,GE Healthcare, Dallas, TX 75390, USA
| | - Jeff Liticker
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jaffar Raza
- Department of Pharmacy Practice, The Texas Tech University Health Sciences Center, Dallas, TX 75216, USA
| | - Ronald G Hall
- Department of Pharmacy Practice, The Texas Tech University Health Sciences Center, Dallas, TX 75216, USA
| | - Craig R Malloy
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Surendra Barshikar
- Department of Physical Medicine & Rehabilitation, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Christopher J Madden
- Department of Neurological Surgery, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jae Mo Park
- Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Department of Electrical and Computer Engineering, The University of Texas at Dallas, Richardson TX 75080, USA
| |
Collapse
|
30
|
Traumatic brain injury modifies synaptic plasticity in newly-generated granule cells of the adult hippocampus. Exp Neurol 2020; 336:113527. [PMID: 33188818 DOI: 10.1016/j.expneurol.2020.113527] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/30/2020] [Accepted: 10/27/2020] [Indexed: 01/11/2023]
Abstract
The hippocampus is vulnerable to traumatic brain injury (TBI), and hippocampal damage is associated with cognitive deficits that are often the hallmark of TBI. Recent studies have found that TBI induces enhanced neurogenesis in the dentate gyrus (DG) of the hippocampus, and this cellular response is related to innate cognitive recovery. However, cellular mechanisms of the role of DG neurogenesis in post-TBI recovery remain unclear. This study investigated changes in long-term potentiation (LTP) within the DG in relation to TBI-induced neurogenesis. Adult male rats received a moderate TBI or sham injury and were sacrificed for brain slice recordings at 30 or 60 days post-injury. Recordings were taken from the medial perforant path input to DG granule cells in the presence or absence of the GABAergic antagonist picrotoxin, reflecting activity of either all DG granule cells or predominately newborn granule cells, respectively. Measurements of LTP observed in the total granule cell population (with picrotoxin) showed a prolonged impairment which worsened between 30 and 60 days post-TBI. Under conditions which predominantly reflected the LTP elicited in newly born granule cells (no picrotoxin), a strikingly different pattern of post-TBI changes was observed, with a time-dependent cycle of functional impairment and recovery. At 30 days after injury this cell population showed little or no LTP, but by 60 days the capacity for LTP of the newly born granule cells was no different from that of sham controls. The time-frame of LTP improvements in the newborn cell population, comparable to that of behavioral recovery reported previously, suggests the unique functional properties of newborn granule cells enable them to contribute to restorative change following brain injury.
Collapse
|
31
|
Crupi R, Cordaro M, Cuzzocrea S, Impellizzeri D. Management of Traumatic Brain Injury: From Present to Future. Antioxidants (Basel) 2020; 9:antiox9040297. [PMID: 32252390 PMCID: PMC7222188 DOI: 10.3390/antiox9040297] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/29/2020] [Accepted: 03/31/2020] [Indexed: 12/12/2022] Open
Abstract
TBI (traumatic brain injury) is a major cause of death among youth in industrialized societies. Brain damage following traumatic injury is a result of direct and indirect mechanisms; indirect or secondary injury involves the initiation of an acute inflammatory response, including the breakdown of the blood–brain barrier (BBB), brain edema, infiltration of peripheral blood cells, and activation of resident immunocompetent cells, as well as the release of numerous immune mediators such as interleukins and chemotactic factors. TBI can cause changes in molecular signaling and cellular functions and structures, in addition to tissue damage, such as hemorrhage, diffuse axonal damages, and contusions. TBI typically disturbs brain functions such as executive actions, cognitive grade, attention, memory data processing, and language abilities. Animal models have been developed to reproduce the different features of human TBI, better understand its pathophysiology, and discover potential new treatments. For many years, the first approach to manage TBI has been treatment of the injured tissue with interventions designed to reduce the complex secondary-injury cascade. Several studies in the literature have stressed the importance of more closely examining injuries, including endothelial, microglia, astroglia, oligodendroglia, and precursor cells. Significant effort has been invested in developing neuroprotective agents. The aim of this work is to review TBI pathophysiology and existing and potential new therapeutic strategies in the management of inflammatory events and behavioral deficits associated with TBI.
Collapse
Affiliation(s)
- Rosalia Crupi
- Department of Veterinary Science, University of Messina, 98168 Messina, Italy;
| | - Marika Cordaro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Via Consolare Valeria 1, 98100 Messina, Italy;
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmacological and Environmental Sciences, Messina University, Viale F. Stagno D’Alcontres 31, 98166 Messina, Italy;
- Department of Pharmacological and Physiological Science, Saint Louis University, Saint Louis, MO 63104, USA
- Correspondence: ; Tel.: +390-906-765-208
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmacological and Environmental Sciences, Messina University, Viale F. Stagno D’Alcontres 31, 98166 Messina, Italy;
| |
Collapse
|
32
|
Zhou C, Chen H, Zheng JF, Guo ZD, Huang ZJ, Wu Y, Zhong JJ, Sun XC, Cheng CJ. Pentraxin 3 contributes to neurogenesis after traumatic brain injury in mice. Neural Regen Res 2020; 15:2318-2326. [PMID: 32594056 PMCID: PMC7749468 DOI: 10.4103/1673-5374.285001] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Emerging evidence indicates that pentraxin 3 is an acute-phase protein that is linked with the immune response to inflammation. It is also a newly discovered marker of anti-inflammatory A2 reactive astrocytes, and potentially has multiple protective effects in stroke; however, its role in the adult brain after traumatic brain injury is unknown. In the present study, a moderate model of traumatic brain injury in mice was established using controlled cortical impact. The models were intraventricularly injected with recombinant pentraxin 3 (the recombinant pentraxin 3 group) or an equal volume of vehicle (the control group). The sham-operated mice underwent craniotomy, but did not undergo the controlled cortical impact. The potential neuroprotective and neuroregenerative roles of pentraxin 3 were investigated on days 14 and 21 after traumatic brain injury. Western blot assay showed that the expression of endogenous pentraxin 3 was increased after traumatic brain injury in mice. Furthermore, the neurological severity test and wire grip test revealed that recombinant pentraxin 3 treatment reduced the neurological severity score and increased the wire grip score, suggesting an improved recovery of sensory-motor functions. The Morris water maze results demonstrated that recombinant pentraxin 3 treatment reduced the latency to the platform, increased the time spent in the correct quadrant, and increased the number of times traveled across the platform, thus suggesting an improved recovery of cognitive function. In addition, to investigate the effects of pentraxin 3 on astrocytes, specific markers of A2 astrocytes were detected in primary astrocyte cultures in vitro using western blot assay. The results demonstrated that pentraxin 3 administration activates A2 astrocytes. To explore the protective mechanisms of pentraxin 3, immunofluorescence staining was used. Intraventricular injection of recombinant pentraxin 3 increased neuronal maintenance in the peri-injured cortex and ipsilateral hippocampus, increased the number of doublecortin-positive neural progenitor cells in the subventricular and subgranular zones, and increased the number of bromodeoxyuridine (proliferation) and neuronal nuclear antigen (mature neuron) double-labeled cells in the hippocampus and peri-injured cortex. Pentraxin 3 administration also increased the number of neurospheres and the number of bromodeoxyuridine and doublecortin double-labeled cells in neurospheres, and enhanced the proliferation of neural progenitor cells in primary neural progenitor cell cultures in vitro. In conclusion, recombinant pentraxin 3 administration activated A2 astrocytes, and consequently improved the recovery of neural function by increasing neuronal survival and enhancing neurogenesis. All experiments were approved by the Animal Ethics Committee of the First Affiliated Hospital of Chongqing Medical University, China on March 1, 2016.
Collapse
Affiliation(s)
- Chao Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Chen
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian-Feng Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zong-Duo Guo
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhi-Jian Huang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Wu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian-Jun Zhong
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao-Chuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chong-Jie Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
33
|
Sun M, McDonald SJ, Brady RD, Collins-Praino L, Yamakawa GR, Monif M, O'Brien TJ, Cloud GC, Sobey CG, Mychasiuk R, Loane DJ, Shultz SR. The need to incorporate aged animals into the preclinical modeling of neurological conditions. Neurosci Biobehav Rev 2019; 109:114-128. [PMID: 31877345 DOI: 10.1016/j.neubiorev.2019.12.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/04/2019] [Accepted: 12/19/2019] [Indexed: 12/14/2022]
Abstract
Neurological conditions such as traumatic brain injury, stroke, Parkinson's disease, epilepsy, multiple sclerosis, and Alzheimer's disease are serious clinical problems that affect millions of people worldwide. The majority of clinical trials for these common conditions have failed, and there is a critical need to understand why treatments in preclinical animal models do not translate to patients. Many patients with these conditions are middle-aged or older, however, the majority of preclinical studies have used only young-adult animals. Considering that aging involves biological changes that are relevant to the pathobiology of neurological diseases, the lack of aged subjects in preclinical research could contribute to translational failures. This paper details how aging affects biological processes involved in neurological conditions, and reviews aging research in the context of traumatic brain injury, stroke, Parkinson's disease, epilepsy, multiple sclerosis, and Alzheimer's disease. We conclude that aging is an important, but often overlooked, factor that influences biology and outcomes in neurological conditions, and provide suggestions to improve our understanding and treatment of these diseases in aged patients.
Collapse
Affiliation(s)
- Mujun Sun
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia; Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC 3086, Australia
| | - Rhys D Brady
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3052, Australia
| | - Lyndsey Collins-Praino
- Department of Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - Mastura Monif
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3052, Australia; Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia
| | - Geoffrey C Cloud
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia; Department of Stroke Services, Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC 3086, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia
| | - David J Loane
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA; School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Ireland
| | - Sandy R Shultz
- Department of Neuroscience, Monash University, Melbourne, VIC 3004, Australia; Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3052, Australia; Department of Neurology, Alfred Health, Melbourne, VIC 3004, Australia.
| |
Collapse
|
34
|
Li L, Chopp M, Ding G, Davoodi-Bojd E, Li Q, Mahmood A, Xiong Y, Jiang Q. Diffuse white matter response in trauma-injured brain to bone marrow stromal cell treatment detected by diffusional kurtosis imaging. Brain Res 2019; 1717:127-135. [PMID: 31009610 PMCID: PMC6571170 DOI: 10.1016/j.brainres.2019.04.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/25/2019] [Accepted: 04/18/2019] [Indexed: 12/14/2022]
Abstract
Diffuse white matter (WM) response to traumatic brain injury (TBI) and transplantation of human bone marrow stromal cells (hMSCs) after the injury were non-invasively and dynamically investigated. Male Wistar rats (300-350 g) subjected to TBI were intravenously injected with 1 ml of saline (n = 10) or with hMSCs in suspension (∼3 × 106 hMSCs, n = 10) 1-week post-TBI. MRI measurements of T2-weighted imaging and diffusional kurtosis imaging (DKI) were acquired on all animals at multiple time points up to 3-months post-injury. Functional outcome was assessed using the Morris water maze test. DKI-derived metrics of fractional anisotropy (FA), axonal water fraction (AWF) and radial kurtosis (RK) longitudinally reveal an evolving pattern of structural alteration post-TBI occurring in the brain region remote from primary impact site. The progressive structural change is characterized by gradual disruption of WM integrity at an early stage (weeks post-TBI), followed by spontaneous recovery at a later stage (months post-TBI). Transplantation of hMSCs post-TBI promotes this structural plasticity as indicated by significantly increased FA and AWF in conjunction with substantially elevated RK at the later stage. Our long-term imaging data demonstrate that hMSC therapy leads to modified temporal profiles of these metrics, inducing an earlier presence of enhanced structural remodeling, which may contribute to improved functional recovery.
Collapse
Affiliation(s)
- Lian Li
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; Department of Physics, Oakland University, Rochester, MI 48309, USA.
| | - Guangliang Ding
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA.
| | | | - Qingjiang Li
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Asim Mahmood
- Department of Neurosurgery, Henry Ford Health System, Detroit, MI 48208, USA.
| | - Ye Xiong
- Department of Neurosurgery, Henry Ford Health System, Detroit, MI 48208, USA.
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA.
| |
Collapse
|
35
|
Głombik K, Trojan E, Kurek A, Budziszewska B, Basta-Kaim A. Inflammatory Consequences of Maternal Diabetes on the Offspring Brain: a Hippocampal Organotypic Culture Study. Neurotox Res 2019; 36:357-375. [PMID: 31197747 PMCID: PMC6616224 DOI: 10.1007/s12640-019-00070-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 05/10/2019] [Accepted: 05/28/2019] [Indexed: 01/09/2023]
Abstract
Gestational diabetes is a disorder associated with abnormal chronic inflammation that poses a risk to the developing fetus. We investigated the effects of experimentally induced diabetes (streptozotocin model) in Wistar female rats on the inflammatory status of the hippocampi of their offspring. Additionally, the impact of antidiabetic drugs (metformin and glyburide) on inflammatory processes was evaluated. Organotypic hippocampal cultures (OHCs) were prepared from the brains of the 7-day-old rat offspring of control and diabetic mother rats. On the 7th day in vitro, the cultures were pretreated with metformin (3 μM) or glyburide (1 μM) and then stimulated for 24 h with lipopolysaccharide (LPS, 1 μg/ml). The OHCs obtained from the offspring of diabetic mothers were characterized by the increased mortality of cells and an enhanced susceptibility to damage caused by LPS. Although we showed that LPS stimulated the secretion of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) in the control and diabetic cultures, the levels of IL-1β and IL-6 in the OHC medium obtained from the offspring of diabetic mothers were more pronounced. In the diabetic cultures, enhanced levels of TLR-4 and the overactivation of the NLRP3 inflammasome were demonstrated. Metformin and glyburide pretreatment normalized the LPS-induced IL-1β secretion in the control and diabetic cultures. Furthermore, glyburide diminished both: LPS-induced IL-6 and TNF-α secretion in the control and diabetic cultures and increased NF-κB p65 subunit phosphorylation. Glyburide also diminished the levels of the NLRP3 subunit and caspase-1, but only in the diabetic cultures. The results showed that maternal diabetes affected inflammatory processes in the offspring brain and increased hippocampal sensitivity to the LPS-induced inflammatory response. The use of antidiabetic agents, especially glyburide, had a beneficial impact on the changes caused by maternal diabetes.
Collapse
Affiliation(s)
- Katarzyna Głombik
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland.
| | - Ewa Trojan
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Anna Kurek
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Bogusława Budziszewska
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| | - Agnieszka Basta-Kaim
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St, 31-343, Kraków, Poland
| |
Collapse
|
36
|
Kumar Sahel D, Kaira M, Raj K, Sharma S, Singh S. Mitochondrial dysfunctioning and neuroinflammation: Recent highlights on the possible mechanisms involved in Traumatic Brain Injury. Neurosci Lett 2019; 710:134347. [PMID: 31229625 DOI: 10.1016/j.neulet.2019.134347] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 06/12/2019] [Accepted: 06/19/2019] [Indexed: 11/18/2022]
Abstract
Traumatic brain injury (TBI) is the injury to the vasculature of brain while trauma caused by physical, chemical and biological stimuli. TBI is the leading cause of mortality and morbidity around the world. In this, primary insult leads to secondary injury through the involvement and initiation of various pathological processes. The most citable includes excitotoxicity, Blood Brain Barrier (BBB) dysfunction, inflammation, mitochondrial dysfunction, oxidative stress, calcium efflux, microglial mediated release of proinflammatory mediators (cytokine, chemokines, interleukin, tissue necrosis factor etc.). The morphological changes in TBI are proportional to mitochondrial dysfunctioning and microglial activation, which play an assorted role in neurodegeneration following traumatic brain injury. It is also assumed that the release of nitric oxide, activation of microglial cells plays a diversive role in maintaining the physiological and pathological balance. This review cites different pathophysiological mechanisms that are involved in progenesis of secondary injury after primary insult. These targets further are useful to explore the deep molecular mechanisms and to analyse the effectiveness of available drugs. Moreover, the present review reflects the underlying inflammatory cascade responsible for neuronal loss and neurological deficit in TBI.
Collapse
Affiliation(s)
- Deepak Kumar Sahel
- Neuroscience Division, Department of Pharmacology, I.S.F. College of Pharmacy, Moga, Punjab, 142001, India
| | - Meenakshi Kaira
- Department of Pharmaceutical Sciences, M.D University, Rohtak, Haryana, 124001, India
| | - Khadga Raj
- Neuroscience Division, Department of Pharmacology, I.S.F. College of Pharmacy, Moga, Punjab, 142001, India
| | - Shakshi Sharma
- Neuroscience Division, Department of Pharmacology, I.S.F. College of Pharmacy, Moga, Punjab, 142001, India
| | - Shamsher Singh
- Neuroscience Division, Department of Pharmacology, I.S.F. College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
37
|
Alhelaly MM, Soliman AM, Khaled A, Ellotf H, Attia MM, Elmaraezy A. Efficacy of tranexamic acid in traumatic brain injury: Updated systematic review and meta-analysis. TRAUMA-ENGLAND 2019. [DOI: 10.1177/1460408619842736] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
| | | | - Anas Khaled
- Damietta Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| | - Hamed Ellotf
- Damietta Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| | | | | |
Collapse
|
38
|
Novgorodov SA, Voltin JR, Wang W, Tomlinson S, Riley CL, Gudz TI. Acid sphingomyelinase deficiency protects mitochondria and improves function recovery after brain injury. J Lipid Res 2019; 60:609-623. [PMID: 30662008 PMCID: PMC6399498 DOI: 10.1194/jlr.m091132] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/11/2019] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the leading causes of disability worldwide and a prominent risk factor for neurodegenerative diseases. The expansion of nervous tissue damage after the initial trauma involves a multifactorial cascade of events, including excitotoxicity, oxidative stress, inflammation, and deregulation of sphingolipid metabolism that further mitochondrial dysfunction and secondary brain damage. Here, we show that a posttranscriptional activation of an acid sphingomyelinase (ASM), a key enzyme of the sphingolipid recycling pathway, resulted in a selective increase of sphingosine in mitochondria during the first week post-TBI that was accompanied by reduced activity of mitochondrial cytochrome oxidase and activation of the Nod-like receptor protein 3 inflammasome. TBI-induced mitochondrial abnormalities were rescued in the brains of ASM KO mice, which demonstrated improved behavioral deficit recovery compared with WT mice. Furthermore, an elevated autophagy in an ASM-deficient brain at the baseline and during the development of secondary brain injury seems to foster the preservation of mitochondria and brain function after TBI. Of note, ASM deficiency attenuated the early stages of reactive astrogliosis progression in an injured brain. These findings highlight the crucial role of ASM in governing mitochondrial dysfunction and brain-function impairment, emphasizing the importance of sphingolipids in the neuroinflammatory response to TBI.
Collapse
Affiliation(s)
- Sergei A Novgorodov
- Departments of Neuroscience Medical University of South Carolina, Charleston, SC 29425
| | - Joshua R Voltin
- Departments of Neuroscience Medical University of South Carolina, Charleston, SC 29425
| | - Wenxue Wang
- Microbiology and Immunology Medical University of South Carolina, Charleston, SC 29425
| | - Stephen Tomlinson
- Microbiology and Immunology Medical University of South Carolina, Charleston, SC 29425
| | | | - Tatyana I Gudz
- Departments of Neuroscience Medical University of South Carolina, Charleston, SC 29425
- Ralph H. Johnson Veterans Affairs Medical Center Charleston, SC 29401
| |
Collapse
|
39
|
Rana A, Singh S, Sharma R, Kumar A. Traumatic Brain Injury Altered Normal Brain Signaling Pathways: Implications for Novel Therapeutics Approaches. Curr Neuropharmacol 2019; 17:614-629. [PMID: 30207236 PMCID: PMC6712292 DOI: 10.2174/1570159x16666180911121847] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 09/01/2018] [Accepted: 09/06/2018] [Indexed: 12/20/2022] Open
Abstract
Traumatic brain injury (TBI) is the main reason of lifelong disability and casualty worldwide. In the United State alone, 1.7 million traumatic events occur yearly, out of which 50,000 results in deaths. Injury to the brain could alter various biological signaling pathways such as excitotoxicity, ionic imbalance, oxidative stress, inflammation, and apoptosis which can result in various neurological disorders such as Psychosis, Depression, Alzheimer disease, Parkinson disease, etc. In literature, various reports have indicated the alteration of these pathways after traumatic brain injury but the exact mechanism is still unclear. Thus, in the first part of this article, we have tried to summarize TBI as a modulator of various neuronal signaling pathways. Currently, very few drugs are available in the market for the treatment of TBI and these drugs only provide the supportive care. Thus, in the second part of the article, based on TBI altered signaling pathways, we have tried to find out potential targets and promising therapeutic approaches in the treatment of TBI.
Collapse
Affiliation(s)
| | | | | | - Anoop Kumar
- Address correspondence to this author at the Department of Pharmacology, Indo-Soviet Friendship College of Pharmacy (ISFCP), Moga, Punjab-142001, India; Tel: +91 636 324200/324201; E-mail:
| |
Collapse
|
40
|
|
41
|
Johnson T, Zhao L, Manuel G, Taylor H, Liu D. Approaches to therapeutic angiogenesis for ischemic heart disease. J Mol Med (Berl) 2018; 97:141-151. [PMID: 30554258 DOI: 10.1007/s00109-018-1729-3] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 11/30/2018] [Accepted: 12/05/2018] [Indexed: 12/13/2022]
Abstract
Ischemic heart disease (IHD) is caused by the narrowing of arteries that work to provide blood, nutrients, and oxygen to the myocardial tissue. The worldwide epidemic of IHD urgently requires innovative treatments despite the significant advances in medical, interventional, and surgical therapies for this disease. Angiogenesis is a physiological and pathophysiological process that initiates vascular growth from pre-existing blood vessels in response to a lack of oxygen. This process occurs naturally over time and has encouraged researchers and clinicians to investigate the outcomes of accelerating or enhancing this angiogenic response as an alternative IHD therapy. Therapeutic angiogenesis has been shown to revascularize ischemic heart tissue, reduce the progression of tissue infarction, and evade the need for invasive surgical procedures or tissue/organ transplants. Several approaches, including the use of proteins, genes, stem/progenitor cells, and various combinations, have been employed to promote angiogenesis. While clinical trials for these approaches are ongoing, microvesicles and exosomes have recently been investigated as a cell-free approach to stimulate angiogenesis and may circumvent limitations of using viable cells. This review summarizes the approaches to accomplish therapeutic angiogenesis for IHD by highlighting the advances and challenges that addresses the applicability of a potential pro-angiogenic medicine.
Collapse
Affiliation(s)
- Takerra Johnson
- Morehouse School of Medicine, Cardiovascular Research Institute, 720 Westview Drive SW, Atlanta, GA, 30310, USA
| | - Lina Zhao
- Morehouse School of Medicine, Cardiovascular Research Institute, 720 Westview Drive SW, Atlanta, GA, 30310, USA
| | - Gygeria Manuel
- Department of Biochemistry, Spelman College, 350 Spelman Lane, Atlanta, GA, 30314, USA
| | - Herman Taylor
- Morehouse School of Medicine, Cardiovascular Research Institute, 720 Westview Drive SW, Atlanta, GA, 30310, USA
| | - Dong Liu
- Morehouse School of Medicine, Cardiovascular Research Institute, 720 Westview Drive SW, Atlanta, GA, 30310, USA.
| |
Collapse
|
42
|
Nasser M, Ballout N, Mantash S, Bejjani F, Najdi F, Ramadan N, Soueid J, Zibara K, Kobeissy F. Transplantation of Embryonic Neural Stem Cells and Differentiated Cells in a Controlled Cortical Impact (CCI) Model of Adult Mouse Somatosensory Cortex. Front Neurol 2018; 9:895. [PMID: 30405520 PMCID: PMC6208009 DOI: 10.3389/fneur.2018.00895] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 10/02/2018] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is a major cause of death worldwide. Depending on the severity of the injury, TBI can reflect a broad range of consequences such as speech impairment, memory disturbances, and premature death. In this study, embryonic neural stem cells (ENSC) were isolated from E14 mouse embryos and cultured to produce neurospheres which were induced to generate differentiated cells (DC). As a cell replacement treatment option, we aimed to transplant ENSC or DC into the adult injured C57BL/6 mouse cortex controlled cortical impact (CCI) model, 7 days post-trauma, in comparison to saline injection (control). The effect of grafted cells on neuroinflammation and neurogenesis was investigated at 1 and 4 weeks post-transplantation. Results showed that microglia were activated following mild CCI, but not enhanced after engraftment of ENSC or DC. Indeed, ipsilateral lesioned somatosensory area expressed high levels of Iba-1+ microglia within the different groups after 1 and 4 weeks. On the other hand, treatment with ENSC or DC demonstrated a significant reduction in astrogliosis. The levels of GFAP expressing astrocytes started decreasing early (1 week) in the ENSC group and then were similarly low at 4 weeks in both ENSC and DC. Moreover, neurogenesis was significantly enhanced in ENSC and DC groups. Indeed, a significant increase in the number of DCX expressing progenitor cells was observed at 1 week in the ENSC group, and in DC and ENSC groups at 4 weeks. Furthermore, the number of mature neuronal cells (NeuN+) significantly increased in DC group at 4 weeks whereas they decreased in ENSC group at 1 week. Therefore, injection of ENSC or DC post-CCI caused decreased astrogliosis and suggested an increased neurogenesis via inducing neural progenitor proliferation and expression rather than neuronal maturation. Thus, ENSC may play a role in replacing lost cells and brain repair following TBI by improving neurogenesis and reducing neuroinflammation, reflecting an optimal environment for transplanted and newly born cells.
Collapse
Affiliation(s)
- Mohammad Nasser
- Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon.,ER045, PRASE, DSST, Lebanese University, Beirut, Lebanon
| | | | - Sarah Mantash
- ER045, PRASE, DSST, Lebanese University, Beirut, Lebanon
| | | | - Farah Najdi
- ER045, PRASE, DSST, Lebanese University, Beirut, Lebanon
| | - Naify Ramadan
- ER045, PRASE, DSST, Lebanese University, Beirut, Lebanon.,Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Jihane Soueid
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Kazem Zibara
- Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon.,ER045, PRASE, DSST, Lebanese University, Beirut, Lebanon
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
43
|
Zhang Y, Chopp M, Rex CS, Simmon VF, Sarraf ST, Zhang ZG, Mahmood A, Xiong Y. A Small Molecule Spinogenic Compound Enhances Functional Outcome and Dendritic Spine Plasticity in a Rat Model of Traumatic Brain Injury. J Neurotrauma 2018; 36:589-600. [PMID: 30014757 DOI: 10.1089/neu.2018.5790] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The tetra (ethylene glycol) derivative of benzothiazole aniline (SPG101) has been shown to improve dendritic spine density and cognitive memory in the triple transgenic mouse model of Alzheimer disease (AD) when administered intraperitoneally. The present study was designed to investigate the therapeutic effects of SPG101 on dendritic spine density and morphology and sensorimotor and cognitive functional recovery in a rat model of traumatic brain injury (TBI) induced by controlled cortical impact (CCI). Young adult male Wistar rats with CCI were randomly divided into the following two groups (n = 7/group): (1) Vehicle, and (2) SPG101. SPG101 (30 mg/kg) dissolved in vehicle (1% dimethyl sulfoxide in phosphate buffered saline) or Vehicle were intraperitoneally administered starting at 1 h post-injury and once daily for the next 34 days. Sensorimotor deficits were assessed using a modified neurological severity score and adhesive removal and foot fault tests. Cognitive function was measured by Morris water maze, novel object recognition (NOR), and three-chamber social recognition tests. The animals were sacrificed 35 days after injury, and their brains were processed for measurement of dendritic spine density and morphology using ballistic dye labeling. Compared with the vehicle treatment, SPG101 treatment initiated 1 h post-injury significantly improved sensorimotor functional recovery (days 7-35, p < 0.0001), spatial learning (days 32-35, p < 0.0001), NOR (days 14 and 35, p < 0.0001), social recognition (days 14 and 35, p < 0.0001). Further, treatment significantly increased dendritic spine density in the injured cortex (p < 0.05), decreased heterogeneous distribution of spine lengths in the injured cortex and hippocampus (p < 0.0001), modifications that are associated with the promotion of spine maturation in these brain regions. In summary, treatment with SPG101 initiated 1 h post-injury and continued for an additional 34 days improves both sensorimotor and cognitive functional recovery, indicating that SPG101 acts as a spinogenic agent and may have potential as a novel treatment of TBI.
Collapse
Affiliation(s)
- Yanlu Zhang
- 1 Department of Neurosurgery, Henry Ford Hospital , Detroit, Michigan
| | - Michael Chopp
- 2 Department of Neurology, Henry Ford Hospital , Detroit, Michigan.,3 Department of Physics, Oakland University , Rochester, Michigan
| | | | | | | | - Zheng Gang Zhang
- 2 Department of Neurology, Henry Ford Hospital , Detroit, Michigan
| | - Asim Mahmood
- 1 Department of Neurosurgery, Henry Ford Hospital , Detroit, Michigan
| | - Ye Xiong
- 1 Department of Neurosurgery, Henry Ford Hospital , Detroit, Michigan
| |
Collapse
|
44
|
Wu H, Li J, Xu D, Zhang Q, Cui T. Growth Differentiation Factor 5 Improves Neurogenesis and Functional Recovery in Adult Mouse Hippocampus Following Traumatic Brain Injury. Front Neurol 2018; 9:592. [PMID: 30083129 PMCID: PMC6064945 DOI: 10.3389/fneur.2018.00592] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/02/2018] [Indexed: 01/01/2023] Open
Abstract
The aim of this study was to investigate the therapeutic effect of growth differentiation factor 5 (GDF-5) on traumatic brain injury (TBI) in mice. We utilized a controlled cortical impact to establish a mouse TBI model, and then stereotaxically administered 25 or 100 ng GDF-5 into the bilateral hippocampal dentate gyrus (DG) of each of the animals. Seven days after the injury, some of the animals were sacrificed for immunohistochemical and immunofluorescence examination of 5-bromo-2'-deoxyuridine (BrdU), Sox-2, doublecortin (DCX) and phosphorylated cAMP response element binding protein (p-CREB). Dendrite quantification was also performed using DCX positive cells. Activation of newborn neurons was assessed 35 days after the injury. The remaining animals were subjected to open field, Y maze and contextual fear conditioning tests 2 months after TBI. As a result, we found that post-injury stereotaxical administration of GDF-5 can improve neural stem cell proliferation and differentiation in the DG of the hippocampus, evidenced by the increase in BrdU, Sox-2, and DCX-labeled cells, as well as the improvement in dendrite arborization and newborn neuron activation in response to GDF-5 treatment. Mechanistically, these effects of GDF-5 may be mediated by the CREB pathway, manifested by the recovery of TBI-induced dephosphorylation of CREB upon GDF-5 administration. Behavioral tests further verified the effects of GDF-5 on improving cognitive and behavioral dysfunction after TBI. Collectively, these results reveal that direct injection of GDF-5 into the hippocampus can stimulate neurogenesis and improve functional recovery in a mouse TBI model, indicating the potential therapeutic effects of GDF-5 on TBI.
Collapse
Affiliation(s)
- Hongjie Wu
- Department of Neurosurgery The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Jing Li
- Department of Neurosurgery The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Dongxiao Xu
- Department of Neurosurgery The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Qiansheng Zhang
- Department of Neurosurgery The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Tao Cui
- Department of Neurosurgery The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
45
|
Shapiro LA. Altered Hippocampal Neurogenesis during the First 7 Days after a Fluid Percussion Traumatic Brain Injury. Cell Transplant 2018; 26:1314-1318. [PMID: 28933222 PMCID: PMC5657729 DOI: 10.1177/0963689717714099] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Traumatic brain injury (TBI) is a devastating disorder causing negative outcomes in millions of people each year. Despite the alarming number of brain injuries and the long-term detrimental outcomes that can be associated with TBI, treatment options are lacking. Extensive investigation is underway, in hopes of identifying effective treatment strategies. Among the most state-of-the-art strategies is cell replacement therapy. TBI is a seemingly good candidate for cell replacement studies because there is often loss of neurons. However, translation of this therapy has not yet been successful. It is possible that a better understanding of endogenous neurogenic mechanisms after TBI could lead to more efficacious study designs using exogenous cell replacement strategies. Therefore, this study was designed to examine the number and migration of immature neurons at 1 and 7 d after a fluid percussion TBI. The results show that the number of immature neurons increases from 7 d after a fluid percussion injury (FPI), and there is ectopic migration of doublecortin (DCX+) immature neurons into the hilar region of the dentate gyrus. These results add important data to the current understanding of the endogenous neurogenic niche after TBI. Follow-up studies are needed to better understand the functional significance of elevated neurogenesis and aberrant migration into the hilus.
Collapse
Affiliation(s)
- Lee A Shapiro
- 1 Department of Surgery, Texas A&M Health Science Center, Temple, TX, USA
| |
Collapse
|
46
|
|
47
|
Chen J, Chopp M. Exosome Therapy for Stroke. Stroke 2018; 49:1083-1090. [PMID: 29669873 PMCID: PMC6028936 DOI: 10.1161/strokeaha.117.018292] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 08/28/2017] [Accepted: 08/30/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Jieli Chen
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (J.C., M.C.)
- Department of Geriatrics, Tianjin Medical University General Hospital, China (J.C.)
- Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, China (J.C.)
| | - Michael Chopp
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (J.C., M.C.)
- Department of Physics, Oakland University, Rochester, MI (M.C.)
| |
Collapse
|
48
|
Erythropoietin Effects on Pathological Changes of Brain Tissues and Motor Balance Functions after Trauma Brain Injury in Animal Model. RAZAVI INTERNATIONAL JOURNAL OF MEDICINE 2017. [DOI: 10.5812/rijm.65025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
49
|
Alali AS, Mukherjee K, McCredie VA, Golan E, Shah PS, Bardes JM, Hamblin SE, Haut ER, Jackson JC, Khwaja K, Patel NJ, Raj SR, Wilson LD, Nathens AB, Patel MB. Beta-blockers and Traumatic Brain Injury: A Systematic Review, Meta-analysis, and Eastern Association for the Surgery of Trauma Guideline. Ann Surg 2017; 266:952-961. [PMID: 28525411 PMCID: PMC5997270 DOI: 10.1097/sla.0000000000002286] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To determine if beta-(β)-blockers improve outcomes after acute traumatic brain injury (TBI). BACKGROUND There have been no new inpatient pharmacologic therapies to improve TBI outcomes in a half-century. Treatment of TBI patients with β-blockers offers a potentially beneficial approach. METHODS Using MEDLINE, EMBASE, and CENTRAL databases, eligible articles for our systematic review and meta-analysis (PROSPERO CRD42016048547) included adult (age ≥ 16 years) blunt trauma patients admitted with TBI. The exposure of interest was β-blocker administration initiated during the hospitalization. Outcomes were mortality, functional measures, quality of life, cardiopulmonary morbidity (e.g., hypotension, bradycardia, bronchospasm, and/or congestive heart failure). Data were analyzed using a random-effects model, and represented by pooled odds ratio (OR) with 95% confidence intervals (CI) and statistical heterogeneity (I). RESULTS Data were extracted from 9 included studies encompassing 2005 unique TBI patients with β-blocker treatment and 6240 unique controls. Exposure to β-blockers after TBI was associated with a reduction of in-hospital mortality (pooled OR 0.39, 95% CI: 0.27-0.56; I = 65%, P < 0.00001). None of the included studies examined functional outcome or quality of life measures, and cardiopulmonary adverse events were rarely reported. No clear evidence of reporting bias was identified. CONCLUSIONS In adults with acute TBI, observational studies reveal a significant mortality advantage with β-blockers; however, quality of evidence is very low. We conditionally recommend the use of in-hospital β-blockers. However, we recommend further high-quality trials to answer questions about the mechanisms of action, effectiveness on subgroups, dose-response, length of therapy, functional outcome, and quality of life after β-blocker use for TBI.
Collapse
Affiliation(s)
- Aziz S. Alali
- Interdepartmental Division of Critical Care, University of Toronto, Toronto, ON, Canada
| | - Kaushik Mukherjee
- Division of Acute Care Surgery, Department of Surgery, Loma Linda University Medical Center, Loma Linda, CA
- Eastern Association for the Surgery of Trauma
| | | | - Eyal Golan
- Interdepartmental Division of Critical Care, University of Toronto, Toronto, ON, Canada
- Department of Critical Care, University Health Network, Toronto, ON, Canada
- Division of Critical Care and Department of Medicine, Mackenzie Health, Toronto, ON, Canada
| | - Prakesh S. Shah
- Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - James M. Bardes
- Department of Surgery, West Virginia University; Department of Surgery, USC+LAC, Los Angeles, CA
- Eastern Association for the Surgery of Trauma
| | - Susan E. Hamblin
- Department of Pharmaceutical Services, Vanderbilt University Medical Center, Nashville, TN
| | - Elliott R. Haut
- Departments of Surgery, Anesthesiology / Critical Care Medicine, and Emergency Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
- Eastern Association for the Surgery of Trauma
| | - James C. Jackson
- Division of Pulmonary and Critical Care Medicine and Center for Health Services Research, Department of Medicine, Vanderbilt University Medical Center; Research Service, Nashville Veterans Affairs Medical Center, Tennessee Valley Healthcare System
| | - Kosar Khwaja
- Departments of Surgery and Critical Care Medicine, McGill University Health Centre, Montreal, QC, Canada
- Eastern Association for the Surgery of Trauma
| | - Nimitt J. Patel
- Division of Trauma, Critical Care, and Burns, Department of Surgery, MetroHealth Medical Center, Cleveland, OH
- Eastern Association for the Surgery of Trauma
| | - Satish R. Raj
- Department of Cardiac Sciences, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada
| | - Laura D. Wilson
- Department of Communication Sciences and Disorders, Oxley College of Health Sciences, The University of Tulsa; Department of Hearing and Speech Sciences, Vanderbilt University School of Medicine
| | - Avery B. Nathens
- Department of Surgery, University of Toronto, Toronto, ON, Canada
- Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Mayur B. Patel
- Eastern Association for the Surgery of Trauma
- Division of Trauma, Emergency General Surgery, and Surgical Critical Care, Departments of Surgery, Neurosurgery, and Hearing and Speech Sciences, Section of Surgical Sciences, Vanderbilt Brain Institute, Vanderbilt Center for Health Services Research, Vanderbilt University Medical Center; Surgical Service, General Surgery Section, Nashville VA Medical Center, Tennessee Valley Healthcare System, US Department of Veterans Affairs, Nashville, TN
| |
Collapse
|
50
|
Zhang H, Wang W, Jiang S, Zhang Y, Heo HY, Wang X, Peng Y, Wang J, Zhou J. Amide proton transfer-weighted MRI detection of traumatic brain injury in rats. J Cereb Blood Flow Metab 2017; 37:3422-3432. [PMID: 28128026 PMCID: PMC5624391 DOI: 10.1177/0271678x17690165] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The purpose of this study was to explore the capability and uniqueness of amide proton transfer-weighted (APTw) imaging in the detection of primary and secondary injury after controlled cortical impact (CCI)-induced traumatic brain injury (TBI) in rats. Eleven adult rats had craniotomy plus CCI surgery under isoflurane anesthesia. Multi-parameter MRI data were acquired at 4.7 T, at eight time points (1, 6 h, and 1, 2, 3, 7, 14, and 28 days after TBI). At one and six hours post-injury, average APTw signal intensities decreased significantly in the impacted and peri-lesional areas due to tissue acidosis. A slightly high APTw signal was seen in the core lesion area with respect to the peri-lesional area, which was due to hemorrhage, as shown by T2*w. After the initial drop, the APTw signals dramatically increased in some peri-lesional areas at two and three days post-injury, likely due to the secondary inflammatory response. The use of APTw MRI has the potential to introduce a novel molecular neuroimaging approach for the simultaneous detection of ischemia, hemorrhage, and neuroinflammation in TBI.
Collapse
Affiliation(s)
- Hong Zhang
- 1 Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, USA.,2 Department of Radiology, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Wenzhu Wang
- 3 Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA.,4 Department of Integrated Chinese and West Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shanshan Jiang
- 1 Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
| | - Yi Zhang
- 1 Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
| | - Hye-Young Heo
- 1 Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
| | - Xianlong Wang
- 1 Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
| | - Yun Peng
- 2 Department of Radiology, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Jian Wang
- 3 Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jinyuan Zhou
- 1 Division of MR Research, Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|