1
|
Wang G, Li Q, Liu S, Li M, Liu B, Zhao T, Liu B, Chen Z. An injectable decellularized extracellular matrix hydrogel with cortical neuron-derived exosomes enhances tissue repair following traumatic spinal cord injury. Mater Today Bio 2024; 28:101250. [PMID: 39318371 PMCID: PMC11421349 DOI: 10.1016/j.mtbio.2024.101250] [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: 04/11/2024] [Revised: 07/11/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024] Open
Abstract
Traumatic spinal cord injury (SCI), known for its limited intrinsic regeneration capacity, often results in considerable neurological impairment. Studies suggest that therapeutic techniques utilizing exosomes (Exo) to promote tissue regeneration and modulate immune responses are promising for SCI treatment. However, combining exosome therapy with biomaterials for SCI treatment is not very effective. This study developed an adhesive hydrogel using exosomes secreted by cortical neurons derived from human induced pluripotent stem cells (iPSCs) and decellularized extracellular matrix (dECM) from human umbilical cord mesenchymal stem cells (hUCMSCs) to enhance motor function recovery post-SCI. In vitro assessments demonstrated the excellent cytocompatibility of the dECM hydrogel. Additionally, the Exo-dECM hydrogel facilitated the polarization of early M2 macrophages, reduced neuronal apoptosis, and established a pro-regenerative microenvironment in a rodent SCI model. Subsequent analyses revealed significant activation of endogenous neural stem cells and promotion of axon regeneration and remyelination at eight weeks post-surgery. The Exo-dECM hydrogel also promoted the functional recovery and preservation of urinary tissue in SCI-afflicted rats. These findings highlighted that the Exo-dECM hydrogel is a promising therapeutic strategy for treating SCI.
Collapse
Affiliation(s)
- Gang Wang
- Cell Therapy Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Qian Li
- Cell Therapy Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Sumei Liu
- Cell Therapy Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Mo Li
- Cell Therapy Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Baoguo Liu
- Cell Therapy Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Tianyao Zhao
- Cell Therapy Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Bochao Liu
- Cell Therapy Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Zhiguo Chen
- Cell Therapy Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| |
Collapse
|
2
|
Hu H, Gao H, Wang K, Jin Z, Zheng W, Wang Q, Yang Y, Yu C, Xu K, Gao C. Effective treatment of traumatic brain injury by injection of a selenium-containing ointment. Acta Biomater 2024; 187:161-171. [PMID: 39236795 DOI: 10.1016/j.actbio.2024.08.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 08/22/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024]
Abstract
Traumatic brain injury (TBI) is an incurable and overwhelming disease accompanied with serve disability and huge financial burden, where the overproduced reactive oxygen species (ROS) can exacerbate the secondary injury, leading to massive apoptosis of neurons. In this study, β-cyclodextrin (CD)-capped hyperbranched polymers containing selenium element (HSE-CD) were crosslinked with CD-modified hyaluronic acid (HA-CD) and amantadine-modified hyaluronic acid (HA-AD) to obtain a ROS-responsive ointment (R-O). The structures of synthesized polymers were characterized with 1H nuclear magnetic resonance, and the properties of ointment were investigated with rheology and antioxidation. Compared to non-ROS-responsive ointment (N-O), the R-O ointment had stronger efficiency in decreasing the ROS level in BV2 cells in vitro. In a controlled rat cortical impact (CCI) model, the R-O ointment could relieve the DNA damage and decrease apoptosis in injured area via reducing the ROS level. Besides, after the R-O treatment, the rats showed significantly less activated astrocytes and microglia, a lower level of pro-inflammatory cytokines and a higher ratio of M2/M1 macrophage and microglia. Moreover, compared to the TBI group the R-O ointment promoted the doublecortin (DCX) expression and tissue structure integrity around the cavity, and promoted the recovery of nerve function post TBI. STATEMENT OF SIGNIFICANCE: Traumatic brain injury (TBI) is an incurable and overwhelming disease, leading to severe disability and huge social burden, where reactive oxygen species (ROS) are considered as one of the most significant factors in the secondary injury of TBI. A ROS responsive supramolecular ointment containing di-selenide bonds was injected in rats with controlled cortical impact. It relieved the DNA damage and decreased apoptosis in the injured area via reducing the ROS levels, downregulated neuroinflammation, and improved neurological recovery of TBI in vivo. This designed self-adaptive biomaterial effectively regulated the pathological microenvironment in injured tissue, and achieved better therapeutic effect.
Collapse
Affiliation(s)
- Haijun Hu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Huan Gao
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou 310027, China
| | - Kai Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Zeyuan Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Weiwei Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Qiaoxuan Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Yufang Yang
- Key Laboratory of Biomedical Engineering of Education Ministry, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Chaonan Yu
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou 310027, China
| | - Kedi Xu
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou 310027, China; Key Laboratory of Biomedical Engineering of Education Ministry, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China.
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China; Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing 312099, China.
| |
Collapse
|
3
|
Liu W, Liu L, Li H, Xie Y, Bai J, Guan J, Qi H, Sun J. Targeted pathophysiological treatment of ischemic stroke using nanoparticle-based drug delivery system. J Nanobiotechnology 2024; 22:499. [PMID: 39164747 PMCID: PMC11337765 DOI: 10.1186/s12951-024-02772-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024] Open
Abstract
Ischemic stroke poses significant challenges in terms of mortality and disability rates globally. A key obstacle to the successful treatment of ischemic stroke lies in the limited efficacy of administering therapeutic agents. Leveraging the unique properties of nanoparticles for brain targeting and crossing the blood-brain barrier, researchers have engineered diverse nanoparticle-based drug delivery systems to improve the therapeutic outcomes of ischemic stroke. This review provides a concise overview of the pathophysiological mechanisms implicated in ischemic stroke, encompassing oxidative stress, glutamate excitotoxicity, neuroinflammation, and cell death, to elucidate potential targets for nanoparticle-based drug delivery systems. Furthermore, the review outlines the classification of nanoparticle-based drug delivery systems according to these distinct physiological processes. This categorization aids in identifying the attributes and commonalities of nanoparticles that target specific pathophysiological pathways in ischemic stroke, thereby facilitating the advancement of nanomedicine development. The review discusses the potential benefits and existing challenges associated with employing nanoparticles in the treatment of ischemic stroke, offering new perspectives on designing efficacious nanoparticles to enhance ischemic stroke treatment outcomes.
Collapse
Affiliation(s)
- Wei Liu
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Lubin Liu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Hong Li
- Clinical Laboratory, Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital), Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, 266033, China
| | - Yutong Xie
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Ju Bai
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Jialiang Guan
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Hongzhao Qi
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China.
| | - Jinping Sun
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
4
|
Li M, Wang X, Qi B, Cui S, Zheng T, Guan Y, Ma L, Liu S, Li Q, Chen Z, Jian F. Treatment of Syringomyelia Characterized by Focal Dilatation of the Central Canal Using Mesenchymal Stem Cells and Neural Stem Cells. Tissue Eng Regen Med 2024; 21:625-639. [PMID: 38578425 PMCID: PMC11087409 DOI: 10.1007/s13770-024-00637-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 02/03/2024] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Syringomyelia is a progressive chronic disease that leads to nerve pain, sensory dissociation, and dyskinesia. Symptoms often do not improve after surgery. Stem cells have been widely explored for the treatment of nervous system diseases due to their immunoregulatory and neural replacement abilities. METHODS In this study, we used a rat model of syringomyelia characterized by focal dilatation of the central canal to explore an effective transplantation scheme and evaluate the effect of mesenchymal stem cells and induced neural stem cells for the treatment of syringomyelia. RESULTS The results showed that cell transplantation could not only promote syrinx shrinkage but also stimulate the proliferation of ependymal cells, and the effect of this result was related to the transplantation location. These reactions appeared only when the cells were transplanted into the cavity. Additionally, we discovered that cell transplantation transformed activated microglia into the M2 phenotype. IGF1-expressing M2 microglia may play a significant role in the repair of nerve pain. CONCLUSION Cell transplantation can promote cavity shrinkage and regulate the local inflammatory environment. Moreover, the proliferation of ependymal cells may indicate the activation of endogenous stem cells, which is important for the regeneration and repair of spinal cord injury.
Collapse
Affiliation(s)
- Mo Li
- Department of Neurosurgery, China International Neuroscience Institute, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Xinyu Wang
- Department of Neurosurgery, China International Neuroscience Institute, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Boling Qi
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Shengyu Cui
- Department of Neurosurgery, China International Neuroscience Institute, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Tianqi Zheng
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Yunqian Guan
- Department of Neurosurgery, China International Neuroscience Institute, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Longbing Ma
- Department of Neurosurgery, China International Neuroscience Institute, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Sumei Liu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, 100053, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Qian Li
- Department of Neurosurgery, China International Neuroscience Institute, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Zhiguo Chen
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Key Laboratory of Neurodegeneration, Ministry of Education, Beijing, 100053, China.
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, China.
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, China.
| | - Fengzeng Jian
- Department of Neurosurgery, China International Neuroscience Institute, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Research Center of Spine and Spinal Cord, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
5
|
Shen J, Gu X, Xiao C, Yan H, Feng Y, Li X. Genome-wide association analysis reveals potential genetic correlation and causality between circulating inflammatory proteins and amyotrophic lateral sclerosis. Aging (Albany NY) 2024; 16:9470-9484. [PMID: 38819224 PMCID: PMC11210256 DOI: 10.18632/aging.205878] [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: 01/26/2024] [Accepted: 04/18/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND Amyotrophic Lateral Sclerosis (ALS), a fatal neurodegenerative disease, continues to elude complete comprehension of its pathological underpinnings. Recent focus on inflammation in ALS pathogenesis prompts this investigation into the genetic correlation and potential causal relationships between circulating inflammatory proteins and ALS. METHODS Genome-wide association study (GWAS) data encompassing 91 circulating inflammatory protein measures from 14,824 individuals of European ancestry, alongside records from 27,205 ALS cases and 110,881 controls, were employed. Assessment of genetic correlation and overlap utilized LD score regression (LDSC), high-definition likelihood (HDL), and genetic analysis integrating pleiotropy and annotation (GPA) methodologies. Identification of shared genetic loci involved pleiotropy analysis, functional mapping and annotation (FUMA), and co-localization analysis. Finally, Mendelian randomization was applied to probe causal relationships between inflammatory proteins and ALS. RESULTS Our investigation revealed significant genetic correlation and overlap between ALS and various inflammatory proteins, including C-C motif chemokine 28, Interleukin-18, C-X-C motif chemokine 1, and Leukemia inhibitory factor receptor (LIFR). Pleiotropy analysis uncovered shared variations at specific genetic loci, some of which bore potential harm. Mendelian randomization analysis suggested that alterations in specific inflammatory protein levels, notably LIFR, could impact ALS risk. CONCLUSIONS Our findings uncover a genetic correlation between certain circulating inflammatory proteins and ALS, suggesting their possible causal involvement in ALS pathogenesis. Moreover, the identification of LIFR as a crucial protein may yield new insights into ALS pathomechanisms and offer a promising avenue for therapeutic interventions. These discoveries provide novel perspectives for advancing the comprehension of ALS pathophysiology and exploring potential therapeutic avenues.
Collapse
Affiliation(s)
- Jing Shen
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou 251221, China
| | - Xiaochu Gu
- Medical Laboratory, Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou 215137, China
| | - Chenxu Xiao
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou 251221, China
| | - Hanfei Yan
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou 251221, China
| | - Yu Feng
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou 251221, China
- The University of New South Wales, Sydney 2052, Australia
- The University of Melbourne, Melbourne 3010, Australia
| | - Xiaowei Li
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou 251221, China
| |
Collapse
|
6
|
Hu Y, Sun Y, Yuan H, Liu J, Chen L, Liu D, Xu Y, Zhou X, Ding L, Zhang Z, Xiong L, Xue L, Wang T. Vof16-miR-185-5p-GAP43 network improves the outcomes following spinal cord injury via enhancing self-repair and promoting axonal growth. CNS Neurosci Ther 2024; 30:e14535. [PMID: 38168094 PMCID: PMC11017428 DOI: 10.1111/cns.14535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/31/2023] [Accepted: 11/04/2023] [Indexed: 01/05/2024] Open
Abstract
INTRODUCTION Self-repair of spinal cord injury (SCI) has been found in humans and experimental animals with partial recovery of neurological functions. However, the regulatory mechanisms underlying the spontaneous locomotion recovery after SCI are elusive. AIMS This study was aimed at evaluating the pathological changes in injured spinal cord and exploring the possible mechanism related to the spontaneous recovery. RESULTS Immunofluorescence staining was performed to detect GAP43 expression in lesion site after spinal cord transection (SCT) in rats. Then RNA sequencing and gene ontology (GO) analysis were employed to predict lncRNA that correlates with GAP43. LncRNA smart-silencing was applied to verify the function of lncRNA vof16 in vitro, and knockout rats were used to evaluate its role in neurobehavioral functions after SCT. MicroRNA sequencing, target scan, and RNA22 prediction were performed to further explore the underlying regulatory mechanisms, and miR-185-5p stands out. A miR-185-5p site-regulated relationship with GAP43 and vof16 was determined by luciferase activity analysis. GAP43-silencing, miR-185-5p-mimic/inhibitor, and miR-185-5p knockout rats were also applied to elucidate their effects on spinal cord neurite growth and neurobehavioral function after SCT. We found that a time-dependent increase of GAP43 corresponded with the limited neurological recovery in rats with SCT. CRNA chip and GO analysis revealed lncRNA vof16 was the most functional in targeting GAP43 in SCT rats. Additionally, silencing vof16 suppressed neurite growth and attenuated the motor dysfunction in SCT rats. Luciferase reporter assay showed that miR-185-5p competitively bound the same regulatory region of vof16 and GAP43. CONCLUSIONS Our data indicated miR-185-5p could be a detrimental factor in SCT, and vof16 may function as a ceRNA by competitively binding miR-185-5p to modulate GAP43 in the process of self-recovery after SCT. Our study revealed a novel vof16-miR-185-5p-GAP43 regulatory network in neurological self-repair after SCT and may underlie the potential treatment target for SCI.
Collapse
Affiliation(s)
- Yue Hu
- Department of Anesthesiology, Institute of Neurological Disease, Translational Neuroscience Center, West China HospitalSichuan UniversityChengduChina
- Department of Anesthesia Operation, The First People's Hospital of Shuangliu DistrictWest China Airport Hospital of Sichuan UniversityChengduChina
| | - Yi‐Fei Sun
- Department of Anesthesiology, Institute of Neurological Disease, Translational Neuroscience Center, West China HospitalSichuan UniversityChengduChina
| | - Hao Yuan
- Laboratory Zoology Department, Institute of NeuroscienceKunming Medical UniversityKunmingChina
| | - Jia Liu
- Laboratory Zoology Department, Institute of NeuroscienceKunming Medical UniversityKunmingChina
| | - Li Chen
- Department of Anesthesiology, Institute of Neurological Disease, Translational Neuroscience Center, West China HospitalSichuan UniversityChengduChina
| | - Dong‐Hui Liu
- Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Yang Xu
- Department of Anesthesiology, Institute of Neurological Disease, Translational Neuroscience Center, West China HospitalSichuan UniversityChengduChina
| | - Xin‐Fu Zhou
- Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Li Ding
- Department of Anesthesiology, Institute of Neurological Disease, Translational Neuroscience Center, West China HospitalSichuan UniversityChengduChina
| | - Ze‐Tao Zhang
- Department of Anesthesiology, Institute of Neurological Disease, Translational Neuroscience Center, West China HospitalSichuan UniversityChengduChina
| | - Liu‐Lin Xiong
- Department of AnesthesiologyAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - Lu‐Lu Xue
- State Key Laboratory of BiotherapySichuan UniversityChengduSichuanChina
| | - Ting‐Hua Wang
- Department of Anesthesiology, Institute of Neurological Disease, Translational Neuroscience Center, West China HospitalSichuan UniversityChengduChina
- Laboratory Zoology Department, Institute of NeuroscienceKunming Medical UniversityKunmingChina
- State Key Laboratory of BiotherapySichuan UniversityChengduSichuanChina
| |
Collapse
|
7
|
Liu Y, Luo Z, Xie Y, Sun Y, Yuan F, Jiang L, Lu H, Hu J. Extracellular vesicles from UTX-knockout endothelial cells boost neural stem cell differentiation in spinal cord injury. Cell Commun Signal 2024; 22:155. [PMID: 38424563 PMCID: PMC10903014 DOI: 10.1186/s12964-023-01434-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/11/2023] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Vascular endothelial cells are pivotal in the pathophysiological progression following spinal cord injury (SCI). The UTX (Ubiquitously Transcribed Tetratripeptide Repeat on Chromosome X) serves as a significant regulator of endothelial cell phenotype. The manipulation of endogenous neural stem cells (NSCs) offers a compelling strategy for the amelioration of SCI. METHODS Two mouse models were used to investigate SCI: NSCs lineage-traced mice and mice with conditional UTX knockout (UTX KO) in endothelial cells. To study the effects of UTX KO on neural differentiation, we harvested extracellular vesicles (EVs) from both UTX KO spinal cord microvascular endothelial cells (SCMECs) and negative control SCMECs. These EVs were then employed to modulate the differentiation trajectory of endogenous NSCs in the SCI model. RESULTS In our NSCs lineage-traced mice model of SCI, a marked decrease in neurogenesis was observed post-injury. Notably, NSCs in UTX KO SCMECs mice showed enhanced neuronal differentiation compared to controls. RNA sequencing and western blot analyses revealed an upregulation of L1 cell adhesion molecule (L1CAM), a gene associated with neurogenesis, in UTX KO SCMECs and their secreted EVs. This aligns with the observed promotion of neurogenesis in UTX KO conditions. In vivo administration of L1CAM-rich EVs from UTX KO SCMECs (KO EVs) to the mice significantly enhanced neural differentiation. Similarly, in vitro exposure of NSCs to KO EVs resulted in increased activation of the Akt signaling pathway, further promoting neural differentiation. Conversely, inhibiting Akt phosphorylation or knocking down L1CAM negated the beneficial effects of KO EVs on NSC neuronal differentiation. CONCLUSIONS In conclusion, our findings substantiate that EVs derived from UTX KO SCMECs can act as facilitators of neural differentiation following SCI. This study not only elucidates a novel mechanism but also opens new horizons for therapeutic interventions in the treatment of SCI. Video Abstract.
Collapse
Affiliation(s)
- Yudong Liu
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zixiang Luo
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yong Xie
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yi Sun
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Feifei Yuan
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Liyuan Jiang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China.
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.
- Hunan Engineering Research Center of Sports and Health, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Hongbin Lu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China.
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.
- Hunan Engineering Research Center of Sports and Health, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Jianzhong Hu
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, China.
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China.
- Hunan Engineering Research Center of Sports and Health, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
8
|
Zhao Y, Ma J, Ding G, Wang Y, Yu H, Cheng X. Astragalus polysaccharides promote neural stem cells-derived oligodendrogenesis through attenuating CD8 +T cell infiltration in experimental autoimmune encephalomyelitis. Int Immunopharmacol 2024; 126:111303. [PMID: 38043269 DOI: 10.1016/j.intimp.2023.111303] [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: 07/25/2023] [Revised: 10/31/2023] [Accepted: 11/24/2023] [Indexed: 12/05/2023]
Abstract
Endogenous neural stem cells (NSCs) have the potential to generate remyelinating oligodendrocytes, which play an important role in multiple sclerosis (MS). However, the differentiation of NSCs into oligodendrocytes is insufficient, which is considered a major cause of remyelination failure. Our previous work reported that Astragalus polysaccharides (APS) had a neuroprotective effect on experimental autoimmune encephalomyelitis (EAE) mice. However, it remains unclear whether APS regulate NSCs differentiation in EAE mice. In this study, our data illustrated that APS administration could promote NSCs in the subventricular zone (SVZ) to differentiate into oligodendrocytes. Furthermore, we found that APS significantly improved neuroinflammation and inhibited CD8+T cell infiltration into SVZ of EAE mice. We also found that MOG35-55-specific CD8+T cells suppressed NSCs differentiation into oligodendrocytes by secreting IFN-γ, and APS facilitated the differentiation of NSCs into oligodendrocytes which was related to decreased IFN-γ secretion. In addition, APS treatment did not show a better effect on the NSCs-derived oligodendrogenesis after CD8+T cell depletion. This present study demonstrated that APS alleviated neuroinflammation and CD8+T cell infiltration into SVZ to induce oligodendroglial differentiation, and thus exerted neuroprotective effect. Our findings revealed that reducing the infiltration of CD8+T cells might contribute to enhancing NSCs-derived neurogenesis. And APS might be a promising drug candidate to treat MS.
Collapse
Affiliation(s)
- Yan Zhao
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Jinyun Ma
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Guiqing Ding
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yuanhua Wang
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Hua Yu
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Xiaodong Cheng
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| |
Collapse
|
9
|
Liu D, Shen H, Zhang K, Shen Y, Wen R, He X, Long G, Li X. Functional Hydrogel Co-Remolding Migration and Differentiation Microenvironment for Severe Spinal Cord Injury Repair. Adv Healthc Mater 2024; 13:e2301662. [PMID: 37937326 DOI: 10.1002/adhm.202301662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/25/2023] [Indexed: 11/09/2023]
Abstract
Spinal cord injury (SCI) activates nestin+ neural stem cells (NSCs), which can be regarded as potential seed cells for neuronal regeneration. However, the lesion microenvironment seriously hinders the migration of the nestin+ cells to the lesion epicenter and their differentiation into neurons to rebuild neural circuits. In this study, a photosensitive hydrogel scaffold is prepared as drug delivery carrier. Genetically engineered SDF1α and NT3 are designed and the scaffold is binary modified to reshape the lesion microenvironment. The binary modified scaffold can effectively induce the migration and neuronal differentiation of nestin+ NSCs in vitro. When implanted into a rat complete SCI model, many of the SCI-activated nestin+ cells migrate into the lesion site and give rise to neurons in short-term. Meanwhile, long-term repair results also show that implantation of the binary modified scaffold can effectively promote the maturation, functionalization and synaptic network reconstruction of neurons in the lesion site. In addition, animals treated with binary scaffold also showed better improvement in motor functions. The therapeutic strategy based on remolding the migration and neuronal differentiation lesion microenvironment provides a new insight into SCI repair by targeting activated nestin+ cells, which exhibits excellent clinical transformation prospects.
Collapse
Affiliation(s)
- Dingyang Liu
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan Province, 410078, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - He Shen
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Kai Zhang
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan Province, 410078, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Yeyu Shen
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan Province, 410078, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Runlin Wen
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan Province, 410078, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Xinghui He
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan Province, 410078, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| | - Ge Long
- Department of Anesthesia, the Third Xiangya Hospital, Central South University, Changsha, Hunan Province, 410078, China
| | - Xing Li
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, Hunan Province, 410078, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China
| |
Collapse
|
10
|
Li Z, Xu P, Shang L, Ma B, Zhang H, Fu L, Ou Y, Mao Y. 3D collagen porous scaffold carrying PLGA-PTX/SDF-1α recruits and promotes neural stem cell differentiation for spinal cord injury repair. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2023; 34:2332-2355. [PMID: 37566099 DOI: 10.1080/09205063.2023.2247715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/26/2023] [Accepted: 08/02/2023] [Indexed: 08/12/2023]
Abstract
Spinal Cord Injury (SCI), one of the major factors of disability, can cause irreversible motor and sensory impairment. There are no effective therapeutic drugs and technologies available in domestic or foreign countries currently. Neural stem cells (NSCs), with the potential for multidirectional differentiation, are a potential treatment for SCI. However, it has been demonstrated that NSCs primarily differentiated into astrocytes rather than neurons due to the inflammatory microenvironment, and the current challenge remains to direct the differentiation of NSCs into neurons in the lesion site. It was reported that the microtubule-stabilizing agent paclitaxel (PTX) was able to promote the differentiation of NSCs into neurons rather than astrocytes after SCI. SDF-1α can recruit NSCs and thus guide the migration of stem cells. In this study, we developed a functional collagen scaffold by loading SDF-1α and nanoparticle-encapsulated PLGA-PTX into a 3D collagen porous scaffold, allowing for slow release of PTX. When the functional scaffolds were implanted into the injury site, it provided a neural regeneration conduit channel for the migration of NSCs and neuronal differentiation. Neural regeneration promoted the recovery of motor function and reduced glial scar formation after SCI. In conclusion, a 3D collagen porous scaffold combined with PLGA-PTX and SDF-1α is a promising therapeutic strategy for SCI repair.
Collapse
Affiliation(s)
- Zhixiang Li
- School of Life Sciences, Bengbu Medical College, Bengbu, China
- Department of Orthopedics and Department of Plastic Surgery, The First Affiliated Hospital, Bengbu Medical College, Bengbu, China
| | - Panpan Xu
- Department of Orthopedics and Department of Plastic Surgery, The First Affiliated Hospital, Bengbu Medical College, Bengbu, China
| | - Lijun Shang
- School of Life Sciences, Bengbu Medical College, Bengbu, China
| | - Bingxu Ma
- Department of Orthopedics and Department of Plastic Surgery, The First Affiliated Hospital, Bengbu Medical College, Bengbu, China
| | - Huihui Zhang
- Department of Oncology, The First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Liangmin Fu
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Yuanyuan Ou
- School of Life Sciences, Bengbu Medical College, Bengbu, China
| | - Yingji Mao
- School of Life Sciences, Bengbu Medical College, Bengbu, China
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| |
Collapse
|
11
|
Miao L, Qing SW, Tao L. Huntingtin-associated protein 1 ameliorates neurological function rehabilitation by facilitating neurite elongation through TrKA-MAPK pathway in mice spinal cord injury. Front Mol Neurosci 2023; 16:1214150. [PMID: 37609072 PMCID: PMC10442162 DOI: 10.3389/fnmol.2023.1214150] [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: 04/29/2023] [Accepted: 07/24/2023] [Indexed: 08/24/2023] Open
Abstract
Aims Huntingtin-associated protein 1 (HAP1) is a neuronal protein closely associated with microtubules and might facilitate neurological function rehabilitation. This study aimed to investigate the effects of HAP1 on SCI and the underlying mechanisms. Methods the spinal cord injury (SCI) mouse model was induced by Allen's method. Then recombinant-HAP1 (r-HAP1) was administrated by intrathecal injection, and the BMS, Thermal nociceptive thresholds, tactile nociceptive thresholds, and neurofibrillary regeneration were identified to inspect the therapy outcome. Then NSCs were isolated from mice on embryonic day 14.5 and induced to differentiate into neurons. The efficiency of axon growth was calculated. Signaling pathway array was conducted to examine the signaling pathways in NSCs treated with r-HAP1. Antagonists and activators of TrkA were used to confirm the role of TrkA of HAP1 intervention both in vitro and in vivo. Results r-HAP1 ameliorates the neurological function rehabilitation after SCI, and benefits the regain of Tuj in injury spinal cord. Also significantly enhances neurite growth during neuronal differentiation of NSCs; Signaling pathway array and Western blot revealed that r-HAP1 significantly activates the phosphorylation of TrkA-MAPK/ERK in NSCs. TrkA selective inhibitor GW441756 blocks r-HAP1 on TrkA-MAPK/ERK signaling pathway and detracts from axonal growth after neuronal differentiation. TrkA selective activator gambogic amide can mimic the function of r-HAP1 by activating the foregoing pathway. ERK activator U-46619 reverses the blocking effect of GW441756 on r-HAP1. Conclusion HAP1 activates the TrkA-MAPK signaling pathway and is conducive to neurite elongation during NSC neuronal differentiation; by which to improve the prognosis of spinal cord injury in mice.
Collapse
Affiliation(s)
- Li Miao
- Department of Orthopedics, Hunan Children's Hospital, Changsha, China
- The School of Pediatrics, Hengyang Medical School, University of South China, Changsha, China
- Key Laboratory of Pediatric Bone Science of Hunan Province, Changsha, China
| | - Sun Wan Qing
- Hunan Rehabilitation Hospital Third Internal Department, Changsha, China
| | - Lu Tao
- Department of Neurosurgery, Changde Hospital, Xiangya School of Medicine, Central South University, Changde, China
| |
Collapse
|
12
|
Ding L, Chu W, Xia Y, Shi M, Li T, Zhou FQ, Deng DYB. UCHL1 facilitates protein aggregates clearance to enhance neural stem cell activation in spinal cord injury. Cell Death Dis 2023; 14:479. [PMID: 37507386 PMCID: PMC10382505 DOI: 10.1038/s41419-023-06003-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 07/12/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023]
Abstract
Activation of endogenous neural stem cells (NSCs) is greatly significant for the adult neurogenesis; however, it is extremely limited in the spinal cord after injury. Recent evidence suggests that accumulation of protein aggregates impairs the ability of quiescent NSCs to activate. Ubiquitin c-terminal hydrolase l-1 (UCHL1), an important deubiquitinating enzyme, plays critical roles in protein aggregations clearance, but its effects on NSC activation remains unknown. Here, we show that UCHL1 promotes NSC activation by clearing protein aggregates through ubiquitin-proteasome approach. Upregulation of UCHL1 facilitated the proliferation of spinal cord NSCs after spinal cord injury (SCI). Based on protein microarray analysis of SCI cerebrospinal fluid, it is further revealed that C3+ neurotoxic reactive astrocytes negatively regulated UCHL1 and proteasome activity via C3/C3aR signaling, led to increased abundances of protein aggregations and decreased NSC proliferation. Furthermore, blockade of reactive astrocytes or C3/C3aR pathway enhanced NSC activation post-SCI by reserving UCHL1 and proteasome functions. Together, this study elucidated a mechanism regulating NSC activation in the adult spinal cord involving the UCHL1-proteasome approach, which may provide potential molecular targets and new insights for NSC fate regulation.
Collapse
Affiliation(s)
- Lu Ding
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Weiwei Chu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Yu Xia
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Ming Shi
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Tian Li
- Obstetrics and Gynecology Department, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China
| | - Feng-Quan Zhou
- Department of Orthopaedic Surgery and Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA.
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| | - David Y B Deng
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
13
|
Shahemi NH, Mahat MM, Asri NAN, Amir MA, Ab Rahim S, Kasri MA. Application of Conductive Hydrogels on Spinal Cord Injury Repair: A Review. ACS Biomater Sci Eng 2023. [PMID: 37364251 DOI: 10.1021/acsbiomaterials.3c00194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Spinal cord injury (SCI) causes severe motor or sensory damage that leads to long-term disabilities due to disruption of electrical conduction in neuronal pathways. Despite current clinical therapies being used to limit the propagation of cell or tissue damage, the need for neuroregenerative therapies remains. Conductive hydrogels have been considered a promising neuroregenerative therapy due to their ability to provide a pro-regenerative microenvironment and flexible structure, which conforms to a complex SCI lesion. Furthermore, their conductivity can be utilized for noninvasive electrical signaling in dictating neuronal cell behavior. However, the ability of hydrogels to guide directional axon growth to reach the distal end for complete nerve reconnection remains a critical challenge. In this Review, we highlight recent advances in conductive hydrogels, including the incorporation of conductive materials, fabrication techniques, and cross-linking interactions. We also discuss important characteristics for designing conductive hydrogels for directional growth and regenerative therapy. We propose insights into electrical conductivity properties in a hydrogel that could be implemented as guidance for directional cell growth for SCI applications. Specifically, we highlight the practical implications of recent findings in the field, including the potential for conductive hydrogels to be used in clinical applications. We conclude that conductive hydrogels are a promising neuroregenerative therapy for SCI and that further research is needed to optimize their design and application.
Collapse
Affiliation(s)
- Nur Hidayah Shahemi
- Faculty of Applied Sciences, Universiti Teknologi MARA, 40450 Shah Alam, Selangor, Malaysia
| | - Mohd Muzamir Mahat
- Faculty of Applied Sciences, Universiti Teknologi MARA, 40450 Shah Alam, Selangor, Malaysia
| | - Nurul Ain Najihah Asri
- Faculty of Applied Sciences, Universiti Teknologi MARA, 40450 Shah Alam, Selangor, Malaysia
| | - Muhammad Abid Amir
- Faculty of Medicine, Sungai Buloh Campus, Universiti Teknologi MARA, 47000 Sungai Buloh, Selangor, Malaysia
| | - Sharaniza Ab Rahim
- Faculty of Medicine, Sungai Buloh Campus, Universiti Teknologi MARA, 47000 Sungai Buloh, Selangor, Malaysia
| | - Mohamad Arif Kasri
- Kulliyyah of Science, International Islamic University Malaysia, 25200 Kuantan, Pahang, Malaysia
| |
Collapse
|
14
|
Ge H, Zhou T, Zhang C, Cun Y, Chen W, Yang Y, Zhang Q, Li H, Zhong J, Zhang X, Feng H, Hu R. Targeting ASIC1a Promotes Neural Progenitor Cell Migration and Neurogenesis in Ischemic Stroke. RESEARCH (WASHINGTON, D.C.) 2023; 6:0105. [PMID: 37275123 PMCID: PMC10234266 DOI: 10.34133/research.0105] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/13/2023] [Indexed: 06/07/2023]
Abstract
Cell replacement therapy using neural progenitor cells (NPCs) has been shown to be an effective treatment for ischemic stroke. However, the therapeutic effect is unsatisfactory due to the imbalanced homeostasis of the local microenvironment after ischemia. Microenvironmental acidosis is a common imbalanced homeostasis in the penumbra and could activate acid-sensing ion channels 1a (ASIC1a), a subunit of proton-gated cation channels following ischemic stroke. However, the role of ASIC1a in NPCs post-ischemia remains elusive. Here, our results indicated that ASIC1a was expressed in NPCs with channel functionality, which could be activated by extracellular acidification. Further evidence revealed that ASIC1a activation inhibited NPC migration and neurogenesis through RhoA signaling-mediated reorganization of filopodia formation, which could be primarily reversed by pharmacological or genetic disruption of ASIC1a. In vivo data showed that the knockout of the ASIC1a gene facilitated NPC migration and neurogenesis in the penumbra to improve behavioral recovery after stroke. Subsequently, ASIC1a gain of function partially abrogated this effect. Moreover, the administration of ASIC1a antagonists (amiloride or Psalmotoxin 1) promoted functional recovery by enhancing NPC migration and neurogenesis. Together, these results demonstrate targeting ASIC1a is a novel strategy potentiating NPC migration toward penumbra to repair lesions following ischemic stroke and even for other neurological diseases with the presence of niche acidosis.
Collapse
Affiliation(s)
- Hongfei Ge
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
- Medical Research Center, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Tengyuan Zhou
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
- Medical Research Center, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Chao Zhang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
- Medical Research Center, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Yupeng Cun
- Pediatric Research Institute,
Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, 400014 Chongqing, China
| | - Weixiang Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Yang Yang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Qian Zhang
- Medical Research Center, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Huanhuan Li
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Jun Zhong
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Xuyang Zhang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Hua Feng
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Rong Hu
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
- Medical Research Center, Southwest Hospital,
Third Military Medical University (Army Medical University), 400038 Chongqing, China
| |
Collapse
|
15
|
Liu S, Ma L, Qi B, Li Q, Chen Z, Jian F. Suppression of TGFβR-Smad3 pathway alleviates the syrinx induced by syringomyelia. Cell Biosci 2023; 13:98. [PMID: 37248485 DOI: 10.1186/s13578-023-01048-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/06/2023] [Indexed: 05/31/2023] Open
Abstract
BACKGROUND Syringomyelia is a cerebrospinal fluid (CSF) disorder resulted in separation of pain and temperature, dilation of central canal and formation of syrinx in central canal. It is unclear about mechanisms of the dilation and syrinx formation. We aimed to investigate roles of ependymal cells lining central canal on the dilation, trying to reduce syrinx formation in central canal. METHODS We employed 78 Sprague-Dawley (SD) rats totally with syringomyelia to detect the contribution of ependymal cells to the dilation of central canal. Immunofluorescence was used to examine the activation of ependymal cells in 54 syringomyelia rat models. BrdU was used to indicate the proliferation of ependymal cells through intraperitoneal administration in 6 syringomyelia rat models. 18 rats with syringomyelia were injected with SIS3, an inhibitor of TGFβR-Smad3, and rats injected with DMSO were used as control. Among the 18 rats, 12 rats were used for observation of syrinx following SIS3 or DMSO administration by using magnetic resonance imaging (MRI) on day 14 and day 30 under syringomyelia without decompression. All the data were expressed as mean ± standard deviation (mean ± SD). Differences between groups were compared using the two-tailed Student's t-test or ANOVA. Differences were considered significant when *p < 0.05. RESULTS Our study showed the dilation and protrusions of central canal on day 5 and enlargement from day 14 after syringomyelia induction in rats with activation of ependymal cells lining central canal. Moreover, the ependymal cells contributed to protrusion formation possibly through migration along with central canal. Furthermore, suppression of TGFβR-Smad3 which was crucial for migration reversed the size of syrnix in central canal without treatment of decompression, suggesting TGFβR-Smad3 signal might be key for dilation of central canal and formation of syrinx. CONCLUSIONS The size of syrinx was decreased after SIS3 administration without decompression. Our study depicted the mechanisms of syrinx formation and suggested TGFβR-Smad3 signal might be key for dilation of central canal and formation of syrinx.
Collapse
Affiliation(s)
- Sumei Liu
- Department of Neurosurgery, China International Neuroscience Institute, Xuanwu Hospital Capital Medical University, 45 Changchun Street, Beijing, 100053, China
- Cell Therapy Center, Xuanwu Hospital Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Longbing Ma
- Department of Neurosurgery, China International Neuroscience Institute, Xuanwu Hospital Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Boling Qi
- Cell Therapy Center, Xuanwu Hospital Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Qian Li
- Department of Neurosurgery, China International Neuroscience Institute, Xuanwu Hospital Capital Medical University, 45 Changchun Street, Beijing, 100053, China
| | - Zhiguo Chen
- Cell Therapy Center, Xuanwu Hospital Capital Medical University, 45 Changchun Street, Beijing, 100053, China.
| | - Fengzeng Jian
- Department of Neurosurgery, China International Neuroscience Institute, Xuanwu Hospital Capital Medical University, 45 Changchun Street, Beijing, 100053, China.
- Spine Center, China International Neuroscience Institute (CHINA-INI), Beijing, China.
- Lab of Spinal Cord Injury and Functional Reconstruction, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Research Center of Spine and Spinal Cord, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China.
- National Center for Neurological Disorders, Beijing, China.
| |
Collapse
|
16
|
Inhibition of Neural Stem Cell Necroptosis Mediated by RIPK1/MLKL Promotes Functional Recovery After SCI. Mol Neurobiol 2023; 60:2135-2149. [PMID: 36602703 DOI: 10.1007/s12035-022-03156-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/26/2022] [Indexed: 01/06/2023]
Abstract
Endogenous neural stem cells (eNSCs) are a new therapeutic strategy for the noninvasive repair of spinal cord injury (SCI). Necroptosis is a necrosome-dependent cell death process that serves as a significant regulatory mechanism in SCI. Current research shows that neurons, oligodendrocytes, and astrocytes all undergo necroptosis after SCI. However, it is unclear whether eNSCs are associated with necroptosis after SCI. By performing immunofluorescence analysis, we found that eNSCs undergo necroptosis during spinal cord injury repair in mice. Our present work demonstrates that receptor-interacting protein kinase 1 (RIPK1)/mixed lineage kinase domain-like protein (MLKL) are involved in necroptosis pathway in SCI mice. In vitro, the necroptosis induced by TNF-α/Smac-mimetic/Z-VAD-FMK (TSZ) treatment regulates phenotype of NSCs. In detail, the proliferative capacity of NSCs was significantly decreased in the presence of continual TSZ treatment, and the transcription of proinflammatory genes was upregulated, while the transcription of neurotrophic factors was inhibited. NSCs exhibited an obvious tendency to differentiate into glial cells under short-duration TSZ stimulation (6 h and 12 h); as the stimulus duration increased (24 h), the differentiation ability of the NSCs was significantly inhibited. These phenotypic changes are not conducive to neural cell survival and neural repair. Moreover, we examined the effect of necroptosis inhibitors on TSZ-treated NSCs. Necrostatin-1 and necrosulfonamide significantly reduced the necroptosis of NSCs after TSZ treatment and improved the phenotypic function of NSCs under TSZ stimulation. In additional in vivo experiments, after 2 weeks of administration, the necroptosis inhibitors reduced the necroptosis of NSCs and improved functional recovery in SCI mice. Taken together, these data indicate that the inhibition of NSC necroptosis with necroptosis inhibitors facilitates survival and phenotype maintenance in vitro and contributes to neuroprotection and repair in vivo. Our findings suggest that blocking necroptosis of eNSCs may be a potential therapeutic strategy for treating SCI.
Collapse
|
17
|
Hachem LD, Hong J, Velumian A, Mothe AJ, Tator CH, Fehlings MG. Excitotoxic glutamate levels drive spinal cord ependymal stem cell proliferation and fate specification through CP-AMPAR signaling. Stem Cell Reports 2023; 18:672-687. [PMID: 36764296 PMCID: PMC10031285 DOI: 10.1016/j.stemcr.2023.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 01/11/2023] [Accepted: 01/11/2023] [Indexed: 02/11/2023] Open
Abstract
The adult spinal cord contains a population of ependymal-derived neural stem/progenitor cells (epNSPCs) that are normally quiescent, but are activated to proliferate, differentiate, and migrate after spinal cord injury. The mechanisms that regulate their response to injury cues, however, remain unknown. Here, we demonstrate that excitotoxic levels of glutamate promote the proliferation and astrocytic fate specification of adult spinal cord epNSPCs. We show that glutamate-mediated calcium influx through calcium-permeable alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors (CP-AMPARs) in concert with Notch signaling increases the proliferation of epNSPCs via pCREB, and induces astrocytic differentiation through Hes1 upregulation. Furthermore, the in vivo targeting of this pathway via positive modulation of AMPARs after spinal cord injury enhances epNSPC proliferation, astrogliogenesis, neurotrophic factor production and increases neuronal survival. Our study uncovers an important mechanism by which CP-AMPARs regulate the growth and phenotype of epNSPCs, which can be targeted therapeutically to harness the regenerative potential of these cells after injury.
Collapse
Affiliation(s)
- Laureen D Hachem
- Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON M5T 2S8, Canada
| | - James Hong
- Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada
| | - Alexander Velumian
- Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON M5T 2S8, Canada
| | - Andrea J Mothe
- Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada
| | - Charles H Tator
- Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON M5T 2S8, Canada.
| | - Michael G Fehlings
- Krembil Research Institute, University Health Network, Toronto, ON M5T 2S8, Canada; Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON M5T 2S8, Canada.
| |
Collapse
|
18
|
Su QS, Zhuang DL, Nasser MI, Sai X, Deng G, Li G, Zhu P. Stem Cell Therapies for Restorative Treatments of Central Nervous System Ischemia-Reperfusion Injury. Cell Mol Neurobiol 2023; 43:491-510. [PMID: 35129759 DOI: 10.1007/s10571-022-01204-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 02/01/2022] [Indexed: 11/27/2022]
Abstract
Ischemic damage to the central nervous system (CNS) is a catastrophic postoperative complication of aortic occlusion subsequent to cardiovascular surgery that can cause brain impairment and sometimes even paraplegia. Over recent years, numerous studies have investigated techniques for protecting and revascularizing the nervous system during intraoperative ischemia; however, owing to a lack of knowledge of the physiological distinctions between the brain and spinal cord, as well as the limited availability of testing techniques and treatments for ischemia-reperfusion injury, the cause of brain and spinal cord ischemia-reperfusion injury remains poorly understood, and no adequate response steps are currently available in the clinic. Given the limited ability of the CNS to repair itself, it is of great clinical value to make full use of the proliferative and differentiation potential of stem cells to repair nerves in degenerated and necrotic regions by stem cell transplantation or mobilization, thereby introducing a novel concept for the treatment of severe CNS ischemia-reperfusion injury. This review summarizes the most recent advances in stem cell therapy for ischemia-reperfusion injury in the brain and spinal cord, aiming to advance basic research and the clinical use of stem cell therapy as a promising treatment for this condition.
Collapse
Affiliation(s)
- Qi-Song Su
- Medical Research Center, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, Guangdong, China.,School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510080, Guangdong, China
| | - Dong-Lin Zhuang
- Medical Research Center, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, Guangdong, China.,College of Medicine, Shantou University, Shantou, 515063, Guangdong, China
| | - Moussa Ide Nasser
- Medical Research Center, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, Guangdong, China
| | - Xiyalatu Sai
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao City, 028000, Inner Mongolia, China
| | - Gang Deng
- Medical Research Center, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, Guangdong, China.,School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
| | - Ge Li
- Medical Research Center, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, Guangdong, China. .,School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510080, Guangdong, China.
| | - Ping Zhu
- Medical Research Center, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, Guangdong, China. .,School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510080, Guangdong, China. .,College of Medicine, Shantou University, Shantou, 515063, Guangdong, China. .,Guangdong Provincial Key Laboratory of Structural Heart Disease, Guangzhou, 510100, Guangdong, China. .,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China. .,Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao City, 028000, Inner Mongolia, China.
| |
Collapse
|
19
|
Hu H, Chen X, Zhao K, Zheng W, Gao C. Recent Advances in Biomaterials-Based Therapies for Alleviation and Regeneration of Traumatic Brain Injury. Macromol Biosci 2023; 23:e2200577. [PMID: 36758541 DOI: 10.1002/mabi.202200577] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/27/2023] [Indexed: 02/11/2023]
Abstract
Traumatic brain injury (TBI), a major public health problem accompanied with numerous complications, usually leads to serve disability and huge financial burden. The adverse and unfavorable pathological environment triggers a series of secondary injuries, resulting in serious loss of nerve function and huge obstacle of endogenous nerve regeneration. With the advances in adaptive tissue regeneration biomaterials, regulation of detrimental microenvironment to reduce the secondary injury and to promote the neurogenesis becomes possible. The adaptive biomaterials could respond and regulate biochemical, cellular, and physiological events in the secondary injury, including excitotoxicity, oxidative stress, and neuroinflammation, to rebuild circumstances suitable for regeneration. In this review, the development of pathology after TBI is discussed, followed by the introduction of adaptive biomaterials based on various pathological characteristics. The adaptive biomaterials carried with neurotrophic factors and stem cells for TBI treatment are then summarized. Finally, the current drawbacks and future perspective of biomaterials for TBI treatment are suggested.
Collapse
Affiliation(s)
- Haijun Hu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xiping Chen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Kefei Zhao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Weiwei Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.,Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China
| |
Collapse
|
20
|
Transplanting neurofibromatosis-1 gene knockout neural stem cells improve functional recovery in rats with spinal cord injury by enhancing the mTORC2 pathway. Exp Mol Med 2022; 54:1766-1777. [PMID: 36241865 PMCID: PMC9636387 DOI: 10.1038/s12276-022-00850-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 05/25/2022] [Accepted: 07/19/2022] [Indexed: 12/29/2022] Open
Abstract
The poor survival and low efficiency of neuronal differentiation limits the therapeutic effects of transplanted neural stem cells in the treatment of spinal cord injury. Neurofibromatosis-1 (NF-1) is a tumor suppressor gene that restricts the rapid and abnormal growth and differentiation of neural cells. In the present study, lentiviral vectors were used to knock out NF-1, Ricotr (the core member of mTORC2) or NF-1+Ricotr in neural stem cells in vitro, and the NF-1, Ricotr or NF-1+Ricotr knockout neural stem cells were transplanted at the lesion site in a rat model of spinal cord injury (SCI). We first demonstrated that targeted knockout of NF-1 had an antiapoptotic effect and improved neuronal differentiation by enhancing the mTORC2/Rictor pathway of neural stem cells in vitro. Subsequently, transplanting NF-1 knockout neural stem cells into the injured site sufficiently promoted the tissue repair and functional recovery of rats with spinal cord injury by enhancing the survival and neuronal differentiation of grafted neural stem cells. Collectively, these findings reveal a prominent role of NF-1 in neural stem cell biology, which is an invaluable step forward in enhancing the benefit of neural stem cell-mediated regenerative cell therapy for spinal cord injury and identifies the transplantation of NF-1 knockout neural stem cells as a promising strategy for spinal cord injury.
Collapse
|
21
|
Guo W, Zhang X, Zhai J, Xue J. The roles and applications of neural stem cells in spinal cord injury repair. Front Bioeng Biotechnol 2022; 10:966866. [PMID: 36105599 PMCID: PMC9465243 DOI: 10.3389/fbioe.2022.966866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 07/28/2022] [Indexed: 12/05/2022] Open
Abstract
Spinal cord injury (SCI), which has no current cure, places a severe burden on patients. Stem cell-based therapies are considered promising in attempts to repair injured spinal cords; such options include neural stem cells (NSCs). NSCs are multipotent stem cells that differentiate into neuronal and neuroglial lineages. This feature makes NSCs suitable candidates for regenerating injured spinal cords. Many studies have revealed the therapeutic potential of NSCs. In this review, we discuss from an integrated view how NSCs can help SCI repair. We will discuss the sources and therapeutic potential of NSCs, as well as representative pre-clinical studies and clinical trials of NSC-based therapies for SCI repair.
Collapse
Affiliation(s)
- Wen Guo
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xindan Zhang
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, China
| | - Jiliang Zhai
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Beijing, China
- *Correspondence: Jiliang Zhai, ; Jiajia Xue,
| | - Jiajia Xue
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, China
- *Correspondence: Jiliang Zhai, ; Jiajia Xue,
| |
Collapse
|
22
|
Xie J, Li J, Ma J, Li M, Wang X, Fu X, Ma Y, Yang H, Li B, Saijilafu. Magnesium Oxide/Poly(l-lactide-co-ε-caprolactone) Scaffolds Loaded with Neural Morphogens Promote Spinal Cord Repair through Targeting the Calcium Influx and Neuronal Differentiation of Neural Stem Cells. Adv Healthc Mater 2022; 11:e2200386. [PMID: 35587044 PMCID: PMC11469078 DOI: 10.1002/adhm.202200386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/22/2022] [Indexed: 11/08/2022]
Abstract
Because of the limited regenerative ability of the central nervous system (CNS), effective treatments for spinal cord injury (SCI) are still lacking. After SCI, neuron loss and axon regeneration failure often result in irreversible functional impairment. The calcium overload induced by the N-methyl-D-aspartate receptor (NMDAR) overactivation is critical for cell death in SCI. It has been reported that the magnesium ion (Mg2+ ) can competitively block the NMDAR and reduce the calcium influx, and that sonic hedgehog (Shh) and retinoic acid (RA) are the critical regulators of neuronal differentiation of endogenous neural stem cells (NSCs). Here, magnesium oxide (MgO)/poly (l-lactide-co-ε-caprolactone) (PLCL) scaffold loaded with purmorphamine (PUR, a Shh signaling agonist) and RA is developed and its feasibility in SCI repair is tested. The results showed that the Mg2+ released from MgO attenuated cell apoptosis by blocking the calcium influx, and the PUR/RA promoted the recruitment and neuronal differentiation of endogenous NSCs, thereby reducing the glial scar formation at the SCI lesion site. Furthermore, implantation of PUR/RA-loaded MgO/PLCL scaffold facilitates the partial recovery of a locomotor function of SCI mouse in vivo. Together, findings from this study imply that PUR/RA-loaded MgO/PLCL scaffold may be a promising biomaterial for the clinical treatment of SCI.
Collapse
Affiliation(s)
- Jile Xie
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow University899 Pinghai RoadSuzhouJiangsu215006China
| | - Jiaying Li
- Orthopaedic InstituteMedical CollegeSoochow University1 Shizi RoadSuzhouJiangsu215006China
| | - Jinjin Ma
- Orthopaedic InstituteMedical CollegeSoochow University1 Shizi RoadSuzhouJiangsu215006China
| | - Meimei Li
- Orthopaedic InstituteMedical CollegeSoochow University1 Shizi RoadSuzhouJiangsu215006China
| | - Xingran Wang
- Orthopaedic InstituteMedical CollegeSoochow University1 Shizi RoadSuzhouJiangsu215006China
| | - Xinya Fu
- Orthopaedic InstituteMedical CollegeSoochow University1 Shizi RoadSuzhouJiangsu215006China
| | - Yanxia Ma
- Orthopaedic InstituteMedical CollegeSoochow University1 Shizi RoadSuzhouJiangsu215006China
| | - Huilin Yang
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow University899 Pinghai RoadSuzhouJiangsu215006China
- Orthopaedic InstituteMedical CollegeSoochow University1 Shizi RoadSuzhouJiangsu215006China
| | - Bin Li
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow University899 Pinghai RoadSuzhouJiangsu215006China
- Orthopaedic InstituteMedical CollegeSoochow University1 Shizi RoadSuzhouJiangsu215006China
| | - Saijilafu
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow University899 Pinghai RoadSuzhouJiangsu215006China
- Orthopaedic InstituteMedical CollegeSoochow University1 Shizi RoadSuzhouJiangsu215006China
| |
Collapse
|
23
|
Hou Y, Luo D, Hou Y, Luan J, Zhan J, Chen Z, E S, Xu L, Lin D. Bu Shen Huo Xue decoction promotes functional recovery in spinal cord injury mice by improving the microenvironment to promote axonal regeneration. Chin Med 2022; 17:85. [PMID: 35820953 PMCID: PMC9277908 DOI: 10.1186/s13020-022-00639-y] [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: 01/10/2022] [Accepted: 06/28/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Bu-Shen-Huo-Xue (BSHX) decoction has been used in the postoperative rehabilitation of patients with spinal cord injury in China. In the present study, we aim to reveal the bioactive compounds in BSHX decoction and comprehensively explore the effects of BSHX decoction and the underlying mechanism in spinal cord injury recovery. METHODS The main chemical constituents in BSHX decoction were determined by UPLC-MS/MS. SCI mice were induced by a pneumatic impact device at T9-T10 level of the vertebra, and treated with BSHX decoction. Basso-Beattie-Bresnahan (BBB) score, footprint analysis, hematoxylin-eosin (H&E) staining, Nissl staining and a series of immunofluorescence staining were performed to investigate the functional recovery, glial scar formation and axon regeneration after BSHX treatment. Immunofluorescent staining of bromodeoxyuridine (BrdU), neuronal nuclei (NeuN) and glial fibrillary acidic protein (GFAP) was performed to evaluate the effect of BSHX decoction on neural stem cells (NSCs) proliferation and differentiation. RESULTS We found that the main compounds in BSHX decoction were Gallic acid, 3,4-Dihydroxybenzaldehyde, (+)-Catechin, Paeoniflorin, Rosmarinic acid, and Diosmetin. BSHX decoction improved the pathological findings in SCI mice through invigorating blood circulation and cleaning blood stasis in the lesion site. In addition, it reduced tissue damage and neuron loss by inhibiting astrocytes activation, and promoting the polarization of microglia towards M2 phenotype. The functional recovery test revealed that BSHX treatment improved the motor function recovery post SCI. CONCLUSIONS Our study provided evidence that BSHX treatment could improve the microenvironment of the injured spinal cord to promote axonal regeneration and functional recovery in SCI mice.
Collapse
Affiliation(s)
- Yonghui Hou
- Department of Orthopedic Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, 510120, Guangdong, People's Republic of China.,Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou, 510405, Guangdong, People's Republic of China.,Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China
| | - Dan Luo
- Department of Orthopedic Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, 510120, Guangdong, People's Republic of China.,Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou, 510405, Guangdong, People's Republic of China.,Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China
| | - Yu Hou
- Department of Orthopedic Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, 510120, Guangdong, People's Republic of China.,Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou, 510405, Guangdong, People's Republic of China.,Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China
| | - Jiyao Luan
- Department of Orthopedic Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, 510120, Guangdong, People's Republic of China.,Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou, 510405, Guangdong, People's Republic of China.,Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China
| | - Jiheng Zhan
- Department of Orthopedic Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, 510120, Guangdong, People's Republic of China.,Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou, 510405, Guangdong, People's Republic of China.,Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China
| | - Zepeng Chen
- Department of Orthopedic Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, 510120, Guangdong, People's Republic of China.,Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou, 510405, Guangdong, People's Republic of China.,Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China
| | - Shunmei E
- Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou, 510405, Guangdong, People's Republic of China.,Department of Laboratory Medicine, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou Higher Education Mega Center, 55 Neihuan Xi Road, Panyu District, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Liangliang Xu
- Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou, 510405, Guangdong, People's Republic of China. .,Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China. .,Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China.
| | - Dingkun Lin
- Department of Orthopedic Surgery, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, 510120, Guangdong, People's Republic of China. .,Guangzhou University of Chinese Medicine, No. 12, Jichang Road, Baiyun District, Guangzhou, 510405, Guangdong, People's Republic of China.
| |
Collapse
|
24
|
Wang Y, Yuan H. Research progress of endogenous neural stem cells in spinal cord injury. IBRAIN 2022; 8:199-209. [PMID: 37786888 PMCID: PMC10529172 DOI: 10.1002/ibra.12048] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 05/06/2022] [Accepted: 05/17/2022] [Indexed: 10/04/2023]
Abstract
Spinal cord injury (SCI) is a severe disabling disease, which mainly manifests as impairments of sensory and motor functions, sexual function, bladder and intestinal functions, respiratory and cardiac functions below the injury plane. In addition, the condition has a profound effect on the mental health of patients, which often results in severe sequelae. Some patients may be paraplegic for life or even die, which places a huge burden on the family and society. There is still no effective treatment for SCI. Studies have confirmed that endogenous neural stem cells (ENSCs), as multipotent neural stem cells, which are located in the ependymal region of the central canal of the adult mammalian spinal cord, are activated after SCI and then differentiate into various nerve cells to promote endogenous repair and regeneration. However, the central canal of the spinal cord is often occluded to varying degrees in adults, and residual ependymal cells cannot be activated and do not proliferate after SCI. Besides, the destruction of the microenvironment after SCI is also an important factor that affects the proliferation and differentiation of ENSCs and spinal cord repair. Therefore, this review describes the role of ENSCs in SCI, in terms of the origin, transformation, treatment, and influencing factors, to provide new ideas for clinical treatment of SCI.
Collapse
Affiliation(s)
- Ya‐Ting Wang
- Department of AnesthesiologySouthwest Medical UniversityLuzhouSichuanChina
| | - Hao Yuan
- Institute of NeuroscienceKunming Medical UniversityKunmingYunnanChina
| |
Collapse
|
25
|
Click chemistry extracellular vesicle/peptide/chemokine nanomissiles for treating central nervous systems injuries. Acta Pharm Sin B 2022; 13:2202-2218. [DOI: 10.1016/j.apsb.2022.06.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 12/19/2022] Open
|
26
|
Jiang Q, Tao B, Gao G, Sun M, Wang H, Li J, Wang Z, Shang A. Filum Terminale: A Comprehensive Review with Anatomical, Pathological, and Surgical Considerations. World Neurosurg 2022; 164:167-176. [PMID: 35500871 DOI: 10.1016/j.wneu.2022.04.098] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/22/2022] [Accepted: 04/23/2022] [Indexed: 11/28/2022]
Abstract
The conus medullaris is the distal tapering end of the spinal cord, and the filum terminale (FT) is regarded as a bundle of non-functional fibrous tissue; therefore, some scholars call it the spinal ligament, while others describe the human FT as "remnants of the spinal cord." It was later found that in the human spinal cord, the FT is composed of an intradural segment and an epidural segment, and the end of the FT is connected to the coccyx periosteum. Because some nerve tissue is also found in the FT, as research progresses, FT may have the potential for transplantation. A lack of exhaustive overviews on the FT in the present literature prompted us to conduct this review. Considering that a current comprehensive review seemed to be the need of the hour, herein, we attempted to summarize previous research and theories on the FT, elucidate its anatomy, and understand its pathological involvement in various diseases.
Collapse
Affiliation(s)
- Qingyu Jiang
- Chinese PLA Medical School, Beijing 100853, China
| | - Benzhang Tao
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing 100853, China; Tianjin Medical University
| | - Gan Gao
- Chinese PLA Medical School, Beijing 100853, China
| | - Mengchun Sun
- Chinese PLA Medical School, Beijing 100853, China; Medical School, Nankai University, Nankai District, Tianjin, China
| | - Hui Wang
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing 100853, China
| | - Junyang Li
- Chinese PLA Medical School, Beijing 100853, China; Medical School, Nankai University, Nankai District, Tianjin, China
| | | | - Aijia Shang
- Department of Neurosurgery, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
27
|
Laycock C, Kieser D, Fitz-Gerald C, Soltani S, Frampton C. A systematic review of large animal and human studies of stem cell therapeutics for acute adult traumatic spinal cord injury. JOURNAL OF ORTHOPAEDICS, TRAUMA AND REHABILITATION 2022. [DOI: 10.1177/22104917221087401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Traumatic spinal cord injury (TSCI) is a devastating condition and the search for a cure remains one of the most tenacious healthcare challenges to date. Current therapies are limited in their efficacy to restore full neurological function – resulting in lifelong disability and loss of autonomy. Whilst there remains a necessity to refine therapeutic protocols, stem cell (SC) studies have shown promise in the mending and re-establishment of the spinal cord neuroanatomy. Objectives: We conducted a systematic review of functional outcomes in stem cell therapeutics over the last three decades in large animals and humans. Methods: Medline, Embase, Cochrane and SCOPUS databases were searched for potentially pertinent articles from 1990 to 2020. Studies published in English were included if the stem cells were directly injected into the intraspinal, epidural or intrathecal compartments within two weeks of a traumatic mechanism of injury, including acute intervertebral disc prolapse. The participants were either large animals – defined as canine, porcine or non-human primate in-vivo models – or human patients. Results: Nine studies were included in this review. Statistically significant improvements in motor function and deep pain perception were seen at 8 weeks to 6 months post-SC injection compared to controls. Limitations: Functional outcomes are variably measured across studies. Almost all studies used experimentally induced trauma, which may not accurately represent the complexity of human spinal cord injury. Due to the exclusion criteria, there were no non-human primate studies included, yet these animal models are considered a closer anatomical match to humans than other large mammals. No human studies were included. Conclusions and Implications: Autologous and allogeneic stem cells have been trialled for the reconstitution of damaged and lost cells, remyelination of axons and remodelling of the pathophysiological microenvironment within the injured spinal cord, with some promising outcome data. This may translate to more successful future Phase I/II human clinical trials into the use of stem cells after TSCI in adults.
Collapse
Affiliation(s)
- Charlotte Laycock
- University of Oxford Medical School, John Radcliffe Hospital, Oxford, UK
| | - David Kieser
- Department of Orthopaedics and Musculoskeletal Medicine, University of Otago, Christchurch School of Medicine, Christchurch, New Zealand
| | - Connor Fitz-Gerald
- Department of Orthopaedics and Musculoskeletal Medicine, University of Otago, Christchurch School of Medicine, Christchurch, New Zealand
| | - Sherry Soltani
- University of Oxford Medical School, John Radcliffe Hospital, Oxford, UK
| | - Chris Frampton
- Department of Orthopaedics and Musculoskeletal Medicine, University of Otago, Christchurch School of Medicine, Christchurch, New Zealand
| |
Collapse
|
28
|
Becker T, Becker CG. Regenerative neurogenesis: the integration of developmental, physiological and immune signals. Development 2022; 149:275248. [PMID: 35502778 PMCID: PMC9124576 DOI: 10.1242/dev.199907] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In fishes and salamanders, but not mammals, neural stem cells switch back to neurogenesis after injury. The signalling environment of neural stem cells is strongly altered by the presence of damaged cells and an influx of immune, as well as other, cells. Here, we summarise our recently expanded knowledge of developmental, physiological and immune signals that act on neural stem cells in the zebrafish central nervous system to directly, or indirectly, influence their neurogenic state. These signals act on several intracellular pathways, which leads to changes in chromatin accessibility and gene expression, ultimately resulting in regenerative neurogenesis. Translational approaches in non-regenerating mammals indicate that central nervous system stem cells can be reprogrammed for neurogenesis. Understanding signalling mechanisms in naturally regenerating species show the path to experimentally promoting neurogenesis in mammals.
Collapse
Affiliation(s)
- Thomas Becker
- Center for Regenerative Therapies at the TU Dresden, Technische Universität Dresden, 01307 Dresden, Germany.,Centre for Discovery Brain Sciences, University of Edinburgh Medical School, Biomedical Science, Edinburgh, EH16 4SB, Scotland
| | - Catherina G Becker
- Center for Regenerative Therapies at the TU Dresden, Technische Universität Dresden, 01307 Dresden, Germany.,Centre for Discovery Brain Sciences, University of Edinburgh Medical School, Biomedical Science, Edinburgh, EH16 4SB, Scotland
| |
Collapse
|
29
|
Coupe D, Bossing T. Insights into nervous system repair from the fruit fly. Neuronal Signal 2022; 6:NS20210051. [PMID: 35474685 PMCID: PMC9008705 DOI: 10.1042/ns20210051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 11/17/2022] Open
Abstract
Millions of people experience injury to the central nervous system (CNS) each year, many of whom are left permanently disabled, providing a challenging hurdle for the field of regenerative medicine. Repair of damage in the CNS occurs through a concerted effort of phagocytosis of debris, cell proliferation and differentiation to produce new neurons and glia, distal axon/dendrite degeneration, proximal axon/dendrite regeneration and axon re-enwrapment. In humans, regeneration is observed within the peripheral nervous system, while in the CNS injured axons exhibit limited ability to regenerate. This has also been described for the fruit fly Drosophila. Powerful genetic tools available in Drosophila have allowed the response to CNS insults to be probed and novel regulators with mammalian orthologs identified. The conservation of many regenerative pathways, despite considerable evolutionary separation, stresses that these signals are principal regulators and may serve as potential therapeutic targets. Here, we highlight the role of Drosophila CNS injury models in providing key insight into regenerative processes by exploring the underlying pathways that control glial and neuronal activation in response to insult, and their contribution to damage repair in the CNS.
Collapse
Affiliation(s)
- David Coupe
- Peninsula Medical School, University of Plymouth, John Bull Building, 16 Research Way, Plymouth PL6 8BU, U.K
| | - Torsten Bossing
- Peninsula Medical School, University of Plymouth, John Bull Building, 16 Research Way, Plymouth PL6 8BU, U.K
| |
Collapse
|
30
|
Chen Z, Zhang H, Fan C, Zhuang Y, Yang W, Chen Y, Shen H, Xiao Z, Zhao Y, Li X, Dai J. Adhesive, Stretchable, and Spatiotemporal Delivery Fibrous Hydrogels Harness Endogenous Neural Stem/Progenitor Cells for Spinal Cord Injury Repair. ACS NANO 2022; 16:1986-1998. [PMID: 34842412 DOI: 10.1021/acsnano.1c06892] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Aligned fibrous hydrogels capable of recruiting endogenous neural stem/progenitor cells (NSPCs) show great promise in spinal cord injury (SCI) repair. However, the hydrogels suffer from severe issues in close contact with the transected nerve stumps and harnessing the NSPC fate in the lesion microenvironment. Herein, we report aligned collagen-fibrin (Col-FB) fibrous hydrogels with stretchable property, adhesive behavior, and stromal cell-derived factor-1α (SDF1α)/paclitaxel (PTX) spatiotemporal delivery capability. The resultant Col-FB fibrous hydrogels exhibited 1.98 times longer elongation at break (230%), 2.55 times lower Young's modulus (17.93 ± 1.16 KPa), and 2.21 times greater adhesive strength (3.45 ± 0.48 KPa) than collagen (Col) fibrous hydrogels. The soft aligned fibrous hydrogels simulate the oriented microstructure and soft tissue feature of a natural spinal cord and provide elasticity and adhesivity to ensure a persistent close contact with host stumps. The repair of complete transection SCI in rats demonstrates that "middle-to-bilateral" SDF1α gradient release induced endogenous NSPC migration to the lesion site in 10 days, and SDF1α/PTX sequential release promoted neuronal differentiation of the recruited NSPCs over 8 weeks, leading to hind limb locomotion recovery. The presented strategy was proved to be efficient for harnessing endogenous NSPCs, which facilitate SCI repair significantly.
Collapse
Affiliation(s)
- Zhenni Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Haimin Zhang
- CAS Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Caixia Fan
- CAS Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yan Zhuang
- CAS Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Wen Yang
- CAS Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yanyan Chen
- CAS Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - He Shen
- CAS Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaoran Li
- Innovation Center for Textile Science and Technology, Donghua University, Shanghai 200051, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
31
|
Zhang J, Hu Q, Jiang X, Wang S, Zhou X, Lu Y, Huang X, Duan H, Zhang T, Ge H, Yu A. Actin Alpha 2 Downregulation Inhibits Neural Stem Cell Proliferation and Differentiation into Neurons through Canonical Wnt/ β-Catenin Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7486726. [PMID: 35186189 PMCID: PMC8850075 DOI: 10.1155/2022/7486726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/27/2021] [Accepted: 01/05/2022] [Indexed: 11/29/2022]
Abstract
Our previous study has shown that actin alpha 2 (ACTA2) is expressed in NSC and ACTA2 downregulation inhibits NSC migration by increasing RhoA expression and decreasing the expression of Rac1 to curb actin filament polymerization. Given that proliferation and differentiation are the two main characteristics of NSC, the role of ACTA2 downregulation in the proliferation and differentiation of NSC remains elusive. Here, the results demonstrated that ACTA2 downregulation using ACTA2 siRNA held the potential of inhibiting NSC proliferation using cell counting kit-8 (CCK8) and immunostaining. Then, our data illustrated that ACTA2 downregulation attenuated NSC differentiation into neurons, while directing NSC into astrocytes and oligodendrocytes using immunostaining and immunoblotting. Thereafter, the results revealed that the canonical Wnt/β-catenin pathway was involved in the effect of ACTA2 downregulation on the proliferation and differentiation of NSC through upregulating p-β-catenin and decreasing β-catenin due to inactivating GSK-3β, while this effect could be partially abolished with administration of CHIR99012, a GSK-3 inhibitor. Collectively, these results indicate that ACTA2 downregulation inhibits NSC proliferation and differentiation into neurons through inactivation of the canonical Wnt/β-catenin pathway. The aim of the present study is to elucidate the role of ACTA2 in proliferation and differentiation of NSC and to provide an intervention target for promoting NSC proliferation and properly directing NSC differentiation.
Collapse
Affiliation(s)
- Ji Zhang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Quan Hu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Xuheng Jiang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Shuhong Wang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Xin Zhou
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Yuanlan Lu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Xiaofei Huang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Haizhen Duan
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Tianxi Zhang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Hongfei Ge
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| | - Anyong Yu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, 563003 Zunyi, Guizhou, China
| |
Collapse
|
32
|
P2Y2 Receptor Mediated Neuronal Regeneration and Angiogenesis to Affect Functional Recovery in Rats with Spinal Cord Injury. Neural Plast 2022; 2022:2191011. [PMID: 35154311 PMCID: PMC8828345 DOI: 10.1155/2022/2191011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 12/07/2021] [Accepted: 12/17/2021] [Indexed: 11/17/2022] Open
Abstract
The aim of this study was to investigate the effect of the P2Y2 receptor (P2Y2R) signaling pathway on neuronal regeneration and angiogenesis during spinal cord injury (SCI). The rats were randomly divided into 3 groups, including the sham+dimethyl sulfoxide (DMSO), SCI+DMSO, and SCI+P2Y2R groups. The SCI animal models were constructed. A locomotor rating scale was used for behavioral assessments. The apoptosis of spinal cord tissues was detected by TUNEL staining. The expression levels of P2Y2R, GFAP, nestin, Tuj1, and CD34 were detected by immunofluorescence staining, and the expression levels of TNF-α, IL-1β, and IL-6 were detected by enzyme-linked immunosorbent assay. The locomotor score in the model group was significantly lower than the sham group. The expression of P2Y2R was increased after SCI. The expression levels of TNF-α, IL-1β, and IL-6 were increased remarkably in the SCI model group compared with the sham group. The P2Y2R inhibitor relieved neuronal inflammation after SCI. Compared with the sham group, the apoptotic rate of spinal cord tissue cells in the model group was significantly increased. The P2Y2R inhibitor reduced the apoptosis of the spinal cord tissue. The expressions of CD34, Tuj1, and nestin in the model group were decreased, while the expressions of GFAP and P2Y2R were increased. The P2Y2R inhibitor reversed their expression levels. The P2Y2R inhibitor could alleviate SCI by relieving the neuronal inflammation, inhibiting the spinal cord tissue apoptosis, and promoting neuronal differentiation and vascular proliferation after SCI. P2Y2R may serve as a target for the treatment of SCI.
Collapse
|
33
|
Mesenchymal Stem Cells in the Treatment of Human Spinal Cord Injury: The Effect on Individual Values of pNF-H, GFAP, S100 Proteins and Selected Growth Factors, Cytokines and Chemokines. Curr Issues Mol Biol 2022; 44:578-596. [PMID: 35723326 PMCID: PMC8929137 DOI: 10.3390/cimb44020040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 11/21/2022] Open
Abstract
At present, there is no effective way to treat the consequences of spinal cord injury (SCI). SCI leads to the death of neural and glial cells and widespread neuroinflammation with persisting for several weeks after the injury. Mesenchymal stem cells (MSCs) therapy is one of the most promising approaches in the treatment of this injury. The aim of this study was to characterize the expression profile of multiple cytokines, chemokines, growth factors, and so-called neuromarkers in the serum of an SCI patient treated with autologous bone marrow-derived MSCs (BM-MSCs). SCI resulted in a significant increase in the levels of neuromarkers and proteins involved in the inflammatory process. BM-MSCs administration resulted in significant changes in the levels of neuromarkers (S100, GFAP, and pNF-H) as well as changes in the expression of proteins and growth factors involved in the inflammatory response following SCI in the serum of a patient with traumatic SCI. Our preliminary results encouraged that BM-MSCs with their neuroprotective and immunomodulatory effects could affect the repair process after injury.
Collapse
|
34
|
Repetitive Trans Spinal Magnetic Stimulation Improves Functional Recovery and Tissue Repair in Contusive and Penetrating Spinal Cord Injury Models in Rats. Biomedicines 2021; 9:biomedicines9121827. [PMID: 34944643 PMCID: PMC8698720 DOI: 10.3390/biomedicines9121827] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 12/22/2022] Open
Abstract
Spinal cord injury (SCI) is an incurable condition in which the brain is disconnected partially or completely from the periphery. Mainly, SCIs are traumatic and are due to traffic, domestic or sport accidents. To date, SCIs are incurable and, most of the time, leave the patients with a permanent loss of sensitive and motor functions. Therefore, for several decades, researchers have tried to develop treatments to cure SCI. Among them, recently, our lab has demonstrated that, in mice, repetitive trans-spinal magnetic stimulation (rTSMS) can, after SCI, modulate the lesion scar and can induce functional locomotor recovery non-invasively. These results are promising; however, before we can translate them to humans, it is important to reproduce them in a more clinically relevant model. Indeed, SCIs do not lead to the same cellular events in mice and humans. In particular, SCIs in humans induce the formation of cystic cavities. That is why we propose here to validate the effects of rTSMS in a rat animal model in which SCI leads to the formation of cystic cavities after penetrating and contusive SCI. To do so, several techniques, including immunohistochemical, behavioral and MRI, were performed. Our results demonstrate that rTSMS, in both SCI models, modulates the lesion scar by decreasing the formation of cystic cavities and by improving axonal survival. Moreover, rTSMS, in both models, enhances functional locomotor recovery. Altogether, our study describes that rTSMS exerts positive effects after SCI in rats. This study is a further step towards the use of this treatment in humans.
Collapse
|
35
|
Nicaise AM, D'Angelo A, Ionescu RB, Krzak G, Willis CM, Pluchino S. The role of neural stem cells in regulating glial scar formation and repair. Cell Tissue Res 2021; 387:399-414. [PMID: 34820704 PMCID: PMC8975756 DOI: 10.1007/s00441-021-03554-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 11/10/2021] [Indexed: 02/06/2023]
Abstract
Glial scars are a common pathological occurrence in a variety of central nervous system (CNS) diseases and injuries. They are caused after severe damage and consist of reactive glia that form a barrier around the damaged tissue that leads to a non-permissive microenvironment which prevents proper endogenous regeneration. While there are a number of therapies that are able to address some components of disease, there are none that provide regenerative properties. Within the past decade, neural stem cells (NSCs) have been heavily studied due to their potent anti-inflammatory and reparative capabilities in disease and injury. Exogenously applied NSCs have been found to aid in glial scar healing by reducing inflammation and providing cell replacement. However, endogenous NSCs have also been found to contribute to the reactive environment by different means. Further understanding how NSCs can be leveraged to aid in the resolution of the glial scar is imperative in the use of these cells as regenerative therapies. To do so, humanised 3D model systems have been developed to study the development and maintenance of the glial scar. Herein, we explore the current work on endogenous and exogenous NSCs in the glial scar as well as the novel 3D stem cell–based technologies being used to model this pathology in a dish.
Collapse
Affiliation(s)
- Alexandra M Nicaise
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, UK.
| | - Andrea D'Angelo
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Rosana-Bristena Ionescu
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Grzegorz Krzak
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Cory M Willis
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Stefano Pluchino
- Department of Clinical Neurosciences and National Institute for Health Research (NIHR) Biomedical Research Centre, University of Cambridge, Cambridge, UK.
| |
Collapse
|
36
|
Rong Z, Huang Y, Cai H, Chen M, Wang H, Liu G, Zhang Z, Wu J. Gut Microbiota Disorders Promote Inflammation and Aggravate Spinal Cord Injury Through the TLR4/MyD88 Signaling Pathway. Front Nutr 2021; 8:702659. [PMID: 34589510 PMCID: PMC8473614 DOI: 10.3389/fnut.2021.702659] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/23/2021] [Indexed: 12/02/2022] Open
Abstract
Background: In spinal cord injury (SCI), systemic inflammation and the death of nerve cells in the spinal cord are life threatening. The connection between gut microbiota and signaling pathways has been a hot research topic in recent years. The Toll-like receptor 4/Myeloid differentiation factor 88 (TLR4/MyD88) signaling pathway is closely related to the inflammatory response. This study explored whether the gut microbiota imbalance could affect the TLR4/MyD88 signaling pathway to regulate SCI to provide a new basis for SCI research and treatment. Methods: An SCI model was constructed to study the influence on the injury of gut microbiota. 16S amplicon sequencing was used to identify the diversity and abundance of gut microbes. Fecal microbiota transplantation was performed in mice with SCI. ELISA was used to detect the serum levels of pro-inflammatory and anti-inflammatory factors in mice. Hematoxylin and eosin staining was used to observe SCI in mice. Immunofluorescence was used to detect the rates of loss glial fibrillary acidic protein (GFAP), neuronal nuclear protein (NeuN), and ionized calcium-binding adapter molecule 1 (IBA1) in the spinal cord as indicators of apoptosis. The expression of the TLR4/MyD88 signaling pathway was detected by qRT-PCR and western blotting. Results: Significant differences were observed in the gut microbiota of SCI mice and normal mice. The gut microbiota of SCI mice was imbalanced. The levels of pro-inflammatory cytokines tumor necrosis factor-α, interleukin (IL)-1β, and IL-6 in SCI mice were increased, as was the level of the toxic induced nitric oxide synthase. The levels of anti-inflammatory factors IL-4, transforming growth factor-β, and IL-10 were decreased, as was the level of arginase-1. The apoptosis rates of GFAP, NeuN, and IBA1 were increased. The TLR4/MyD88 signaling pathway was activated. In the SCI group, inflammation increased after fecal transplantation, apoptosis of GFAP, NeuN, and IBA1 increased, and SCI was more serious. Conclusion: The TLR4/MyD88 signaling pathway promotes the death of nerve cells by inducing inflammation. Gut microbiota dysregulation can lead to aggravated SCI by activating the TLR4/MyD88 signaling pathway.
Collapse
Affiliation(s)
- Zijie Rong
- Department of Spine Surgery, Huizhou Municipal Central Hospital, Huizhou, China.,Orthopaedic Institute, Huizhou Municipal Central Hospital, Huizhou, China
| | - Yuliang Huang
- Orthopaedic Institute, Huizhou Municipal Central Hospital, Huizhou, China.,Department of Orthopaedics, Huizhou Municipal Central Hospital, Huizhou, China
| | - Honghua Cai
- Department of Spine Surgery, Huizhou Municipal Central Hospital, Huizhou, China.,Orthopaedic Institute, Huizhou Municipal Central Hospital, Huizhou, China
| | - Min Chen
- Department of Spine Surgery, Huizhou Municipal Central Hospital, Huizhou, China.,Orthopaedic Institute, Huizhou Municipal Central Hospital, Huizhou, China
| | - Hao Wang
- Department of Spine Surgery, Huizhou Municipal Central Hospital, Huizhou, China.,Orthopaedic Institute, Huizhou Municipal Central Hospital, Huizhou, China
| | - Guihua Liu
- Department of Spine Surgery, Huizhou Municipal Central Hospital, Huizhou, China.,Orthopaedic Institute, Huizhou Municipal Central Hospital, Huizhou, China
| | - Zhiwen Zhang
- Orthopaedic Institute, Huizhou Municipal Central Hospital, Huizhou, China.,Department of Orthopaedics, Huizhou Municipal Central Hospital, Huizhou, China
| | - Jiawen Wu
- Department of Spine Surgery, The People's Hospital of Longhua, Shenzhen Longhua Clinical Medical College of Guangdong Medical University, Shenzhen, China
| |
Collapse
|
37
|
Fang C, Sun J, Wei L, Gao F, Qian J. Oscillating field stimulation promotes recovery from spinal cord injury in rats by regulating the differentiation of endogenous neural stem cells. Exp Ther Med 2021; 22:979. [PMID: 34345261 PMCID: PMC8311232 DOI: 10.3892/etm.2021.10411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 06/26/2020] [Indexed: 12/25/2022] Open
Abstract
The mammalian spinal cord (SC) has a limited self-repair capacity and exogenous treatments are yet to produce substantial functional recovery following SC injury (SCI). The SC contains endogenous neural stem cells (NSCs) with multi-lineage differentiation potential and it may be possible to restore function via interventions that promote NSC differentiation following SCI. Oscillating field stimulation (OFS) has been reported to regulate the Wnt signaling pathway, a known modulator of NSC differentiation. However, the effects of OFS on NSC differentiation following SCI and associated functional recovery have not been previously examined. In the current study, the Basso-Beattie-Bresnahan (BBB) score was used to assess locomotion recovery following SCI in rats and immunofluorescence double-staining was used to examine the regeneration of neurons and oligodendrocytes derived from NSCs. Furthermore, Nissl staining was performed to assess the viability and survival of neurons following SCI, while recovery of the myelin sheath was examined by uranium-lead staining under transmission electron microscopy. OFS delivered via an implanted stimulator enhanced the differentiation of NSCs into neurons and oligodendrocytes and accelerated the regeneration of myelinated axons. Additionally, BBB scores revealed superior locomotion recovery in OFS-treated rats compared with SCI controls. Collectively, these results indicated that OFS may be a feasible strategy to promote SCI recovery by regulating the differentiation of endogenous NSCs.
Collapse
Affiliation(s)
- Chao Fang
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Jian Sun
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Laifu Wei
- Department of Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230031, P.R. China
| | - Fei Gao
- Department of Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230031, P.R. China
| | - Jun Qian
- Department of Spine Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230031, P.R. China
| |
Collapse
|
38
|
Shu J, Cheng F, Gong Z, Ying L, Wang C, Yu C, Zhou X, Xiao M, Wang J, Xia K, Huang X, Tao Y, Shi K, Liu Y, Liang C, Chen Q, Feng X, Li F. Transplantation Strategies for Spinal Cord Injury Based on Microenvironment Modulation. Curr Stem Cell Res Ther 2021; 15:522-530. [PMID: 32316901 DOI: 10.2174/1574888x15666200421112622] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/27/2020] [Accepted: 03/02/2020] [Indexed: 12/20/2022]
Abstract
Spinal cord injury (SCI) is different from peripheral nerve injury; it results in devastating and permanent damage to the spine, leading to severe motor, sensory and autonomic dysfunction. SCI produces a complex microenvironment that can result in hemorrhage, inflammation and scar formation. Not only does it significantly limit regeneration, but it also challenges a multitude of transplantation strategies. In order to promote regeneration, researchers have recently begun to focus their attention on strategies that manipulate the complicated microenvironment produced by SCI. And some have achieved great therapeutic effects. Hence, reconstructing an appropriate microenvironment after transplantation could be a potential therapeutic solution for SCI. In this review, first, we aim to summarize the influential compositions of the microenvironment and their different effects on regeneration. Second, we highlight recent research that used various transplantation strategies to modulate different microenvironments produced by SCI in order to improve regeneration. Finally, we discuss future transplantation strategies regarding SCI.
Collapse
Affiliation(s)
- Jiawei Shu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Feng Cheng
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Zhe Gong
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Liwei Ying
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Chenggui Wang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Chao Yu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Xiaopeng Zhou
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Mu Xiao
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingkai Wang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Kaishun Xia
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Xianpeng Huang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Yiqing Tao
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Kesi Shi
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Yuemei Liu
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chengzhen Liang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Qixin Chen
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Xinhua Feng
- The MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Fangcai Li
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| |
Collapse
|
39
|
Induction of Neurogenesis and Angiogenesis in a Rat Hemisection Spinal Cord Injury Model With Combined Neural Stem Cell, Endothelial Progenitor Cell, and Biomimetic Hydrogel Matrix Therapy. Crit Care Explor 2021; 3:e0436. [PMID: 34151277 PMCID: PMC8205216 DOI: 10.1097/cce.0000000000000436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Acute spinal cord injury is a devastating injury that may lead to loss of independent function. Stem-cell therapies have shown promise; however, a clinically efficacious stem-cell therapy has yet to be developed. Functionally, endothelial progenitor cells induce angiogenesis, and neural stem cells induce neurogenesis. In this study, we explored using a multimodal therapy combining endothelial progenitor cells with neural stem cells encapsulated in a bioactive biomimetic hydrogel matrix to facilitate stem cell-induced neurogenesis and angiogenesis in a rat hemisection spinal cord injury model. DESIGN Laboratory experimentation. SETTING University laboratory. SUBJECTS Female Fischer 344 rats. INTERVENTIONS Three groups of rats: 1) control, 2) biomimetic hydrogel therapy, and 3) combined neural stem cell, endothelial progenitor cell, biomimetic hydrogel therapy underwent right-sided spinal cord hemisection at T9-T10. The blinded Basso, Beattie, and Bresnahan motor score was obtained weekly; after 4 weeks, observational histologic analysis of the injured spinal cords was completed. MEASUREMENTS AND MAIN RESULTS Blinded Basso, Beattie, and Bresnahan motor score of the hind limb revealed significantly improved motor function in rats treated with combined neural stem cell, endothelial progenitor cell, and biomimetic hydrogel therapy (p < 0.05) compared with the control group. The acellular biomimetic hydrogel group did not demonstrate a significant improvement in motor function compared with the control group. Immunohistochemistry evaluation of the injured spinal cords demonstrated de novo neurogenesis and angiogenesis in the combined neural stem cell, endothelial progenitor cell, and biomimetic hydrogel therapy group, whereas, in the control group, a gap or scar was found in the injured spinal cord. CONCLUSIONS This study demonstrates proof of concept that multimodal therapy with endothelial progenitor cells and neural stem cells combined with a bioactive biomimetic hydrogel can be used to induce de novo CNS tissue in an injured rat spinal cord.
Collapse
|
40
|
Li X, Gao Y, Tian F, Du R, Yuan Y, Li P, Liu F, Wang C. miR-31 promotes neural stem cell proliferation and restores motor function after spinal cord injury. Exp Biol Med (Maywood) 2021; 246:1274-1286. [PMID: 33715531 PMCID: PMC8371310 DOI: 10.1177/1535370221997071] [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: 08/26/2020] [Accepted: 02/01/2021] [Indexed: 01/17/2023] Open
Abstract
This study aims to examine whether miR-31 promotes endogenous NSC proliferation and be used for spinal cord injury management. In the present study, the morpholino knockdown of miR-31 induced abnormal neuronal apoptosis in zebrafish, resulting in impaired development of the tail. miR-31 agomir transfection in NSCs increased Nestin expression and decreased ChAT and GFAP expression levels. miR-31 induced the proliferation of mouse NSCs by upregulating the Notch signaling pathway, and more NSCs entered G1; Notch was inhibited by miR-31 inactivation. Injection of a miR-31 agomir into mouse models of spinal cord injury could effectively restore motor functions after spinal cord injury, which was achieved by promoting the proliferation of endogenous NSCs. After the injection of a miR-31 agomir in spinal cord injury mice, the expression of Nestin and GFAP increased, while GFAP expression decreased. In conclusion, the zebrafish experiments prove that a lack of miR-31 will block nervous system development. In spinal cord injury mouse models, miR-31 overexpression might promote spinal cord injury repair.
Collapse
Affiliation(s)
- Xiao Li
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| | - Yuantao Gao
- Queen Mary School, Nanchang University, Nanchang 330000, China
| | - Feng Tian
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| | - Ruochen Du
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| | - Yitong Yuan
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| | - Pengfei Li
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| | - Fang Liu
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| | - Chunfang Wang
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| |
Collapse
|
41
|
Delarue Q, Robac A, Massardier R, Marie JP, Guérout N. Comparison of the effects of two therapeutic strategies based on olfactory ensheathing cell transplantation and repetitive magnetic stimulation after spinal cord injury in female mice. J Neurosci Res 2021; 99:1835-1849. [PMID: 33960512 PMCID: PMC8359979 DOI: 10.1002/jnr.24836] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 12/11/2022]
Abstract
Spinal cord injury (SCI) is a debilitating condition, which leads to a permanent loss of functions below the injury site. The events which take place after SCI are characterized by cellular death, release of inhibitory factors, and inflammation. Many therapies have been studied to cure SCI, among them magnetic stimulation aims to reduce the secondary damages in particular by decreasing apoptosis, while, cellular transplantation promotes neuroregeneration by enhancing axonal regrowth. In the present study, we compared individually primary olfactory ensheathing cell (OEC) transplantation and repetitive trans‐spinal magnetic stimulation (rTSMS) and then, we combined these two therapeutic approaches on tissue repair and functional recovery after SCI. To do so, SCIs were performed at Th10 level on female C57BL/6 mice, which were randomized into four groups: SCI, SCI + primary bOECs, SCI + STM, SCI + primary bulbar olfactory ensheathing cells (bOECs) + stimulation (STM). On these animals bioluminescence, immunohistological, and behavioral experiments were performed after SCI. Our results show that rTSMS has beneficial effect on the modulation of spinal scar by reducing fibrosis, demyelination, and microglial cell activation and by increasing the astroglial component of the scar, while, primary bOEC transplantation decreases microglial reactivity. At the opposite, locotronic experiments show that both treatments induce functional recovery. We did not observed any additional effect by combining the two therapeutic approaches. Taken together, the present study indicates that primary bOEC transplantation and rTSMS treatment act through different mechanisms after SCI to induce functional recovery. In our experimental paradigm, the combination of the two therapies does not induce any additional benefit.
Collapse
Key Words
- RRID:AB_10563302: PDGFRβ, Abcam, ab91066
- RRID:AB_10643424: PE, poly4064, BioLegend, 406408
- RRID:AB_2313568: Jackson ImmunoResearch, 711-166-152
- RRID:AB_2340667: Jackson ImmunoResearch, 712-165-153
- RRID:AB_2340812: Jackson ImmunoResearch, 715-165-140
- RRID:AB_2715913: Alexa 488, MRG2b-85, BioLegend
- RRID:AB_306827: p75, Abcam, ab8874
- RRID:AB_476889: GFAP Cy3-conjugated Sigma-Aldrich, C9205
- RRID:AB_777165:P DGFRβAbcam ab32570
- RRID:AB_839504: Iba1, Wako, 019-19741
- RRID:AB_94975: MBP, Millipore, MAB386
- RRID:IMSR_JAX:008450: L2G85Chco+/+ (FVB-Tg(CAG-luc,-GFP)L2G85Chco/J)
- glial scar
- magnetic stimulation
- olfactory ensheathing cells and neuroregeneration
- rehabilitation
- spinal cord injury
Collapse
Affiliation(s)
- Quentin Delarue
- Normandie Univ, UNIROUEN, EA3830-GRHV, Rouen, France.,Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Amandine Robac
- Normandie Univ, UNIROUEN, EA3830-GRHV, Rouen, France.,Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Romane Massardier
- Normandie Univ, UNIROUEN, EA3830-GRHV, Rouen, France.,Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Jean-Paul Marie
- Normandie Univ, UNIROUEN, EA3830-GRHV, Rouen, France.,Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Nicolas Guérout
- Normandie Univ, UNIROUEN, EA3830-GRHV, Rouen, France.,Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| |
Collapse
|
42
|
Wang D, Wang K, Liu Z, Wang Z, Wu H. Valproic Acid Labeled Chitosan Nanoparticles Promote the Proliferation and Differentiation of Neural Stem Cells After Spinal Cord Injury. Neurotox Res 2021; 39:456-466. [PMID: 33247828 DOI: 10.1007/s12640-020-00304-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/05/2020] [Accepted: 10/21/2020] [Indexed: 12/12/2022]
Abstract
Chitosan nanoparticles and valproic acid are demonstrated as the protective agents in the treatment of spinal cord injury (SCI). However, the effects of valproic acid-labeled chitosan nanoparticles (VA-CN) on endogenous spinal cord neural stem cells (NSCs) following SCI and the underlying mechanisms involved remain to be elucidated. In this study, the VA-CN was constructed and the effects of VA-CN on NSCs were assessed in a rat model of SCI. We found VA-CN treatment promoted recovery of the tissue and locomotive function following SCI. Moreover, administration of VA-CN significantly enhanced neural stem cell proliferation and the expression levels of neurotrophic factors following SCI. Furthermore, administration of VA-CN led to a decrease in the number of microglia following SCI. In addition, VA-CN treatment significantly increased the Tuj 1- positive cells in the spinal cord of the SCI rats, suggesting that VA-CN could enhance the differentiation of NSCs following SCI. In conclusion, these results demonstrated that VA-CN could improve the functional and histological recovery through promoting the proliferation and differentiation of NSCs following SCI, which would provide a newly potential therapeutic manner for the treatment of SCI.
Collapse
Affiliation(s)
- Dimin Wang
- School of Medicine, Zhejiang University, Hangzhou, China
- College of Basic Medical Sciences, Second Military Medical University, Shanghai, China
| | - Kai Wang
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Zhenlei Liu
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Zonglin Wang
- College of Basic Medical Sciences, Second Military Medical University, Shanghai, China
| | - Hao Wu
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China.
| |
Collapse
|
43
|
Wang S, He Y, Zhang H, Chen L, Cao L, Yang L, Wang C, Pan Y, Tang Q, Tan W, Dou X, Li Q. The Neural Stem Cell Properties of PKD2L1 + Cerebrospinal Fluid-Contacting Neurons in vitro. Front Cell Neurosci 2021; 15:630882. [PMID: 33790741 PMCID: PMC8005615 DOI: 10.3389/fncel.2021.630882] [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: 11/18/2020] [Accepted: 02/16/2021] [Indexed: 01/20/2023] Open
Abstract
Cerebrospinal fluid-touching neurons (CSF-cNs) exist in the region surrounding the central canal of the spinal cord, which locate in the adult neurogenic niche. Previous research showed that CSF-cNs expressed the molecular markers of immature neural cells in vivo. Here, we explored the potential of CSF-cNs as neural stem cell in intro. We first found that PKD2L1+ CSF-cNs, isolating by FACS using the molecular marker PKD2L1 of CSF-cNs, expressed neural stem cells markers like Nestin, Sox2, and GFAP by immunofluorescence staining. PKD2L1+ CSF-cNs were able to form neurospheres and passaged in vitro. Immunofluorescence staining showed that the neurospheres forming by PKD2L1+ CSF-cNs also expressed neural stem cell markers Nestin, Sox2 and GFAP. The neurospheres expressed proliferation markers Ki67 and PCNA by immunofluorescence staining, indicating that the neurospheres forming by PKD2L1+ CSF-cNs were proliferative. The neurospheres, forming by CSF-cNs, had the ability of differentiation into neurons, astrocytes, and oligodendrocytes. Collectively, our data suggested that PKD2L1+ CSF-cNs have the properties of neural stem cells in vitro and may provide a promising approach for the repair of spinal cord injury.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Orthopedics Traumatic, The Affiliated Hospital of Guizhou Medical University, Guiyang City, China.,School of Clinical Medicine, Guizhou Medical University, Guiyang, China.,Shandong Juxian People's Hospital, Rizhao City, China
| | - Yuqi He
- Department of Orthopedics Traumatic, The Affiliated Hospital of Guizhou Medical University, Guiyang City, China.,School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Huiqian Zhang
- Department of Orthopedics Traumatic, The Affiliated Hospital of Guizhou Medical University, Guiyang City, China.,School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Li Chen
- Department of Orthopedics Traumatic, The Affiliated Hospital of Guizhou Medical University, Guiyang City, China.,School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Liang Cao
- Department of Orthopedics Traumatic, The Affiliated Hospital of Guizhou Medical University, Guiyang City, China.,School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Leiluo Yang
- Department of Orthopedics Traumatic, The Affiliated Hospital of Guizhou Medical University, Guiyang City, China
| | - Chunqing Wang
- Department of Orthopedics Traumatic, The Affiliated Hospital of Guizhou Medical University, Guiyang City, China
| | - Yujie Pan
- Department of Orthopedics Traumatic, The Affiliated Hospital of Guizhou Medical University, Guiyang City, China
| | - Qian Tang
- Department of Orthopedics Traumatic, The Affiliated Hospital of Guizhou Medical University, Guiyang City, China
| | - Wei Tan
- Department of Orthopedics Traumatic, The Affiliated Hospital of Guizhou Medical University, Guiyang City, China
| | - Xiaowei Dou
- Clinical Research Center, Affiliated Hospital of Guizhou Medical University, Guiyang City, China
| | - Qing Li
- Department of Orthopedics Traumatic, The Affiliated Hospital of Guizhou Medical University, Guiyang City, China
| |
Collapse
|
44
|
Amini S, Salehi H, Setayeshmehr M, Ghorbani M. Natural and synthetic polymeric scaffolds used in peripheral nerve tissue engineering: Advantages and disadvantages. POLYM ADVAN TECHNOL 2021. [DOI: 10.1002/pat.5263] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Shahram Amini
- Department of Anatomical Sciences and Molecular Biology, School of Medicine Isfahan University of Medical Sciences hezarjerib Isfahan Iran
- Student Research Committee Baqiyatallah University of Medical Sciences Tehran Iran
| | - Hossein Salehi
- Department of Anatomical Sciences and Molecular Biology, School of Medicine Isfahan University of Medical Sciences hezarjerib Isfahan Iran
| | - Mohsen Setayeshmehr
- Department of Biomaterials, Tissue Engineering and Nanotechnology, School of Advanced Technologies in Medicine Isfahan University of Medical Sciences Isfahan Iran
| | - Masoud Ghorbani
- Applied Biotechnology Research Center Baqiyatallah University of Medical Sciences Tehran Iran
| |
Collapse
|
45
|
High mobility group box 1 promotes the differentiation of spinal ependymal cells into astrocytes rather than neurons. Neuroreport 2021; 32:399-406. [PMID: 33661806 DOI: 10.1097/wnr.0000000000001609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Spinal ependymal cells are involved in proliferation, differentiation and migration after spinal cord injury (SCI) and represent an endogenous source of repair cells for treating SCI. However, 95% of activated ependymal cells eventually differentiate into astrocytes after SCI and ultimately contribute more than half of the new astrocytes that form glial scars in vivo. The factors that regulate the fate of ependymal cells after SCI remain unclear. High mobility group box 1 (HMGB1) is regarded as an important proinflammatory factor in nerve injury, and recent studies have shown that HMGB1 can regulate the fate of stem cells after injury. In this study, we investigated whether HMGB1 released from reactive astrocytes after SCI regulates the proliferation and differentiation of ependymal cells in vitro. Ependymal cells extracted and cultured from the spinal cord of mice were separately treated with astrocyte culture medium (ACM), IL-1β, ACM (IL-1β) and the HMGB1 protein, and the proliferation and differentiation of ependymal cells were detected. Additionally, an HMGB1-neutralizing antibody (anti-HMGB1) was added to further verify the regulatory effect of HMGB1 on ependymal cells. The results showed that HMGB1 released from reactive astrocytes promoted ependymal cell differentiation into astrocytes and inhibited ependymal cell differentiation into neurons in vitro; however, the effect disappeared after the addition of anti-HMGB1. HMGB1 had no significant effect on ependymal cell proliferation. Our findings demonstrate that HMGB1 can regulate the differentiation of ependymal cells after SCI. These results provide a new strategy for the treatment of SCI.
Collapse
|
46
|
Hemani S, Lane O, Agarwal S, Yu SP, Woodbury A. Systematic Review of Erythropoietin (EPO) for Neuroprotection in Human Studies. Neurochem Res 2021; 46:732-739. [PMID: 33521906 DOI: 10.1007/s11064-021-03242-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 11/29/2022]
Abstract
Erythropoietin (EPO) is an exciting neurotherapeutic option. Despite its potential, concerns exist regarding the potential for thrombosis and adverse events with EPO administration in normonemic adults. Systematic review of literature using PRISMA guidelines to examine the application and risks of EPO as a treatment option for neuroprotection in normonemic adults. Independent, systematic searches were performed in July 2019. PubMed (1960-2019) and the Cochrane Controlled Trials Register (1960-2019) were screened. Search terms included erythropoietin, neuroprotection, and humans. The PubMed search resulted in the following search strategy: ("erythropoietin" [MeSH Terms] OR "erythropoietin" [All Fields] OR "epoetin alfa" [MeSH Terms] OR ("epoetin" [All Fields] AND "alfa" [All Fields]) OR "epoetin alfa" [All Fields]) AND ("neuroprotection" [MeSH Terms] OR "neuroprotection" [All Fields]) AND "humans" [MeSH Terms]. PubMed, Cochrane Controlled Trials Register, and articles based on prior searches yielded 388 citations. 50 studies were included, comprising of 4351 patients. There were 13 studies that noted adverse effects from EPO. Three attributed serious adverse effects to EPO and complications were statistically significant. Two of these studies related the adverse events to the co-administration of EPO with tPA. Minor adverse effects associated with the EPO group included nausea, pyrexia, headache, generalized weakness and superficial phlebitis. Most published studies focus on spinal cord injury, peri-surgical outcomes and central effects of EPO. We found no studies to date evaluating the role of EPO in post-operative pain. Future trials could evaluate this application in persistent post-surgical pain and in the peri-operative period.
Collapse
Affiliation(s)
- Salman Hemani
- Division of Pain Medicine, Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Olabisi Lane
- Division of Pain Medicine, Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Sunil Agarwal
- Division of Pain Medicine, Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Shan Ping Yu
- Division of Pain Medicine, Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Atlanta Veterans Affairs Healthcare System, Decatur, GA, 30033, USA
| | - Anna Woodbury
- Division of Pain Medicine, Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Atlanta Veterans Affairs Healthcare System, Decatur, GA, 30033, USA
| |
Collapse
|
47
|
Ge H, Zhang C, Yang Y, Chen W, Zhong J, Fang X, Jiang X, Tan L, Zou Y, Hu R, Chen Y, Feng H. Ambroxol Upregulates Glucocerebrosidase Expression to Promote Neural Stem Cells Differentiation Into Neurons Through Wnt/β-Catenin Pathway After Ischemic Stroke. Front Mol Neurosci 2021; 13:596039. [PMID: 33551744 PMCID: PMC7855720 DOI: 10.3389/fnmol.2020.596039] [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: 08/28/2020] [Accepted: 12/22/2020] [Indexed: 12/29/2022] Open
Abstract
Ischemic stroke has been becoming one of the leading causes resulting in mortality and adult long-term disability worldwide. Post-stroke pneumonia is a common complication in patients with ischemic stroke and always associated with 1-year mortality. Though ambroxol therapy often serves as a supplementary treatment for post-stroke pneumonia in ischemic stroke patients, its effect on functional recovery and potential mechanism after ischemic stroke remain elusive. In the present study, the results indicated that administration of 70 mg/kg and 100 mg/kg enhanced functional recovery by virtue of decreasing infarct volume. The potential mechanism, to some extent, was due to promoting NSCs differentiation into neurons and interfering NSCs differentiation into astrocytes through increasing GCase expression to activate Wnt/β-catenin signaling pathway in penumbra after ischemic stroke, which advanced basic knowledge of ambroxol in regulating NSCs differentiation and provided a feasible therapy for ischemic stroke treatment, even in other brain disorders in clinic.
Collapse
Affiliation(s)
- Hongfei Ge
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chao Zhang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yang Yang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Weixiang Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jun Zhong
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xuanyu Fang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xuheng Jiang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Liang Tan
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yongjie Zou
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Department of Neurosurgery, Hospital of People's Liberation Army, Nanchang, China
| | - Rong Hu
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yujie Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hua Feng
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
48
|
Ahuja CS, Mothe A, Khazaei M, Badhiwala JH, Gilbert EA, van der Kooy D, Morshead CM, Tator C, Fehlings MG. The leading edge: Emerging neuroprotective and neuroregenerative cell-based therapies for spinal cord injury. Stem Cells Transl Med 2020; 9:1509-1530. [PMID: 32691994 PMCID: PMC7695641 DOI: 10.1002/sctm.19-0135] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/01/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injuries (SCIs) are associated with tremendous physical, social, and financial costs for millions of individuals and families worldwide. Rapid delivery of specialized medical and surgical care has reduced mortality; however, long-term functional recovery remains limited. Cell-based therapies represent an exciting neuroprotective and neuroregenerative strategy for SCI. This article summarizes the most promising preclinical and clinical cell approaches to date including transplantation of mesenchymal stem cells, neural stem cells, oligodendrocyte progenitor cells, Schwann cells, and olfactory ensheathing cells, as well as strategies to activate endogenous multipotent cell pools. Throughout, we emphasize the fundamental biology of cell-based therapies, critical features in the pathophysiology of spinal cord injury, and the strengths and limitations of each approach. We also highlight salient completed and ongoing clinical trials worldwide and the bidirectional translation of their findings. We then provide an overview of key adjunct strategies such as trophic factor support to optimize graft survival and differentiation, engineered biomaterials to provide a support scaffold, electrical fields to stimulate migration, and novel approaches to degrade the glial scar. We also discuss important considerations when initiating a clinical trial for a cell therapy such as the logistics of clinical-grade cell line scale-up, cell storage and transportation, and the delivery of cells into humans. We conclude with an outlook on the future of cell-based treatments for SCI and opportunities for interdisciplinary collaboration in the field.
Collapse
Affiliation(s)
- Christopher S. Ahuja
- Division of Neurosurgery, Department of SurgeryUniversity of TorontoTorontoOntarioCanada
- Institute of Medical ScienceUniversity of TorontoTorontoOntarioCanada
- Department of Genetics and DevelopmentKrembil Research Institute, UHNTorontoOntarioCanada
| | - Andrea Mothe
- Department of Genetics and DevelopmentKrembil Research Institute, UHNTorontoOntarioCanada
| | - Mohamad Khazaei
- Department of Genetics and DevelopmentKrembil Research Institute, UHNTorontoOntarioCanada
| | - Jetan H. Badhiwala
- Division of Neurosurgery, Department of SurgeryUniversity of TorontoTorontoOntarioCanada
| | - Emily A. Gilbert
- Division of Anatomy, Department of SurgeryUniversity of TorontoTorontoOntarioCanada
| | - Derek van der Kooy
- Department of Molecular GeneticsUniversity of TorontoTorontoOntarioCanada
| | - Cindi M. Morshead
- Institute of Medical ScienceUniversity of TorontoTorontoOntarioCanada
- Division of Anatomy, Department of SurgeryUniversity of TorontoTorontoOntarioCanada
- Institute of Biomaterials and Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
| | - Charles Tator
- Division of Neurosurgery, Department of SurgeryUniversity of TorontoTorontoOntarioCanada
- Institute of Medical ScienceUniversity of TorontoTorontoOntarioCanada
- Department of Genetics and DevelopmentKrembil Research Institute, UHNTorontoOntarioCanada
| | - Michael G. Fehlings
- Division of Neurosurgery, Department of SurgeryUniversity of TorontoTorontoOntarioCanada
- Institute of Medical ScienceUniversity of TorontoTorontoOntarioCanada
- Department of Genetics and DevelopmentKrembil Research Institute, UHNTorontoOntarioCanada
| |
Collapse
|
49
|
Progress in Stem Cell Therapy for Spinal Cord Injury. Stem Cells Int 2020; 2020:2853650. [PMID: 33204276 PMCID: PMC7661146 DOI: 10.1155/2020/2853650] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 10/04/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
Background Spinal cord injury (SCI) is one of the serious neurological diseases that occur in young people with high morbidity and disability. However, there is still a lack of effective treatments for it. Stem cell (SC) treatment of SCI has gradually become a new research hotspot over the past decades. This article is aimed at reviewing the research progress of SC therapy for SCI. Methods Review the literature and summarize the effects, strategies, related mechanisms, safety, and clinical application of different SC types and new approaches in combination with SC in SCI treatment. Results A large number of studies have focused on SC therapy for SCI, most of which showed good effects. The common SC types for SCI treatment include mesenchymal stem cells (MSCs), hematopoietic stem cells (HSCs), neural stem cells (NSCs), induced pluripotent stem cells (iPSCs), and embryonic stem cells (ESCs). The modes of treatment include in vivo and in vitro induction. The pathways of transplantation consist of intravenous, transarterial, nasal, intraperitoneal, intrathecal, and intramedullary injections. Most of the SC treatments for SCI use a number of cells ranging from tens of thousands to millions. Early or late SC administration, application of immunosuppressant or not are still controversies. Potential mechanisms of SC therapy include tissue repair and replacement, neurotrophy, and regeneration and promotion of angiogenesis, antiapoptosis, and anti-inflammatory. Common safety issues include thrombosis and embolism, tumorigenicity and instability, infection, high fever, and even death. Recently, some new approaches, such as the pharmacological activation of endogenous SCs, biomaterials, 3D print, and optogenetics, have been also developed, which greatly improved the application of SC therapy for SCI. Conclusion Most studies support the effects of SC therapy on SCI, while a few studies do not. The cell types, mechanisms, and strategies of SC therapy for SCI are very different among studies. In addition, the safety cannot be ignored, and more clinical trials are required. The application of new technology will promote SC therapy of SCI.
Collapse
|
50
|
Cai Y, Liu W, Lian L, Xu Y, Bai X, Xu S, Zhang J. Stroke treatment: Is exosome therapy superior to stem cell therapy? Biochimie 2020; 179:190-204. [PMID: 33010339 DOI: 10.1016/j.biochi.2020.09.025] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 09/24/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023]
Abstract
Stroke is one of the most common causes of disability and death, and currently, ideal clinical treatment is lacking. Stem cell transplantation is a widely-used treatment approach for stroke. When compared with other types of stem cells, bone marrow mesenchymal stem cells (BMSCs) have been widely studied because of their many advantages. The paracrine effect is the primary mechanism for stem cells to play their role, and exosomes play an essential role in the paracrine effect. When compared with cell therapy, cell-free exosome therapy can prevent many risks and difficulties, and therefore, represents a promising and novel approach for treatment. In this study, we reviewed the research progress in the application of BMSCs-derived exosomes (BMSCs-exos) and BMSCs in the treatment of stroke. In addition, the advantages and disadvantages of cell therapy and cell-free exosome therapy were described, and the possible factors that hinder the introduction of these two treatments into the clinic were analyzed. Furthermore, we reviewed the current optimization methods of cell therapy and cell-free exosome therapy. Taken together, we hypothesize that cell-free exosome therapy will have excellent research prospects in the future, and therefore, it is worth further exploring. There are still some issues that need to be further addressed. For example, differences between the in vivo microenvironment and in vitro culture conditions will affect the paracrine effect of stem cells. Most importantly, we believe that more preclinical and clinical design studies are required to compare the efficacy of stem cells and exosomes.
Collapse
Affiliation(s)
- Yichen Cai
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Wanying Liu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Lu Lian
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Yingzhi Xu
- Beijing University of Chinese Medicine (BUCM), Beijing, China
| | - Xiaodan Bai
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Shixin Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China.
| | - Junping Zhang
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China; Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, 300193, China.
| |
Collapse
|