1
|
Kim H, Kim BJ, Koh S, Cho HJ, Jin X, Kim BG, Choi JY. High mobility group box 1 in the central nervous system: regeneration hidden beneath inflammation. Neural Regen Res 2025; 20:107-115. [PMID: 38767480 PMCID: PMC11246138 DOI: 10.4103/nrr.nrr-d-23-01964] [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: 11/29/2023] [Revised: 01/31/2024] [Accepted: 03/04/2024] [Indexed: 05/22/2024] Open
Abstract
High-mobility group box 1 was first discovered in the calf thymus as a DNA-binding nuclear protein and has been widely studied in diverse fields, including neurology and neuroscience. High-mobility group box 1 in the extracellular space functions as a pro-inflammatory damage-associated molecular pattern, which has been proven to play an important role in a wide variety of central nervous system disorders such as ischemic stroke, Alzheimer's disease, frontotemporal dementia, Parkinson's disease, multiple sclerosis, epilepsy, and traumatic brain injury. Several drugs that inhibit high-mobility group box 1 as a damage-associated molecular pattern, such as glycyrrhizin, ethyl pyruvate, and neutralizing anti-high-mobility group box 1 antibodies, are commonly used to target high-mobility group box 1 activity in central nervous system disorders. Although it is commonly known for its detrimental inflammatory effect, high-mobility group box 1 has also been shown to have beneficial pro-regenerative roles in central nervous system disorders. In this narrative review, we provide a brief summary of the history of high-mobility group box 1 research and its characterization as a damage-associated molecular pattern, its downstream receptors, and intracellular signaling pathways, how high-mobility group box 1 exerts the repair-favoring roles in general and in the central nervous system, and clues on how to differentiate the pro-regenerative from the pro-inflammatory role. Research targeting high-mobility group box 1 in the central nervous system may benefit from differentiating between the two functions rather than overall suppression of high-mobility group box 1.
Collapse
Affiliation(s)
- Hanki Kim
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, South Korea
| | - Bum Jun Kim
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, South Korea
| | - Seungyon Koh
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, South Korea
- Department of Neurology, Ajou University School of Medicine, Suwon, South Korea
| | - Hyo Jin Cho
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
| | - Xuelian Jin
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
- Department of Geriatrics, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, Jiangsu Province, China
| | - Byung Gon Kim
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
- Department of Neurology, Ajou University School of Medicine, Suwon, South Korea
| | - Jun Young Choi
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
- Department of Neurology, Ajou University School of Medicine, Suwon, South Korea
| |
Collapse
|
2
|
Tan X, Li X, Li R, Meng W, Xie Z, Li J, Pang Y, Huang G, Li L, Li H. β-hydroxybutyrate alleviates neurological deficits by restoring glymphatic and inflammation after subarachnoid hemorrhage in mice. Exp Neurol 2024; 378:114819. [PMID: 38763355 DOI: 10.1016/j.expneurol.2024.114819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/07/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Both glymphatic system dysfunction and inflammatory response aggravate neurological dysfunction after subarachnoid hemorrhage (SAH). Studies have shown that β-hydroxybutyrate (BHB) may mitigate painful diabetic neuropathy (PDN) by upregulating SNTA1 expression and reinstating AQP4 polarity. However, the potential of BHB to ameliorate glymphatic system function and inflammatory response in SAH mice remains uncertain. METHODS The SAH models were constructed by injection of arterial blood into cisterna Magana. Three groups of C57 mice were randomly assigned: Sham, SAH, and BHB. All mice were subjected to neurological function assessment, western blot, immunofluorescence double staining, and RNA-seq. Glymphatic system function was examined with tracer and immunofluorescence double staining, and the differential genes were examined with RNA-seq. In addition, the expression of related inflammation was detected. RESULTS Compared with the SAH group, BHB reinstated AQP4 polarization by upregulating SNTA1 protein to enhance the glymphatic system. According to RNA-seq, the different genes were primarily connected to microglia activation, astrocytes, and inflammation. Western blot and immunofluorescence further confirmed that the related inflammatory protein expression levels were upregulated. BHB attenuated neuroinflammation after SAH. Ultimately, it can mitigate the neurological deficits in SAH mice. CONCLUSION The study reveals a novel mechanism that BHB treatment mitigates neurologic impairment in SAH mice. We propose that BHB may play a neuroprotective effect by enhancing glymphatic system function and attenuating neuroinflammatory subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Xiaoli Tan
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Xiaohong Li
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Ruhua Li
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Weiting Meng
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Zhuoxi Xie
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Jing Li
- Department of Cardiology, The 924th Hospital of Chinese People's Liberation Army Joint Service Support Force, Guilin, China
| | - Yeyu Pang
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Guilan Huang
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Li Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, China.
| | - Hao Li
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin 541001, China.
| |
Collapse
|
3
|
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
|
4
|
Li Z, Yu S, Li L, Zhou C, Wang L, Tang S, Gu N, Zhang Z, Huang Z, Chen H, Tang W, Wang Y, Yang X, Sun X, Yan J. TREM2 alleviates white matter injury after traumatic brain injury in mice might be mediated by regulation of DHCR24/LXR pathway in microglia. Clin Transl Med 2024; 14:e1665. [PMID: 38649789 PMCID: PMC11035381 DOI: 10.1002/ctm2.1665] [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: 11/23/2023] [Revised: 03/26/2024] [Accepted: 04/04/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND White matter injury (WMI) is an important pathological process after traumatic brain injury (TBI). The correlation between white matter functions and the myeloid cells expressing triggering receptor-2 (TREM2) has been convincingly demonstrated. Moreover, a recent study revealed that microglial sterol metabolism is crucial for early remyelination after demyelinating diseases. However, the potential roles of TREM2 expression and microglial sterol metabolism in WMI after TBI have not yet been explored. METHODS Controlled cortical injury was induced in both wild-type (WT) and TREM2 depletion (TREM2 KO) mice to simulate clinical TBI. COG1410 was used to upregulate TREM2, while PLX5622 and GSK2033 were used to deplete microglia and inhibit the liver X receptor (LXR), respectively. Immunofluorescence, Luxol fast blue staining, magnetic resonance imaging, transmission electron microscopy, and oil red O staining were employed to assess WMI after TBI. Neurological behaviour tests and electrophysiological recordings were utilized to evaluate cognitive functions following TBI. Microglial cell sorting and transcriptomic sequencing were utilized to identify alterations in microglial sterol metabolism-related genes, while western blot was conducted to validate the findings. RESULTS TREM2 expressed highest at 3 days post-TBI and was predominantly localized to microglial cells within the white matter. Depletion of TREM2 worsened aberrant neurological behaviours, and this phenomenon was mediated by the exacerbation of WMI, reduced renewal of oligodendrocytes, and impaired phagocytosis ability of microglia after TBI. Subsequently, the upregulation of TREM2 alleviated WMI, promoted oligodendrocyte regeneration, and ultimately facilitated the recovery of neurological behaviours after TBI. Finally, the expression of DHCR24 increased in TREM2 KO mice after TBI. Interestingly, TREM2 inhibited DHCR24 and upregulated members of the LXR pathway. Moreover, LXR inhibition could partially reverse the effects of TREM2 upregulation on electrophysiological activities. CONCLUSIONS We demonstrate that TREM2 has the potential to alleviate WMI following TBI, possibly through the DHCR24/LXR pathway in microglia.
Collapse
Affiliation(s)
- Zhao Li
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Emergency DepartmentChengdu First People's HospitalChengduChina
| | - Shenghui Yu
- Emergency DepartmentChengdu First People's HospitalChengduChina
| | - Lin Li
- Department of NeurosurgeryChongqing University Cancer HospitalChongqingChina
| | - Chao Zhou
- Emergency DepartmentChengdu First People's HospitalChengduChina
| | - Lin Wang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Department of NeurosurgeryNanchong Central HospitalThe Second Clinical Medical College of North Sichuan Medical CollegeNanchongChina
| | - Shuang Tang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Department of NeurosurgerySuining Central HospitalSuiningChina
| | - Nina Gu
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Zhaosi Zhang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Zhijian Huang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Hong Chen
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Wei Tang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Yingwen Wang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xiaomin Yang
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Xiaochuan Sun
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Jin Yan
- Department of NeurosurgeryThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
5
|
Peper CJ, Kilgore MD, Jiang Y, Xiu Y, Xia W, Wang Y, Shi M, Zhou D, Dumont AS, Wang X, Liu N. Tracing the path of disruption: 13C isotope applications in traumatic brain injury-induced metabolic dysfunction. CNS Neurosci Ther 2024; 30:e14693. [PMID: 38544365 PMCID: PMC10973562 DOI: 10.1111/cns.14693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/25/2024] [Accepted: 03/12/2024] [Indexed: 05/14/2024] Open
Abstract
Cerebral metabolic dysfunction is a critical pathological hallmark observed in the aftermath of traumatic brain injury (TBI), as extensively documented in clinical investigations and experimental models. An in-depth understanding of the bioenergetic disturbances that occur following TBI promises to reveal novel therapeutic targets, paving the way for the timely development of interventions to improve patient outcomes. The 13C isotope tracing technique represents a robust methodological advance, harnessing biochemical quantification to delineate the metabolic trajectories of isotopically labeled substrates. This nuanced approach enables real-time mapping of metabolic fluxes, providing a window into the cellular energetic state and elucidating the perturbations in key metabolic circuits. By applying this sophisticated tool, researchers can dissect the complexities of bioenergetic networks within the central nervous system, offering insights into the metabolic derangements specific to TBI pathology. Embraced by both animal studies and clinical research, 13C isotope tracing has bolstered our understanding of TBI-induced metabolic dysregulation. This review synthesizes current applications of isotope tracing and its transformative potential in evaluating and addressing the metabolic sequelae of TBI.
Collapse
Affiliation(s)
- Charles J. Peper
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Mitchell D. Kilgore
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Yinghua Jiang
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Yuwen Xiu
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Winna Xia
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Yingjie Wang
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Mengxuan Shi
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Di Zhou
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Aaron S. Dumont
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Xiaoying Wang
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
- Neuroscience Program, Tulane Brain InstituteTulane UniversityNew OrleansLouisianaUSA
| | - Ning Liu
- Clinical Neuroscience Research Center, Departments of Neurosurgery and NeurologyTulane University School of MedicineNew OrleansLouisianaUSA
- Neuroscience Program, Tulane Brain InstituteTulane UniversityNew OrleansLouisianaUSA
- Tulane University Translational Sciences InstituteNew OrleansLouisianaUSA
| |
Collapse
|
6
|
Wang J, Wang L, Wu Q, Cai Y, Cui C, Yang M, Sun B, Mao L, Wang Y. Interleukin-4 Modulates Neuroinflammation by Inducing Phenotypic Transformation of Microglia Following Subarachnoid Hemorrhage. Inflammation 2024; 47:390-403. [PMID: 37898992 PMCID: PMC10799105 DOI: 10.1007/s10753-023-01917-z] [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: 08/31/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 10/31/2023]
Abstract
Neuroinflammation, a key pathological feature following subarachnoid hemorrhage (SAH), can be therapeutically targeted by inhibiting microglia M1 polarization and promoting phenotypic transformation to M2 microglia. Interleukin-4 (IL-4) is a pleiotropic cytokine known to its regulation of physiological functions of the central nervous system (CNS) and mediate neuroinflammatory processes. However, its specific role in neuroinflammation and microglia responses following SAH remains unexplored. In this investigation, we established both in vivo and in vitro SAH models and employed a comprehensive array of assessments, including ELISA, neurofunctional profiling, immunofluorescence staining, qRT-PCR, determination of phagocytic capacity, and RNA-Seq analyses. The findings demonstrate an elevated expression of IL-4 within cerebrospinal fluid (CSF) subsequent to SAH. Furthermore, exogenous administration of IL-4 ameliorates post-SAH neurofunctional deficits, attenuates cellular apoptosis, fosters M2 microglia phenotype conversion, and mitigates neuroinflammatory responses. The RNA-Seq analysis signifies that IL-4 governs the modulation of neuroinflammation in microglia within an in vitro SAH model through intricate cascades of signaling pathways, encompassing interactions between cytokines and cytokine receptors. These discoveries not only augment comprehension of the neuropathogenesis associated with post-SAH neuroinflammation but also present novel therapeutic targets for the management thereof.
Collapse
Affiliation(s)
- Jing Wang
- Medical College of Qingdao University, Qingdao, Shandong, 266021, China
- Institute for Neurological Research, School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, The Second Affiliated Hospital, Taian, Shandong, 271000, China
| | - Lili Wang
- Institute for Neurological Research, School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, The Second Affiliated Hospital, Taian, Shandong, 271000, China
| | - Qingjian Wu
- Department of Emergency, Jining No. 1 People's Hospital, No. 6, Jiankang Road, Jining, Shandong Province, 272011, China
| | - Yichen Cai
- Institute for Neurological Research, School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, The Second Affiliated Hospital, Taian, Shandong, 271000, China
| | - Chengfu Cui
- Cheeloo College of Medicine, Shandong University, Jinan, 250100, Shandong, China
| | - Ming Yang
- Department of Ultrasonic Diagnosis and Treatment, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Baoliang Sun
- Medical College of Qingdao University, Qingdao, Shandong, 266021, China.
- Institute for Neurological Research, School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, The Second Affiliated Hospital, Taian, Shandong, 271000, China.
| | - Leilei Mao
- Institute for Neurological Research, School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, The Second Affiliated Hospital, Taian, Shandong, 271000, China.
| | - Yuan Wang
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
| |
Collapse
|
7
|
Wang J, Zhu M, Sun J, Feng L, Yang M, Sun B, Mao L. Gene therapy of adeno-associated virus (AAV) vectors in preclinical models of ischemic stroke. CNS Neurosci Ther 2023; 29:3725-3740. [PMID: 37551863 PMCID: PMC10651967 DOI: 10.1111/cns.14392] [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: 06/09/2023] [Revised: 07/15/2023] [Accepted: 07/27/2023] [Indexed: 08/09/2023] Open
Abstract
Stroke has been associated with devastating clinical outcomes, with current treatment strategies proving largely ineffective. Therefore, there is a need to explore alternative treatment options for addressing post-stroke functional deficits. Gene therapy utilizing adeno-associated viruses (AAVs) as a critical gene vector delivering genes to the central nervous system (CNS) gene delivery has emerged as a promising approach for treating various CNS diseases. This review aims to provide an overview of the biological characteristics of AAV vectors and the therapeutic advancements observed in preclinical models of ischemic stroke. The study further investigates the potential of manipulating AAV vectors in preclinical applications, emphasizing the challenges and prospects in the selection of viral vectors, drug delivery strategies, immune reactions, and clinical translation.
Collapse
Affiliation(s)
- Jing Wang
- Medical College of Qingdao UniversityQingdaoChina
- Institute for Neurological Research, The Second Affiliated HospitalSchool of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical SciencesTaianChina
| | - Mengna Zhu
- Institute for Neurological Research, The Second Affiliated HospitalSchool of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical SciencesTaianChina
| | - Jingyi Sun
- Department of Spinal SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanChina
| | - Lina Feng
- Institute for Neurological Research, The Second Affiliated HospitalSchool of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical SciencesTaianChina
| | - Mingfeng Yang
- Institute for Neurological Research, The Second Affiliated HospitalSchool of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical SciencesTaianChina
| | - Baoliang Sun
- Medical College of Qingdao UniversityQingdaoChina
- Institute for Neurological Research, The Second Affiliated HospitalSchool of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical SciencesTaianChina
| | - Leilei Mao
- Institute for Neurological Research, The Second Affiliated HospitalSchool of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical SciencesTaianChina
| |
Collapse
|
8
|
Wang Z, Nie X, Gao F, Tang Y, Ma Y, Zhang Y, Gao Y, Yang C, Ding J, Wang X. Increasing brain N-acetylneuraminic acid alleviates hydrocephalus-induced neurological deficits. CNS Neurosci Ther 2023; 29:3183-3198. [PMID: 37222223 PMCID: PMC10580356 DOI: 10.1111/cns.14253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 03/27/2023] [Accepted: 04/01/2023] [Indexed: 05/25/2023] Open
Abstract
AIMS This metabolomic study aimed to evaluate the role of N-acetylneuraminic acid (Neu5Ac) in the neurological deficits of normal pressure hydrocephalus (NPH) and its potential therapeutic effect. METHODS We analyzed the metabolic profiles of NPH using cerebrospinal fluid with multivariate and univariate statistical analyses in a set of 42 NPH patients and 38 controls. We further correlated the levels of differential metabolites with severity-related clinical parameters, including the normal pressure hydrocephalus grading scale (NPHGS). We then established kaolin-induced hydrocephalus in mice and treated them using N-acetylmannosamine (ManNAc), a precursor of Neu5Ac. We examined brain Neu5Ac, astrocyte polarization, demyelination, and neurobehavioral outcomes to explore its therapeutic effect. RESULTS Three metabolites were significantly altered in NPH patients. Only decreased Neu5Ac levels were correlated with NPHGS scores. Decreased brain Neu5Ac levels have been observed in hydrocephalic mice. Increasing brain Neu5Ac by ManNAc suppressed the activation of astrocytes and promoted their transition from A1 to A2 polarization. ManNAc also attenuated the periventricular white matter demyelination and improved neurobehavioral outcomes in hydrocephalic mice. CONCLUSION Increasing brain Neu5Ac improved the neurological outcomes associated with the regulation of astrocyte polarization and the suppression of demyelination in hydrocephalic mice, which may be a potential therapeutic strategy for NPH.
Collapse
Affiliation(s)
- Zhangyang Wang
- Department of Neurology, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Xiaoqun Nie
- CAS Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant SciencesChinese Academy of Sciences (CAS)ShanghaiChina
| | - Fang Gao
- CAS Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant SciencesChinese Academy of Sciences (CAS)ShanghaiChina
| | - Yanmin Tang
- Department of Neurology, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Yuanyuan Ma
- Department of Neurology, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Yiying Zhang
- Department of Neurology, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Yanqin Gao
- Department of the State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Chen Yang
- CAS Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant SciencesChinese Academy of Sciences (CAS)ShanghaiChina
| | - Jing Ding
- Department of Neurology, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Xin Wang
- Department of Neurology, Zhongshan HospitalFudan UniversityShanghaiChina
- Department of the State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
| |
Collapse
|
9
|
Datta S, Lin F, Jones LD, Pingle SC, Kesari S, Ashili S. Traumatic brain injury and immunological outcomes: the double-edged killer. Future Sci OA 2023; 9:FSO864. [PMID: 37228857 PMCID: PMC10203904 DOI: 10.2144/fsoa-2023-0037] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/20/2023] [Indexed: 05/27/2023] Open
Abstract
Traumatic brain injury (TBI) is a significant cause of mortality and morbidity worldwide resulting from falls, car accidents, sports, and blast injuries. TBI is characterized by severe, life-threatening consequences due to neuroinflammation in the brain. Contact and collision sports lead to higher disability and death rates among young adults. Unfortunately, no therapy or drug protocol currently addresses the complex pathophysiology of TBI, leading to the long-term chronic neuroinflammatory assaults. However, the immune response plays a crucial role in tissue-level injury repair. This review aims to provide a better understanding of TBI's immunobiology and management protocols from an immunopathological perspective. It further elaborates on the risk factors, disease outcomes, and preclinical studies to design precisely targeted interventions for enhancing TBI outcomes.
Collapse
Affiliation(s)
- Souvik Datta
- Rhenix Lifesciences, 237 Arsha Apartments, Kalyan Nagar, Hyderabad, TG 500038, India
| | - Feng Lin
- CureScience, 5820 Oberlin Drive #202, San Diego, CA 92121, USA
| | | | | | - Santosh Kesari
- Saint John's Cancer Institute, Santa Monica, CA 90404, USA
| | | |
Collapse
|
10
|
Takenaka T, Ohnishi Y, Yamamoto M, Setoyama D, Kishima H. Glycolytic System in Axons Supplement Decreased ATP Levels after Axotomy of the Peripheral Nerve. eNeuro 2023; 10:ENEURO.0353-22.2023. [PMID: 36894321 PMCID: PMC10035771 DOI: 10.1523/eneuro.0353-22.2023] [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: 08/30/2022] [Revised: 02/04/2023] [Accepted: 02/25/2023] [Indexed: 03/11/2023] Open
Abstract
Wallerian degeneration (WD) occurs in the early stages of numerous neurologic disorders, and clarifying WD pathology is crucial for the advancement of neurologic therapies. ATP is acknowledged as one of the key pathologic substances in WD. The ATP-related pathologic pathways that regulate WD have been defined. The elevation of ATP levels in axon contributes to delay WD and protects axons. However, ATP is necessary for the active processes to proceed WD, given that WD is stringently managed by auto-destruction programs. But little is known about the bioenergetics during WD. In this study, we made sciatic nerve transection models for GO-ATeam2 knock-in rats and mice. We presented the spatiotemporal ATP distribution in the injured axons with in vivo ATP imaging systems, and investigated the metabolic source of ATP in the distal nerve stump. A gradual decrease in ATP levels was observed before the progression of WD. In addition, the glycolytic system and monocarboxylate transporters (MCTs) were activated in Schwann cells following axotomy. Interestingly, in axons, we found the activation of glycolytic system and the inactivation of the tricarboxylic acid (TCA) cycle. Glycolytic inhibitors, 2-deoxyglucose (2-DG) and MCT inhibitors, a-cyano-4-hydroxycinnamic acid (4-CIN) decreased ATP and enhanced WD progression, whereas mitochondrial pyruvate carrier (MPC) inhibitors (MSDC-0160) did not change. Finally, ethyl pyruvate (EP) increased ATP levels and delayed WD. Together, our findings suggest that glycolytic system, both in Schwann cells and axons, is the main source of maintaining ATP levels in the distal nerve stump.
Collapse
Affiliation(s)
- Tomofumi Takenaka
- Department of neurosurgery, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
- Department of Research Promotion and Management, National Cerebral and Cardiovascular Center, Osaka, 564-8565, Japan
| | - Yuichiro Ohnishi
- Department of Research Promotion and Management, National Cerebral and Cardiovascular Center, Osaka, 564-8565, Japan
- Department of Neurosurgery, Osaka Gyoumeikan Hospital, Osaka, 554-0012, Japan
| | - Masamichi Yamamoto
- Department of Research Promotion and Management, National Cerebral and Cardiovascular Center, Osaka, 564-8565, Japan
| | - Daiki Setoyama
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Haruhiko Kishima
- Department of neurosurgery, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| |
Collapse
|
11
|
Sezer C, Zırh S, Gokten M, Sezer A, Acıkalın R, Bilgin E, Zırh EB. Neuroprotective Effects of Milrinone on Acute Traumatic Brain Injury. World Neurosurg 2023; 170:e558-e567. [PMID: 36403936 DOI: 10.1016/j.wneu.2022.11.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Traumatic brain injury is still an important health problem worldwide. Traumatic brain injury not only causes direct mechanical damage to the brain but also induces biochemical changes that lead to secondary nerve cell loss. In this study, we investigated the neuroprotective effect of milrinone after traumatic brain injury (TBI) in a rat model. METHODS Forty male Wistar albino rats, were used. Rats were divided into 4 groups: 1) sham, 2) TBI, 3) TBI + Ringers, and 4) TBI + Milrinone. In group 1 (sham), only craniotomy was performed. In group 2 (TBI), TBI was performed after craniotomy. In group 3 (TBI + Ringer), TBI was performed after craniotomy and intraperitoneal Ringers solution was given immediately afterward. Group 4 (TBI + Milrinone), TBI was performed after craniotomy, and milrinone was given 1.0 mg/kg milrinone intraperitoneally directly (0.5 mg/kg milrinone intraperitoneally again 24 hours, 48 hours, and 72 hours after trauma). Tests were performed for neurological and neurobehavioral functions. Immunohistochemistry and histopathology studies were performed. RESULTS In group 4 compared with group 2 and group 3 groups, tests for neurological functions and neurobehavioral functions were significantly better. In the milrinone treatment used in group 4, plasma and brain tissue tumor necrosis factor, 8-OH 2-deoxyguanosine , and interleukin 6 levels were significantly decreased, and increased plasma and tissue IL-10 levels were detected. Histopathological spinal cord injury and apoptotic index increased in groups 2 and 3, while significantly decreasing in group 4. CONCLUSIONS This study shows for the first time that the anti-inflammatory, antioxidant and antiapoptotic properties of milrinone may be neuroprotective after TBI.
Collapse
Affiliation(s)
- Can Sezer
- Department of Neurosurgery, University of Health Sciences, Adana City Training and Research Hospital, Adana, Turkey.
| | - Selim Zırh
- Department of Histology, Binali Yıldırım University, Erzincan, Turkey
| | - Murat Gokten
- Department of Neurosurgery, Corlu State Hospital, Tekirdag, Turkey
| | - Aykut Sezer
- Department of Neurosurgery, Dr. Ersin Arslan Training and Research Hospital, Gaziantep, Turkey
| | - Rıdvan Acıkalın
- Department of Neurosurgery, Medical Park Mersin Hospital, Mersin, Turkey
| | - Emre Bilgin
- Department of Neurosurgery, University of Health Sciences, Adana City Training and Research Hospital, Adana, Turkey
| | - Elham Bahador Zırh
- Department of Histology, TOBB University of Economics and Technology, Ankara, Turkey
| |
Collapse
|
12
|
Zhang Y, Lan J, Zhao D, Ruan C, Zhou J, Tan H, Bao Y. Netrin-1 upregulates GPX4 and prevents ferroptosis after traumatic brain injury via the UNC5B/Nrf2 signaling pathway. CNS Neurosci Ther 2022; 29:216-227. [PMID: 36468399 PMCID: PMC9804081 DOI: 10.1111/cns.13997] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/04/2022] [Accepted: 09/09/2022] [Indexed: 12/12/2022] Open
Abstract
AIM We aimed to investigate the regulatory role of Netrin-1 (NTN1) in ferroptosis after traumatic brain injury (TBI) in mice. METHODS We assessed the expression pattern of NTN1 by RT-PCR, western blot, and immunofluorescence after establishing the TBI model in mice. After treatment with NTN1 shRNA or recombinant NTN1, we determined the biochemical and morphological changes associated with ferroptosis and netrin-1-related pathways. We used Nissl staining to assess lesion volume and Morris water maze and beam-walking test to evaluate ethological manifestation. RESULTS The mRNA and protein levels of NTN1 were upregulated after TBI. The application of NTN1 shRNA increased the number of FJB positive cells, malondialdehyde (MDA), and reactive oxygen species (ROSs) levels. However, the application of NTN1 recombinant had the opposite effect. Furthermore, knockdown or inhibition of GPX4, Nrf2, and UNC5B counteracted the effects of NTN1 recombinant. Intravenous injection of NTN1 recombinant reduced neuronal loss after CCI and improved motor and cognitive function. CONCLUSION NTN1 had a neuroprotective effect after TBI and inhibited ferroptosis via activating the UNC5B/Nrf2 pathway. These findings may provide potential therapeutic strategies for TBI.
Collapse
Affiliation(s)
- Yuanda Zhang
- Department of Neurosurgery, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Jin Lan
- Department of Neurosurgery, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Dongxu Zhao
- Department of Neurosurgery, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Cijie Ruan
- Department of Neurosurgery, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Jue Zhou
- Department of Neurosurgery, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Haoyuan Tan
- Department of Neurosurgery, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Yinghui Bao
- Department of Neurosurgery, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
13
|
Gao X, Zhang H, Peng Z, Zhuang Z, Li W. Elevated Level of Cerebrospinal Fluid Pyruvate Dehydrogenase Kinase 4 Is a Predictive Biomarker of Clinical Outcome after Subarachnoid Hemorrhage. Brain Sci 2022; 12:1507. [PMID: 36358433 PMCID: PMC9688583 DOI: 10.3390/brainsci12111507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/24/2022] [Accepted: 11/04/2022] [Indexed: 07/06/2024] Open
Abstract
Subarachnoid hemorrhage (SAH) is a central nervous system disease with high mortality and morbidity. Some independent factors valuable for prognosis prediction in patients with SAH are still lacking. In our earlier study, we found that PDK4 exerts a protective effect after SAH, primarily by reducing oxidative stress and neuronal death via the ROS/ASK1/p38 signaling pathway. Therefore, we investigated the changes in the level of pyruvate dehydrogenase kinase 4 (PDK4) in patients after subarachnoid hemorrhage (SAH) and analyzed the value of the cerebrospinal fluid (CSF) PDK4 level in predicting the prognoses of patients with SAH after interventional embolization surgery. Some knee arthritis subjects who needed surgery were recruited as a control group. The results showed that PDK4 expression was elevated in the CSF of SAH patients compared with that of controls. PDK4 levels in CSF (OR = 4.525; 95% CI: 1.135-18.038; p = 0.032), time to surgery (OR = 0.795; 95% CI: 0.646-0.977; p = 0.029), and initial GCS scores (OR = 2.758; 95% CI: 0.177-43.106; p = 0.469) were independent prognostic risk factors for SAH patients after surgery. The receiver operating characteristic (ROC) curve showed PDK4 levels in CSF had a higher predictive value. Thus, PDK4 in CSF could be an independent prognostic risk factor for SAH patients after surgery. PDK4 has the potential to serve as a new therapeutic target and biomarker for use in the diagnosis of SAH severity and the prediction of recovery.
Collapse
Affiliation(s)
- Xuan Gao
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300333, China
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Huasheng Zhang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Zheng Peng
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Zong Zhuang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Wei Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| |
Collapse
|
14
|
Fan R, Wang L, Botchway BOA, Zhang Y, Liu X. Protective role of ethyl pyruvate in spinal cord injury by inhibiting the high mobility group box-1/toll-like receptor4/nuclear factor-kappa B signaling pathway. Front Mol Neurosci 2022; 15:1013033. [PMID: 36187352 PMCID: PMC9524569 DOI: 10.3389/fnmol.2022.1013033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Spinal cord injury (SCI) is a high incident rate of central nervous system disease that usually causes paralysis below the injured level. The occurrence of chronic inflammation with the axonal regeneration difficulties are the underlying barriers for the recovery of SCI patients. Current studies have paid attention to controlling the instigative and developmental process of neuro-inflammation. Ethyl pyruvate, as a derivative of pyruvate, has strong anti-inflammatory and neuroprotective functions. Herein, we reviewed the recent studies of ethyl pyruvate and high mobility group box-1 (HMGB1). We think HMGB1 that is one of the main nuclear protein mediators to cause an inflammatory response. This protein induces astrocytic activation, and promotes glial scar formation. Interestingly, ethyl pyruvate has potent inhibitory effects on HMGB1 protein, as it inhibits chronic inflammatory response by modulating the HMGB1/TLR4/NF-κB signaling pathway. This paper discusses the potential mechanism of ethyl pyruvate in inhibiting chronic inflammation after SCI. Ethyl pyruvate can be a prospective therapeutic agent for SCI.
Collapse
Affiliation(s)
- Ruihua Fan
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, China
- School of Life Sciences, Shaoxing University, Shaoxing, China
| | - Lvxia Wang
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, China
- School of Life Sciences, Shaoxing University, Shaoxing, China
| | | | - Yong Zhang
- School of Life Sciences, Shaoxing University, Shaoxing, China
| | - Xuehong Liu
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, China
- School of Life Sciences, Shaoxing University, Shaoxing, China
- *Correspondence: Xuehong Liu, ; orcid.org/0000-0003-4325-6762
| |
Collapse
|
15
|
Chen Q, Cai Y, Zhu X, Wang J, Gao F, Yang M, Mao L, Zhang Z, Sun B. Edaravone Dexborneol Treatment Attenuates Neuronal Apoptosis and Improves Neurological Function by Suppressing 4-HNE-Associated Oxidative Stress After Subarachnoid Hemorrhage. Front Pharmacol 2022; 13:848529. [PMID: 35529450 PMCID: PMC9068884 DOI: 10.3389/fphar.2022.848529] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
Edaravone dexborneol is a novel neuroprotective drug that comprises edaravone and (+)-borneol in a 4:1 ratio. Phase II and III studies have demonstrated that Chinese patients treated with edaravone dexborneol within 48 h of AIS onset have better functional outcomes than those treated with edaravone alone. However, the effect of edaravone dexborneol on subarachnoid hemorrhage (SAH) has not yet been elucidated. This study aimed to investigate the therapeutic effects of edaravone dexborneol on SAH-induced brain injury and long-term behavioral deficits and to explore the possible mechanisms. The experimental rat SAH model was induced by an intraluminal puncture of the left middle cerebral artery (MCA). Edaravone dexborneol or edaravone at a clinical dose was infused into the tail vein for 3 days post-SAH surgery. Behavioral outcomes were assessed by a modified Garcia scoring system and rotarod, foot-fault, and corner tests. Immunofluorescence, Western blot, and ELISA methods were used to evaluate neuronal damage and oxidative stress. Our results showed that a post-SAH therapeutic regimen with edaravone dexborneol helped improve neurological function up to 21 days after SAH surgery and demonstrated a greater beneficial effect than edaravone alone, accompanied by an obvious inhibition of neuronal apoptosis in the CA1 hippocampus and basal cortex regions. Mechanistically, edaravone dexborneol not only suppressed the lipid peroxidation product malondialdehyde (MDA) but also improved the total antioxidant capability (TAC) 3 days after SAH. Notably, edaravone dexborneol treatment significantly inhibited the expression of another lipid peroxidation product, 4-hydroxynonenal (4-HNE), in the CA1 hippocampus and basal cortex, which are vital participants in the process of neuronal oxidative damage and death after SAH because of their acute cytotoxicity. Together, our results demonstrate that edaravone dexborneol confers neuroprotection and stabilizes long-term behavioral ability after SAH injury, possibly by suppressing 4-HNE-associated oxidative stress. These results may help develop new clinical strategies for SAH treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Leilei Mao
- The Second Affiliated Hospital, Brain Science Institute, School of Basic Medical Sciences of Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Zongyong Zhang
- The Second Affiliated Hospital, Brain Science Institute, School of Basic Medical Sciences of Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Baoliang Sun
- The Second Affiliated Hospital, Brain Science Institute, School of Basic Medical Sciences of Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| |
Collapse
|
16
|
Wei P, Wang K, Luo C, Huang Y, Misilimu D, Wen H, Jin P, Li C, Gong Y, Gao Y. Cordycepin confers long-term neuroprotection via inhibiting neutrophil infiltration and neuroinflammation after traumatic brain injury. J Neuroinflammation 2021; 18:137. [PMID: 34130727 PMCID: PMC8207641 DOI: 10.1186/s12974-021-02188-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 05/30/2021] [Indexed: 02/06/2023] Open
Abstract
Background The secondary injury caused by traumatic brain injury (TBI), especially white matter injury (WMI), is highly sensitive to neuroinflammation, which further leads to unfavored long-term outcomes. Although the cross-talk between the three active events, immune cell infiltration, BBB breakdown, and proinflammatory microglial/macrophage polarization, plays a role in the vicious cycle, its mechanisms are not fully understood. It has been reported that cordycepin, an extract from Cordyceps militaris, can inhibit TBI-induced neuroinflammation although the long-term effects of cordycepin remain unknown. Here, we report our investigation of cordycepin’s long-term neuroprotective function and its underlying immunological mechanism. Methods TBI mice model was established with a controlled cortical impact (CCI) method. Cordycepin was intraperitoneally administered twice daily for a week. Neurological outcomes were assessed by behavioral tests, including grid walking test, cylinder test, wire hang test, and rotarod test. Immunofluorescence staining, transmission electron microscopy, and electrophysiology recording were employed to assess histological and functional lesions. Quantitative-PCR and flow cytometry were used to detect neuroinflammation. The tracers of Sulfo-NHS-biotin and Evans blue were assessed for the blood-brain barrier (BBB) leakage. Western blot and gelatin zymography were used to analyze protein activity or expression. Neutrophil depletion in vivo was performed via using Ly6G antibody intraperitoneal injection. Results Cordycepin administration ameliorated long-term neurological deficits and reduced neuronal tissue loss in TBI mice. Meanwhile, the long-term integrity of white matter was also preserved, which was revealed in multiple dimensions, such as morphology, histology, ultrastructure, and electrical conductivity. Cordycepin administration inhibited microglia/macrophage pro-inflammatory polarization and promoted anti-inflammatory polarization after TBI. BBB breach was attenuated by cordycepin administration at 3 days after TBI. Cordycepin suppressed the activities of MMP-2 and MMP-9 and the neutrophil infiltration at 3 days after TBI. Moreover, neutrophil depletion provided a cordycepin-like effect, and cordycepin administration united with neutrophil depletion did not show a benefit of superposition. Conclusions The long-term neuroprotective function of cordycepin via suppressing neutrophil infiltration after TBI, thereby preserving BBB integrity and changing microglia/macrophage polarization. These findings provide significant clinical potentials to improve the quality of life for TBI patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02188-x.
Collapse
Affiliation(s)
- Pengju Wei
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Ke Wang
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Chen Luo
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yichen Huang
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Dilidaer Misilimu
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Huimei Wen
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Peng Jin
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Chuhua Li
- School of Life Science, South China Normal University, Guangzhou, 510631, China
| | - Ye Gong
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| | - Yanqin Gao
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
17
|
Lu F, Cao J, Su Q, Zhao Q, Wang H, Guan W, Zhou W. Recent Advances in Fluorescence Imaging of Traumatic Brain Injury in Animal Models. Front Mol Biosci 2021; 8:660993. [PMID: 34124151 PMCID: PMC8194861 DOI: 10.3389/fmolb.2021.660993] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the top three specific neurological disorders, requiring reliable, rapid, and sensitive imaging of brain vessels, tissues, and cells for effective diagnosis and treatment. Although the use of medical imaging such as computed tomography (CT) and magnetic resonance imaging (MRI) for the TBI detection is well established, the exploration of novel TBI imaging techniques is of great interest. In this review, recent advances in fluorescence imaging for the diagnosis and evaluation of TBI are summarized and discussed in three sections: imaging of cerebral vessels, imaging of brain tissues and cells, and imaging of TBI-related biomarkers. Design strategies for probes and labels used in TBI fluorescence imaging are also described in detail to inspire broader applications. Moreover, the multimodal TBI imaging platforms combining MRI and fluorescence imaging are also briefly introduced. It is hoped that this review will promote more studies on TBI fluorescence imaging, and enable its use for clinical diagnosis as early as possible, helping TBI patients get better treatment and rehabilitation.
Collapse
Affiliation(s)
- Fei Lu
- Department of Rehabilitation Medicine, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, China
| | - Jiating Cao
- Department of Chemistry, Capital Normal University, Beijing, China
| | - Qinglun Su
- Department of Rehabilitation Medicine, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, China
| | - Qin Zhao
- Department of Rehabilitation Medicine, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, China
| | - Huihai Wang
- Department of Rehabilitation Medicine, The First People's Hospital of Lianyungang, The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, China
| | - Weijiang Guan
- State Key Laboratory of Chemical Resource Engineering, College of Chemistry, Beijing University of Chemical Technology, Beijing, China
| | - Wenjuan Zhou
- Department of Chemistry, Capital Normal University, Beijing, China
| |
Collapse
|
18
|
Chen C, Li P. Neurovascular unit protection—novel therapeutic targets and strategies. CNS Neurosci Ther 2021. [PMCID: PMC7804891 DOI: 10.1111/cns.13588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Chen Chen
- Department of Anesthesiology State Key Laboratory of Oncogenes and Related Genes Shanghai Cancer InstituteRenji HospitalSchool of Medicine Shanghai Jiaotong University Shanghai China
| | - Peiying Li
- Department of Anesthesiology State Key Laboratory of Oncogenes and Related Genes Shanghai Cancer InstituteRenji HospitalSchool of Medicine Shanghai Jiaotong University Shanghai China
| |
Collapse
|