1
|
Jiang T, Li Y, Liu H, Sun Y, Zhang H, Zhang Q, Tang S, Niu X, Du H, Yu Y, Yue H, Guo Y, Chen Y, Xu F. Blood-brain barrier disruption and neuroinflammation in the hippocampus of a cardiac arrest porcine model: Single-cell RNA sequencing analysis. Neural Regen Res 2026; 21:742-755. [PMID: 40146000 DOI: 10.4103/nrr.nrr-d-24-01269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202602000-00043/figure1/v/2025-05-05T160104Z/r/image-tiff Global brain ischemia and neurological deficit are consequences of cardiac arrest that lead to high mortality. Despite advancements in resuscitation science, our limited understanding of the cellular and molecular mechanisms underlying post-cardiac arrest brain injury have hindered the development of effective neuroprotective strategies. Previous studies primarily focused on neuronal death, potentially overlooking the contributions of non-neuronal cells and intercellular communication to the pathophysiology of cardiac arrest-induced brain injury. To address these gaps, we hypothesized that single-cell transcriptomic analysis could uncover previously unidentified cellular subpopulations, altered cell communication networks, and novel molecular mechanisms involved in post-cardiac arrest brain injury. In this study, we performed a single-cell transcriptomic analysis of the hippocampus from pigs with ventricular fibrillation-induced cardiac arrest at 6 and 24 hours following the return of spontaneous circulation, and from sham control pigs. Sequencing results revealed changes in the proportions of different cell types, suggesting post-arrest disruption in the blood-brain barrier and infiltration of neutrophils. These results were validated through western blotting, quantitative reverse transcription-polymerase chain reaction, and immunofluorescence staining. We also identified and validated a unique subcluster of activated microglia with high expression of S100A8, which increased over time following cardiac arrest. This subcluster simultaneously exhibited significant M1/M2 polarization and expressed key functional genes related to chemokines and interleukins. Additionally, we revealed the post-cardiac arrest dysfunction of oligodendrocytes and the differentiation of oligodendrocyte precursor cells into oligodendrocytes. Cell communication analysis identified enhanced post-cardiac arrest communication between neutrophils and microglia that was mediated by neutrophil-derived resistin, driving pro-inflammatory microglial polarization. Our findings provide a comprehensive single-cell map of the post-cardiac arrest hippocampus, offering potential novel targets for neuroprotection and repair following cardiac arrest.
Collapse
Affiliation(s)
- Tangxing Jiang
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yaning Li
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Hehui Liu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yijun Sun
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Huidan Zhang
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Qirui Zhang
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Shuyao Tang
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Xu Niu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Han Du
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yinxia Yu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Hongwei Yue
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yunyun Guo
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yuguo Chen
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Feng Xu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- Medical and Pharmaceutical Basic Research Innovation Center of Emergency and Critical Care Medicine, China's Ministry of Education, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
2
|
Chen J, Li Y, Quan X, Chen J, Han Y, Yang L, Zhou M, Mok GSP, Wang R, Zhao Y. Utilizing engineered extracellular vesicles as delivery vectors in the management of ischemic stroke: a special outlook on mitochondrial delivery. Neural Regen Res 2025; 20:2181-2198. [PMID: 39101653 PMCID: PMC11759020 DOI: 10.4103/nrr.nrr-d-24-00243] [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: 02/28/2024] [Revised: 06/03/2024] [Accepted: 06/22/2024] [Indexed: 08/06/2024] Open
Abstract
Ischemic stroke is a secondary cause of mortality worldwide, imposing considerable medical and economic burdens on society. Extracellular vesicles, serving as natural nano-carriers for drug delivery, exhibit excellent biocompatibility in vivo and have significant advantages in the management of ischemic stroke. However, the uncertain distribution and rapid clearance of extracellular vesicles impede their delivery efficiency. By utilizing membrane decoration or by encapsulating therapeutic cargo within extracellular vesicles, their delivery efficacy may be greatly improved. Furthermore, previous studies have indicated that microvesicles, a subset of large-sized extracellular vesicles, can transport mitochondria to neighboring cells, thereby aiding in the restoration of mitochondrial function post-ischemic stroke. Small extracellular vesicles have also demonstrated the capability to transfer mitochondrial components, such as proteins or deoxyribonucleic acid, or their sub-components, for extracellular vesicle-based ischemic stroke therapy. In this review, we undertake a comparative analysis of the isolation techniques employed for extracellular vesicles and present an overview of the current dominant extracellular vesicle modification methodologies. Given the complex facets of treating ischemic stroke, we also delineate various extracellular vesicle modification approaches which are suited to different facets of the treatment process. Moreover, given the burgeoning interest in mitochondrial delivery, we delved into the feasibility and existing research findings on the transportation of mitochondrial fractions or intact mitochondria through small extracellular vesicles and microvesicles to offer a fresh perspective on ischemic stroke therapy.
Collapse
Affiliation(s)
- Jiali Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Yiyang Li
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Xingping Quan
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Jinfen Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Yan Han
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Li Yang
- Department of Pharmacy, Hunan Provincial People’s Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Manfei Zhou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Greta Seng Peng Mok
- Department of Electrical and Computer Engineering, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Ruibing Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao Special Administrative Region, China
| | - Yonghua Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macao Special Administrative Region, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macao Special Administrative Region, China
| |
Collapse
|
3
|
Chen J, Li C, Hong J, Zhao F, Zhang J, Yang M, Liang S, Wen H. High-Frequency repetitive transcranial magnetic stimulation enhances white matter integrity in a rat model of ischemic stroke: A diffusion tensor imaging study using tract-based spatial statistics. Neuroimage 2025; 311:121204. [PMID: 40222496 DOI: 10.1016/j.neuroimage.2025.121204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/15/2025] Open
Abstract
Ischemic stroke leads to white matter damage and neurological deficits. Previous studies have revealed that high-frequency repetitive transcranial magnetic stimulation (HF-rTMS) has beneficial effects on white matter reorganization and neurological recovery after stroke. However, the characteristics of poststroke white matter repair after treatment with HF-rTMS remain unclear. Therefore, this study used diffusion tensor imaging (DTI) to investigate the impact of HF-rTMS on white matter integrity following middle cerebral artery occlusion (MCAO) in a rat model. The modified neurological severity score (mNSS) and T2-weighted imaging data were used to assess neurological function and infarct size. We used a tract-based spatial statistics (TBSS) approach to analyze changes in fractional anisotropy (FA) across various white matter tracts. Furthermore, we performed Luxol fast blue (LFB) staining and transmission electron microscopy (TEM) to detect white matter and myelin damage. The results revealed that compared with the tMCAO group, the tMCAO+rTMS group presented a significant decrease in infarct size and the mNSS, as well as significantly greater FA values, mostly in the left external capsule, left internal capsule, left optic tract, left deep cerebral white matter, left stria terminalis and right external capsule. The LFB staining and electron microscopy results are consistent with the DTI results. These findings suggest that HF-rTMS contributes to the recovery of white matter integrity and neurological function. This study underscores the importance of HF-rTMS as a noninvasive intervention for enhancing poststroke neurological recovery by improving white matter integrity.
Collapse
Affiliation(s)
- Jiemei Chen
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China; Department of Rehabilitation Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - Chao Li
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Jiena Hong
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Fei Zhao
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Jiantao Zhang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Man Yang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Shengxiang Liang
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province 350122, China; Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian Province 350122, China.
| | - Hongmei Wen
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China.
| |
Collapse
|
4
|
Wang T, Wang X, Liu S, Li M, Wan K, Zheng J, Liao K, Wang J, Zou K, Wang L, Xu H, Lei W, Chen G, Li W. Transcription Factor-Based Gene Therapy Enables Functional Repair of Rat Following Chronic Ischemic Stroke. CNS Neurosci Ther 2025; 31:e70448. [PMID: 40401537 PMCID: PMC12096174 DOI: 10.1111/cns.70448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/21/2025] [Accepted: 05/07/2025] [Indexed: 05/23/2025] Open
Abstract
OBJECTIVE In vivo transcription factor (TF) -mediated gene therapy through astrocyte-to-neuron (AtN) conversion has shown therapeutic effects on rodent and non-human primate cortical ischemic injury in the subacute phase. However, in the clinic, subcortical regions including striatum as well as white matter are vulnerable regions of stroke, with millions of patients beyond subacute phase. In this study, we investigate whether TF-mediated AtN conversion therapy can be extended to treat chronic-phase ischemic stroke involving subcortical regions (e.g., striatum) and white matter, beyond cortical injuries. METHODS Rat middle cerebral artery occlusion (MCAO)-like models were established to induce broad ischemic injuries including cortical and striatal regions. Then multiple rounds of TF-mediated gene therapy treatments through adeno-associated virus (AAV) system to cover the large-scaled infarct areas were conducted in the chronic phase of the stroke models. Magnetic resonance imaging (MRI), [18F] FDG-PET/CT, behavioral tests, immunohistochemistry and bulk-RNA seq were applied to evaluate the AtN conversion, tissue repair and functional recovery. RESULTS Our results revealed that administrated in the chronic phase of ischemic stroke, TF-mediated gene therapy can efficiently regenerate new neurons in both cortical and striatal regions, and promote tissue repair in both grey and white matter. Compared with single round of AAV administration, multiple rounds of treatment regenerated more neurons and led to a significant functional recovery. CONCLUSIONS Our study demonstrates that TF-mediated gene therapy has a broad therapeutic time window and can be applied multiple rounds to treat severe ischemic stroke, making it an attractive therapeutic intervention in the chronic phase after stroke, when current approaches are largely ineffective.
Collapse
Affiliation(s)
- Tao Wang
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Xu Wang
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Shanggong Liu
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Menglei Li
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Kaiying Wan
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Jiajun Zheng
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Kai Liao
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non‐Human Primate Research, GHM Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Jinyu Wang
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Kaiming Zou
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Lu Wang
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non‐Human Primate Research, GHM Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Hao Xu
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non‐Human Primate Research, GHM Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Wenliang Lei
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsJinan UniversityGuangzhouChina
- Department of Nuclear Medicine and PET/CT‐MRI CenterThe First Affiliated Hospital of Jinan University & Institute of Molecular and Functional Imaging, Jinan UniversityGuangzhouChina
| | - Gong Chen
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsJinan UniversityGuangzhouChina
- Department of Nuclear Medicine and PET/CT‐MRI CenterThe First Affiliated Hospital of Jinan University & Institute of Molecular and Functional Imaging, Jinan UniversityGuangzhouChina
| | - Wen Li
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsJinan UniversityGuangzhouChina
- Department of Nuclear Medicine and PET/CT‐MRI CenterThe First Affiliated Hospital of Jinan University & Institute of Molecular and Functional Imaging, Jinan UniversityGuangzhouChina
| |
Collapse
|
5
|
Wang J, Gao Y, Wang B, Zhang C, Yuan Y, Xu R, Ji H, Zhang X. Low-Intensity Pulsed Ultrasound Promotes Oligodendrocyte Maturation and Remyelination by Down-regulating the Interleukin-17A/Notch1 Signaling Pathway in Mice with Ischemic Stroke. RESEARCH (WASHINGTON, D.C.) 2025; 8:0676. [PMID: 40290135 PMCID: PMC12022504 DOI: 10.34133/research.0676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025]
Abstract
Increasing evidence indicates that oligodendrocyte (OL) numbers and myelin as a dynamic cellular compartment perform a key role in the maintenance of neuronal function. Inhibiting white matter (WM) demyelination or promoting remyelination has garnered interest for its potential therapeutic strategy against ischemic stroke. Our previous work has shown that low-intensity pulsed ultrasound (LIPUS) could improve stroke recovery. However, it is unclear whether LIPUS can maintain WM integrity early after stroke or promote late WM repair. This study evaluated the efficacy of LIPUS on WM repair and long-term neurologic recovery after stroke. Male adult C57BL/6 mice underwent a focal cerebral ischemia model and were randomized to receive ultrasound stimulation (30 min once daily for 14 days). The effect of LIPUS on sensorimotor function was assessed by modified neurological severity score, rotarod test, grip strength test, and gait analysis up to 28 days after stroke. We found that ischemic stroke-induced WM damage was severe on day 7 and partially recovered on day 28. LIPUS prevented neuronal and oligodendrocyte progenitor cell (OPC) death during the acute phase of stroke (d7), protected WM integrity, and reduced brain atrophy and tissue damage during the recovery phase (d28). To further confirm the effect of LIPUS on remyelination, we assessed the proliferation and differentiation of OPCs. We found that LIPUS did not increase the number of OPCs (PDGFRα+ or NG2+), but markedly increased the number of newly produced mature OLs (APC+) and myelin protein levels. Mechanistically, LIPUS may promote OL maturation and remyelination by down-regulating the interleukin-17A/Notch1 signaling pathway. In summary, LIPUS can protect OLs and neurons early after stroke and promote long-term WM repair and functional recovery. LIPUS will be a viable strategy for the treatment of ischemic stroke in the future.
Collapse
Affiliation(s)
- Jingjing Wang
- Department of Neurology,
Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, China
- Key Laboratory of Clinical Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Yuxiao Gao
- Department of Neurology,
Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, China
- Key Laboratory of Clinical Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Bin Wang
- Department of Neurology,
Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, China
- Key Laboratory of Clinical Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Cong Zhang
- Department of Neurology,
Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, China
- Key Laboratory of Clinical Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Yi Yuan
- School of Electrical Engineering,
Yanshan University, Qinhuangdao 066004, China
- Key Laboratory of Intelligent Rehabilitation and Neuromodulation of Hebei Province,
Yanshan University, Qinhuangdao 066004, China
| | - Renhao Xu
- Department of Neurology,
Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, China
- Key Laboratory of Clinical Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Hui Ji
- Department of Neurology,
Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, China
- Key Laboratory of Clinical Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Xiangjian Zhang
- Department of Neurology,
Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, China
- Key Laboratory of Clinical Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| |
Collapse
|
6
|
Bernier LP, Hefendehl JK, Scott RW, Tung LW, Lewis CA, Soliman H, Simm S, Dissing-Olesen L, Hofmann J, Guo D, DeMeglio M, Rossi FM, Underhill TM, MacVicar BA. Brain pericytes and perivascular fibroblasts are stromal progenitors with dual functions in cerebrovascular regeneration after stroke. Nat Neurosci 2025; 28:517-535. [PMID: 39962273 DOI: 10.1038/s41593-025-01872-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 12/18/2024] [Indexed: 03/12/2025]
Abstract
Functional revascularization is key to stroke recovery and requires remodeling and regeneration of blood vessels around which is located the brain's only stromal compartment. Stromal progenitor cells (SPCs) are critical for tissue regeneration following injury in many organs, yet their identity in the brain remains elusive. Here we show that the perivascular niche of brain SPCs includes pericytes, venular smooth muscle cells and perivascular fibroblasts that together help cerebral microvasculature regenerate following experimental stroke. Ischemic injury triggers amplification of pericytes and perivascular fibroblasts in the infarct region where they associate with endothelial cells inside a reactive astrocyte border. Fate-tracking of Hic1+ SPCs uncovered a transient functional and transcriptional phenotype of stroke-activated pericytes and perivascular fibroblasts. Both populations of these cells remained segregated, displaying distinct angiogenic and fibrogenic profiles. Therefore, pericytes and perivascular fibroblasts are distinct subpopulations of SPCs in the adult brain that coordinate revascularization and scar formation after injury.
Collapse
Affiliation(s)
- Louis-Philippe Bernier
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, British Colombia, Canada.
| | - Jasmin K Hefendehl
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, British Colombia, Canada
- Goethe University Frankfurt, Institute for Cell biology and Neuroscience, Buchmann Institute for Molecular Life Sciences, Frankfurt am Main, Germany
| | - R Wilder Scott
- University of British Columbia, Biomedical Research Centre, Vancouver, British Colombia, Canada
- Dept. of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Colombia, Canada
| | - Lin Wei Tung
- University of British Columbia, Biomedical Research Centre, Vancouver, British Colombia, Canada
| | - Coral-Ann Lewis
- University of British Columbia, Biomedical Research Centre, Vancouver, British Colombia, Canada
| | - Hesham Soliman
- University of British Columbia, Biomedical Research Centre, Vancouver, British Colombia, Canada
| | - Stefan Simm
- University Medicine Greifswald, Institute for Bioinformatics, Greifswald, Germany
| | - Lasse Dissing-Olesen
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, British Colombia, Canada
| | - Jan Hofmann
- Goethe University Frankfurt, Institute for Cell biology and Neuroscience, Buchmann Institute for Molecular Life Sciences, Frankfurt am Main, Germany
| | - David Guo
- University of British Columbia, Biomedical Research Centre, Vancouver, British Colombia, Canada
| | - Murphy DeMeglio
- Goethe University Frankfurt, Institute for Cell biology and Neuroscience, Buchmann Institute for Molecular Life Sciences, Frankfurt am Main, Germany
| | - Fabio M Rossi
- University of British Columbia, Biomedical Research Centre, Vancouver, British Colombia, Canada
| | - T Michael Underhill
- University of British Columbia, Biomedical Research Centre, Vancouver, British Colombia, Canada
- Dept. of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Colombia, Canada
| | - Brian A MacVicar
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, British Colombia, Canada.
| |
Collapse
|
7
|
Jiang H, Zhang C, Lin M, Yin Y, Deng S, Liu W, Zhuo B, Tian G, Du Y, Meng Z. Deciphering the mechanistic impact of acupuncture on the neurovascular unit in acute ischemic stroke: Insights from basic research in a narrative review. Ageing Res Rev 2024; 101:102536. [PMID: 39384155 DOI: 10.1016/j.arr.2024.102536] [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: 08/18/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Ischemic stroke(IS), a severe acute cerebrovascular disease, not only imposes a heavy economic burden on society but also presents numerous challenges in treatment. During the acute phase, while thrombolysis and thrombectomy serve as primary treatments, these approaches are restricted by a narrow therapeutic window. During rehabilitation, commonly used neuroprotective agents struggle with their low drug delivery efficiency and inadequate preclinical testing, and the long-term pharmacological and toxicity effects of nanomedicines remain undefined. Meanwhile, acupuncture as a therapeutic approach is widely acknowledged for its effectiveness in treating IS and has been recommended by the World Health Organization (WHO) as an alternative and complementary therapy, even though its exact mechanisms remain unclear. This review aims to summarize the known mechanisms of acupuncture and electroacupuncture (EA) in the treatment of IS. Research shows that acupuncture treatment mainly protects the neurovascular unit through five mechanisms: 1) reducing neuronal apoptosis and promoting neuronal repair and proliferation; 2) maintaining the integrity of the blood-brain barrier (BBB); 3) inhibiting the overactivation and polarization imbalance of microglia; 4) regulating the movement of vascular smooth muscle (VSM) cells; 5) promoting the proliferation of oligodendrocyte precursors. Through an in-depth analysis, this review reveals the multi-level, multi-dimensional impact of acupuncture treatment on the neurovascular unit (NVU) following IS, providing stronger evidence and a theoretical basis for its clinical application.
Collapse
Affiliation(s)
- Hailun Jiang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Chao Zhang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Mengxuan Lin
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yu Yin
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shizhe Deng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Wei Liu
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Bifang Zhuo
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Guang Tian
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yuzheng Du
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Zhihong Meng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
8
|
Martín-Lopez G, Mallavibarrena PR, Villa-Gonzalez M, Vidal N, Pérez-Alvarez MJ. The dynamics of oligodendrocyte populations following permanent ischemia promotes long-term spontaneous remyelination of damaged area. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167270. [PMID: 38823461 DOI: 10.1016/j.bbadis.2024.167270] [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: 12/04/2023] [Revised: 05/10/2024] [Accepted: 05/25/2024] [Indexed: 06/03/2024]
Abstract
Stroke is a major public health concern, with limited clinically approved interventions available to enhance sensorimotor recovery beyond reperfusion. Remarkably, spontaneous recovery is observed in certain stroke patients, suggesting the existence of a brain self-repair mechanism not yet fully understood. In a rat model of permanent cerebral ischemia, we described an increase in oligodendrocytes expressing 3RTau in damaged area. Considering that restoration of myelin integrity ameliorates symptoms in many neurodegenerative diseases, here we hypothesize that this cellular response could trigger remyelination. Our results revealed after ischemia an early recruitment of OPCs to damaged area, followed by their differentiation into 3RTau+ pre-myelinating cells and subsequent into remyelinating oligodendrocytes. Using rat brain slices and mouse primary culture we confirmed the presence of 3RTau in pre-myelinating and a subset of mature oligodendrocytes. The myelin status analysis confirmed long-term remyelination in the damaged area. Postmortem samples from stroke subjects showed a reduction in oligodendrocytes, 3RTau+ cells, and myelin complexity in subcortical white matter. In conclusion, the dynamics of oligodendrocyte populations after ischemia reveals a spontaneous brain self-repair mechanism which restores the functionality of neuronal circuits long-term by remyelination of damaged area. This is evidenced by the improvement of sensorimotor functions in ischemic rats. A deep understanding of this mechanism could be valuable in the search for alternative oligodendrocyte-based, therapeutic interventions to reduce the effects of stroke.
Collapse
Affiliation(s)
- Gerardo Martín-Lopez
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Paula R Mallavibarrena
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Mario Villa-Gonzalez
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Noemi Vidal
- Departamento de Patología, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, 08907 Barcelona, Spain
| | - Maria José Pérez-Alvarez
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain; Instituto Universitario de Biología Molecular (IUBM), Universidad Autónoma de Madrid, Centro de Biología Molecular Severo Ochoa (CBM), Departamento de Neuropatología Molecular UAM-CSIC, 28049 Madrid, Spain.
| |
Collapse
|
9
|
Wu CYC, Zhang Y, Xu L, Huang Z, Zou P, Clemons GA, Li C, Citadin CT, Zhang Q, Lee RHC. The role of serum/glucocorticoid-regulated kinase 1 in brain function following cerebral ischemia. J Cereb Blood Flow Metab 2024; 44:1145-1162. [PMID: 38235747 PMCID: PMC11179613 DOI: 10.1177/0271678x231224508] [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: 08/15/2023] [Revised: 11/30/2023] [Accepted: 12/05/2023] [Indexed: 01/19/2024]
Abstract
Cardiopulmonary arrest (CA) is a major cause of death/disability in the U.S. with poor prognosis and survival rates. Current therapeutic challenges are physiologically complex because they involve hypoperfusion (decreased cerebral blood flow), neuroinflammation, and mitochondrial dysfunction. We previously discovered novel serum/glucocorticoid-regulated kinase 1 (SGK1) is highly expressed in brain of neurons that are susceptible to ischemia (hippocampus and cortex). We inhibited SGK1 and utilized pharmacological (specific inhibitor, GSK650394) and neuron-specific genetic approaches (shRNA) in rodent models of CA to determine if SGK1 is responsible for hypoperfusion, neuroinflammation, mitochondrial dysfunctional, and neurological deficits after CA. Inhibition of SGK1 alleviated cortical hypoperfusion and neuroinflammation (via Iba1, GFAP, and cytokine array). Treatment with GSK650394 enhanced mitochondrial function (via Seahorse respirometry) in the hippocampus 3 and 7 days after CA. Neuronal injury (via MAP2, dMBP, and Golgi staining) in the hippocampus and cortex was observed 7 days after CA but ameliorated with SGK1-shRNA. Moreover, SGK1 mediated neuronal injury by regulating the Ndrg1-SOX10 axis. Finally, animals subjected to CA exhibited learning/memory, motor, and anxiety deficits after CA, whereas SGK1 inhibition via SGK1-shRNA improved neurocognitive function. The present study suggests the fundamental roles of SGK1 in brain circulation and neuronal survival/death in cerebral ischemia-related diseases.
Collapse
Affiliation(s)
- Celeste Yin-Chieh Wu
- Stroke Center for Research, Louisiana State University Health, Shreveport, LA, USA
- Department of Neurology, Louisiana State University Health, Shreveport, LA, USA
| | - Yulan Zhang
- Stroke Center for Research, Louisiana State University Health, Shreveport, LA, USA
- Department of Neurology, Louisiana State University Health, Shreveport, LA, USA
| | - Li Xu
- Stroke Center for Research, Louisiana State University Health, Shreveport, LA, USA
- Department of Neurology, Louisiana State University Health, Shreveport, LA, USA
| | - Zhihai Huang
- Stroke Center for Research, Louisiana State University Health, Shreveport, LA, USA
- Department of Neurology, Louisiana State University Health, Shreveport, LA, USA
| | - Peibin Zou
- Stroke Center for Research, Louisiana State University Health, Shreveport, LA, USA
- Department of Neurology, Louisiana State University Health, Shreveport, LA, USA
| | - Garrett A Clemons
- Department of Cellular Biology and Anatomy, Louisiana State University Health, Shreveport, LA, USA
| | - Chun Li
- Stroke Center for Research, Louisiana State University Health, Shreveport, LA, USA
- Department of Neurology, Louisiana State University Health, Shreveport, LA, USA
| | - Cristiane T Citadin
- Department of Cellular Biology and Anatomy, Louisiana State University Health, Shreveport, LA, USA
| | - Quanguang Zhang
- Stroke Center for Research, Louisiana State University Health, Shreveport, LA, USA
- Department of Neurology, Louisiana State University Health, Shreveport, LA, USA
| | - Reggie Hui-Chao Lee
- Stroke Center for Research, Louisiana State University Health, Shreveport, LA, USA
- Department of Neurology, Louisiana State University Health, Shreveport, LA, USA
| |
Collapse
|
10
|
Valcárcel-Hernández V, Mayerl S, Guadaño-Ferraz A, Remaud S. Thyroid hormone action in adult neurogliogenic niches: the known and unknown. Front Endocrinol (Lausanne) 2024; 15:1347802. [PMID: 38516412 PMCID: PMC10954857 DOI: 10.3389/fendo.2024.1347802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/08/2024] [Indexed: 03/23/2024] Open
Abstract
Over the last decades, thyroid hormones (THs) signaling has been established as a key signaling cue for the proper maintenance of brain functions in adult mammals, including humans. One of the most fascinating roles of THs in the mature mammalian brain is their ability to regulate adult neurogliogenic processes. In this respect, THs control the generation of new neuronal and glial progenitors from neural stem cells (NSCs) as well as their final differentiation and maturation programs. In this review, we summarize current knowledge on the cellular organization of adult rodent neurogliogenic niches encompassing well-established niches in the subventricular zone (SVZ) lining the lateral ventricles, the hippocampal subgranular zone (SGZ), and the hypothalamus, but also less characterized niches in the striatum and the cerebral cortex. We then discuss critical questions regarding how THs availability is regulated in the respective niches in rodents and larger mammals as well as how modulating THs availability in those niches interferes with lineage decision and progression at the molecular, cellular, and functional levels. Based on those alterations, we explore the novel therapeutic avenues aiming at harnessing THs regulatory influences on neurogliogenic output to stimulate repair processes by influencing the generation of either new neurons (i.e. Alzheimer's, Parkinson's diseases), oligodendrocytes (multiple sclerosis) or both (stroke). Finally, we point out future challenges, which will shape research in this exciting field in the upcoming years.
Collapse
Affiliation(s)
- Victor Valcárcel-Hernández
- Laboratory Molecular Physiology and Adaptation, CNRS UMR 7221, Department Adaptations of Life, Muséum National d’Histoire Naturelle, Paris, France
| | - Steffen Mayerl
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ana Guadaño-Ferraz
- Department of Neurological Diseases and Aging, Instituto de Investigaciones Biomédicas Sols-Morreale, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Sylvie Remaud
- Laboratory Molecular Physiology and Adaptation, CNRS UMR 7221, Department Adaptations of Life, Muséum National d’Histoire Naturelle, Paris, France
| |
Collapse
|
11
|
Yang X, Chang L, Liu Z, Geng X, Wang R, Yin X, Fan W, Zhao BQ. Neddylation in the chronically hypoperfused corpus callosum: MLN4924 reduces blood-brain barrier injury via ERK5/KLF2 signaling. Exp Neurol 2024; 371:114587. [PMID: 37914067 DOI: 10.1016/j.expneurol.2023.114587] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/13/2023] [Accepted: 10/25/2023] [Indexed: 11/03/2023]
Abstract
Blood-brain barrier (BBB) breakdown and cerebrovascular dysfunction may contribute to the pathology in white matter lesions and consequent cognitive decline caused by cerebral hypoperfusion. Neddylation is the process of attaching a ubiquitin-like molecule NEDD8 (neuronal precursor cell-expressed developmentally downregulated protein 8) to specific targets. By modifying protein substrates, neddylation plays critical roles in various important biological processes. However, whether neddylation influences the pathogenesis of hypoperfused brain remains unclear. In the present study, cerebral hypoperfusion-induced white matter lesions were produced by bilateral common carotid artery stenosis in mice. The function of the neddylation pathway, BBB integrity, cerebrovascular dysfunction, myelin density in the corpus callosum and cognitive function were determined. We show that NEDD8 conjugation aberrantly amplified in microvascular endothelium in the corpus callosum following cerebral hypoperfusion. MLN4924, a small-molecule inhibitor of NEDD8-activating enzyme currently in clinical trials, preserved BBB integrity, attenuated glial activation and enhanced oligodendrocyte differentiation, and reduced hypoperfusion-induced white matter lesions in the corpus callosum and thus improved cognitive performance via inactivating cullin-RING E3 ligase (CRL). Administration of MLN4924 caused the accumulation of ERK5 and KLF2. The ERK5 inhibitor BIX 02189, down-regulated MLN4924-induced activation of KLF2 and reversed MLN4924-mediated increase in pericyte coverage and junctional proteins. Furthermore, BIX 02189 blocked MLN4924-afforded protection against BBB disruption and white matter lesions in the corpus callosum. Collectively, our results revealed that neddylation impairs vascular function and thus exacerbated the pathology of hypoperfused brain and that inhibition of neddylation with MLN4924 may offer novel therapeutic opportunities for cerebral hypoperfusion-associated cognitive impairment.
Collapse
Affiliation(s)
- Xing Yang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Luping Chang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Zhongwang Liu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Xue Geng
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Ranran Wang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Xuhui Yin
- Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Wenying Fan
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| | - Bing-Qiao Zhao
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
12
|
Wang J, Ding X, Li C, Huang C, Ke C, Xu C, Wan C. Early exercise intervention promotes myelin repair in the brains of ischemic rats by inhibiting the MEK/ERK pathway. Transl Neurosci 2024; 15:20220335. [PMID: 38511170 PMCID: PMC10951688 DOI: 10.1515/tnsci-2022-0335] [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: 11/11/2023] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 03/22/2024] Open
Abstract
Our previous studies have shown that early exercise intervention after stroke increases neural activity and synaptic plasticity and promotes the recovery of nerve fiber bundle integrity in the brain. However, the effect of exercise on the repair of myelin in the brain and the related mechanism are still unclear. In this study, we randomly divided the rats into three groups. Before and after 28 days of intervention, body weight, nerve function, the infarct size, white matter fiber bundle integrity, and nerve myelin structure and function were observed by measuring body weight, analysis of modified neurological severity score, CatWalk gait analysis, MRI, luxol fast blue staining, immunofluorescence, and transmission electron microscopy. Changes in the expression of proteins in the MEK/ERK pathway were assessed. The results showed that early exercise intervention resulted in neurological recovery, decreased the infarct volume and increased nerve fiber integrity, the myelin coverage area, myelin basic protein (MBP) fluorescence intensity expression, and myelin thickness. Furthermore, the expression level of MBP was significantly increased after early exercise intervention, while the expression levels of p-MEK1/2 and p-ERK1/2 were significantly reduced. In the cell study, MBP expression levels were significantly higher in the oxygen and glucose deprivation and administration group.In summary, early exercise intervention after stroke can promote myelin repair by inhibiting the MEK/ERK signaling pathway.
Collapse
Affiliation(s)
- Junyi Wang
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Xinyu Ding
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Chen Li
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Chuan Huang
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Changkai Ke
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Chunlei Xu
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Chunxiao Wan
- Department of Physical Medicine and Rehabilitation, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| |
Collapse
|
13
|
Zhang Z, Shu X, Cao Q, Xu L, Wang Z, Li C, Xia S, Shao P, Bao X, Sun L, Xu Y, Xu Y. Compound from Magnolia officinalis Ameliorates White Matter Injury by Promoting Oligodendrocyte Maturation in Chronic Cerebral Ischemia Models. Neurosci Bull 2023; 39:1497-1511. [PMID: 37291477 PMCID: PMC10533772 DOI: 10.1007/s12264-023-01068-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 02/20/2023] [Indexed: 06/10/2023] Open
Abstract
Chronic cerebral hypoperfusion leads to white matter injury (WMI), which subsequently causes neurodegeneration and even cognitive impairment. However, due to the lack of treatment specifically for WMI, novel recognized and effective therapeutic strategies are urgently needed. In this study, we found that honokiol and magnolol, two compounds derived from Magnolia officinalis, significantly facilitated the differentiation of primary oligodendrocyte precursor cells (OPCs) into mature oligodendrocytes, with a more prominent effect of the former compound. Moreover, our results demonstrated that honokiol treatment improved myelin injury, induced mature oligodendrocyte protein expression, attenuated cognitive decline, promoted oligodendrocyte regeneration, and inhibited astrocytic activation in the bilateral carotid artery stenosis model. Mechanistically, honokiol increased the phosphorylation of serine/threonine kinase (Akt) and mammalian target of rapamycin (mTOR) by activating cannabinoid receptor 1 during OPC differentiation. Collectively, our study indicates that honokiol might serve as a potential treatment for WMI in chronic cerebral ischemia.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Xin Shu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Qian Cao
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Lushan Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Zibu Wang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Chenggang Li
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Shengnan Xia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China
| | - Pengfei Shao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China
| | - Xinyu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China
| | - Liang Sun
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Yuhao Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China.
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School and State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China.
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China.
- Nanjing Neurology Medical Center, Nanjing, 210008, China.
| |
Collapse
|
14
|
Lu W, Chen Z, Wen J. The role of RhoA/ROCK pathway in the ischemic stroke-induced neuroinflammation. Biomed Pharmacother 2023; 165:115141. [PMID: 37437375 DOI: 10.1016/j.biopha.2023.115141] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 07/14/2023] Open
Abstract
It is widely known that ischemic stroke is the prominent cause of death and disability. To date, neuroinflammation following ischemic stroke represents a complex event, which is an essential process and affects the prognosis of both experimental stroke animals and stroke patients. Intense neuroinflammation occurring during the acute phase of stroke contributes to neuronal injury, BBB breakdown, and worse neurological outcomes. Inhibition of neuroinflammation may be a promising target in the development of new therapeutic strategies. RhoA is a small GTPase protein that activates a downstream effector, ROCK. The up-regulation of RhoA/ROCK pathway possesses important roles in promoting the neuroinflammation and mediating brain injury. In addition, nuclear factor-kappa B (NF-κB) is another vital regulator of ischemic stroke-induced neuroinflammation through regulating the functions of microglial cells and astrocytes. After stroke onset, the microglial cells and astrocytes are activated and undergo the morphological and functional changes, thereby deeply participate in a complicated neuroinflammation cascade. In this review, we focused on the relationship among RhoA/ROCK pathway, NF-κB and glial cells in the neuroinflammation following ischemic stroke to reveal new strategies for preventing the intense neuroinflammation.
Collapse
Affiliation(s)
- Weizhuo Lu
- Medical Branch, Hefei Technology College, Hefei, China
| | - Zhiwu Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
15
|
Mihailova V, Stoyanova II, Tonchev AB. Glial Populations in the Human Brain Following Ischemic Injury. Biomedicines 2023; 11:2332. [PMID: 37760773 PMCID: PMC10525766 DOI: 10.3390/biomedicines11092332] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 09/29/2023] Open
Abstract
There is a growing interest in glial cells in the central nervous system due to their important role in maintaining brain homeostasis under physiological conditions and after injury. A significant amount of evidence has been accumulated regarding their capacity to exert either pro-inflammatory or anti-inflammatory effects under different pathological conditions. In combination with their proliferative potential, they contribute not only to the limitation of brain damage and tissue remodeling but also to neuronal repair and synaptic recovery. Moreover, reactive glial cells can modulate the processes of neurogenesis, neuronal differentiation, and migration of neurons in the existing neural circuits in the adult brain. By discovering precise signals within specific niches, the regulation of sequential processes in adult neurogenesis holds the potential to unlock strategies that can stimulate the generation of functional neurons, whether in response to injury or as a means of addressing degenerative neurological conditions. Cerebral ischemic stroke, a condition falling within the realm of acute vascular disorders affecting the circulation in the brain, stands as a prominent global cause of disability and mortality. Extensive investigations into glial plasticity and their intricate interactions with other cells in the central nervous system have predominantly relied on studies conducted on experimental animals, including rodents and primates. However, valuable insights have also been gleaned from in vivo studies involving poststroke patients, utilizing highly specialized imaging techniques. Following the attempts to map brain cells, the role of various transcription factors in modulating gene expression in response to cerebral ischemia is gaining increasing popularity. Although the results obtained thus far remain incomplete and occasionally ambiguous, they serve as a solid foundation for the development of strategies aimed at influencing the recovery process after ischemic brain injury.
Collapse
Affiliation(s)
- Victoria Mihailova
- Department of Anatomy and Cell Biology, Faculty of Medicine, Medical University Varna, 9000 Varna, Bulgaria; (I.I.S.); (A.B.T.)
| | | | | |
Collapse
|
16
|
Werner L, Gliem M, Rychlik N, Pavic G, Reiche L, Kirchhoff F, Silva Oliveira Junior M, Gruchot J, Meuth SG, Küry P, Göttle P. A Novel Ex Vivo Model to Study Therapeutic Treatments for Myelin Repair following Ischemic Damage. Int J Mol Sci 2023; 24:10972. [PMID: 37446147 DOI: 10.3390/ijms241310972] [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: 05/31/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Stroke is a major reason for persistent disability due to insufficient treatment strategies beyond reperfusion, leading to oligodendrocyte death and axon demyelination, persistent inflammation and astrogliosis in peri-infarct areas. After injury, oligodendroglial precursor cells (OPCs) have been shown to compensate for myelin loss and prevent axonal loss through the replacement of lost oligodendrocytes, an inefficient process leaving axons chronically demyelinated. Phenotypic screening approaches in demyelinating paradigms revealed substances that promote myelin repair. We established an ex vivo adult organotypic coronal slice culture (OCSC) system to study repair after stroke in a resource-efficient way. Post-photothrombotic OCSCs can be manipulated for 8 d by exposure to pharmacologically active substances testing remyelination activity. OCSCs were isolated from a NG2-CreERT2-td-Tomato knock-in transgenic mouse line to analyze oligodendroglial fate/differentiation and kinetics. Parbendazole boosted differentiation of NG2+ cells and stabilized oligodendroglial fate reflected by altered expression of associated markers PDGFR-α, CC1, BCAS1 and Sox10 and GFAP. In vitro scratch assay and chemical ischemia confirmed the observed effects upon parbendazole treatment. Adult OCSCs represent a fast, reproducible, and quantifiable model to study OPC differentiation competence after stroke. Pharmacological stimulation by means of parbendazole promoted OPC differentiation.
Collapse
Affiliation(s)
- Luisa Werner
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Michael Gliem
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Nicole Rychlik
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Goran Pavic
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Laura Reiche
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, 66424 Homburg, Germany
| | | | - Joel Gruchot
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Peter Göttle
- Department of Neurology, Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany
| |
Collapse
|
17
|
Gómez-Pinedo U, Matías-Guiu JA, Ojeda-Hernandez D, de la Fuente-Martin S, Kamal OMF, Benito-Martin MS, Selma-Calvo B, Montero-Escribano P, Matías-Guiu J. In Vitro Effects of Methylprednisolone over Oligodendroglial Cells: Foresight to Future Cell Therapies. Cells 2023; 12:1515. [PMID: 37296635 PMCID: PMC10252523 DOI: 10.3390/cells12111515] [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: 04/09/2023] [Revised: 05/28/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
The implantation of oligodendrocyte precursor cells may be a useful therapeutic strategy for targeting remyelination. However, it is yet to be established how these cells behave after implantation and whether they retain the capacity to proliferate or differentiate into myelin-forming oligodendrocytes. One essential issue is the creation of administration protocols and determining which factors need to be well established. There is controversy around whether these cells may be implanted simultaneously with corticosteroid treatment, which is widely used in many clinical situations. This study assesses the influence of corticosteroids on the capacity for proliferation and differentiation and the survival of human oligodendroglioma cells. Our findings show that corticosteroids reduce the capacity of these cells to proliferate and to differentiate into oligodendrocytes and decrease cell survival. Thus, their effect does not favour remyelination; this is consistent with the results of studies with rodent cells. In conclusion, protocols for the administration of oligodendrocyte lineage cells with the aim of repopulating oligodendroglial niches or repairing demyelinated axons should not include corticosteroids, given the evidence that the effects of these drugs may undermine the objectives of cell transplantation.
Collapse
Affiliation(s)
- Ulises Gómez-Pinedo
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain; (D.O.-H.); (S.d.l.F.-M.); (O.M.-F.K.); (M.S.B.-M.); (B.S.-C.); (J.M.-G.)
| | - Jordi A. Matías-Guiu
- Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.M.-G.); (P.M.-E.)
| | - Denise Ojeda-Hernandez
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain; (D.O.-H.); (S.d.l.F.-M.); (O.M.-F.K.); (M.S.B.-M.); (B.S.-C.); (J.M.-G.)
| | - Sarah de la Fuente-Martin
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain; (D.O.-H.); (S.d.l.F.-M.); (O.M.-F.K.); (M.S.B.-M.); (B.S.-C.); (J.M.-G.)
| | - Ola Mohamed-Fathy Kamal
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain; (D.O.-H.); (S.d.l.F.-M.); (O.M.-F.K.); (M.S.B.-M.); (B.S.-C.); (J.M.-G.)
| | - Maria Soledad Benito-Martin
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain; (D.O.-H.); (S.d.l.F.-M.); (O.M.-F.K.); (M.S.B.-M.); (B.S.-C.); (J.M.-G.)
| | - Belen Selma-Calvo
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain; (D.O.-H.); (S.d.l.F.-M.); (O.M.-F.K.); (M.S.B.-M.); (B.S.-C.); (J.M.-G.)
| | - Paloma Montero-Escribano
- Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.M.-G.); (P.M.-E.)
| | - Jorge Matías-Guiu
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain; (D.O.-H.); (S.d.l.F.-M.); (O.M.-F.K.); (M.S.B.-M.); (B.S.-C.); (J.M.-G.)
- Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.M.-G.); (P.M.-E.)
| |
Collapse
|
18
|
Lee JH, Kim EJ. A Comprehensive Review of the Effects of Extracorporeal Shock Wave Therapy on Stroke Patients: Balance, Pain, Spasticity. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59050857. [PMID: 37241089 DOI: 10.3390/medicina59050857] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023]
Abstract
Stroke remains a leading cause of disability worldwide, with survivors often experiencing impairments in balance, pain, spasticity, and control that limit their ability to perform daily living activities. Extracorporeal shock wave therapy (ESWT) has emerged as a potential treatment modality to improve these outcomes in stroke patients. This review aims to provide a comprehensive examination of the effects of ESWT on stroke patients, focusing on the theoretical background, balance, pain reduction, muscle spasticity and control, and upper and lower extremities. This study reviewed the use of ESWT in treating balance, pain, and spasticity in stroke patients, focusing on articles published in PubMed between January 2003 and January 2023. Systematic reviews related to stroke were used to provide an overview of stroke, and a total of 33 articles related to balance, pain, and spasticity were selected. ESWT has several shock wave generation methods and application methods, and it has been shown to have positive therapeutic effects on various aspects of rehabilitation for stroke patients, such as improving balance, reducing pain, decreasing muscle spasticity and increasing control, and enhancing functional activities of the upper and lower extremities. The efficacy of ESWT may vary depending on the patient's condition, application method, and treatment area. Therefore, it is important to apply ESWT according to the individual characteristics of each patient in clinical practice to maximize its potential benefits.
Collapse
Affiliation(s)
- Jung-Ho Lee
- Department of Physical Therapy, Kyungdong University, 815, Gyeonhwon-ro, Munmak-eup, Wonju-si 26495, Gang-won-do, Republic of Korea
| | - Eun-Ja Kim
- Department of Physical Therapy, Kyungdong University, 815, Gyeonhwon-ro, Munmak-eup, Wonju-si 26495, Gang-won-do, Republic of Korea
| |
Collapse
|
19
|
Shibahara T, Nakamura K, Wakisaka Y, Shijo M, Yamanaka K, Takashima M, Takaki H, Hidaka M, Kitazono T, Ago T. PDGFR β-positive cell-mediated post-stroke remodeling of fibronectin and laminin α2 for tissue repair and functional recovery. J Cereb Blood Flow Metab 2023; 43:518-530. [PMID: 36514952 PMCID: PMC10063838 DOI: 10.1177/0271678x221145092] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Post-stroke intra-infarct repair promotes peri-infarct neural reorganization leading to functional recovery. Herein, we examined the remodeling of extracellular matrix proteins (ECM) that constitute the intact basal membrane after permanent middle cerebral artery occlusion (pMCAO) in mice. Among ECM, collagen type IV remained localized on small vessel walls surrounding CD31-positive endothelial cells within infarct areas. Fibronectin was gradually deposited from peri-infarct areas to the ischemic core, in parallel with the accumulation of PDGFRβ-positive cells. Cultured PDGFRβ-positive pericytes produced fibronectin, which was enhanced by the treatment with PDGF-BB. Intra-infarct deposition of fibronectin was significantly attenuated in pericyte-deficient Pdgfrb+/-mice. Phagocytic activity of macrophages against myelin debris was significantly enhanced on fibronectin-coated dishes. In contrast, laminin α2, produced by GFAP- and aquaporin 4-positive astrocytes, accumulated strongly in the boundary of peri-infarct areas. Pericyte-conditioned medium increased the expression of laminin α2 in cultured astrocytes, partly through TGFβ1. Laminin α2 increased the differentiation of oligodendrocyte precursor cells into oligodendrocytes and the expression of myelin-associated proteins. Peri-infarct deposition of laminin α2 was significantly reduced in Pdgfrb+/-mice, with attenuated oligodendrogenesis in peri-infarct areas. Collectively, intra-infarct PDGFRβ-positive cells may orchestrate post-stroke remodeling of key ECM that create optimal environments promoting clearance of myelin debris and peri-infarct oligodendrogenesis.
Collapse
Affiliation(s)
- Tomoya Shibahara
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kuniyuki Nakamura
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshinobu Wakisaka
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masahiro Shijo
- Department of Internal Medicine, Fukuoka Dental College Medical and Dental Hospital, Fukuoka, Japan
| | - Kei Yamanaka
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masamitsu Takashima
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hayato Takaki
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaoki Hidaka
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuro Ago
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
20
|
Hu X, Geng P, Zhao X, Wang Q, Liu C, Guo C, Dong W, Jin X. The NG2-glia is a potential target to maintain the integrity of neurovascular unit after acute ischemic stroke. Neurobiol Dis 2023; 180:106076. [PMID: 36921779 DOI: 10.1016/j.nbd.2023.106076] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/07/2023] [Accepted: 03/07/2023] [Indexed: 03/18/2023] Open
Abstract
The neurovascular unit (NVU) plays a critical role in health and disease. In the current review, we discuss the critical role of a class of neural/glial antigen 2 (NG2)-expressing glial cells (NG2-glia) in regulating NVU after acute ischemic stroke (AIS). We first introduce the role of NG2-glia in the formation of NVU during development as well as aging-induced damage to NVU and accompanying NG2-glia change. We then discuss the reciprocal interactions between NG2-glia and the other component cells of NVU, emphasizing the factors that could influence NG2-glia. Damage to the NVU integrity is the pathological basis of edema and hemorrhagic transformation, the most dreaded complication after AIS. The role of NG2-glia in AIS-induced NVU damage and the effect of NG2-glia transplantation on AIS-induced NVU damage are summarized. We next discuss the role of NG2-glia and the effect of NG2-glia transplantation in oligodendrogenesis and white matter repair as well as angiogenesis which is associated with the outcome of the patients after AIS. Finally, we review the current strategies to promote NG2-glia proliferation and differentiation and propose to use the dental pulp stem cells (DPSC)-derived exosome as a promising strategy to reduce AIS-induced injury and promote repair through maintaining the integrity of NVU by regulating endogenous NG2-glia proliferation and differentiation.
Collapse
Affiliation(s)
- Xiaoyan Hu
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Panpan Geng
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Xiaoyun Zhao
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Qian Wang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Changqing Liu
- Department of Neurosurgery, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Chun Guo
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, UK
| | - Wen Dong
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Xinchun Jin
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China; Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China.
| |
Collapse
|
21
|
Fessel J. Supplementary Pharmacotherapy for the Behavioral Abnormalities Caused by Stressors in Humans, Focused on Post-Traumatic Stress Disorder (PTSD). J Clin Med 2023; 12:1680. [PMID: 36836215 PMCID: PMC9967886 DOI: 10.3390/jcm12041680] [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: 01/14/2023] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023] Open
Abstract
Used as a supplement to psychotherapy, pharmacotherapy that addresses all of the known metabolic and genetic contributions to the pathogenesis of psychiatric conditions caused by stressors would require an inordinate number of drugs. Far simpler is to address the abnormalities caused by those metabolic and genetic changes in the cell types of the brain that mediate the behavioral abnormality. Relevant data regarding the changed brain cell types are described in this article and are derived from subjects with the paradigmatic behavioral abnormality of PTSD and from subjects with traumatic brain injury or chronic traumatic encephalopathy. If this analysis is correct, then therapy is required that benefits all of the affected brain cell types; those are astrocytes, oligodendrocytes, synapses and neurons, endothelial cells, and microglia (the pro-inflammatory (M1) subtype requires switching to the anti-inflammatory (M2) subtype). Combinations are advocated using several drugs, erythropoietin, fluoxetine, lithium, and pioglitazone, that benefit all of the five cell types, and that should be used to form a two-drug combination, suggested as pioglitazone with either fluoxetine or lithium. Clemastine, fingolimod, and memantine benefit four of the cell types, and one chosen from those could be added to the two-drug combination to form a three-drug combination. Using low doses of chosen drugs will limit both toxicity and drug-drug interactions. A clinical trial is required to validate both the advocated concept and the choice of drugs.
Collapse
Affiliation(s)
- Jeffrey Fessel
- Department of Medicine, University of California, 2069 Filbert Street, San Francisco, CA 94123, USA
| |
Collapse
|
22
|
Song W, Yao Y, Zhang H, Hao X, Zhou L, Song Z, Wei T, Chi T, Liu P, Ji X, Zou L. Sigma-1 Receptor Activation Improves Oligodendrogenesis and Promotes White-Matter Integrity after Stroke in Mice with Diabetic Mellitus. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28010390. [PMID: 36615583 PMCID: PMC9823930 DOI: 10.3390/molecules28010390] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/26/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023]
Abstract
Diabetes mellitus (DM) is a major risk factor for stroke and exacerbates white-matter damage in focal cerebral ischemia. Our previous study showed that the sigma-1 receptor agonist PRE084 ameliorates bilateral common-carotid-artery occlusion-induced brain damage in mice. However, whether this protective effect can extend to white matter remains unclear. In this study, C57BL/6 mice were treated with high-fat diets (HFDs) combined with streptozotocin (STZ) injection to mimic type 2 diabetes mellitus (T2DM). Focal cerebral ischemia in T2DM mice was established via injection of the vasoconstrictor peptide endothelin-1 (ET-1) into the hippocampus. Three different treatment plans were used in this study. In one plan, 1 mg/kg of PRE084 (intraperitoneally) was administered for 7 d before ET-1 injection; the mice were sacrificed 24 h after ET-1 injection. In another plan, PRE084 treatment was initiated 24 h after ET-1 injection and lasted for 7 d. In the third plan, PRE084 treatment was initiated 24 h after ET-1 injection and lasted for 21 d. The Y-maze, novel object recognition, and passive avoidance tests were used to assess neurobehavioral outcomes. We found no cognitive dysfunction or white-matter damage 24 h after ET-1 injection. However, 7 and 21 d after ET-1 injection, the mice showed significant cognitive impairment and white-matter damage. Only PRE084 treatment for 21 d could improve this white-matter injury; increase axon and myelin density; decrease demyelination; and increase the expressions of myelin regulator 2'-3'-cyclic nucleotide 3'-phosphodiesterase (CNpase) and myelin oligodendrocyte protein (MOG) (which was expressed by mature oligodendrocytes), the number of nerve/glial-antigen 2 (NG2)-positive cells, and the expression of platelet-derived growth factor receptor-alpha (PDGFRα), all of which were expressed by oligodendrocyte progenitor cells in mice with diabetes and focal cerebral ischemia. These results indicate that maybe there was more severe white-matter damage in the focal cerebral ischemia of the diabetic mice than in the mice with normal blood glucose levels. Long-term sigma-1 receptor activation may promote oligodendrogenesis and white-matter functional recovery in patients with stroke and with diabetes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Peng Liu
- Correspondence: (P.L.); (X.J.); (L.Z.); Tel.: +86-24-23986260 (P.L.)
| | - Xuefei Ji
- Correspondence: (P.L.); (X.J.); (L.Z.); Tel.: +86-24-23986260 (P.L.)
| | - Libo Zou
- Correspondence: (P.L.); (X.J.); (L.Z.); Tel.: +86-24-23986260 (P.L.)
| |
Collapse
|
23
|
Liu L, Liu J, Li M, Lyu J, Su W, Feng S, Ji X. Selective brain hypothermia attenuates focal cerebral ischemic injury and improves long-term neurological outcome in aged female mice. CNS Neurosci Ther 2022; 29:129-139. [PMID: 36341958 PMCID: PMC9804044 DOI: 10.1111/cns.14017] [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: 06/24/2022] [Revised: 10/09/2022] [Accepted: 10/20/2022] [Indexed: 11/09/2022] Open
Abstract
AIMS This study aimed to investigate the effects of mild selective brain hypothermia on aged female ischemic mice. METHODS A distal middle cerebral artery occlusion (dMCAO) model was established in aged female mice, who were then subjected to mild selective brain hypothermia immediately after the dMCAO procedure. Neurological behavioral examinations were conducted prior to and up to 35 days post-ischemia. Infarct volume, brain atrophy, pro-inflammation, and anti-inflammation microglia/macrophages phenotype and white matter injury were evaluated by immunofluorescence staining. Correlations between neurological behaviors and histological parameters were evaluated by Pearson product linear regression analysis. RESULTS Sensorimotor and cognitive function tests confirmed the protective effect of mild selective brain hypothermia in elderly female ischemic mice. In addition, hypothermia decreased the infarct volume and brain atrophy induced by focal cerebral ischemia. Furthermore, hypothermia alleviated ischemia-induced short-term and long-term white matter injury, which was correlated with behavioral deficits. Finally, hypothermia suppressed the harmful immunological response by promoting the transformation of pro-inflammatory microglia/macrophages to anti-inflammatory phenotype. This polarization was negatively correlated with neuronal loss and white matter injury. CONCLUSION Mild selective brain hypothermia promoted long-term functional recovery by alleviating white matter damage in an aged female mouse model of ischemia.
Collapse
Affiliation(s)
- Liqiang Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Jia Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Ming Li
- Beijing Institute of Geriatrics, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Junxuan Lyu
- Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Wei Su
- Department of Neurosurgery, Beijing Tsing Hua Chang Gung Hospital, School of Clinical MedicineTsing Hua UniversityBeijingChina
| | - Shejun Feng
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina,Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
24
|
Zhang Z, Lv M, Zhou X, Cui Y. Roles of peripheral immune cells in the recovery of neurological function after ischemic stroke. Front Cell Neurosci 2022; 16:1013905. [PMID: 36339825 PMCID: PMC9634819 DOI: 10.3389/fncel.2022.1013905] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/03/2022] [Indexed: 10/15/2023] Open
Abstract
Stroke is a leading cause of mortality and long-term disability worldwide, with limited spontaneous repair processes occurring after injury. Immune cells are involved in multiple aspects of ischemic stroke, from early damage processes to late recovery-related events. Compared with the substantial advances that have been made in elucidating how immune cells modulate acute ischemic injury, the understanding of the impact of the immune system on functional recovery is limited. In this review, we summarized the mechanisms of brain repair after ischemic stroke from both the neuronal and non-neuronal perspectives, and we review advances in understanding of the effects on functional recovery after ischemic stroke mediated by infiltrated peripheral innate and adaptive immune cells, immune cell-released cytokines and cell-cell interactions. We also highlight studies that advance our understanding of the mechanisms underlying functional recovery mediated by peripheral immune cells after ischemia. Insights into these processes will shed light on the double-edged role of infiltrated peripheral immune cells in functional recovery after ischemic stroke and provide clues for new therapies for improving neurological function.
Collapse
Affiliation(s)
- Zhaolong Zhang
- Department of Interventional Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Mengfei Lv
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, China
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
| | - Xin Zhou
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, China
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
| | - Yu Cui
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, China
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
25
|
Li C, Xie Z, Xing Z, Zhu H, Zhou W, Xie S, Zhang Z, Li MH. The Notch Signaling Pathway Regulates Differentiation of NG2 Cells into Oligodendrocytes in Demyelinating Diseases. Cell Mol Neurobiol 2022; 42:1-11. [PMID: 33826017 PMCID: PMC11421596 DOI: 10.1007/s10571-021-01089-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/29/2021] [Indexed: 12/13/2022]
Abstract
NG2 cells are highly proliferative glial cells that can self-renew or differentiate into oligodendrocytes, promoting remyelination. Following demyelination, the proliferative and differentiation potentials of NG2 cells increase rapidly, enhancing their differentiation into functional myelinating cells. Levels of the transcription factors Olig1 and Olig2 increase during the differentiation of NG2 cells and play important roles in the development and repair of oligodendrocytes. However, the ability to generate new oligodendrocytes is hampered by injury-related factors (e.g., myelin fragments, Wnt and Notch signaling components), leading to failed differentiation and maturation of NG2 cells into oligodendrocytes. Here, we review Notch signaling as a negative regulator of oligodendrocyte differentiation and discuss the extracellular ligands, intracellular pathways, and key transcription factors involved.
Collapse
Affiliation(s)
- Chengcai Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, No. 17 Yongwai Zheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Zhiping Xie
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, No. 17 Yongwai Zheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Zelong Xing
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, No. 17 Yongwai Zheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Huaxin Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, No. 17 Yongwai Zheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Wu Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, No. 17 Yongwai Zheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Shenke Xie
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, No. 17 Yongwai Zheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Zhixiong Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, No. 17 Yongwai Zheng Street, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Mei-Hua Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, No. 17 Yongwai Zheng Street, Nanchang, 330006, Jiangxi, People's Republic of China.
| |
Collapse
|
26
|
Gu L, Sun M, Li R, Tao Y, Luo X, Zhang X, Yuan Y, Xie Z. Microglial pyroptosis: Therapeutic target in secondary brain injury following intracerebral hemorrhage. Front Cell Neurosci 2022; 16:971469. [PMID: 36159393 PMCID: PMC9507402 DOI: 10.3389/fncel.2022.971469] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/25/2022] [Indexed: 11/25/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a major cerebrovascular illness that causes substantial neurological sequelae and dysfunction caused by secondary brain injury (SBI), and there are no effective therapies to mitigate the disability. Microglia, the brain-resident macrophage, participates in the primary inflammatory response, and activation of microglia to an M1-like phenotype largely takes place in the acute phase following ICH. A growing body of research suggests that the pathophysiology of SBI after ICH is mediated by an inflammatory response mediated by microglial-pyroptotic inflammasomes, while inhibiting the activation of microglial pyroptosis could suppress the inflammatory cascade reaction, thus attenuating the brain injury after ICH. Pyroptosis is characterized by rapid plasma membrane disruption, followed by the release of cellular contents and pro-inflammatory mediators. In this review, we outline the molecular mechanism of microglial pyroptosis and summarize the up-to-date evidence of its involvement in the pathological process of ICH, and highlight microglial pyroptosis-targeted strategies that have the potential to cure intracerebral hemorrhage. This review contributes to a better understanding of the function of microglial pyroptosis in ICH and assesses it as a possible therapeutic target.
Collapse
|
27
|
Tarricone G, Carmagnola I, Chiono V. Tissue-Engineered Models of the Human Brain: State-of-the-Art Analysis and Challenges. J Funct Biomater 2022; 13:146. [PMID: 36135581 PMCID: PMC9501967 DOI: 10.3390/jfb13030146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/03/2022] [Accepted: 09/06/2022] [Indexed: 11/26/2022] Open
Abstract
Neurological disorders affect billions of people across the world, making the discovery of effective treatments an important challenge. The evaluation of drug efficacy is further complicated because of the lack of in vitro models able to reproduce the complexity of the human brain structure and functions. Some limitations of 2D preclinical models of the human brain have been overcome by the use of 3D cultures such as cell spheroids, organoids and organs-on-chip. However, one of the most promising approaches for mimicking not only cell structure, but also brain architecture, is currently represented by tissue-engineered brain models. Both conventional (particularly electrospinning and salt leaching) and unconventional (particularly bioprinting) techniques have been exploited, making use of natural polymers or combinations between natural and synthetic polymers. Moreover, the use of induced pluripotent stem cells (iPSCs) has allowed the co-culture of different human brain cells (neurons, astrocytes, oligodendrocytes, microglia), helping towards approaching the central nervous system complexity. In this review article, we explain the importance of in vitro brain modeling, and present the main in vitro brain models developed to date, with a special focus on the most recent advancements in tissue-engineered brain models making use of iPSCs. Finally, we critically discuss achievements, main challenges and future perspectives.
Collapse
Affiliation(s)
- Giulia Tarricone
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- PolitoBioMedLab, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principle in Teaching and Research, Centro 3R, 56122 Pisa, Italy
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy
- Department of Chemistry and Industrial Chemistry, University of Genova, Via Dodecaneso 31, 16146 Genova, Italy
| | - Irene Carmagnola
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- PolitoBioMedLab, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principle in Teaching and Research, Centro 3R, 56122 Pisa, Italy
| | - Valeria Chiono
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- PolitoBioMedLab, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- Interuniversity Center for the Promotion of the 3Rs Principle in Teaching and Research, Centro 3R, 56122 Pisa, Italy
| |
Collapse
|
28
|
Zhang Z, Li X, Zhou H, Zhou J. NG2-glia crosstalk with microglia in health and disease. CNS Neurosci Ther 2022; 28:1663-1674. [PMID: 36000202 PMCID: PMC9532922 DOI: 10.1111/cns.13948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 11/30/2022] Open
Abstract
Neurodegenerative diseases are increasingly becoming a global problem. However, the pathological mechanisms underlying neurodegenerative diseases are not fully understood. NG2‐glia abnormalities and microglia activation are involved in the development and/or progression of neurodegenerative disorders, such as multiple sclerosis, Alzheimer's disease, Parkinson's disease, and cerebrovascular diseases. In this review, we summarize the present understanding of the interaction between NG2‐glia and microglia in physiological and pathological states and discuss unsolved questions concerning their fate and potential fate. First, we introduce the NG2‐glia and microglia in health and disease. Second, we formulate the interaction between NG2‐glia and microglia. NG2‐glia proliferation, migration, differentiation, and apoptosis are influenced by factors released from the microglia. On the other hand, NG2‐glia also regulate microglia actions. We conclude that NG2‐glia and microglia are important immunomodulatory cells in the brain. Understanding the interaction between NG2‐glia and microglia will help provide a novel method to modulate myelination and treat neurodegenerative disorders.
Collapse
Affiliation(s)
- Zuo Zhang
- National Drug Clinical Trial Institution, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Xiaolong Li
- National Drug Clinical Trial Institution, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Hongli Zhou
- National Drug Clinical Trial Institution, Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Jiyin Zhou
- National Drug Clinical Trial Institution, Second Affiliated Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
29
|
Li Q, Lou J, Yang T, Wei Z, Li S, Zhang F. Ischemic Preconditioning Induces Oligodendrogenesis in Mouse Brain: Effects of Nrf2 Deficiency. Cell Mol Neurobiol 2022; 42:1859-1873. [PMID: 33666795 PMCID: PMC11421701 DOI: 10.1007/s10571-021-01068-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 02/23/2021] [Indexed: 10/22/2022]
Abstract
Ischemic preconditioning (IPC) is an approach of protection against cerebral ischemia by inducing endogenous cytoprotective machinery. However, few studies in neurogenesis and oligodendrogenesis after IPC have been reported, especially the latter. The purpose of this study is to test our hypothesis that IPC may also induce cell proliferation and oligodendrogenesis in the subventricular zone and striatum, as well as to investigate the effect of nuclear factor erythroid 2-related factor 2 (Nrf2) on oligodendrogenesis. IPC was induced in mice by 12-min ischemia through the occlusion of the middle cerebral artery. Newly generated cells were labeled with 5-bromo-2'-deoxyuridine. Our findings demonstrated that IPC stimulated the proliferation of neural stem cells in the subventricular zone, promoted the generation of oligodendrocyte precursor cells in the striatum and corpus callosum/external capsule (CC/EC), and stimulated oligodendrocyte precursor cells differentiation into oligodendrocytes in the striatum and the CC/EC. Furthermore, we describe a crucial role for Nrf2 in IPC-induced oligodendrogenesis in the subventricular zone, striatum, and CC/EC and show for the first time that Nrf2 promoted the migration and differentiation of oligodendrocyte precursor cells into oligodendrocytes in the striatum and CC/EC. Our data imply that IPC stimulates the oligodendrogenesis in the brain and that Nrf2 signaling may contribute to the oligodendrogenesis.
Collapse
Affiliation(s)
- Qianqian Li
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jiyu Lou
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tuo Yang
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zhishuo Wei
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Senmiao Li
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Feng Zhang
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
30
|
Raffaele S, Fumagalli M. Dynamics of Microglia Activation in the Ischemic Brain: Implications for Myelin Repair and Functional Recovery. Front Cell Neurosci 2022; 16:950819. [PMID: 35899017 PMCID: PMC9309466 DOI: 10.3389/fncel.2022.950819] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Ischemic stroke is a neurological disorder representing a leading cause of death and permanent disability world-wide, for which effective regenerative treatments are missing. Oligodendrocyte degeneration and consequent myelin disruption are considered major contributing factors to stroke-associated neurological deficits. Therefore, fostering myelin reconstruction by oligodendrocyte precursor cells (OPCs) has emerged as a promising therapeutic approach to enhance functional recovery in stroke patients. A pivotal role in regulating remyelination is played by microglia, the resident immune cells of the brain. Early after stroke, microglial cells exert beneficial functions, promoting OPC recruitment toward the ischemic lesion and preserving myelin integrity. However, the protective features of microglia are lost during disease progression, contributing to remyelination failure. Unveiling the mechanisms driving the pro-remyelination properties of microglia may provide important opportunities for both reducing myelin damage and promoting its regeneration. Here, we summarize recent evidence describing microglia activation kinetics in experimental models of ischemic injury, focusing on the contribution of these innate immune cells to myelin damage and repair. Some molecular signals regulating the pro-regenerative functions of microglia after stroke have been highlighted to provide new possible therapeutic targets involved in the protective functions of these cells. Finally, we analyzed the impact of microglia-to-OPCs communication via extracellular vesicles on post-stroke remyelination and functional recovery. The results collected in this review underline the importance of supporting the pro-remyelination functions of microglial cells after stroke.
Collapse
|
31
|
Bub A, Brenna S, Alawi M, Kügler P, Gui Y, Kretz O, Altmeppen H, Magnus T, Puig B. Multiplexed mRNA analysis of brain-derived extracellular vesicles upon experimental stroke in mice reveals increased mRNA content with potential relevance to inflammation and recovery processes. Cell Mol Life Sci 2022; 79:329. [PMID: 35639208 PMCID: PMC9156510 DOI: 10.1007/s00018-022-04357-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/05/2022] [Accepted: 05/09/2022] [Indexed: 12/14/2022]
Abstract
Extracellular vesicles (EVs) are lipid bilayer-enclosed structures that represent newly discovered means for cell-to-cell communication as well as promising disease biomarkers and therapeutic tools. Apart from proteins, lipids, and metabolites, EVs can deliver genetic information such as mRNA, eliciting a response in the recipient cells. In the present study, we have analyzed the mRNA content of brain-derived EVs (BDEVs) isolated 72 h after experimental stroke in mice and compared them to controls (shams) using nCounter® Nanostring panels, with or without prior RNA isolation. We found that both panels show similar results when comparing upregulated mRNAs in stroke. Notably, the highest upregulated mRNAs were related to processes of stress and immune system responses, but also to anatomical structure development, cell differentiation, and extracellular matrix organization, thus indicating that regenerative mechanisms already take place at this time-point. The five top overrepresented mRNAs in stroke mice were confirmed by RT-qPCR and, interestingly, found to be full-length. We could reveal that the majority of the mRNA cargo in BDEVs was of microglial origin and predominantly present in small BDEVs (≤ 200 nm in diameter). However, the EV population with the highest increase in the total BDEVs pool at 72 h after stroke was of oligodendrocytic origin. Our study shows that nCounter® panels are a good tool to study mRNA content in tissue-derived EVs as they can be carried out even without previous mRNA isolation, and that the mRNA cargo of BDEVs indicates a possible participation in inflammatory but also recovery processes after stroke.
Collapse
Affiliation(s)
- Annika Bub
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Santra Brenna
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malik Alawi
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Paul Kügler
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yuqi Gui
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Oliver Kretz
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hermann Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Berta Puig
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
32
|
Zhang Y, Lian L, Fu R, Liu J, Shan X, Jin Y, Xu S. Microglia: The Hub of Intercellular Communication in Ischemic Stroke. Front Cell Neurosci 2022; 16:889442. [PMID: 35518646 PMCID: PMC9062186 DOI: 10.3389/fncel.2022.889442] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/30/2022] [Indexed: 12/14/2022] Open
Abstract
Communication between microglia and other cells has recently been at the forefront of research in central nervous system (CNS) disease. In this review, we provide an overview of the neuroinflammation mediated by microglia, highlight recent studies of crosstalk between microglia and CNS resident and infiltrating cells in the context of ischemic stroke (IS), and discuss how these interactions affect the course of IS. The in-depth exploration of microglia-intercellular communication will be beneficial for therapeutic tools development and clinical translation for stroke control.
Collapse
Affiliation(s)
- Yunsha Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, China
| | - Lu Lian
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rong Fu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jueling Liu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoqian Shan
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yang Jin
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, China
| | - Shixin Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| |
Collapse
|
33
|
Sun Y, Langer HF. Platelets, Thromboinflammation and Neurovascular Disease. Front Immunol 2022; 13:843404. [PMID: 35309326 PMCID: PMC8930842 DOI: 10.3389/fimmu.2022.843404] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 02/07/2022] [Indexed: 12/18/2022] Open
Abstract
The brain and spinal cord are immune-privileged organs, but in the disease state protection mechanisms such as the blood brain barrier (BBB) are ineffective or overcome by pathological processes. In neuroinflammatory diseases, microglia cells and other resident immune cells contribute to local vascular inflammation and potentially a systemic inflammatory response taking place in parallel. Microglia cells interact with other cells impacting on the integrity of the BBB and propagate the inflammatory response through the release of inflammatory signals. Here, we discuss the activation and response mechanisms of innate and adaptive immune processes in response to neuroinflammation. Furthermore, the clinical importance of neuroinflammatory mediators and a potential translational relevance of involved mechanisms are addressed also with focus on non-classical immune cells including microglia cells or platelets. As illustrative examples, novel agents such as Anfibatide or Revacept, which result in reduced recruitment and activation of platelets, a subsequently blunted activation of the coagulation cascade and further inflammatory process, demonstrating that mechanisms of neuroinflammation and thrombosis are interconnected and should be further subject to in depth clinical and basic research.
Collapse
Affiliation(s)
- Ying Sun
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
- University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Harald F. Langer
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
- University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany
- *Correspondence: Harald F. Langer,
| |
Collapse
|
34
|
Guo YS, Yuan M, Han Y, Shen XY, Gao ZK, Bi X. Effects of enriched environment on microglia and functional white matter recovery in rats with post stroke cognitive impairment. Neurochem Int 2022; 154:105295. [PMID: 35121010 DOI: 10.1016/j.neuint.2022.105295] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 12/11/2021] [Accepted: 01/27/2022] [Indexed: 01/02/2023]
Abstract
BACKGROUND White matter damage is an important contributor to cognitive impairment after stroke. This study was designed to explore the beneficial effects of enriched environment (EE) on white matter recovery and cognitive dysfunction after stroke, and further explore the potential mechanism of EE on white matter recovery from the perspective of microglia and microglia-mediated neuroinflammation. METHODS Male SD rats underwent middle cerebral artery occlusion(MCAO) or sham surgery. During the MCAO operation, a laser Doppler blood flow meter was used to monitor the blood flow to ensure the success of the model. At 72 hours after the operation, 3 rats were selected for TTC staining to identify the infarct size. One week after surgery, the rats were randomly assigned into four different groups-MCAO+standard environment (SE), MCAO+enriched environment(EE), Sham+SE and Sham+EE for 4 weeks. At four weeks after MCAO surgery, neurological function deficiency condition and cognitive function were assessed using Longa score and Morris Water Maze prior to euthanasia. The loss or regeneration of myelin was stained with LFB, the expression of myelin regeneration-related protein and microglia protein was quantified by western blot and immunofluorescence, and the level of inflammatory factors was measured by ELISA. RESULTS EE treatment remarkably decreased the neurological deficit score, ameliorated the cognitive functional deficit in MCAO rats. Furthermore, EE alleviated white matter lesions and demyelination, increased myelin basic protein expression and decreased the number of activated microglia in the hippocampus of MCAO rats. In addition, ELISA analysis indicated that EE decreased the level of IL-1β, IL-6, which further suggests that EE may reduce the level of pro-inflammatory factors by affecting the expression of microglia marker, IBA1, provide a benefit physiological environment for myelin recovery, and improve post stroke cognitive impairment. CONCLUSIONS Our results suggest that exposure to EE substantially reduced the damage to brain tissue caused by activation of microglia activation, decreased the level of pro-inflammatory cytokins, which may induced by microglia, protected and promote white matter recovery to improve cognitive function after stroke. Our findings also indicate exposure to EE is beneficial for patients with white matter impairment characterised by white matter disease-related inflammation.
Collapse
Affiliation(s)
- Yi-Sha Guo
- Department of Rehabilitation Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China; Shanghai University of Sport, Shanghai, 200438, China
| | - Mei Yuan
- Department of Rehabilitation Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China; Shanghai University of Sport, Shanghai, 200438, China
| | - Yu Han
- Department of Rehabilitation Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China; Shanghai University of Sport, Shanghai, 200438, China
| | - Xin-Ya Shen
- Department of Rehabilitation Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China; Shanghai University of Traditionary Chinese Medicine, Shanghai, 201203, China
| | - Zhen-Kun Gao
- Department of Rehabilitation Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China; Shanghai University of Traditionary Chinese Medicine, Shanghai, 201203, China
| | - Xia Bi
- Department of Rehabilitation Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China; Shanghai University of Sport, Shanghai, 200438, China.
| |
Collapse
|
35
|
Lee HJ, Jung DH, Kim NK, Shin HK, Choi BT. Effects of electroacupuncture on the functionality of NG2-expressing cells in perilesional brain tissue of mice following ischemic stroke. Neural Regen Res 2021; 17:1556-1565. [PMID: 34916441 PMCID: PMC8771106 DOI: 10.4103/1673-5374.330611] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Neural/glial antigen 2 (NG2)-expressing cells has multipotent stem cell activity under cerebral ischemia. Our study examined the effects of electroacupuncture (EA) therapy (2 Hz, 1 or 3 mA, 20 minutes) at the Sishencong acupoint on motor function after ischemic insult in the brain by investigating the rehabilitative potential of NG2-derived cells in a mouse model of ischemic stroke. EA stimulation alleviated motor deficits caused by ischemic stroke, and 1 mA EA stimulation was more efficacious than 3 mA EA stimulation or positive control treatment with edaravone, a free radical scavenger. The properties of NG2-expressing cells were altered with 1 mA EA stimulation, enhancing their survival in perilesional brain tissue via reduction of tumor necrosis factor alpha expression. EA stimulation robustly activated signaling pathways related to proliferation and survival of NG2-expressing cells and increased the expression of neurotrophic factors such as brain-derived neurotrophic factor, tumor growth factor beta, and neurotrophin 3. In the perilesional striatum, EA stimulation greatly increased the number of NG2-expressing cells double-positive for oligodendrocyte, endothelial cell, and microglia/macrophage markers (CC1, CD31, and CD68). EA therapy also greatly activated brain-derived neurotrophic factor/tropomyosin receptor kinase B and glycogen synthase kinase 3 beta signaling. Our results indicate that EA therapy may prevent functional loss at the perilesional site by enhancing survival and differentiation of NG2-expressing cells via the activation of brain-derived neurotrophic factor -induced signaling, subsequently ameliorating motor dysfunction. The animal experiments were approved by the Animal Ethics Committee of Pusan National University (approval Nos. PNU2019-2199 and PNU2019-2884) on April 8, 2019 and June 19, 2019.
Collapse
Affiliation(s)
- Hong Ju Lee
- Department of Korean Medical Science, School of Korean Medicine; Graduate Training Program of Korean Medicine for Healthy Aging, Pusan National University, Yangsan, Republic of Korea
| | - Da Hee Jung
- Department of Korean Medical Science, School of Korean Medicine; Graduate Training Program of Korean Medicine for Healthy Aging, Pusan National University, Yangsan, Republic of Korea
| | - Nam Kwen Kim
- Department of Korean Ophthalmology, Otolaryngology and Dermatology, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Hwa Kyoung Shin
- Department of Korean Medical Science, School of Korean Medicine; Graduate Training Program of Korean Medicine for Healthy Aging, Pusan National University, Yangsan, Republic of Korea
| | - Byung Tae Choi
- Department of Korean Medical Science, School of Korean Medicine; Graduate Training Program of Korean Medicine for Healthy Aging, Pusan National University, Yangsan, Republic of Korea
| |
Collapse
|
36
|
Alia C, Cangi D, Massa V, Salluzzo M, Vignozzi L, Caleo M, Spalletti C. Cell-to-Cell Interactions Mediating Functional Recovery after Stroke. Cells 2021; 10:3050. [PMID: 34831273 PMCID: PMC8623942 DOI: 10.3390/cells10113050] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/27/2021] [Accepted: 11/02/2021] [Indexed: 12/22/2022] Open
Abstract
Ischemic damage in brain tissue triggers a cascade of molecular and structural plastic changes, thus influencing a wide range of cell-to-cell interactions. Understanding and manipulating this scenario of intercellular connections is the Holy Grail for post-stroke neurorehabilitation. Here, we discuss the main findings in the literature related to post-stroke alterations in cell-to-cell interactions, which may be either detrimental or supportive for functional recovery. We consider both neural and non-neural cells, starting from astrocytes and reactive astrogliosis and moving to the roles of the oligodendrocytes in the support of vulnerable neurons and sprouting inhibition. We discuss the controversial role of microglia in neural inflammation after injury and we conclude with the description of post-stroke alterations in pyramidal and GABAergic cells interactions. For all of these sections, we review not only the spontaneous evolution in cellular interactions after ischemic injury, but also the experimental strategies which have targeted these interactions and that are inspiring novel therapeutic strategies for clinical application.
Collapse
Affiliation(s)
- Claudia Alia
- Neuroscience Institute, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (V.M.); (M.S.); (M.C.); (C.S.)
| | - Daniele Cangi
- Department of Neurosciences, Psychology, Drugs and Child Health Area, School of Psychology, University of Florence, 50121 Florence, Italy;
| | - Verediana Massa
- Neuroscience Institute, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (V.M.); (M.S.); (M.C.); (C.S.)
| | - Marco Salluzzo
- Neuroscience Institute, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (V.M.); (M.S.); (M.C.); (C.S.)
- Department of Neurosciences, Psychology, Drugs and Child Health Area, School of Psychology, University of Florence, 50121 Florence, Italy;
| | - Livia Vignozzi
- Department of Biomedical Sciences, University of Padua, Viale G. Colombo 3, 35121 Padua, Italy;
| | - Matteo Caleo
- Neuroscience Institute, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (V.M.); (M.S.); (M.C.); (C.S.)
- Department of Biomedical Sciences, University of Padua, Viale G. Colombo 3, 35121 Padua, Italy;
| | - Cristina Spalletti
- Neuroscience Institute, National Research Council (CNR), Via G. Moruzzi 1, 56124 Pisa, Italy; (V.M.); (M.S.); (M.C.); (C.S.)
| |
Collapse
|
37
|
Damian C, Ghuman H, Mauney C, Azar R, Reinartz J, Badylak SF, Modo M. Post-Stroke Timing of ECM Hydrogel Implantation Affects Biodegradation and Tissue Restoration. Int J Mol Sci 2021; 22:ijms222111372. [PMID: 34768800 PMCID: PMC8583606 DOI: 10.3390/ijms222111372] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/13/2021] [Accepted: 10/17/2021] [Indexed: 01/01/2023] Open
Abstract
Extracellular matrix (ECM) hydrogel promotes tissue regeneration in lesion cavities after stroke. However, a bioscaffold's regenerative potential needs to be considered in the context of the evolving pathological environment caused by a stroke. To evaluate this key issue in rats, ECM hydrogel was delivered to the lesion core/cavity at 7-, 14-, 28-, and 90-days post-stroke. Due to a lack of tissue cavitation 7-days post-stroke, implantation of ECM hydrogel did not achieve a sufficient volume and distribution to warrant comparison with the other time points. Biodegradation of ECM hydrogel implanted 14- and 28-days post-stroke were efficiently (80%) degraded by 14-days post-bioscaffold implantation, whereas implantation 90-days post-stroke revealed only a 60% decrease. Macrophage invasion was robust at 14- and 28-days post-stroke but reduced in the 90-days post-stroke condition. The pro-inflammation (M1) and pro-repair (M2) phenotype ratios were equivalent at all time points, suggesting that the pathological environment determines macrophage invasion, whereas ECM hydrogel defines their polarization. Neural cells (neural progenitors, neurons, astrocytes, oligodendrocytes) were found at all time points, but a 90-days post-stroke implantation resulted in reduced densities of mature phenotypes. Brain tissue restoration is therefore dependent on an efficient delivery of a bioscaffold to a tissue cavity, with 28-days post-stroke producing the most efficient biodegradation and tissue regeneration, whereas by 90-days post-stroke, these effects are significantly reduced. Improving our understanding of how the pathological environment influences biodegradation and the tissue restoration process is hence essential to devise engineering strategies that could extend the therapeutic window for bioscaffolds to repair the damaged brain.
Collapse
Affiliation(s)
- Corina Damian
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA; (C.D.); (C.M.)
| | - Harmanvir Ghuman
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA; (H.G.); (R.A.); (S.F.B.)
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Carrinton Mauney
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA; (C.D.); (C.M.)
| | - Reem Azar
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA; (H.G.); (R.A.); (S.F.B.)
| | - Janina Reinartz
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA 15260, USA;
| | - Stephen F. Badylak
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA; (H.G.); (R.A.); (S.F.B.)
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Michel Modo
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA; (H.G.); (R.A.); (S.F.B.)
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA 15260, USA;
- Correspondence: ; Tel.: +1-(412)-383-7200
| |
Collapse
|
38
|
Intranasal Administration of Undifferentiated Oligodendrocyte Lineage Cells as a Potential Approach to Deliver Oligodendrocyte Precursor Cells into Brain. Int J Mol Sci 2021; 22:ijms221910738. [PMID: 34639079 PMCID: PMC8509516 DOI: 10.3390/ijms221910738] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 12/14/2022] Open
Abstract
Oligodendrocyte precursor cell (OPC) migration is a mechanism involved in remyelination; these cells migrate from niches in the adult CNS. However, age and disease reduce the pool of OPCs; as a result, the remyelination capacity of the CNS decreases over time. Several experimental studies have introduced OPCs to the brain via direct injection or intrathecal administration. In this study, we used the nose-to brain pathway to deliver oligodendrocyte lineage cells (human oligodendroglioma (HOG) cells), which behave similarly to OPCs in vitro. To this end, we administered GFP-labelled HOG cells intranasally to experimental animals, which were subsequently euthanised at 30 or 60 days. Our results show that the intranasal route is a viable route to the CNS and that HOG cells administered intranasally migrate preferentially to niches of OPCs (clusters created during embryonic development and adult life). Our study provides evidence, albeit limited, that HOG cells either form clusters or adhere to clusters of OPCs in the brains of experimental animals.
Collapse
|
39
|
Hernández IH, Villa-González M, Martín G, Soto M, Pérez-Álvarez MJ. Glial Cells as Therapeutic Approaches in Brain Ischemia-Reperfusion Injury. Cells 2021; 10:1639. [PMID: 34208834 PMCID: PMC8305833 DOI: 10.3390/cells10071639] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/24/2021] [Accepted: 06/26/2021] [Indexed: 02/07/2023] Open
Abstract
Ischemic stroke is the second cause of mortality and the first cause of long-term disability constituting a serious socioeconomic burden worldwide. Approved treatments include thrombectomy and rtPA intravenous administration, which, despite their efficacy in some cases, are not suitable for a great proportion of patients. Glial cell-related therapies are progressively overcoming inefficient neuron-centered approaches in the preclinical phase. Exploiting the ability of microglia to naturally switch between detrimental and protective phenotypes represents a promising therapeutic treatment, in a similar way to what happens with astrocytes. However, the duality present in many of the roles of these cells upon ischemia poses a notorious difficulty in disentangling the precise pathways to target. Still, promoting M2/A2 microglia/astrocyte protective phenotypes and inhibiting M1/A1 neurotoxic profiles is globally rendering promising results in different in vivo models of stroke. On the other hand, described oligodendrogenesis after brain ischemia seems to be strictly beneficial, although these cells are the less studied players in the stroke paradigm and negative effects could be described for oligodendrocytes in the next years. Here, we review recent advances in understanding the precise role of mentioned glial cell types in the main pathological events of ischemic stroke, including inflammation, blood brain barrier integrity, excitotoxicity, reactive oxygen species management, metabolic support, and neurogenesis, among others, with a special attention to tested therapeutic approaches.
Collapse
Affiliation(s)
- Ivó H Hernández
- Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Mario Villa-González
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Gerardo Martín
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Manuel Soto
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - María José Pérez-Álvarez
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
40
|
Gou X, Xu D, Li F, Hou K, Fang W, Li Y. Pyroptosis in stroke-new insights into disease mechanisms and therapeutic strategies. J Physiol Biochem 2021; 77:511-529. [PMID: 33942252 DOI: 10.1007/s13105-021-00817-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 04/13/2021] [Indexed: 02/07/2023]
Abstract
Stroke is a common disease with high mortality and disability worldwide. Different forms of cell deaths, including apoptosis and necrosis, occur in ischemic or hemorrhagic brain tissue, among which pyroptosis, a newly discovered inflammation-related programmed cell death, is generally divided into two main pathways, the canonical inflammasome pathway and the non-canonical inflammasome pathway. Caspase-mediated pyroptosis requires the assembly of inflammasomes such as NLRP3, which leads to the release of inflammatory cytokines IL-1β and IL-18 through the pores formed in the plasma membrane by GSDMD followed by neuroinflammation. Recently, pyroptosis and its relationship with inflammation have attracted more and more attention in the study of cerebral ischemia or hemorrhage. In addition, many inhibitors of pyroptosis targeting caspase, NLRP3, and the upstream pathway have been found to reduce brain tissue damage after stroke. In this review, we mainly introduce the pathology of stroke, the molecular mechanism, and process of pyroptosis, as well as the pivotal roles of pyroptosis in stroke, in order to provide new insights for the treatment of stroke.
Collapse
Affiliation(s)
- Xue Gou
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, 210009, China
| | - Dan Xu
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, 210009, China
| | - Fengyang Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, 210009, China
| | - Kai Hou
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, 210009, China.,Department of Pharmacy, Zhongda Hospital, Southeast University, Nanjing, China
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, 210009, China.
| | - Yunman Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
41
|
Nozohouri S, Vaidya B, Abbruscato TJ. Exosomes in Ischemic Stroke. Curr Pharm Des 2021; 26:5533-5545. [PMID: 32534564 DOI: 10.2174/1381612826666200614180253] [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: 01/30/2020] [Accepted: 05/05/2020] [Indexed: 12/14/2022]
Abstract
Ischemic stroke, a leading cause of mortality, results in severe neurological outcomes in the patients. Effective stroke therapies may significantly decrease the extent of injury. For this purpose, novel and efficient drug delivery strategies need to be developed. Among a myriad of therapeutic and drug delivery techniques, exosomes have shown promising results in ischemic stroke either by their intrinsic therapeutic characteristics, which can result in angiogenesis and neurogenesis or by acting as competent, biocompatible drug delivery vehicles to transport neurotherapeutic agents into the brain. In this review, we have discussed different methods of exosome isolation and cargo loading techniques, advantages and disadvantages of using exosomes as a drug delivery carrier and the therapeutic applications of exosomes with a focus on ischemic stroke therapy.
Collapse
Affiliation(s)
- Saeideh Nozohouri
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX-79106, United States
| | - Bhuvaneshwar Vaidya
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX-79106, United States
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX-79106, United States
| |
Collapse
|
42
|
Zheng J, Zhang T, Han S, Liu C, Liu M, Li S, Li J. Activin A improves the neurological outcome after ischemic stroke in mice by promoting oligodendroglial ACVR1B-mediated white matter remyelination. Exp Neurol 2021; 337:113574. [PMID: 33345977 DOI: 10.1016/j.expneurol.2020.113574] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/08/2020] [Accepted: 12/16/2020] [Indexed: 02/07/2023]
Abstract
Activin A plays important roles in ischemic injury and white matter remyelination, but its mechanisms are unclear. In this study, the adult male C57BL/6 J mice were used to establish the model of 1 h middle cerebral artery occlusion/reperfusion (MCAO/R) 1 d to 28 d-induced ischemic stroke in vivo. We found that the neurological outcome was positively correlated with the levels of myelin associated proteins (include MAG, CNPase, MOG and MBP, n = 6 per group) both in corpus callosum and internal capsule of mice with ischemic stroke. The dynamic changes of Luxol fast blue (LFB) staining intensity, oligodendrocyte (CC1+) and proliferated oligodendrocyte precursor (Ki67+/PDGFRα+) cell numbers indicated demyelination and spontaneous remyelination occurred in the corpus callosum of mice after 1 h MCAO/R 1 d-28 d (n = 6 per group). Activin receptor type I (ACVR1) inhibitor SB431542 aggregated neurological deficits, and reduced MAG, MOG and MBP protein levels of mice with ischemic stroke (n = 6 per group). Meanwhile, recombinant mouse (rm) Activin A enhanced the neurological function recovery, MAG, MOG and MBP protein levels of mice with 1 h MCAO/R 28 d. In addition, the injection of AAV-based ACVR1B shRNA with Olig2 promoter could reverse rmActivin A-induced the increases of CC1+ cell number, LFB intensity, MAG, MOG and MBP protein levels in the corpus callosum (n = 6 per group), and neurological function recovery (n = 10 per group) of mice with 1 h MCAO/R 28 d. These results suggested that Activin A improves the neurological outcome through promoting oligodendroglial ACVR1B-mediated white matter remyelination of mice with ischemic stroke, which may provide a potential therapeutic strategy for ischemic stroke.
Collapse
Affiliation(s)
- Jiayin Zheng
- Department of Neurobiology and Center of Stroke, Beijing Institute for Brain Disorders, School of Basic Medical Science, Capital Medical University, Beijing 100069, PR China
| | - Teng Zhang
- Department of Laboratory Animal Sciences, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China
| | - Song Han
- Department of Neurobiology and Center of Stroke, Beijing Institute for Brain Disorders, School of Basic Medical Science, Capital Medical University, Beijing 100069, PR China
| | - Cui Liu
- Department of Neurobiology and Center of Stroke, Beijing Institute for Brain Disorders, School of Basic Medical Science, Capital Medical University, Beijing 100069, PR China
| | - Meilian Liu
- Department of Neurobiology and Center of Stroke, Beijing Institute for Brain Disorders, School of Basic Medical Science, Capital Medical University, Beijing 100069, PR China
| | - Shujuan Li
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, PR China.
| | - Junfa Li
- Department of Neurobiology and Center of Stroke, Beijing Institute for Brain Disorders, School of Basic Medical Science, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
43
|
Go V, Sarikaya D, Zhou Y, Bowley BGE, Pessina MA, Rosene DL, Zhang ZG, Chopp M, Finklestein SP, Medalla M, Buller B, Moore TL. Extracellular vesicles derived from bone marrow mesenchymal stem cells enhance myelin maintenance after cortical injury in aged rhesus monkeys. Exp Neurol 2021; 337:113540. [PMID: 33264634 PMCID: PMC7946396 DOI: 10.1016/j.expneurol.2020.113540] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 11/05/2020] [Accepted: 11/24/2020] [Indexed: 12/20/2022]
Abstract
Cortical injury, such as stroke, causes neurotoxic cascades that lead to rapid death and/or damage to neurons and glia. Axonal and myelin damage in particular, are critical factors that lead to neuronal dysfunction and impair recovery of function after injury. These factors can be exacerbated in the aged brain where white matter damage is prevalent. Therapies that can ameliorate myelin damage and promote repair by targeting oligodendroglia, the cells that produce and maintain myelin, may facilitate recovery after injury, especially in the aged brain where these processes are already compromised. We previously reported that a novel therapeutic, Mesenchymal Stem Cell derived extracellular vesicles (MSC-EVs), administered intravenously at both 24 h and 14 days after cortical injury, reduced microgliosis (Go et al. 2019), reduced neuronal pathology (Medalla et al. 2020), and improved motor recovery (Moore et al. 2019) in aged female rhesus monkeys. Here, we evaluated the effect of MSC-EV treatment on changes in oligodendrocyte maturation and associated myelin markers in the sublesional white matter using immunohistochemistry, confocal microscopy, stereology, qRT-PCR, and ELISA. Compared to vehicle control monkeys, EV-treated monkeys showed a reduction in the density of damaged oligodendrocytes. Further, EV-treatment was associated with enhanced myelin maintenance, evidenced by upregulation of myelin-related genes and increases in actively myelinating oligodendrocytes in sublesional white matter. These changes in myelination correlate with the rate of motor recovery, suggesting that improved myelin maintenance facilitates this recovery. Overall, our results suggest that EVs act on oligodendrocytes to support myelination and improves functional recovery after injury in the aged brain. SIGNIFICANCE: We previously reported that EVs facilitate recovery of function after cortical injury in the aged monkey brain, while also reducing neuronal pathology (Medalla et al. 2020) and microgliosis (Go et al. 2019). However, the effect of injury and EVs on oligodendrocytes and myelination has not been characterized in the primate brain (Dewar et al. 1999; Sozmen et al. 2012; Zhang et al. 2013). In the present study, we assessed changes in myelination after cortical injury in aged monkeys. Our results show, for the first time, that MSC-EVs support recovery of function after cortical injury by enhancing myelin maintenance in the aged primate brain.
Collapse
Affiliation(s)
- Veronica Go
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, United States.
| | - Deniz Sarikaya
- Research Center for Translational Medicine, Koç University School of Medicine, Turkey
| | - Yuxin Zhou
- Department of Anatomy & Neurobiology, Boston University School of Medicine, United States
| | - Bethany G E Bowley
- Department of Anatomy & Neurobiology, Boston University School of Medicine, United States
| | - Monica A Pessina
- Department of Anatomy & Neurobiology, Boston University School of Medicine, United States
| | - Douglas L Rosene
- Department of Anatomy & Neurobiology, Boston University School of Medicine, United States; Yerkes National Primate Research Center, Emory University, United States; Center for Systems Neuroscience, Boston University, United States
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Health Systems, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Health Systems, United States; Department of Physics, Oakland University, United States
| | - Seth P Finklestein
- Department of Neurology, Massachusetts General Hospital, United States; Stemetix, Inc., United States
| | - Maria Medalla
- Department of Anatomy & Neurobiology, Boston University School of Medicine, United States; Center for Systems Neuroscience, Boston University, United States
| | - Benjamin Buller
- Department of Neurology, Henry Ford Health Systems, United States
| | - Tara L Moore
- Department of Anatomy & Neurobiology, Boston University School of Medicine, United States; Center for Systems Neuroscience, Boston University, United States
| |
Collapse
|
44
|
Cellular Mechanisms Participating in Brain Repair of Adult Zebrafish and Mammals after Injury. Cells 2021; 10:cells10020391. [PMID: 33672842 PMCID: PMC7917790 DOI: 10.3390/cells10020391] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/28/2021] [Accepted: 02/05/2021] [Indexed: 12/12/2022] Open
Abstract
Adult neurogenesis is an evolutionary conserved process occurring in all vertebrates. However, striking differences are observed between the taxa, considering the number of neurogenic niches, the neural stem cell (NSC) identity, and brain plasticity under constitutive and injury-induced conditions. Zebrafish has become a popular model for the investigation of the molecular and cellular mechanisms involved in adult neurogenesis. Compared to mammals, the adult zebrafish displays a high number of neurogenic niches distributed throughout the brain. Furthermore, it exhibits a strong regenerative capacity without scar formation or any obvious disabilities. In this review, we will first discuss the similarities and differences regarding (i) the distribution of neurogenic niches in the brain of adult zebrafish and mammals (mainly mouse) and (ii) the nature of the neural stem cells within the main telencephalic niches. In the second part, we will describe the cascade of cellular events occurring after telencephalic injury in zebrafish and mouse. Our study clearly shows that most early events happening right after the brain injury are shared between zebrafish and mouse including cell death, microglia, and oligodendrocyte recruitment, as well as injury-induced neurogenesis. In mammals, one of the consequences following an injury is the formation of a glial scar that is persistent. This is not the case in zebrafish, which may be one of the main reasons that zebrafish display a higher regenerative capacity.
Collapse
|
45
|
Cellular and Molecular Mechanisms of R/S-Roscovitine and CDKs Related Inhibition under Both Focal and Global Cerebral Ischemia: A Focus on Neurovascular Unit and Immune Cells. Cells 2021; 10:cells10010104. [PMID: 33429982 PMCID: PMC7827530 DOI: 10.3390/cells10010104] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/29/2020] [Accepted: 01/05/2021] [Indexed: 12/29/2022] Open
Abstract
Ischemic stroke is the second leading cause of death worldwide. Following ischemic stroke, Neurovascular Unit (NVU) inflammation and peripheral leucocytes infiltration are major contributors to the extension of brain lesions. For a long time restricted to neurons, the 10 past years have shown the emergence of an increasing number of studies focusing on the role of Cyclin-Dependent Kinases (CDKs) on the other cells of NVU, as well as on the leucocytes. The most widely used CDKs inhibitor, (R)-roscovitine, and its (S) isomer both decreased brain lesions in models of global and focal cerebral ischemia. We previously showed that (S)-roscovitine acted, at least, by modulating NVU response to ischemia. Interestingly, roscovitine was shown to decrease leucocytes-mediated inflammation in several inflammatory models. Specific inhibition of roscovitine majors target CDK 1, 2, 5, 7, and 9 showed that these CDKs played key roles in inflammatory processes of NVU cells and leucocytes after brain lesions, including ischemic stroke. The data summarized here support the investigation of roscovitine as a potential therapeutic agent for the treatment of ischemic stroke, and provide an overview of CDK 1, 2, 5, 7, and 9 functions in brain cells and leucocytes during cerebral ischemia.
Collapse
|
46
|
Villa Gonzalez M, Pérez-Álvarez MJ. A 3R-Tau-mediated mechanism in oligodendrocytes: could it be the key for neuroprotection after stroke? Neural Regen Res 2021; 16:2401-2402. [PMID: 33907017 PMCID: PMC8374573 DOI: 10.4103/1673-5374.313027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Mario Villa Gonzalez
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid; Centro de Biología Molecular "Severo Ochoa", Departamento de Neuropatología Molecular CSIC-UAM, Madrid, Spain
| | - Maria José Pérez-Álvarez
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid; Centro de Biología Molecular "Severo Ochoa", Departamento de Neuropatología Molecular CSIC-UAM; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
47
|
Raffaele S, Gelosa P, Bonfanti E, Lombardi M, Castiglioni L, Cimino M, Sironi L, Abbracchio MP, Verderio C, Fumagalli M. Microglial vesicles improve post-stroke recovery by preventing immune cell senescence and favoring oligodendrogenesis. Mol Ther 2020; 29:1439-1458. [PMID: 33309882 DOI: 10.1016/j.ymthe.2020.12.009] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/10/2020] [Accepted: 12/06/2020] [Indexed: 12/11/2022] Open
Abstract
Contrasting myelin damage through the generation of new myelinating oligodendrocytes represents a promising approach to promote functional recovery after stroke. Here, we asked whether activation of microglia and monocyte-derived macrophages affects the regenerative process sustained by G protein-coupled receptor 17 (GPR17)-expressing oligodendrocyte precursor cells (OPCs), a subpopulation of OPCs specifically reacting to ischemic injury. GPR17-iCreERT2:CAG-eGFP reporter mice were employed to trace the fate of GPR17-expressing OPCs, labeled by the green fluorescent protein (GFP), after permanent middle cerebral artery occlusion. By microglia/macrophages pharmacological depletion studies, we show that innate immune cells favor GFP+ OPC reaction and limit myelin damage early after injury, whereas they lose their pro-resolving capacity and acquire a dystrophic "senescent-like" phenotype at later stages. Intracerebral infusion of regenerative microglia-derived extracellular vesicles (EVs) restores protective microglia/macrophages functions, limiting their senescence during the post-stroke phase, and enhances the maturation of GFP+ OPCs at lesion borders, resulting in ameliorated neurological functionality. In vitro experiments show that EV-carried transmembrane tumor necrosis factor (tmTNF) mediates the pro-differentiating effects on OPCs, with future implications for regenerative therapies.
Collapse
Affiliation(s)
- Stefano Raffaele
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Paolo Gelosa
- IRCCS Centro Cardiologico Monzino, 20138 Milan, Italy
| | - Elisabetta Bonfanti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | | | - Laura Castiglioni
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Mauro Cimino
- Department of Biomolecular Sciences, Università degli Studi di Urbino, 61029 Urbino, Italy
| | - Luigi Sironi
- IRCCS Centro Cardiologico Monzino, 20138 Milan, Italy; Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Maria P Abbracchio
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | | | - Marta Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy.
| |
Collapse
|
48
|
Wu D, Fu Y, Wu L, Huber M, Chen J, Yao T, Zhang M, Wu C, Song M, He X, Li S, Zhang Y, Li S, Ding Y, Ji X. Reperfusion plus Selective Intra-arterial Cooling (SI-AC) Improve Recovery in a Nonhuman Primate Model of Stroke. Neurotherapeutics 2020; 17:1931-1939. [PMID: 32710291 PMCID: PMC7851312 DOI: 10.1007/s13311-020-00895-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Early reperfusion is increasingly prioritized in ischemic stroke care, but outcomes remain suboptimal. Therefore, there is an urgent need to find neuroprotective approaches that can be combined with reperfusion to maximize efficacy. Here, the neuroprotective mechanisms behind therapeutic hypothermia were evaluated in a monkey model of ischemic stroke. Focal ischemia was induced in adult rhesus monkeys by placing autologous clots in the middle cerebral artery. Monkeys were treated with tissue plasminogen activator (t-PA) alone or t-PA plus selective intra-arterial cooling (SI-AC). Serial MRI scans and functional deficit were evaluated after ischemia. Histopathology and immunohistochemistry analysis were performed after the final MRI scan. t-PA plus SI-AC treatment led to a higher rate of MRI tissue rescue, and significantly improved neurologic deficits and daily activity scores compared with t-PA alone. In peri-infarct areas, higher fractional anisotropy values and greater fiber numbers were observed in models receiving t-PA plus SI-AC. Histological findings indicated that myelin damage, spheroids, and spongiosis were significantly ameliorated in models receiving SI-AC treatment. White matter integrity was also improved by SI-AC based on immunochemical staining. Our study demonstrates that SI-AC can be effectively combined with t-PA to improve both structural and functional recovery in a monkey model of focal ischemia. These findings provide proof-of-concept that it may be feasible to add neuroprotective agents as adjunctive treatments to reperfusion therapy for stroke.
Collapse
Affiliation(s)
- Di Wu
- Department of neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
| | - Yongjuan Fu
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Longfei Wu
- Department of neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Mitchell Huber
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jian Chen
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Tianqi Yao
- Department of neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Mo Zhang
- Department of Radiology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Chuanjie Wu
- Department of neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Ming Song
- National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Xiaoduo He
- Department of neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Sijie Li
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China
| | - Yongbiao Zhang
- Interdisciplinary Innovation Institute of Medicine and Engineering, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Shengli Li
- Department of Laboratory Animal Science, Capital Medical University, Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xunming Ji
- Department of neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China.
- Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.
| |
Collapse
|
49
|
Wang C, Chopp M, Huang R, Li C, Zhang Y, Golembieski W, Lu M, Hazan Z, Zhang ZG, Zhang L. Delayed (21 Days) Post Stroke Treatment With RPh201, a Botany-Derived Compound, Improves Neurological Functional Recovery in a Rat Model of Embolic Stroke. Front Neurosci 2020; 14:813. [PMID: 32848574 PMCID: PMC7412960 DOI: 10.3389/fnins.2020.00813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/10/2020] [Indexed: 11/13/2022] Open
Abstract
Background Despite the recent advances in the acute stroke care, treatment options for long-term disability are limited. RPh201 is a botany-derived bioactive compound that has been shown to exert beneficial effects in various experimental models of neural injury. The present study evaluated the effect of delayed RPh201 treatment on long term functional recovery after stroke. Methods Adult male Wistar rats subjected to embolic middle cerebral artery occlusion (MCAO) were randomized into the following experimental groups (n = 20/group): (1) RPh201 treatment, and (2) Vehicle (cottonseed oil). RPh201 (20 μl) or Vehicle were subcutaneously administered twice a week for 16 consecutive weeks starting at 21 days after MCAO. An array of behavioral tests was performed up to120 days after MCAO. Results Ischemic rats treated with RPh201 exhibited significant (p < 0.05) improvement of neurological function measured by adhesive removal test, foot-fault test, and modified neurological severity score at 90 and 120 days after MCAO. Immunohistochemistry analysis showed that RPh201 treatment robustly increased neurofilament heavy chain positive axons and myelin basic protein densities in the peri-infarct area by 61% and 31%, respectively, when compared to the Vehicle treatment, which were further confirmed by Western blot analysis. The RPh201 treatment did not reduce infarct volume. Conclusion Our data demonstrated that RPh201 has a therapeutic effect on improvement of functional recovery in male ischemic rats even when the treatment was initiated 21 days post stroke. Enhanced axonal and myelination densities by RPh201 in ischemic brain may contribute to improved stroke recovery.
Collapse
Affiliation(s)
- Chunyang Wang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States.,Department of Physics, Oakland University, Rochester, MI, United States
| | - Rui Huang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Chao Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Yi Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | | | - Mei Lu
- Department of Biostatistics and Research Epidemiology, Henry Ford Hospital, Detroit, MI, United States
| | | | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Li Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| |
Collapse
|
50
|
Li L, Li R, Zacharek A, Wang F, Landschoot-Ward J, Chopp M, Chen J, Cui X. ABCA1/ApoE/HDL Signaling Pathway Facilitates Myelination and Oligodendrogenesis after Stroke. Int J Mol Sci 2020; 21:ijms21124369. [PMID: 32575457 PMCID: PMC7352241 DOI: 10.3390/ijms21124369] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 02/07/2023] Open
Abstract
ATP-binding cassette transporter A1 (ABCA1) plays an important role in the regulation of apolipoprotein E (ApoE) and the biogenesis of high-density lipoprotein (HDL) cholesterol in the mammalian brain. Cholesterol is a major source for myelination. Here, we investigate whether ABCA1/ApoE/HDL contribute to myelin repair and oligodendrogenesis in the ischemic brain after stroke. Specific brain ABCA1-deficient (ABCA1-B/-B) and ABCA1-floxed (ABCA1fl/fl) control mice were subjected to permanent distal middle-cerebral-artery occlusion (dMCAo) and were intracerebrally administered (1) artificial mouse cerebrospinal fluid (CSF) as vehicle control, (2) human plasma HDL3, and (3) recombined human ApoE2 starting 24 h after dMCAo for 14 days. All stroke mice were sacrificed 21 days after dMCAo. The ABCA1-B/-B–dMCAo mice exhibit significantly reduced myelination and oligodendrogenesis in the ischemic brain as well as decreased functional outcome 21 days after stroke compared with ABCA1fl/fl mice; administration of human ApoE2 or HDL3 in the ischemic brain significantly attenuates the deficits in myelination and oligodendrogenesis in ABCA1-B/-B–dMCAo mice ( p < 0.05, n = 9/group). In vitro, ABCA1-B/-B reduces ApoE expression and decreases primary oligodendrocyte progenitor cell (OPC) migration and oligodendrocyte maturation; HDL3 and ApoE2 treatment significantly reverses ABCA1-B/-B-induced reduction in OPC migration and oligodendrocyte maturation. Our data indicate that the ABCA1/ApoE/HDL signaling pathway contributes to myelination and oligodendrogenesis in the ischemic brain after stroke.
Collapse
Affiliation(s)
- Li Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA; (L.L.); (R.L.); (A.Z.); (F.W.); (J.L.-W.); (M.C.); (J.C.)
| | - Rongwen Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA; (L.L.); (R.L.); (A.Z.); (F.W.); (J.L.-W.); (M.C.); (J.C.)
| | - Alex Zacharek
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA; (L.L.); (R.L.); (A.Z.); (F.W.); (J.L.-W.); (M.C.); (J.C.)
| | - Fengjie Wang
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA; (L.L.); (R.L.); (A.Z.); (F.W.); (J.L.-W.); (M.C.); (J.C.)
| | - Julie Landschoot-Ward
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA; (L.L.); (R.L.); (A.Z.); (F.W.); (J.L.-W.); (M.C.); (J.C.)
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA; (L.L.); (R.L.); (A.Z.); (F.W.); (J.L.-W.); (M.C.); (J.C.)
- Department of Physics, Oakland University, Rochester, MI 48309, USA
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA; (L.L.); (R.L.); (A.Z.); (F.W.); (J.L.-W.); (M.C.); (J.C.)
| | - Xu Cui
- Department of Neurology, Henry Ford Hospital, Detroit, MI 48202, USA; (L.L.); (R.L.); (A.Z.); (F.W.); (J.L.-W.); (M.C.); (J.C.)
- Correspondence: ; Tel.: 01-313-916-2864
| |
Collapse
|