1
|
Li W, Su D, Li X, Lu K, Huang Q, Zheng J, Luo X, Chen G, Fan X. Identification of the core regulatory program driving NEUROD1-induced neuronal reprogramming. Cell Rep 2025; 44:115523. [PMID: 40173039 DOI: 10.1016/j.celrep.2025.115523] [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: 06/28/2024] [Revised: 02/03/2025] [Accepted: 03/14/2025] [Indexed: 04/04/2025] Open
Abstract
NEUROD1 (ND1)-induced astrocyte-to-neuron (AtN) conversion shows promise for treating neurological disorders. To gain insight into the molecular mechanisms of neuronal reprogramming, we established an in vitro system using primary cortical astrocyte cultures from postnatal rats and employed single-cell and multiomics sequencing. Our findings indicate that the initial cultures primarily consisted of immature astrocytes (ImAs), with potentially a minor presence of radial glial cells. The ImAs initially went through an intermediate state, activating both astrocyte and neural progenitor genes. Subsequently, they mimic in vivo neurogenesis to acquire mature neuronal characteristics. We show that ND1 acted as a pioneer factor that reshapes the chromatin landscape of astrocytes to that of neurons. This restructuring promotes the expression of neurogenic genes via inducing H3K27ac modification. Through integrative analysis of various ND1-induced neuronal specification systems, we identified 25 ND1 targets, including Hes6, as key regulators. Thus, our work highlights the key role of ND1 and its downstream regulators in neuronal reprogramming.
Collapse
Affiliation(s)
- Wen Li
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China; Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Dan Su
- GMU-GIBH Joint School of Life Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou 510005, China; The Bioland Laboratory, Guangzhou 510700, China
| | - Xining Li
- GMU-GIBH Joint School of Life Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou 510005, China; The Bioland Laboratory, Guangzhou 510700, China
| | - Kang Lu
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China
| | - Qingpei Huang
- GMU-GIBH Joint School of Life Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou 510005, China; The Bioland Laboratory, Guangzhou 510700, China
| | - Jiajun Zheng
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China; Department of Anesthesiology, Guangzhou First People's Hospital, Guangzhou 510180, China
| | - Xiaopeng Luo
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China
| | - Gong Chen
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China; Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China.
| | - Xiaoying Fan
- GMU-GIBH Joint School of Life Sciences, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou Medical University, Guangzhou 510005, China; The Bioland Laboratory, Guangzhou 510700, China.
| |
Collapse
|
2
|
Cai Y, Lin Z, Shen X, Li M, Xing L, Yang T, Chen G. Effect of microglial Pd1 on glial scar formation after spinal cord injury in mice. J Biol Chem 2025; 301:108489. [PMID: 40209954 DOI: 10.1016/j.jbc.2025.108489] [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: 03/10/2025] [Revised: 03/27/2025] [Accepted: 04/02/2025] [Indexed: 04/12/2025] Open
Abstract
The cross talk between microglia and astrocytes following spinal cord injury (SCI) greatly decides the prognosis. However, a comprehensive understanding of the molecular mechanisms by which microglia regulate astrocytic activity post-SCI is lacking. Programmed cell death protein 1 (Pdcd1, Pd1) plays a crucial role in modulating immune responses by exerting suppressive effects on microglia and peripheral immune cells within the central nervous system. Previous studies have shown the involvement of Pd1 in the pathogenesis of SCI; however, the role of microglial Pd1 in astrocytic activation and the following glial scar formation remains elusive. Here, we demonstrated that the pharmacological depletion of microglia using minocycline decreased the expression of tumor necrosis factor-alpha and interleukin-6 while concurrently increasing the expression of interleukin-10 following SCI, thereby facilitating motor function recovery in mice. We observed an increase in Pd1 expression in the injured spinal cord after SCI, with precise localization of Pd1 within microglia. Based on Pd1 knockout (KO) mice, we further revealed that Pd1 deficiency disrupted glial scar formation, leading to increased inflammation, impeded nerve regeneration, enlarged tissue damage, and compromised functional recovery following SCI. In vitro study showed that siRNA-mediated inhibition of Pd1 in microglia followed by lipopolysaccharide treatment significantly inhibited astrocyte migration and upregulated the secretion of tumor necrosis factor-alpha and CXCL9 from microglia, indicating that microglial Pd1 regulates glial scar formation through modulating the inflammatory microenvironment. Our study gains a new mechanistic insight into how microglial Pd1 decides the fate of SCI and promotes microglial Pd1 as a promising therapeutic target for SCI.
Collapse
Affiliation(s)
- Yunyun Cai
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, China
| | - Zhihao Lin
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong, Jiangsu Province, China
| | - Xin Shen
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, China
| | - Ming Li
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, China
| | - Lingyan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong, Jiangsu Province, China
| | - Tuo Yang
- Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Gang Chen
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, China; Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong, Jiangsu Province, China; Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.
| |
Collapse
|
3
|
Huang Y, Chen Z, Chen J, Liu J, Qiu C, Liu Q, Zhang L, Zhu G, Ma X, Sun S, Shi YS, Wan G. Direct reprogramming of fibroblasts into spiral ganglion neurons by defined transcription factors. Cell Prolif 2025; 58:e13775. [PMID: 39551613 PMCID: PMC11969255 DOI: 10.1111/cpr.13775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/19/2024] Open
Abstract
Degeneration of the cochlear spiral ganglion neurons (SGNs) is one of the major causes of sensorineural hearing loss and significantly impacts the outcomes of cochlear implantation. Functional regeneration of SGNs holds great promise for treating sensorineural hearing loss. In this study, we systematically screened 33 transcriptional regulators implicated in neuronal and SGN fate. Using gene expression array and principal component analyses, we identified a sequential combination of Ascl1, Pou4f1 and Myt1l (APM) in promoting functional reprogramming of SGNs. The neurons induced by APM expressed mature neuronal and SGN lineage-specific markers, displayed mature SGN-like electrophysiological characteristics and exhibited single-cell transcriptomes resembling the endogenous SGNs. Thus, transcription factors APM may serve as novel candidates for direct reprogramming of SGNs and hearing recovery due to SGN damages.
Collapse
Affiliation(s)
- Yuhang Huang
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
| | - Zhen Chen
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
| | - Jiang Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Department of Neurology, The Affiliated Drum Tower Hospital of Medical School and Institute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjingChina
| | - Jingyue Liu
- National Institute of Biological SciencesBeijingChina
- Tsinghua Institute of Multidisciplinary Biomedical ResearchTsinghua UniversityBeijingChina
| | - Cui Qiu
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
| | - Qing Liu
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Linqing Zhang
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
| | - Guang‐Jie Zhu
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Xiaofeng Ma
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| | - Shuohao Sun
- National Institute of Biological SciencesBeijingChina
- Tsinghua Institute of Multidisciplinary Biomedical ResearchTsinghua UniversityBeijingChina
| | - Yun Stone Shi
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Guangdong Institute of Intelligence Science and TechnologyZhuhaiChina
| | - Guoqiang Wan
- MOE Key Laboratory of Model Animal for Disease Study, Department of Otolaryngology Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), The Affiliated Drum Tower Hospital of Medical School and the Model Animal Research Center of Medical SchoolNanjing UniversityNanjingChina
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, National Resource Center for Mutant Mice of ChinaNanjing UniversityNanjingChina
- Research Institute of OtolaryngologyNanjingChina
| |
Collapse
|
4
|
Dhar A, Moinuddin FM, Zamanian CA, Sharar AD, Dominari A, Graepel S, Windebank AJ, Bydon M. SOX Genes in Spinal Cord Injury: Redefining Neural Stem Cell Regeneration Strategies. Mol Neurobiol 2025:10.1007/s12035-025-04882-w. [PMID: 40156684 DOI: 10.1007/s12035-025-04882-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
The study design is literature review. The sex-determining region Y gene (SRY)-related high mobility group box (HMG)-box (SOX) gene family has primarily been associated with neural development and sex determination and is a key component of human embryonic development. Recent studies on zebrafish models have demonstrated that the unique ability of the latter for central nervous tissue (CNS) repair following injury is largely mediated by SOX genes. Given that efforts aimed at the structural regeneration and functional restoration of neural tissue still represent a major therapeutic challenge in patients suffering CNS injury, these findings have initiated a discussion regarding the development of novel therapeutic strategies for SCI focusing on neural tissue regeneration. Spinal cord injury (SCI), in particular, represents a field that could greatly benefit from studies related to the function of the SOX genes. Neuro-informatics Laboratory, Mayo Clinic, Rochester, MN. A literature review was conducted, with a focus on SOX gene that has been described in the experimental studies of SCI. In this review, the existing evidence linking the SOX gene family to the pathophysiology of SCI is summarized, and future research steps regarding the potential implications of the SOX genes in neurological recovery following SCI are discussed, especially focusing on highlighting potential therapeutic targets. The potential implications of the latter could play a crucial role in future efforts to advance the treatment approaches to SCI.
Collapse
Affiliation(s)
- Ashis Dhar
- Mayo Clinic Neuro-Informatics Laboratory, Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - F M Moinuddin
- Mayo Clinic Neuro-Informatics Laboratory, Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - Cameron A Zamanian
- Mayo Clinic Neuro-Informatics Laboratory, Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - Ahnaf Dil Sharar
- Mayo Clinic Neuro-Informatics Laboratory, Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - Asimina Dominari
- Mayo Clinic Neuro-Informatics Laboratory, Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - Stephen Graepel
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Mohamad Bydon
- Mayo Clinic Neuro-Informatics Laboratory, Department of Neurosurgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
5
|
Lee DH, Cao D, Moon Y, Chen C, Liu NK, Xu XM, Wu W. Enhancement of motor functional recovery in thoracic spinal cord injury: voluntary wheel running versus forced treadmill exercise. Neural Regen Res 2025; 20:836-844. [PMID: 38886956 PMCID: PMC11433897 DOI: 10.4103/nrr.nrr-d-23-01585] [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: 09/19/2023] [Revised: 01/03/2024] [Accepted: 02/19/2024] [Indexed: 06/20/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202503000-00028/figure1/v/2024-06-17T092413Z/r/image-tiff Spinal cord injury necessitates effective rehabilitation strategies, with exercise therapies showing promise in promoting recovery. This study investigated the impact of rehabilitation exercise on functional recovery and morphological changes following thoracic contusive spinal cord injury. After a 7-day recovery period after spinal cord injury, mice were assigned to either a trained group (10 weeks of voluntary running wheel or forced treadmill exercise) or an untrained group. Bi-weekly assessments revealed that the exercise-trained group, particularly the voluntary wheel exercise subgroup, displayed significantly improved locomotor recovery, more plasticity of dopaminergic and serotonin modulation compared with the untrained group. Additionally, exercise interventions led to gait pattern restoration and enhanced transcranial magnetic motor-evoked potentials. Despite consistent injury areas across groups, exercise training promoted terminal innervation of descending axons. In summary, voluntary wheel exercise shows promise for enhancing outcomes after thoracic contusive spinal cord injury, emphasizing the role of exercise modality in promoting recovery and morphological changes in spinal cord injuries. Our findings will influence future strategies for rehabilitation exercises, restoring functional movement after spinal cord injury.
Collapse
Affiliation(s)
- Do-Hun Lee
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Dan Cao
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Younghye Moon
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Chen Chen
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nai-Kui Liu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Wei Wu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
6
|
Tang Z, Lu H, Yang X, Wu M, Yang J, Li S, Liu H, Zhou J, Tang B, Du X, Xu F, Shao Y, Wang J. Single-cell RNA sequencing provides new insights into the interaction between astrocytes and neurons after spinal cord injury in mice. Biochem Biophys Rep 2025; 41:101917. [PMID: 39896108 PMCID: PMC11787598 DOI: 10.1016/j.bbrep.2025.101917] [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: 10/09/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 02/04/2025] Open
Abstract
Background Spinal cord injury (SCI) is a devastating neurological disease in which astrocytes play a central role. Understanding the relationship between different subtypes of astrocytes and neuron subtypes during the progression of SCI is critical to understanding the disease. Methods and results In this study, single-cell RNA sequencing (scRNA-seq) was used to analyze the transcriptome data of acute, subacute and intermediate stages of SCI in mice as well as normal tissues. Different subtypes of astrocytes and neuronal cells were identified and their dynamic changes and functionalities during the development of SCI. An intriguing discovery was the identification of a specific subtype of astrocytes characterized by unique expression of Gap43, Vim, Aldoc, and Mt1. This subtype of cells shows similarities in gene expression with neurons and potentially transitioned into neurons during the course of SCI. Furthermore, we have uncovered the important role of the glycolytic pathway in this cellular transformation process. Furthermore, through cellular interaction analysis, we validated pathways (mdk-ptprz1,ptn-ptprz1,ptn-sdc3) associated with the potential conversion of these specific cell subsets into neurons. Finally, these cells were observed by fluorescence microscopy and critical gene expressions were validated by Western blot. Conclusions The results of this study not only deepen our understanding of the mechanisms underlying SCI, but also provide new insights and opportunities for the development of novel therapeutic strategies and interventions.
Collapse
Affiliation(s)
- Zhi Tang
- Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Hengyang Lu
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, China
- The PRC Ministry of Education Engineering Research Center of Intelligent Technology for Healthcare, Wuxi, Jiangsu 214122, China
| | - Xiao Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Mao Wu
- Department of Orthopaedics and Traumatology, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Junfeng Yang
- Department of Orthopaedics and Traumatology, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Shaoshuo Li
- Department of Orthopaedics and Traumatology, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Heng Liu
- Wuxi City Binhu Traditional Chinese Medicine Hospital, China
| | - Junkang Zhou
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, China
| | - Bin Tang
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, China
| | - Xinyao Du
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Fei Xu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yang Shao
- Department of Orthopaedics and Traumatology, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Jianwei Wang
- Department of Orthopaedics and Traumatology, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| |
Collapse
|
7
|
Mseis-Jackson N, Jiang M, Sharma M, Ranchod A, Williams C, Chen X, Li H. Dynamic regulation of NeuroD1 expression level by a novel viral construct during astrocyte-to-neuron reprogramming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.17.638625. [PMID: 40027739 PMCID: PMC11870611 DOI: 10.1101/2025.02.17.638625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Astrocyte-to-neuron reprogramming presents a viable approach for regenerative medicine. The reprogramming factor NeuroD1 has demonstrated capability of neuronal reprogramming with high efficiency both in culture and in the injured central nervous system. High level of NeuroD1 expression is required to break down the cellular identity barrier for a successful reprogramming, and yet persistence of this high level drives the reprogrammed neurons primarily to glutamatergic subtype. This is consistent with the critical role of NeuroD1 in determination of glutamatergic neuronal lineage during development. However, diversified neuronal subtypes are needed to establish appropriate neuronal connectivity in disease/injury conditions. We reason that continuously high level of NeuroD1 expression forces the reprogrammed neurons into glutamatergic subtype, and that reducing NeuroD1 level after reprogramming may allow generation of neurons with diversified subtypes. For this purpose, we engineered a novel viral expression vector by which NeuroD1 expression can be dynamically regulated during the reprogramming process. Specifically, the target site of a neuron-specific microRNA (miR-124) is incorporated in the expression system. Therefore, this novel construct would still achieve a high NeuroD1 expression level in astrocytes for reprogramming to occur and yet reduce its level in the reprogrammed neurons by suppression of endogenous miR-124. In this study, we demonstrated that this construct elicits a dynamic gene expression pattern with much reduced level of NeuroD1 at later stages of neuronal reprogramming. We also showed that this construct still retains relatively high reprogramming efficiency and can generate mature neurons with an enhanced GABAergic neuronal phenotype.
Collapse
|
8
|
Dokukin NV, Chudakova DA, Shkap MO, Kovalchuk AM, Kibirsky PD, Baklaushev VP. Direct Neural Reprogramming in situ: Existing Approaches and Their Optimization. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:214-230. [PMID: 40254400 DOI: 10.1134/s000629792460426x] [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: 11/25/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 04/22/2025]
Abstract
Direct in situ neuronal reprogramming (transdifferentiation) of glial cells (astrocytes and microglia) has attracted a significant interest as a potential approach for the treatment of a wide range of neurodegenerative diseases and damages of the central nervous system (CNS). The nervous system of higher mammals has a very limited capacity for repair. Disruption of CNS functioning due to traumatic injuries or neurodegenerative processes can significantly affect the quality of patients' life, lead to motor and cognitive impairments, and result in disability and, in some cases, death. Restoration of lost neurons in situ via direct reprogramming of glial cells without the intermediate stage of pluripotency seems to be the most attractive approach from the viewpoint of translational biomedicine. The ability of astroglia to actively proliferate in response to the damage of neural tissue supports the idea that these neuron-like cells, which are already present at the lesion site, are good candidates for transdifferentiation into neurons, considering that the possibility of direct neuronal reprogramming of astrocytes both in vitro and in vivo have demonstrated in many independent studies. Overexpression of proneuronal transcription factors, e.g., neurogenic differentiation factors 1-4 (NeuroD1-4), Neurogenin 2 (NeuroG2), Ascl1 (Achaete-Scute homolog 1), and Dlx2 (distal-less homeobox 2), including pioneer transcription factors that recognize target sequences in the compacted chromatin and activate transcription of silent genes, has already been proven as a potential therapeutic strategy. Other strategies, such as microRNA-mediated suppression of activity of PTB and REST transcription factors and application of small molecules or various biomaterials, are also utilized in neuronal reprogramming. However, the efficiency of direct in situ reprogramming is limited by a number of factors, including cell specificity of transgene delivery systems and promoters, brain regions in which transdifferentiation occurs, factors affecting cell metabolism, microenvironment, etc. Reprogramming in situ, which takes place in the presence of a large number of different cell types, requires monitoring and precise phenotypic characterization of subpopulations of cells undergoing transdifferentiation in order to confirm the reprogramming of the astroglia into neurons and subsequent integration of these neurons into the CNS. Here, we discussed the most efficient strategies of neuronal reprogramming and technologies used to visualize the transdifferentiation process, with special focus on the obstacles to efficient neuronal conversion, as well as approaches to overcome them.
Collapse
Affiliation(s)
- Nikita V Dokukin
- Federal Center for Brain and Neurotechnology, Federal Medical and Biological Agency of Russia, Moscow, 117513, Russia
| | - Daria A Chudakova
- Federal Center for Brain and Neurotechnology, Federal Medical and Biological Agency of Russia, Moscow, 117513, Russia
- National Medical Research Center of Children's Health, Ministry of Health of the Russian Federation, Moscow, 119991, Russia
| | - Matvey O Shkap
- Federal Center for Brain and Neurotechnology, Federal Medical and Biological Agency of Russia, Moscow, 117513, Russia
| | - Anna M Kovalchuk
- Federal Center for Brain and Neurotechnology, Federal Medical and Biological Agency of Russia, Moscow, 117513, Russia
| | - Pavel D Kibirsky
- Federal Center for Brain and Neurotechnology, Federal Medical and Biological Agency of Russia, Moscow, 117513, Russia
| | - Vladimir P Baklaushev
- Federal Center for Brain and Neurotechnology, Federal Medical and Biological Agency of Russia, Moscow, 117513, Russia.
- Federal Scientific and Clinical Center for Specialized Types of Medical Care and Medical Technologies, Federal Medical and Biological Agency of Russia, Moscow, 115682, Russia
- Research Institute of Pulmonology, Federal Medical and Biological Agency of Russia, Moscow, 115682, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| |
Collapse
|
9
|
Niceforo A, Zholudeva LV, Fernandes S, Shah Y, Lane MA, Qiang L. Challenges and Efficacy of Astrocyte-to-Neuron Reprogramming in Spinal Cord Injury: In Vitro Insights and In Vivo Outcomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.03.25.586619. [PMID: 38585866 PMCID: PMC10996511 DOI: 10.1101/2024.03.25.586619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Traumatic spinal cord injury (SCI) leads to the disruption of neural pathways, causing loss of neural cells, with subsequent reactive gliosis and tissue scarring that limit endogenous repair. One potential therapeutic strategy to address this is to target reactive scar-forming astrocytes with direct cellular reprogramming to convert them into neurons, by overexpression of neurogenic transcription factors. Here we used lentiviral constructs to overexpress Ascl1 or a combination of microRNAs (miRs) miR124, miR9/9*and NeuroD1 transfected into cultured and in vivo astrocytes. In vitro experiments revealed cortically-derived astrocytes display a higher efficiency (70%) of reprogramming to neurons than spinal cord-derived astrocytes. In a rat cervical SCI model, the same strategy induced only limited reprogramming of astrocytes. Delivery of reprogramming factors did not significantly affect patterns of breathing under baseline and hypoxic conditions, but significant differences in average diaphragm amplitude were seen in the reprogrammed groups during eupneic breathing, hypoxic, and hypercapnic challenges. These results show that while cellular reprogramming can be readily achieved in carefully controlled in vitro conditions, achieving a similar degree of successful reprogramming in vivo is challenging and may require additional steps.
Collapse
Affiliation(s)
- Alessia Niceforo
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, 19129, USA
- Marion Murray Spinal Cord Research Center, College of Medicine, Drexel University, Philadelphia, PA, 19129, USA
| | | | - Silvia Fernandes
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, 19129, USA
- Marion Murray Spinal Cord Research Center, College of Medicine, Drexel University, Philadelphia, PA, 19129, USA
| | - Yashvi Shah
- College of Medicine, Drexel University, Philadelphia, PA, 19104, USA
| | - Michael A. Lane
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, 19129, USA
- Marion Murray Spinal Cord Research Center, College of Medicine, Drexel University, Philadelphia, PA, 19129, USA
| | - Liang Qiang
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, 19129, USA
- Marion Murray Spinal Cord Research Center, College of Medicine, Drexel University, Philadelphia, PA, 19129, USA
| |
Collapse
|
10
|
d'Orange M, Lentini C, Heinrich C. Retrovirus-Mediated Reprogramming of Endogenous Hippocampal Glia into GABAergic Induced Neurons. Methods Mol Biol 2025; 2899:199-219. [PMID: 40067626 DOI: 10.1007/978-1-0716-4386-0_14] [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] [Indexed: 05/13/2025]
Abstract
Lineage reprogramming of glial cells into induced neurons (iNs) has emerged as an innovative strategy to replace neurons lost due to injury or neurological diseases. Here, we describe a step-by-step protocol to induce in vivo conversion of reactive glial cells, proliferating within the injured hippocampus, into mature and functional GABAergic iNs through retrovirus-mediated expression of two neurogenic fate determinants (Ascl1 and Dlx2). We have previously applied this method to study the integration and functional impact of GABAergic iNs in epileptic mice (Lentini et al., Cell Stem Cell 28:2104-2121.e10, 2021). We successfully generated GABAergic iNs that exhibited substantial integration within pathological circuits, leading to a significant reduction in epileptic seizures.
Collapse
Affiliation(s)
- Marie d'Orange
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM, Stem Cell and Brain Research Institute U1208, Bron, France
| | - Célia Lentini
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM, Stem Cell and Brain Research Institute U1208, Bron, France
| | - Christophe Heinrich
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM, Stem Cell and Brain Research Institute U1208, Bron, France.
| |
Collapse
|
11
|
Yin Z, Kang J, Xu H, Huo S, Xu H. Recent progress of principal techniques used in the study of Müller glia reprogramming in mice. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:30. [PMID: 39663301 PMCID: PMC11635068 DOI: 10.1186/s13619-024-00211-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024]
Abstract
In zebrafish, Müller glia (MG) cells retain the ability to proliferate and de-differentiate into retinal progenitor-like cells, subsequently differentiating into retinal neurons that can replace those damaged or lost due to retinal injury. In contrast, the reprogramming potential of MG in mammals has been lost, with these cells typically responding to retinal damage through gliosis. Considerable efforts have been dedicated to achieving the reprogramming of MG cells in mammals. Notably, significant advancements have been achieved in reprogramming MG cells in mice employing various methodologies. At the same time, some inevitable challenges have hindered identifying accurate MG cell reprogramming rather than the illusion, let alone improving the reprogramming efficiency and maturity of daughter cells. Recently, several strategies, including lineage tracking, multi-omics techniques, and functional analysis, have been developed to investigate the MG reprogramming process in mice. This review summarizes both the advantages and limitations of these novel strategies for analyzing MG reprogramming in mice, offering insights into enhancing the reliability and efficiency of MG reprogramming.
Collapse
Affiliation(s)
- Zhiyuan Yin
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P.R. China
| | - Jiahui Kang
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P.R. China
| | - Haoan Xu
- School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Shujia Huo
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P.R. China.
| | - Haiwei Xu
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P.R. China.
| |
Collapse
|
12
|
Yang J. Partial Cell Fate Transitions to Promote Cardiac Regeneration. Cells 2024; 13:2002. [PMID: 39682750 PMCID: PMC11640292 DOI: 10.3390/cells13232002] [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: 10/23/2024] [Revised: 11/24/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Heart disease, including myocardial infarction (MI), remains a leading cause of morbidity and mortality worldwide, necessitating the development of more effective regenerative therapies. Direct reprogramming of cardiomyocyte-like cells from resident fibroblasts offers a promising avenue for myocardial regeneration, but its efficiency and consistency in generating functional cardiomyocytes remain limited. Alternatively, reprogramming induced cardiac progenitor cells (iCPCs) could generate essential cardiac lineages, but existing methods often involve complex procedures. These limitations underscore the need for advanced mechanistic insights and refined reprogramming strategies to improve reparative outcomes in the heart. Partial cellular fate transitions, while still a relatively less well-defined area and primarily explored in longevity and neurobiology, hold remarkable promise for cardiac repair. It enables the reprogramming or rejuvenation of resident cardiac cells into a stem or progenitor-like state with enhanced cardiogenic potential, generating the reparative lineages necessary for comprehensive myocardial recovery while reducing safety risks. As an emerging strategy, partial cellular fate transitions play a pivotal role in reversing myocardial infarction damage and offer substantial potential for therapeutic innovation. This review will summarize current advances in these areas, including recent findings involving two transcription factors that critically regulate stemness and cardiogenesis. It will also explore considerations for further refining these approaches to enhance their therapeutic potential and safety.
Collapse
Affiliation(s)
- Jianchang Yang
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
13
|
McDowall S, Bagda V, Hodgetts S, Mastaglia F, Li D. Controversies and insights into PTBP1-related astrocyte-neuron transdifferentiation: neuronal regeneration strategies for Parkinson's and Alzheimer's disease. Transl Neurodegener 2024; 13:59. [PMID: 39627843 PMCID: PMC11613593 DOI: 10.1186/s40035-024-00450-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/04/2024] [Indexed: 12/06/2024] Open
Abstract
Promising therapeutic strategies are being explored to replace or regenerate the neuronal populations that are lost in patients with neurodegenerative disorders. Several research groups have attempted direct reprogramming of astrocytes into neurons by manipulating the expression of polypyrimidine tract-binding protein 1 (PTBP1) and claimed putative converted neurons to be functional, which led to improved disease outcomes in animal models of several neurodegenerative disorders. However, a few other studies reported data that contradict these claims, raising doubt about whether PTBP1 suppression truly reprograms astrocytes into neurons and the therapeutic potential of this approach. This review discusses recent advances in regenerative therapeutics including stem cell transplantations for central nervous system disorders, with a particular focus on Parkinson's and Alzheimer's diseases. We also provide a perspective on this controversy by considering that astrocyte heterogeneity may be the key to understanding the discrepancy in published studies, and that certain subpopulations of these glial cells may be more readily converted into neurons.
Collapse
Affiliation(s)
- Simon McDowall
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
- School of Human Sciences, The University of Western Australia, Crawley, Perth, WA, Australia
- Department of Anatomy and Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Vaishali Bagda
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
| | - Stuart Hodgetts
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
- School of Human Sciences, The University of Western Australia, Crawley, Perth, WA, Australia
| | - Frank Mastaglia
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.
| | - Dunhui Li
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia.
- Centre for Neuromuscular and Neurological Disorders, Nedlands, WA, Australia.
- Department of Neurology and Stephen and Denise Adams Center for Parkinson's Disease Research, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
14
|
Wei H, Wu JQ. Glial progenitor heterogeneity and plasticity in the adult spinal cord. Neural Regen Res 2024; 19:2567-2568. [PMID: 38808984 PMCID: PMC11168497 DOI: 10.4103/nrr.nrr-d-23-01988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/29/2024] [Accepted: 02/21/2024] [Indexed: 05/30/2024] Open
Affiliation(s)
- Haichao Wei
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX, USA; MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Jia Qian Wu
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX, USA; MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
15
|
Rigo YR, Benvenutti R, Portela LV, Strogulski NR. Neurogenic potential of NG2 in neurotrauma: a systematic review. Neural Regen Res 2024; 19:2673-2683. [PMID: 38595286 PMCID: PMC11168526 DOI: 10.4103/nrr.nrr-d-23-01031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 12/20/2023] [Accepted: 02/07/2024] [Indexed: 04/11/2024] Open
Abstract
Regenerative approaches towards neuronal loss following traumatic brain or spinal cord injury have long been considered a dogma in neuroscience and remain a cutting-edge area of research. This is reflected in a large disparity between the number of studies investigating primary and secondary injury as therapeutic targets in spinal cord and traumatic brain injuries. Significant advances in biotechnology may have the potential to reshape the current state-of-the-art and bring focus to primary injury neurotrauma research. Recent studies using neural-glial factor/antigen 2 (NG2) cells indicate that they may differentiate into neurons even in the developed brain. As these cells show great potential to play a regenerative role, studies have been conducted to test various manipulations in neurotrauma models aimed at eliciting a neurogenic response from them. In the present study, we systematically reviewed the experimental protocols and findings described in the scientific literature, which were peer-reviewed original research articles (1) describing preclinical experimental studies, (2) investigating NG2 cells, (3) associated with neurogenesis and neurotrauma, and (4) in vitro and/or in vivo, available in PubMed/MEDLINE, Web of Science or SCOPUS, from 1998 to 2022. Here, we have reviewed a total of 1504 papers, and summarized findings that ultimately suggest that NG2 cells possess an inducible neurogenic potential in animal models and in vitro. We also discriminate findings of NG2 neurogenesis promoted by different pharmacological and genetic approaches over functional and biochemical outcomes of traumatic brain injury and spinal cord injury models, and provide mounting evidence for the potential benefits of manipulated NG2 cell ex vivo transplantation in primary injury treatment. These findings indicate the feasibility of NG2 cell neurogenesis strategies and add new players in the development of therapeutic alternatives for neurotrauma.
Collapse
Affiliation(s)
- Yuri R. Rigo
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Radharani Benvenutti
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Luis V. Portela
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Nathan R. Strogulski
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, University of Dublin, Dublin, Ireland
| |
Collapse
|
16
|
Liu Y, Wei C, Yang Y, Zhu Z, Ren Y, Pi R. In situ chemical reprogramming of astrocytes into neurons: A new hope for the treatment of central neurodegenerative diseases? Eur J Pharmacol 2024; 982:176930. [PMID: 39179093 DOI: 10.1016/j.ejphar.2024.176930] [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: 05/16/2024] [Revised: 07/18/2024] [Accepted: 08/21/2024] [Indexed: 08/26/2024]
Abstract
Central neurodegenerative disorders (e.g. Alzheimer's disease (AD) and Parkinson's disease (PD)) are tightly associated with extensive neuron loss. Current therapeutic interventions merely mitigate the symptoms of these diseases, falling short of addressing the fundamental issue of neuron loss. Cell reprogramming, involving the transition of a cell from one gene expression profile to another, has made significant strides in the conversion between diverse somatic cell types. This advancement has been facilitated by gene editing techniques or the synergistic application of small molecules, enabling the conversion of glial cells into functional neurons. Despite this progress, the potential for in situ reprogramming of astrocytes in treating neurodegenerative disorders faces challenges such as immune rejection and genotoxicity. A novel avenue emerges through chemical reprogramming of astrocytes utilizing small molecules, circumventing genotoxic effects and unlocking substantial clinical utility. Recent studies have successfully demonstrated the in situ conversion of astrocytes into neurons using small molecules. Nonetheless, these findings have sparked debates, encompassing queries regarding the origin of newborn neurons, pivotal molecular targets, and alterations in metabolic pathways. This review succinctly delineates the background of astrocytes reprogramming, meticulously surveys the principal classes of small molecule combinations employed thus far, and examines the complex signaling pathways they activate. Finally, this article delves into the potential vistas awaiting exploration in the realm of astrocytes chemical reprogramming, heralding a promising future for advancing our understanding and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yuan Liu
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Cailv Wei
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yang Yang
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zeyu Zhu
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yu Ren
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Rongbiao Pi
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of Guangdong, Shenzhen, 518107, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
17
|
Marichal N, Péron S, Beltrán Arranz A, Galante C, Franco Scarante F, Wiffen R, Schuurmans C, Karow M, Gascón S, Berninger B. Reprogramming astroglia into neurons with hallmarks of fast-spiking parvalbumin-positive interneurons by phospho-site-deficient Ascl1. SCIENCE ADVANCES 2024; 10:eadl5935. [PMID: 39454007 PMCID: PMC11506222 DOI: 10.1126/sciadv.adl5935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 09/19/2024] [Indexed: 10/27/2024]
Abstract
Cellular reprogramming of mammalian glia to an induced neuronal fate holds the potential for restoring diseased brain circuits. While the proneural factor achaete-scute complex-like 1 (Ascl1) is widely used for neuronal reprogramming, in the early postnatal mouse cortex, Ascl1 fails to induce the glia-to-neuron conversion, instead promoting the proliferation of oligodendrocyte progenitor cells (OPC). Since Ascl1 activity is posttranslationally regulated, here, we investigated the consequences of mutating six serine phospho-acceptor sites to alanine (Ascl1SA6) on lineage reprogramming in vivo. Ascl1SA6 exhibited increased neurogenic activity in the glia of the early postnatal mouse cortex, an effect enhanced by coexpression of B cell lymphoma 2 (Bcl2). Genetic fate-mapping revealed that most induced neurons originated from astrocytes, while only a few derived from OPCs. Many Ascl1SA6/Bcl2-induced neurons expressed parvalbumin and were capable of high-frequency action potential firing. Our study demonstrates the authentic conversion of astroglia into neurons featuring subclass hallmarks of cortical interneurons, advancing our scope of engineering neuronal fates in the brain.
Collapse
Affiliation(s)
- Nicolás Marichal
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Sophie Péron
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- Institute of Physiological Chemistry, University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Ana Beltrán Arranz
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Chiara Galante
- Institute of Physiological Chemistry, University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Franciele Franco Scarante
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Rebecca Wiffen
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Carol Schuurmans
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Marisa Karow
- Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sergio Gascón
- Department of Molecular, Cellular and Developmental Neurobiology, Cajal Institute – CSIC, Madrid, Spain
| | - Benedikt Berninger
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- Institute of Physiological Chemistry, University Medical Center Johannes Gutenberg University, Mainz, Germany
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- The Francis Crick Institute, London, UK
- Focus Program Translational Neuroscience, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
18
|
Liu MH, Xu YG, Bai XN, Lin JH, Xiang ZQ, Wang T, Xu L, Chen G. Efficient Dlx2-mediated astrocyte-to-neuron conversion and inhibition of neuroinflammation by NeuroD1. Dev Neurobiol 2024; 84:274-290. [PMID: 39034481 DOI: 10.1002/dneu.22951] [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: 08/21/2023] [Revised: 06/05/2024] [Accepted: 07/01/2024] [Indexed: 07/23/2024]
Abstract
In vivo astrocyte-to-neuron (AtN) conversion induced by overexpression of neural transcriptional factors has great potential for neural regeneration and repair. Here, we demonstrate that a single neural transcriptional factor, Dlx2, converts mouse striatal astrocytes into neurons in a dose-dependent manner. Lineage-tracing studies in Aldh1l1-CreERT2 mice confirm that Dlx2 can convert striatal astrocytes into DARPP32+ and Ctip2+ medium spiny neurons (MSNs). Time-course studies reveal a gradual conversion from astrocytes to neurons in 1 month, with a distinct intermediate state in between astrocytes and neurons. Interestingly, when Dlx2-infected astrocytes start to lose astrocytic markers, the other local astrocytes proliferate to maintain astrocytic levels in the converted areas. Unexpectedly, although Dlx2 efficiently reprograms astrocytes into neurons in the gray matter striatum, it also induces partial reprogramming of astrocytes in the white matter corpus callosum. Such partial reprogramming of white matter astrocytes is associated with neuroinflammation, which can be suppressed by the addition of NeuroD1. Our results highlight the importance of investigating AtN conversion in both the gray matter and white matter to thoroughly evaluate therapeutic potentials. This study also unveils the critical role of anti-inflammation by NeuroD1 during AtN conversion.
Collapse
Affiliation(s)
- Min-Hui Liu
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
- VIB Center for Brain and Disease, KU Leuven, Herestraat 49, Leuven, Belgium
| | - Yu-Ge Xu
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Xiao-Ni Bai
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Jian-Hua Lin
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Zong-Qin Xiang
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
- Department of Neurosurgery, the First Affiliated Hospital, Jinan University, Guangzhou, Guangdong Province, China
| | - Tao Wang
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Liang Xu
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Gong Chen
- Guangdong-HongKong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| |
Collapse
|
19
|
Li Y, Luo W, Meng C, Shi K, Gu R, Cui S. Exosomes as promising bioactive materials in the treatment of spinal cord injury. Stem Cell Res Ther 2024; 15:335. [PMID: 39334506 PMCID: PMC11438208 DOI: 10.1186/s13287-024-03952-5] [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: 07/31/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Patients with spinal cord injury (SCI) have permanent devastating motor and sensory disabilities. Secondary SCI is known for its complex progression and presents with sophisticated aberrant inflammation, vascular changes, and secondary cellular dysfunction, which aggravate the primary damage. Since their initial discovery, the potent neuroprotective effects and powerful delivery abilities of exosomes (Exos) have been reported in different research fields, including SCI. In this study, we summarize therapeutic advances related to the application of Exos in preclinical animal studies. Subsequently, we discuss the mechanisms of action of Exos derived from diverse cell types, including neurogenesis, angiogenesis, blood-spinal cord barrier preservation, anti-apoptosis, and anti-inflammatory potential. We also evaluate the relationship between the Exo delivery cargo and signaling pathways. Finally, we discuss the challenges and advantages of using Exos to offer innovative insights regarding the development of efficient clinical strategies for SCI.
Collapse
Affiliation(s)
- Yueying Li
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, No. 126 Xiantai Street, Changchun, Jilin, 130033, P.R. China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, No. 126 Xiantai Street, Changchun, Jilin, 130033, P.R. China
| | - Wenqi Luo
- Department of Orthopaedic Surgery, China-Japan Union Hospital of Jilin University, No. 126 Xiantai Street, Changchun, Jilin, 130033, P.R. China
| | - Chuikai Meng
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, No. 126 Xiantai Street, Changchun, Jilin, 130033, P.R. China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, No. 126 Xiantai Street, Changchun, Jilin, 130033, P.R. China
| | - Kaiyuan Shi
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, No. 126 Xiantai Street, Changchun, Jilin, 130033, P.R. China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, No. 126 Xiantai Street, Changchun, Jilin, 130033, P.R. China
| | - Rui Gu
- Department of Orthopaedic Surgery, China-Japan Union Hospital of Jilin University, No. 126 Xiantai Street, Changchun, Jilin, 130033, P.R. China.
| | - Shusen Cui
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, No. 126 Xiantai Street, Changchun, Jilin, 130033, P.R. China.
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, No. 126 Xiantai Street, Changchun, Jilin, 130033, P.R. China.
| |
Collapse
|
20
|
Serrano C, Cananzi S, Shen T, Wang LL, Zhang CL. Simple and highly specific targeting of resident microglia with adeno-associated virus. iScience 2024; 27:110706. [PMID: 39297168 PMCID: PMC11407971 DOI: 10.1016/j.isci.2024.110706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/28/2024] [Accepted: 08/07/2024] [Indexed: 09/21/2024] Open
Abstract
Microglia, as the immune cells of the central nervous system (CNS), play dynamic roles in both healthy and diseased conditions. The ability to genetically target microglia using viruses is crucial for understanding their functions and advancing microglia-based treatments. We here show that resident microglia can be simply and specifically targeted using adeno-associated virus (AAV) vectors containing a 466-bp DNA fragment from the human IBA1 (hIBA1) promoter. This targeting approach is applicable to both resting and reactive microglia. When combining the short hIBA1 promoter with the target sequence of miR124, up to 98% of transduced cells are identified as microglia. Such a simple and highly specific microglia-targeting strategy may be further optimized for research and therapeutics.
Collapse
Affiliation(s)
- Carolina Serrano
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sergio Cananzi
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tianjin Shen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lei-Lei Wang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chun-Li Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
21
|
Fukui Y, Morihara R, Hu X, Nakano Y, Yunoki T, Takemoto M, Abe K, Yamashita T. Suppression of PTBP1 in hippocampal astrocytes promotes neurogenesis and ameliorates recognition memory in mice with cerebral ischemia. Sci Rep 2024; 14:20521. [PMID: 39227632 PMCID: PMC11372044 DOI: 10.1038/s41598-024-71212-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 08/26/2024] [Indexed: 09/05/2024] Open
Abstract
The therapeutic potential of suppressing polypyrimidine tract-binding protein 1 (Ptbp1) messenger RNA by viral transduction in a post-stroke dementia mouse model has not yet been examined. In this study, 3 days after cerebral ischemia, we injected a viral vector cocktail containing adeno-associated virus (AAV)-pGFAP-mCherry and AAV-pGFAP-CasRx (control vector) or a cocktail of AAV-pGFAP-mCherry and AAV-pGFAP-CasRx-SgRNA-(Ptbp1) (1:5, 1.0 × 1011 viral genomes) into post-stroke mice via the tail vein. We observed new mCherry/NeuN double-positive neuron-like cells in the hippocampus 56 days after cerebral ischemia. A portion of mCherry/GFAP double-positive astrocyte-like glia could have been converted into new mCherry/NeuN double-positive neuron-like cells with morphological changes. The new neuronal cells integrated into the dentate gyrus and recognition memory was significantly ameliorated. These results demonstrated that the in vivo conversion of hippocampal astrocyte-like glia into functional new neurons by the suppression of Ptbp1 might be a therapeutic strategy for post-stroke dementia.
Collapse
Affiliation(s)
- Yusuke Fukui
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikatacho, Kitaku, Okayama, 700-8558, Japan
| | - Ryuta Morihara
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikatacho, Kitaku, Okayama, 700-8558, Japan
| | - Xinran Hu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikatacho, Kitaku, Okayama, 700-8558, Japan
| | - Yumiko Nakano
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikatacho, Kitaku, Okayama, 700-8558, Japan
| | - Taijun Yunoki
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikatacho, Kitaku, Okayama, 700-8558, Japan
| | - Mami Takemoto
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikatacho, Kitaku, Okayama, 700-8558, Japan
| | - Koji Abe
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikatacho, Kitaku, Okayama, 700-8558, Japan
| | - Toru Yamashita
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikatacho, Kitaku, Okayama, 700-8558, Japan.
| |
Collapse
|
22
|
Talifu Z, Zhang C, Xu X, Pan Y, Ke H, Li Z, Liu W, Du H, Wang X, Gao F, Yang D, Jing Y, Yu Y, Du L, Li J. Neuronal repair after spinal cord injury by in vivo astrocyte reprogramming mediated by the overexpression of NeuroD1 and Neurogenin-2. Biol Res 2024; 57:53. [PMID: 39135103 PMCID: PMC11318173 DOI: 10.1186/s40659-024-00534-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/05/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND As a common disabling disease, irreversible neuronal death due to spinal cord injury (SCI) is the root cause of functional impairment; however, the capacity for neuronal regeneration in the developing spinal cord tissue is limited. Therefore, there is an urgent need to investigate how defective neurons can be replenished and functionally integrated by neural regeneration; the reprogramming of intrinsic cells into functional neurons may represent an ideal solution. METHODS A mouse model of transection SCI was prepared by forceps clamping, and an adeno-associated virus (AAV) carrying the transcription factors NeuroD1 and Neurogenin-2(Ngn2) was injected in situ into the spinal cord to specifically overexpress these transcription factors in astrocytes close to the injury site. 5-bromo-2´-deoxyuridine (BrdU) was subsequently injected intraperitoneally to continuously track cell regeneration, neuroblasts and immature neurons marker expression, neuronal regeneration, and glial scar regeneration. In addition, immunoprotein blotting was used to measure the levels of transforming growth factor-β (TGF-β) pathway-related protein expression. We also evaluated motor function, sensory function, and the integrity of the blood-spinal cord barrier(BSCB). RESULTS The in situ overexpression of NeuroD1 and Ngn2 in the spinal cord was achieved by specific AAV vectors. This intervention led to a significant increase in cell regeneration and the proportion of cells with neuroblasts and immature neurons cell properties at the injury site(p < 0.0001). Immunofluorescence staining identified astrocytes with neuroblasts and immature neurons cell properties at the site of injury while neuronal marker-specific staining revealed an increased number of mature astrocytes at the injury site. Behavioral assessments showed that the intervention did not improve The BMS (Basso mouse scale) score (p = 0.0726) and gait (p > 0.05), although the treated mice had more sensory sensitivity and greater voluntary motor ability in open field than the non-intervention mice. We observed significant repair of the BSCB at the center of the injury site (p < 0.0001) and a significant improvement in glial scar proliferation. Electrophysiological assessments revealed a significant improvement in spinal nerve conduction (p < 0.0001) while immunostaining revealed that the levels of TGF-β protein at the site of injury in the intervention group were lower than control group (p = 0.0034); in addition, P70 s6 and PP2A related to the TGF-β pathway showed ascending trend (p = 0.0036, p = 0.0152 respectively). CONCLUSIONS The in situ overexpression of NeuroD1 and Ngn2 in the spinal cord after spinal cord injury can reprogram astrocytes into neurons and significantly enhance cell regeneration at the injury site. The reprogramming of astrocytes can lead to tissue repair, thus improving the reduced threshold and increasing voluntary movements. This strategy can also improve the integrity of the blood-spinal cord barrier and enhance nerve conduction function. However, the simple reprogramming of astrocytes cannot lead to significant improvements in the striding function of the lower limbs.
Collapse
Affiliation(s)
- Zuliyaer Talifu
- School of Rehabilitation, Capital Medical University; Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center; Chinese Institute of Rehabilitation Science; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China
- University of Health and Rehabilitation Sciences, Shandong, 266113, China
| | - Chunjia Zhang
- Department of Rehabilitation Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Xin Xu
- School of Rehabilitation, Capital Medical University; Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center; Chinese Institute of Rehabilitation Science; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
- University of Health and Rehabilitation Sciences, Shandong, 266113, China
- Cheeloo College of Medicine, Shandong University, Shandong Province, Jinan, 250100, China
| | - Yunzhu Pan
- School of Rehabilitation, Capital Medical University; Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center; Chinese Institute of Rehabilitation Science; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
- University of Health and Rehabilitation Sciences, Shandong, 266113, China
| | - Han Ke
- Cheeloo College of Medicine, Shandong University, Shandong Province, Jinan, 250100, China
| | - Zehui Li
- School of Rehabilitation, Capital Medical University; Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center; Chinese Institute of Rehabilitation Science; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
| | - Wubo Liu
- Cheeloo College of Medicine, Shandong University, Shandong Province, Jinan, 250100, China
| | - Huayong Du
- School of Rehabilitation, Capital Medical University; Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center; Chinese Institute of Rehabilitation Science; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
| | - Xiaoxin Wang
- School of Rehabilitation, Capital Medical University; Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center; Chinese Institute of Rehabilitation Science; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
| | - Feng Gao
- School of Rehabilitation, Capital Medical University; Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center; Chinese Institute of Rehabilitation Science; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
| | - Degang Yang
- School of Rehabilitation, Capital Medical University; Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center; Chinese Institute of Rehabilitation Science; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
| | - Yingli Jing
- School of Rehabilitation, Capital Medical University; Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center; Chinese Institute of Rehabilitation Science; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
| | - Yan Yu
- School of Rehabilitation, Capital Medical University; Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center; Chinese Institute of Rehabilitation Science; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
| | - Liangjie Du
- School of Rehabilitation, Capital Medical University; Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center; Chinese Institute of Rehabilitation Science; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China
| | - Jianjun Li
- School of Rehabilitation, Capital Medical University; Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research Center; Chinese Institute of Rehabilitation Science; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, China.
- University of Health and Rehabilitation Sciences, Shandong, 266113, China.
- Cheeloo College of Medicine, Shandong University, Shandong Province, Jinan, 250100, China.
| |
Collapse
|
23
|
Li ST, Wan Y, Chen L, Ding Y. Advances in neuronal reprogramming for neurodegenerative diseases: Strategies, controversies, and opportunities. Exp Neurol 2024; 378:114817. [PMID: 38763354 DOI: 10.1016/j.expneurol.2024.114817] [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] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
Neuronal death is often observed in central nervous system injuries and neurodegenerative diseases. The mammalian central nervous system manifests limited neuronal regeneration capabilities, and traditional cell therapies are limited in their potential applications due to finite cell sources and immune rejection. Neuronal reprogramming has emerged as a novel technology, in which non-neuronal cells (e.g. glial cells) are transdifferentiated into mature neurons. This process results in relatively minimal immune rejection. The present review discuss the latest progress in this cutting-edge field, including starter cell selection, innovative technical strategies and methods of neuronal reprogramming for neurodegenerative diseases, as well as the potential problems and controversies. The further development of neuronal reprogramming technology may pave the way for novel therapeutic strategies in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Si-Tong Li
- Department of Histology and Embryology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Yue Wan
- Department of Histology and Embryology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Li Chen
- Department of Histology and Embryology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Yan Ding
- Department of Histology and Embryology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
24
|
Mseis-Jackson N, Sharma M, Li H. Controlling the Expression Level of the Neuronal Reprogramming Factors for a Successful Reprogramming Outcome. Cells 2024; 13:1223. [PMID: 39056804 PMCID: PMC11274869 DOI: 10.3390/cells13141223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024] Open
Abstract
Neuronal reprogramming is a promising approach for making major advancement in regenerative medicine. Distinct from the approach of induced pluripotent stem cells, neuronal reprogramming converts non-neuronal cells to neurons without going through a primitive stem cell stage. In vivo neuronal reprogramming brings this approach to a higher level by changing the cell fate of glial cells to neurons in neural tissue through overexpressing reprogramming factors. Despite the ongoing debate over the validation and interpretation of newly generated neurons, in vivo neuronal reprogramming is still a feasible approach and has the potential to become clinical treatment with further optimization and refinement. Here, we discuss the major neuronal reprogramming factors (mostly pro-neurogenic transcription factors during development), especially the significance of their expression levels during neurogenesis and the reprogramming process focusing on NeuroD1. In the developing central nervous system, these pro-neurogenic transcription factors usually elicit distinct spatiotemporal expression patterns that are critical to their function in generating mature neurons. We argue that these dynamic expression patterns may be similarly needed in the process of reprogramming adult cells into neurons and further into mature neurons with subtype identities. We also summarize the existing approaches and propose new ones that control gene expression levels for a successful reprogramming outcome.
Collapse
Affiliation(s)
- Natalie Mseis-Jackson
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Mehek Sharma
- Department of Biological Sciences, College of Science & Mathematics, Augusta University, Augusta, GA 30912, USA;
| | - Hedong Li
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| |
Collapse
|
25
|
Liang S, Zhou J, Yu X, Lu S, Liu R. Neuronal conversion from glia to replenish the lost neurons. Neural Regen Res 2024; 19:1446-1453. [PMID: 38051886 PMCID: PMC10883502 DOI: 10.4103/1673-5374.386400] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 08/16/2023] [Indexed: 12/07/2023] Open
Abstract
ABSTRACT Neuronal injury, aging, and cerebrovascular and neurodegenerative diseases such as cerebral infarction, Alzheimer's disease, Parkinson's disease, frontotemporal dementia, amyotrophic lateral sclerosis, and Huntington's disease are characterized by significant neuronal loss. Unfortunately, the neurons of most mammals including humans do not possess the ability to self-regenerate. Replenishment of lost neurons becomes an appealing therapeutic strategy to reverse the disease phenotype. Transplantation of pluripotent neural stem cells can supplement the missing neurons in the brain, but it carries the risk of causing gene mutation, tumorigenesis, severe inflammation, and obstructive hydrocephalus induced by brain edema. Conversion of neural or non-neural lineage cells into functional neurons is a promising strategy for the diseases involving neuron loss, which may overcome the above-mentioned disadvantages of neural stem cell therapy. Thus far, many strategies to transform astrocytes, fibroblasts, microglia, Müller glia, NG2 cells, and other glial cells to mature and functional neurons, or for the conversion between neuronal subtypes have been developed through the regulation of transcription factors, polypyrimidine tract binding protein 1 (PTBP1), and small chemical molecules or are based on a combination of several factors and the location in the central nervous system. However, some recent papers did not obtain expected results, and discrepancies exist. Therefore, in this review, we discuss the history of neuronal transdifferentiation, summarize the strategies for neuronal replenishment and conversion from glia, especially astrocytes, and point out that biosafety, new strategies, and the accurate origin of the truly converted neurons in vivo should be focused upon in future studies. It also arises the attention of replenishing the lost neurons from glia by gene therapies such as up-regulation of some transcription factors or down-regulation of PTBP1 or drug interference therapies.
Collapse
Affiliation(s)
- Shiyu Liang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Jing Zhou
- Department of Geriatric Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Xiaolin Yu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Shuai Lu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Ruitian Liu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
26
|
Finkel Z, Esteban F, Rodriguez B, Clifford T, Joseph A, Alostaz H, Dalmia M, Gutierrez J, Tamasi MJ, Zhang SM, Simone J, Petekci H, Nath S, Escott M, Garg SK, Gormley AJ, Kumar S, Gulati S, Cai L. AAV6 mediated Gsx1 expression in neural stem progenitor cells promotes neurogenesis and restores locomotor function after contusion spinal cord injury. Neurotherapeutics 2024; 21:e00362. [PMID: 38664194 PMCID: PMC11452562 DOI: 10.1016/j.neurot.2024.e00362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/29/2024] [Accepted: 04/11/2024] [Indexed: 05/05/2024] Open
Abstract
Genomic screened homeobox 1 (Gsx1 or Gsh1) is a neurogenic transcription factor required for the generation of excitatory and inhibitory interneurons during spinal cord development. In the adult, lentivirus (LV) mediated Gsx1 expression promotes neural regeneration and functional locomotor recovery in a mouse model of lateral hemisection spinal cord injury (SCI). The LV delivery method is clinically unsafe due to insertional mutations to the host DNA. In addition, the most common clinical case of SCI is contusion/compression. In this study, we identify that adeno-associated virus serotype 6 (AAV6) preferentially infects neural stem/progenitor cells (NSPCs) in the injured spinal cord. Using a rat model of contusion SCI, we demonstrate that AAV6 mediated Gsx1 expression promotes neurogenesis, increases the number of neuroblasts/immature neurons, restores excitatory/inhibitory neuron balance and serotonergic neuronal activity through the lesion core, and promotes locomotor functional recovery. Our findings support that AAV6 preferentially targets NSPCs for gene delivery and confirmed Gsx1 efficacy in clinically relevant rat model of contusion SCI.
Collapse
Affiliation(s)
- Zachary Finkel
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, NJ 08854, USA
| | - Fatima Esteban
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, NJ 08854, USA
| | - Brianna Rodriguez
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, NJ 08854, USA
| | - Tanner Clifford
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, NJ 08854, USA
| | - Adelina Joseph
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, NJ 08854, USA
| | - Hani Alostaz
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, NJ 08854, USA
| | - Mridul Dalmia
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, NJ 08854, USA
| | - Juan Gutierrez
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, NJ 08854, USA; University of California, Santa Barbara, CA 93106, USA
| | - Matthew J Tamasi
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, NJ 08854, USA
| | - Samuel Ming Zhang
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, NJ 08854, USA
| | - Jonah Simone
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, NJ 08854, USA
| | - Hafize Petekci
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, NJ 08854, USA
| | - Susmita Nath
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, NJ 08854, USA
| | - Miriam Escott
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, NJ 08854, USA
| | - Shivam Kumar Garg
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, NJ 08854, USA
| | - Adam J Gormley
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, NJ 08854, USA
| | - Suneel Kumar
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, NJ 08854, USA
| | - Sonia Gulati
- NeuroNovus Therapeutics Inc., 135 E 57th St., New York, NY 10022, USA
| | - Li Cai
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd, Piscataway, NJ 08854, USA; NeuroNovus Therapeutics Inc., 135 E 57th St., New York, NY 10022, USA.
| |
Collapse
|
27
|
Li H, Zhuang Y, Zhang B, Xu X, Liu B. Application of Lineage Tracing in Central Nervous System Development and Regeneration. Mol Biotechnol 2024; 66:1552-1562. [PMID: 37335434 PMCID: PMC11217125 DOI: 10.1007/s12033-023-00769-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 05/09/2023] [Indexed: 06/21/2023]
Abstract
The central nervous system (CNS) is a complicated neural network. The origin and evolution of functional neurons and glia cells remain unclear, as do the cellular alterations that occur during the course of cerebral disease rehabilitation. Lineage tracing is a valuable method for tracing specific cells and achieving a better understanding of the CNS. Recently, various technological breakthroughs have been made in lineage tracing, such as the application of various combinations of fluorescent reporters and advances in barcode technology. The development of lineage tracing has given us a deeper understanding of the normal physiology of the CNS, especially the pathological processes. In this review, we summarize these advances of lineage tracing and their applications in CNS. We focus on the use of lineage tracing techniques to elucidate the process CNS development and especially the mechanism of injury repair. Deep understanding of the central nervous system will help us to use existing technologies to diagnose and treat diseases.
Collapse
Affiliation(s)
- Hao Li
- Department of Neurosurgery, Beijing Tian tan Hospital, Capital Medical University, Beijing, China
| | - Yuan Zhuang
- Department of Neurosurgery, Beijing Tian tan Hospital, Capital Medical University, Beijing, China
| | - Bin Zhang
- Department of Intensive Care Unit, Beijing Tian tan Hospital, Capital Medical University, Beijing, China
| | - Xiaojian Xu
- Beijing Key Laboratory of Central Nervous System Injury, Department of Neurotrauma, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Baiyun Liu
- Department of Neurosurgery, Beijing Tian tan Hospital, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Central Nervous System Injury, Department of Neurotrauma, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
- Center for Nerve Injury and Repair, Beijing Institute of Brain Disorders, Beijing, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
28
|
Wei J, Wang M, Li S, Han R, Xu W, Zhao A, Yu Q, Li H, Li M, Chi G. Reprogramming of astrocytes and glioma cells into neurons for central nervous system repair and glioblastoma therapy. Biomed Pharmacother 2024; 176:116806. [PMID: 38796971 DOI: 10.1016/j.biopha.2024.116806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024] Open
Abstract
Central nervous system (CNS) damage is usually irreversible owing to the limited regenerative capability of neurons. Following CNS injury, astrocytes are reactively activated and are the key cells involved in post-injury repair mechanisms. Consequently, research on the reprogramming of reactive astrocytes into neurons could provide new directions for the restoration of neural function after CNS injury and in the promotion of recovery in various neurodegenerative diseases. This review aims to provide an overview of the means through which reactive astrocytes around lesions can be reprogrammed into neurons, to elucidate the intrinsic connection between the two cell types from a neurogenesis perspective, and to summarize what is known about the neurotranscription factors, small-molecule compounds and MicroRNA that play major roles in astrocyte reprogramming. As the malignant proliferation of astrocytes promotes the development of glioblastoma multiforme (GBM), this review also examines the research advances on and the theoretical basis for the reprogramming of GBM cells into neurons and discusses the advantages of such approaches over traditional treatment modalities. This comprehensive review provides new insights into the field of GBM therapy and theoretical insights into the mechanisms of neurological recovery following neurological injury and in GBM treatment.
Collapse
Affiliation(s)
- Junyuan Wei
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Miaomiao Wang
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Shilin Li
- School of Public Health, Jilin University, Changchun 130021, China.
| | - Rui Han
- Department of Neurovascular Surgery, First Hospital of Jilin University, 1xinmin Avenue, Changchun, Jilin Province 130021, China.
| | - Wenhong Xu
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Anqi Zhao
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Qi Yu
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Haokun Li
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Meiying Li
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Guangfan Chi
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
29
|
Marangon D, Castro e Silva JH, Cerrato V, Boda E, Lecca D. Oligodendrocyte Progenitors in Glial Scar: A Bet on Remyelination. Cells 2024; 13:1024. [PMID: 38920654 PMCID: PMC11202012 DOI: 10.3390/cells13121024] [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: 05/07/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) represent a subtype of glia, giving rise to oligodendrocytes, the myelin-forming cells in the central nervous system (CNS). While OPCs are highly proliferative during development, they become relatively quiescent during adulthood, when their fate is strictly influenced by the extracellular context. In traumatic injuries and chronic neurodegenerative conditions, including those of autoimmune origin, oligodendrocytes undergo apoptosis, and demyelination starts. Adult OPCs become immediately activated; they migrate at the lesion site and proliferate to replenish the damaged area, but their efficiency is hampered by the presence of a glial scar-a barrier mainly formed by reactive astrocytes, microglia and the deposition of inhibitory extracellular matrix components. If, on the one hand, a glial scar limits the lesion spreading, it also blocks tissue regeneration. Therapeutic strategies aimed at reducing astrocyte or microglia activation and shifting them toward a neuroprotective phenotype have been proposed, whereas the role of OPCs has been largely overlooked. In this review, we have considered the glial scar from the perspective of OPCs, analysing their behaviour when lesions originate and exploring the potential therapies aimed at sustaining OPCs to efficiently differentiate and promote remyelination.
Collapse
Affiliation(s)
- Davide Marangon
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (D.M.); (J.H.C.e.S.)
| | - Juliana Helena Castro e Silva
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (D.M.); (J.H.C.e.S.)
| | - Valentina Cerrato
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126 Turin, Italy; (V.C.); (E.B.)
- Neuroscience Institute Cavalieri Ottolenghi, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Enrica Boda
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126 Turin, Italy; (V.C.); (E.B.)
- Neuroscience Institute Cavalieri Ottolenghi, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Davide Lecca
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (D.M.); (J.H.C.e.S.)
| |
Collapse
|
30
|
Fischer G, Bättig L, Stienen MN, Curt A, Fehlings MG, Hejrati N. Advancements in neuroregenerative and neuroprotective therapies for traumatic spinal cord injury. Front Neurosci 2024; 18:1372920. [PMID: 38812974 PMCID: PMC11133582 DOI: 10.3389/fnins.2024.1372920] [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: 01/18/2024] [Accepted: 04/10/2024] [Indexed: 05/31/2024] Open
Abstract
Traumatic spinal cord injuries (SCIs) continue to be a major healthcare concern, with a rising prevalence worldwide. In response to this growing medical challenge, considerable scientific attention has been devoted to developing neuroprotective and neuroregenerative strategies aimed at improving the prognosis and quality of life for individuals with SCIs. This comprehensive review aims to provide an up-to-date and thorough overview of the latest neuroregenerative and neuroprotective therapies currently under investigation. These strategies encompass a multifaceted approach that include neuropharmacological interventions, cell-based therapies, and other promising strategies such as biomaterial scaffolds and neuro-modulation therapies. In addition, the review discusses the importance of acute clinical management, including the role of hemodynamic management as well as timing and technical aspects of surgery as key factors mitigating the secondary injury following SCI. In conclusion, this review underscores the ongoing scientific efforts to enhance patient outcomes and quality of life, focusing on upcoming strategies for the management of traumatic SCI. Each section provides a working knowledge of the fundamental preclinical and patient trials relevant to clinicians while underscoring the pathophysiologic rationale for the therapies.
Collapse
Affiliation(s)
- Gregor Fischer
- Department of Neurosurgery, Cantonal Hospital St.Gallen, Medical School of St.Gallen, St.Gallen, Switzerland
- Spine Center of Eastern Switzerland, Cantonal Hospital St.Gallen, Medical School of St.Gallen, St.Gallen, Switzerland
| | - Linda Bättig
- Department of Neurosurgery, Cantonal Hospital St.Gallen, Medical School of St.Gallen, St.Gallen, Switzerland
- Spine Center of Eastern Switzerland, Cantonal Hospital St.Gallen, Medical School of St.Gallen, St.Gallen, Switzerland
| | - Martin N. Stienen
- Department of Neurosurgery, Cantonal Hospital St.Gallen, Medical School of St.Gallen, St.Gallen, Switzerland
- Spine Center of Eastern Switzerland, Cantonal Hospital St.Gallen, Medical School of St.Gallen, St.Gallen, Switzerland
| | - Armin Curt
- Spinal Cord Injury Center, University Hospital Balgrist, Zurich, Switzerland
| | - Michael G. Fehlings
- Division of Neurosurgery and Spine Program, Department of Surgery, University of Toronto, Toronto, ON, Canada
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Nader Hejrati
- Department of Neurosurgery, Cantonal Hospital St.Gallen, Medical School of St.Gallen, St.Gallen, Switzerland
- Spine Center of Eastern Switzerland, Cantonal Hospital St.Gallen, Medical School of St.Gallen, St.Gallen, Switzerland
| |
Collapse
|
31
|
Tan Z, Qin S, Liu H, Huang X, Pu Y, He C, Yuan Y, Su Z. Small molecules reprogram reactive astrocytes into neuronal cells in the injured adult spinal cord. J Adv Res 2024; 59:111-127. [PMID: 37380102 PMCID: PMC11081968 DOI: 10.1016/j.jare.2023.06.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/23/2023] [Accepted: 06/25/2023] [Indexed: 06/30/2023] Open
Abstract
INTRODUCTION Ectopic expression of transcription factor-mediated in vivo neuronal reprogramming provides promising strategy to compensate for neuronal loss, while its further clinical application may be hindered by delivery and safety concerns. As a novel and attractive alternative, small molecules may offer a non-viral and non-integrative chemical approach for reprogramming cell fates. Recent definitive evidences have shown that small molecules can convert non-neuronal cells into neurons in vitro. However, whether small molecules alone can induce neuronal reprogramming in vivo remains largely unknown. OBJECTIVES To identify chemical compounds that can induce in vivo neuronal reprogramming in the adult spinal cord. METHODS Immunocytochemistry, immunohistochemistry, qRT-PCR and fate-mapping are performed to analyze the role of small molecules in reprogramming astrocytes into neuronal cells in vitro and in vivo. RESULTS By screening, we identify a chemical cocktail with only two chemical compounds that can directly and rapidly reprogram cultured astrocytes into neuronal cells. Importantly, this chemical cocktail can also successfully trigger neuronal reprogramming in the injured adult spinal cord without introducing exogenous genetic factors. These chemically induced cells showed typical neuronal morphologies and neuron-specific marker expression and could become mature and survive for more than 12 months. Lineage tracing indicated that the chemical compound-converted neuronal cells mainly originated from post-injury spinal reactive astrocytes. CONCLUSION Our proof-of-principle study demonstrates that in vivo glia-to-neuron conversion can be manipulated in a chemical compound-based manner. Albeit our current chemical cocktail has a lowreprogramming efficiency, it will bring in vivo cell fate reprogramming closer to clinical application in brain and spinal cord repair. Future studies should focus on further refining our chemical cocktail and reprogramming approach to boost the reprogramming efficiency.
Collapse
Affiliation(s)
- Zijian Tan
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai 200433, China
| | - Shangyao Qin
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai 200433, China
| | - Hong Liu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai 200433, China
| | - Xiao Huang
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai 200433, China
| | - Yingyan Pu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai 200433, China
| | - Cheng He
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai 200433, China
| | - Yimin Yuan
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai 200433, China.
| | - Zhida Su
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
32
|
Umeyama T, Matsuda T, Nakashima K. Lineage Reprogramming: Genetic, Chemical, and Physical Cues for Cell Fate Conversion with a Focus on Neuronal Direct Reprogramming and Pluripotency Reprogramming. Cells 2024; 13:707. [PMID: 38667322 PMCID: PMC11049106 DOI: 10.3390/cells13080707] [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: 04/02/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Although lineage reprogramming from one cell type to another is becoming a breakthrough technology for cell-based therapy, several limitations remain to be overcome, including the low conversion efficiency and subtype specificity. To address these, many studies have been conducted using genetics, chemistry, physics, and cell biology to control transcriptional networks, signaling cascades, and epigenetic modifications during reprogramming. Here, we summarize recent advances in cellular reprogramming and discuss future directions.
Collapse
Affiliation(s)
- Taichi Umeyama
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | - Taito Matsuda
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | | |
Collapse
|
33
|
Wu P, Xu C, Zou X, Yang K, Xu Y, Li X, Li X, Wang Z, Luo Z. Capacitive-Coupling-Responsive Hydrogel Scaffolds Offering Wireless In Situ Electrical Stimulation Promotes Nerve Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310483. [PMID: 38198600 DOI: 10.1002/adma.202310483] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/28/2023] [Indexed: 01/12/2024]
Abstract
Electrical stimulation (ES) has shown beneficial effects in repairing injured tissues. However, current ES techniques that use tissue-traversing leads and bulky external power suppliers have significant limitations in translational medicine. Hence, exploring noninvasive in vivo ES to provide controllable electrical cues in tissue engineering is an imminent necessity. Herein, a conductive hydrogel with in situ electrical generation capability as a biodegradable regeneration scaffold and wireless ES platform for spinal cord injury (SCI) repair is demonstrated. When a soft insulated metal plate is placed on top of the injury site as a wireless power transmitter, the conductive hydrogel implanted at the injury site can serve as a wireless power receiver, and the capacitive coupling between the receiver and transmitter can generate an alternating current in the hydrogel scaffold owing to electrostatic induction effect. In a complete transection model of SCI rats, the implanted conductive hydrogels with capacitive-coupling in situ ES enhance functional recovery and neural tissue repair by promoting remyelination, accelerating axon regeneration, and facilitating endogenous neural stem cell differentiation. This facile wireless-powered electroactive-hydrogel strategy thus offers on-demand in vivo ES with an adjustable timeline, duration, and strength and holds great promise in translational medicine.
Collapse
Affiliation(s)
- Ping Wu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Chao Xu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xianghui Zou
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Kun Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Yanping Xu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xueyao Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Xiaokun Li
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhouguang Wang
- National Key Laboratory of Macromolecular Drug Development and Manufacturing, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zhiqiang Luo
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| |
Collapse
|
34
|
Gerber YN, Perrin FE. In vivo astrocyte reprogramming following spinal cord injury. Neural Regen Res 2024; 19:487-488. [PMID: 37721266 PMCID: PMC10581580 DOI: 10.4103/1673-5374.380893] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/08/2023] [Accepted: 06/20/2023] [Indexed: 09/19/2023] Open
Affiliation(s)
| | - Florence E. Perrin
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France
- Institut Universitaire de France (IUF), Paris, France
| |
Collapse
|
35
|
Tai W, Du X, Chen C, Xu XM, Zhang CL, Wu W. NG2 glia reprogramming induces robust axonal regeneration after spinal cord injury. iScience 2024; 27:108895. [PMID: 38318363 PMCID: PMC10839253 DOI: 10.1016/j.isci.2024.108895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/04/2023] [Accepted: 01/09/2024] [Indexed: 02/07/2024] Open
Abstract
Spinal cord injury (SCI) often leads to neuronal loss, axonal degeneration, and behavioral dysfunction. We recently show that in vivo reprogramming of NG2 glia produces new neurons, reduces glial scaring, and ultimately leads to improved function after SCI. By examining endogenous neurons, we here unexpectedly uncover that NG2 glia reprogramming also induces robust axonal regeneration of the corticospinal tract and serotonergic neurons. Such reprogramming-induced axonal regeneration may contribute to the reconstruction of neural networks essential for behavioral recovery.
Collapse
Affiliation(s)
- Wenjiao Tai
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiaolong Du
- Department of Neurological Surgery, Spinal Cord and Brain Injury Research Group, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Chen Chen
- Department of Neurological Surgery, Spinal Cord and Brain Injury Research Group, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Xiao-Ming Xu
- Department of Neurological Surgery, Spinal Cord and Brain Injury Research Group, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Chun-Li Zhang
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wei Wu
- Department of Neurological Surgery, Spinal Cord and Brain Injury Research Group, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
36
|
Cho HE, Lee S, Seo JH, Kang SW, Choi WA, Cho SR. In Vivo Reprogramming Using Yamanaka Factors in the CNS: A Scoping Review. Cells 2024; 13:343. [PMID: 38391956 PMCID: PMC10886652 DOI: 10.3390/cells13040343] [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: 01/08/2024] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024] Open
Abstract
Central nervous system diseases, particularly neurodegenerative disorders, pose significant challenges in medicine. These conditions, characterized by progressive neuronal loss, have remained largely incurable, exacting a heavy toll on individuals and society. In recent years, in vivo reprogramming using Yamanaka factors has emerged as a promising approach for central nervous system regeneration. This technique involves introducing transcription factors, such as Oct4, Sox2, Klf4, and c-Myc, into adult cells to induce their conversion into neurons. This review summarizes the current state of in vivo reprogramming research in the central nervous system, focusing on the use of Yamanaka factors. In vivo reprogramming using Yamanaka factors has shown promising results in several animal models of central nervous system diseases. Studies have demonstrated that this approach can promote the generation of new neurons, improve functional outcomes, and reduce scar formation. However, there are still several challenges that need to be addressed before this approach can be translated into clinical practice. These challenges include optimizing the efficiency of reprogramming, understanding the cell of origin for each transcription factor, and developing methods for reprogramming in non-subventricular zone areas. Further research is needed to overcome the remaining challenges, but this approach has the potential to revolutionize the way we treat central nervous system disorders.
Collapse
Affiliation(s)
- Han Eol Cho
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (H.E.C.); (S.-W.K.)
- Department of Rehabilitation Medicine, Gangnam Severance Hospital, Seoul 06229, Republic of Korea
| | - Siwoo Lee
- Graduate Program of Biomedical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
- Department of Rehabilitation Medicine, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
| | - Jung Hwa Seo
- Department of Rehabilitation Medicine, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
- Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Seong-Woong Kang
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (H.E.C.); (S.-W.K.)
- Department of Rehabilitation Medicine, Gangnam Severance Hospital, Seoul 06229, Republic of Korea
| | - Won Ah Choi
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (H.E.C.); (S.-W.K.)
- Department of Rehabilitation Medicine, Gangnam Severance Hospital, Seoul 06229, Republic of Korea
| | - Sung-Rae Cho
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul 06229, Republic of Korea; (H.E.C.); (S.-W.K.)
- Graduate Program of Biomedical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
- Department of Rehabilitation Medicine, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea;
- Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
37
|
Sichani AS, Khoddam S, Shakeri S, Tavakkoli Z, Jafroodi AR, Dabbaghipour R, Sisakht M, Fallahi J. Partial Reprogramming as a Method for Regenerating Neural Tissues in Aged Organisms. Cell Reprogram 2024; 26:10-23. [PMID: 38381402 DOI: 10.1089/cell.2023.0123] [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] [Indexed: 02/22/2024] Open
Abstract
Aging causes numerous age-related diseases, leading the human species to death. Nevertheless, rejuvenating strategies based on cell epigenetic modifications are a possible approach to counteract disease progression while getting old. Cell reprogramming of adult somatic cells toward pluripotency ought to be a promising tool for age-related diseases. However, researchers do not have control over this process as cells lose their fate, and cause potential cancerous cells or unexpected cell phenotypes. Direct and partial reprogramming were introduced in recent years with distinctive applications. Although direct reprogramming makes cells lose their identity, it has various applications in regeneration medicine. Temporary and regulated in vivo overexpression of Yamanaka factors has been shown in several experimental contexts to be achievable and is used to rejuvenate mice models. This regeneration can be accomplished by altering the epigenetic adult cell signature to the signature of a younger cell. The greatest advantage of partial reprogramming is that this method does not allow cells to lose their identity when they are resetting their epigenetic clock. It is a regimen of short-term Oct3/4, Sox2, Klf4, and c-Myc expression in vivo that prevents full reprogramming to the pluripotent state and avoids both tumorigenesis and the presence of unwanted undifferentiated cells. We know that many neurological age-related diseases, such as Alzheimer's disease, stroke, dementia, and Parkinson's disease, are the main cause of death in the last decades of life. Therefore, scientists have a special tendency regarding neuroregeneration methods to increase human life expectancy.
Collapse
Affiliation(s)
- Ali Saber Sichani
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Somayeh Khoddam
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shayan Shakeri
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Tavakkoli
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Arad Ranji Jafroodi
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Dabbaghipour
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohsen Sisakht
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jafar Fallahi
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
38
|
Liu Y, Zhao Y, Liao X, Zhou S, Guo X, Yang L, Lv B. PD-1 deficiency aggravates spinal cord injury by regulating the reprogramming of NG2 glia and activating the NgR/RhoA/ROCK signaling pathway. Cell Signal 2024; 114:110978. [PMID: 37972801 DOI: 10.1016/j.cellsig.2023.110978] [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: 07/09/2023] [Revised: 10/24/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
Spinal cord injury (SCI) is a devastating disorder and a leading cause of disability in adults worldwide. Multiple studies have reported the upregulation of programmed cell death 1 (PD-1) following SCI. However, the underlying mechanism of PD-1 deficiency in SCI is not well established. Therefore, we aimed to investigate the role and potential mechanism of PD-1 in SCI pathogenesis. PD-1 Knockout (KO) SCI mouse model was established, and PD-1 expression was evaluated in tissue samples by western blot assay. We then used a series of function gain-and-loss assays to determine the role of PD-1 in SCI pathogenesis. Moreover, mechanistic assays were performed to explore the association between PD-1, neuron-glia antigen-2 (NG2) glia cells, and miR-23b-5p and then investigated the involved signaling pathway. Results illustrated that PD-1 deficiency enhanced the inflammatory response, neuron loss, and functional impairment induced by SCI. We found that NG2 glia depletion aggravated inflammation, reduced neural survival, and suppressed locomotor recovery in murine SCI model. Further analysis indicated that NG2+ cells were increased in the spinal cord of SCI mice, and PD-1 deficiency increased the number of NG2+ cells by activating the Nogo receptor/ras homolog family member A/Rho kinase (NgR/RhoA/ROCK) signaling. Mechanistically, miR-23b-5p was identified as the negative regulator of PD-1 in NG2 glia. MiR-23b-5p deficiency reduced the expression of inflammatory cytokines, enhanced neural survival, and promoted locomotor recovery in SCI mice, which was counteracted by PD-1 deficiency. In conclusion, PD-1 deficiency exacerbates SCI in vivo by regulating reprogramming of NG2 glia and activating the NgR/RhoA/ROCK signaling.
Collapse
Affiliation(s)
- Yang Liu
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Yin Zhao
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Xinyuan Liao
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Shengyuan Zhou
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Xiang Guo
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Lili Yang
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Bitao Lv
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China.
| |
Collapse
|
39
|
Janeckova L, Knotek T, Kriska J, Hermanova Z, Kirdajova D, Kubovciak J, Berkova L, Tureckova J, Camacho Garcia S, Galuskova K, Kolar M, Anderova M, Korinek V. Astrocyte-like subpopulation of NG2 glia in the adult mouse cortex exhibits characteristics of neural progenitor cells. Glia 2024; 72:245-273. [PMID: 37772368 DOI: 10.1002/glia.24471] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 09/05/2023] [Accepted: 09/05/2023] [Indexed: 09/30/2023]
Abstract
Glial cells expressing neuron-glial antigen 2 (NG2), also known as oligodendrocyte progenitor cells (OPCs), play a critical role in maintaining brain health. However, their ability to differentiate after ischemic injury is poorly understood. The aim of this study was to investigate the properties and functions of NG2 glia in the ischemic brain. Using transgenic mice, we selectively labeled NG2-expressing cells and their progeny in both healthy brain and after focal cerebral ischemia (FCI). Using single-cell RNA sequencing, we classified the labeled glial cells into five distinct subpopulations based on their gene expression patterns. Additionally, we examined the membrane properties of these cells using the patch-clamp technique. Of the identified subpopulations, three were identified as OPCs, whereas the fourth subpopulation had characteristics indicative of cells likely to develop into oligodendrocytes. The fifth subpopulation of NG2 glia showed astrocytic markers and had similarities to neural progenitor cells. Interestingly, this subpopulation was present in both healthy and post-ischemic tissue; however, its gene expression profile changed after ischemia, with increased numbers of genes related to neurogenesis. Immunohistochemical analysis confirmed the temporal expression of neurogenic genes and showed an increased presence of NG2 cells positive for Purkinje cell protein-4 at the periphery of the ischemic lesion 12 days after FCI, as well as NeuN-positive NG2 cells 28 and 60 days after injury. These results suggest the potential development of neuron-like cells arising from NG2 glia in the ischemic tissue. Our study provides insights into the plasticity of NG2 glia and their capacity for neurogenesis after stroke.
Collapse
Affiliation(s)
- Lucie Janeckova
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Tomas Knotek
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
- Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jan Kriska
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Zuzana Hermanova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
- Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Denisa Kirdajova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Kubovciak
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Linda Berkova
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jana Tureckova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Sara Camacho Garcia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Katerina Galuskova
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Michal Kolar
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czech Republic
| | - Vladimir Korinek
- Laboratory of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
40
|
Wu Y, Meng X, Cheng WY, Yan Z, Li K, Wang J, Jiang T, Zhou F, Wong KH, Zhong C, Dong Y, Gao S. Can pluripotent/multipotent stem cells reverse Parkinson's disease progression? Front Neurosci 2024; 18:1210447. [PMID: 38356648 PMCID: PMC10864507 DOI: 10.3389/fnins.2024.1210447] [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: 04/22/2023] [Accepted: 01/02/2024] [Indexed: 02/16/2024] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by continuous and selective degeneration or death of dopamine neurons in the midbrain, leading to dysfunction of the nigrostriatal neural circuits. Current clinical treatments for PD include drug treatment and surgery, which provide short-term relief of symptoms but are associated with many side effects and cannot reverse the progression of PD. Pluripotent/multipotent stem cells possess a self-renewal capacity and the potential to differentiate into dopaminergic neurons. Transplantation of pluripotent/multipotent stem cells or dopaminergic neurons derived from these cells is a promising strategy for the complete repair of damaged neural circuits in PD. This article reviews and summarizes the current preclinical/clinical treatments for PD, their efficacies, and the advantages/disadvantages of various stem cells, including pluripotent and multipotent stem cells, to provide a detailed overview of how these cells can be applied in the treatment of PD, as well as the challenges and bottlenecks that need to be overcome in future translational studies.
Collapse
Affiliation(s)
- Yongkang Wu
- Key Laboratory of Adolescent Health Evaluation and Sports Intervention, Ministry of Education, East China Normal University, Shanghai, China
| | - Xiangtian Meng
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wai-Yin Cheng
- Research Institute for Future Food, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Zhichao Yan
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Keqin Li
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jian Wang
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tianfang Jiang
- Department of Neurology, Shanghai Eighth People’s Hospital Affiliated to Jiangsu University, Shanghai, China
| | - Fei Zhou
- Department of Neurology, Third Affiliated Hospital of Navy Military Medical University, Shanghai, China
| | - Ka-Hing Wong
- Research Institute for Future Food, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Chunlong Zhong
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yi Dong
- Key Laboratory of Adolescent Health Evaluation and Sports Intervention, Ministry of Education, East China Normal University, Shanghai, China
| | - Shane Gao
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
41
|
Huang L, Lai X, Liang X, Chen J, Yang Y, Xu W, Qin Q, Qin R, Huang X, Xie M, Chen L. A promise for neuronal repair: reprogramming astrocytes into neurons in vivo. Biosci Rep 2024; 44:BSR20231717. [PMID: 38175538 PMCID: PMC10830445 DOI: 10.1042/bsr20231717] [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: 10/02/2023] [Revised: 12/13/2023] [Accepted: 01/02/2024] [Indexed: 01/05/2024] Open
Abstract
Massive loss of neurons following brain injury or disease is the primary cause of central nervous system dysfunction. Recently, much research has been conducted on how to compensate for neuronal loss in damaged parts of the nervous system and thus restore functional connectivity among neurons. Direct somatic cell differentiation into neurons using pro-neural transcription factors, small molecules, or microRNAs, individually or in association, is the most promising form of neural cell replacement therapy available. This method provides a potential remedy for cell loss in a variety of neurodegenerative illnesses, and the development of reprogramming technology has made this method feasible. This article provides a comprehensive review of reprogramming, including the selection and methods of reprogramming starting cell populations as well as the signaling methods involved in this process. Additionally, we thoroughly examine how reprogramming astrocytes into neurons can be applied to treat stroke and other neurodegenerative diseases. Finally, we discuss the challenges of neuronal reprogramming and offer insights about the field.
Collapse
Affiliation(s)
- Lijuan Huang
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xinyu Lai
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xiaojun Liang
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Jiafeng Chen
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yue Yang
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Wei Xu
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Qingchun Qin
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Rongxing Qin
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Xiaoying Huang
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Minshan Xie
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Li Chen
- Department of Neurology, the First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
| |
Collapse
|
42
|
Qin B, Hu XM, Huang YX, Yang RH, Xiong K. A New Paradigm in Spinal Cord Injury Therapy: from Cell-free Treatment to Engineering Modifications. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:656-673. [PMID: 37076458 DOI: 10.2174/1871527322666230418090857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 01/20/2023] [Accepted: 02/06/2023] [Indexed: 04/21/2023]
Abstract
Spinal cord injury (SCI) is an intractable and poorly prognostic neurological disease, and current treatments are still unable to cure it completely and avoid sequelae. Extracellular vesicles (EVs), as important carriers of intercellular communication and pharmacological effects, are considered to be the most promising candidates for SCI therapy because of their low toxicity and immunogenicity, their ability to encapsulate endogenous bioactive molecules (e.g., proteins, lipids, and nucleic acids), and their ability to cross the blood-brain/cerebrospinal barriers. However, poor targeting, low retention rate, and limited therapeutic efficacy of natural EVs have bottlenecked EVs-based SCI therapy. A new paradigm for SCI treatment will be provided by engineering modified EVs. Furthermore, our limited understanding of the role of EVs in SCI pathology hinders the rational design of novel EVbased therapeutic approaches. In this study, we review the pathophysiology after SCI, especially the multicellular EVs-mediated crosstalk; briefly describe the shift from cellular to cell-free therapies for SCI treatment; discuss and analyze the issues related to the route and dose of EVs administration; summarize and present the common strategies for EVs drug loading in the treatment of SCI and point out the shortcomings of these drug loading methods; finally, we analyze and highlight the feasibility and advantages of bio-scaffold-encapsulated EVs for SCI treatment, providing scalable insights into cell-free therapy for SCI.
Collapse
Affiliation(s)
- Bo Qin
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi, 435003, China
| | - Xi-Min Hu
- Clinical Medicine Eight-year Program, 02 Class, 17 Grade, Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Yan-Xia Huang
- Health Management Center, the Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Rong-Hua Yang
- Department of Burn and Plastic Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
- Hunan Key Laboratory of Ophthalmology, Changsha, 410008, China
- Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou, 571199, China
| |
Collapse
|
43
|
Kao Y, Zhu H, Yang Y, Shen W, Song W, Zhang R, Liu Y, Liu H, Kong X. CREB1 Facilitates GABAergic Neural Differentiation of Human Mesenchymal Stem Cells through BRN2 for Pain Alleviation and Locomotion Recovery after Spinal Cord Injury. Cells 2023; 13:67. [PMID: 38201271 PMCID: PMC10778540 DOI: 10.3390/cells13010067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/28/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
The transplantation of GABAergic neuron cells has been reported to alleviate nerve pain and improve motor function after spinal cord injury (SCI). However, human mesenchymal stem cell (hMSC) differentiation into GABAergic neuron cells in a sufficient quantity remains to be accomplished. From a database screening, cAMP-responsive element-binding protein 1 (CREB1) was chosen as a potential modulator due to its critical role in the protein-protein interaction of genes related to GABAergic neural differentiation. Here, CREB1 was overexpressed in transfected hMSCs, where CREB1 could induce differentiation into GABAergic neuron cells with an upregulation of Map2 and GAD1 by 2- and 3.4-fold, respectively. Additionally, GABAergic neural differentiation was enhanced, while Notch signaling was inhibited, and BRN2 transcriptional activation played an important role in neuronal maturation. Moreover, transfected hMSCs injected into immunocompromised mice caused by CsA exhibited the neuronal markers Tuj1 and Map2 via the intraspinal route, suggesting an improvement in survival and neural differentiation. Significantly, improvement in both BMS scores (6.2 ± 1.30 vs. 4 ± 0) and thermal hyperalgesia latency (7.74 ± 2.36 s vs. 4.52 ± 0.39 s) was seen compared with the SCI naïve treatment at 4 weeks post-transplantation. Our study demonstrates that CREB1 is crucial in generating induced GABAergic neuron cells (iGNs) originating from hMSCs. Transplanting iGNs to injured spinal cord provides a promising strategy for alleviating neuropathic pain and locomotion recovery after SCI.
Collapse
Affiliation(s)
- Yanbing Kao
- Orthopedic Research Center of Qilu Hospital, Shandong University, Jinan 250100, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
| | - Hanming Zhu
- Orthopedic Research Center of Qilu Hospital, Shandong University, Jinan 250100, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
| | - Yu Yang
- Orthopedic Research Center of Qilu Hospital, Shandong University, Jinan 250100, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
| | - Wenyuan Shen
- Orthopedic Research Center of Qilu Hospital, Shandong University, Jinan 250100, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
| | - Wei Song
- Orthopedic Research Center of Qilu Hospital, Shandong University, Jinan 250100, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
| | - Renjie Zhang
- Orthopedic Research Center of Qilu Hospital, Shandong University, Jinan 250100, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
| | - Yanchun Liu
- Orthopedic Research Center of Qilu Hospital, Shandong University, Jinan 250100, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
| | - Haoyun Liu
- Orthopedic Research Center of Qilu Hospital, Shandong University, Jinan 250100, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
| | - Xiaohong Kong
- Orthopedic Research Center of Qilu Hospital, Shandong University, Jinan 250100, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
| |
Collapse
|
44
|
Serrano C, Cananzi S, Shen T, Wang LL, Zhang CL. Simple and Highly Specific Targeting of Resident Microglia with Adeno-Associated Virus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.12.571321. [PMID: 38168285 PMCID: PMC10760038 DOI: 10.1101/2023.12.12.571321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Microglia, as the immune cells of the central nervous system (CNS), play dynamic roles in both health and diseased conditions. The ability to genetically target microglia using viruses is crucial for understanding their functions and advancing microglia-based treatments. We here show that resident microglia can be simply and specifically targeted using adeno-associated virus (AAV) vectors containing a 466-bp DNA fragment from the human IBA1 (hIBA1) promoter. This targeting approach is applicable to both resting and reactive microglia. When combining the short hIBA1 promoter with the target sequence of miR124, up to 95% of transduced cells are identified as microglia. Such a simple and highly specific microglia-targeting strategy may be further optimized for research and therapeutics.
Collapse
Affiliation(s)
- Carolina Serrano
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sergio Cananzi
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tianjin Shen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lei-Lei Wang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chun-Li Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
45
|
Moon S, Ito Y. Vasculature cells control neuroglial co-localization and synaptic connection in a central nervous system tissue mimic system. Hum Cell 2023; 36:1938-1947. [PMID: 37470936 DOI: 10.1007/s13577-023-00955-x] [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: 05/31/2023] [Accepted: 07/13/2023] [Indexed: 07/21/2023]
Abstract
Despite the development of neural tissue differentiation methods using a wide variety of stem cells and compartments, there is no standardized strategy for establishing synapses. As the neuronal network is developed in parallel with blood vessel angiogenesis in the central nervous system (CNS) from the embryonic period, we examined neuron-astrocyte-vasculature interactions to understand the effect of the vasculature on the development and stabilization of neurological morphogenesis. We generated a cellular co-culture module targeting the CNS that was embedded in a collagen-based extracellular matrix (ECM) gel. Our neuron-astrocyte-vascular complex module identified the neurological co-localization effect by endothelial cells, as well as the pericyte-induced improvement of synaptic connections. Furthermore, it was suggested that the PDGF, BDNF, IGF, and WNT/BMP pathways were upregulated in synaptic connections enhanced conditions, which are composed of neurexin. These results suggest that the integrity of the vasculature cells in the CNS is important for the establishment of neuronal networks and for synapse connection.
Collapse
Affiliation(s)
- SongHo Moon
- Faculty of Life and Environmental Sciences (Bioindustrial Sciences), University of Tsukuba, 1-1-1 Tenno-Dai, Tsukuba, Ibaraki, 305-8972, Japan
| | - Yuzuru Ito
- Faculty of Life and Environmental Sciences (Bioindustrial Sciences), University of Tsukuba, 1-1-1 Tenno-Dai, Tsukuba, Ibaraki, 305-8972, Japan.
- Life Science Development Department, Frontier Business Division, Chiyoda Corporation, Yokohama, Kanagawa, Japan.
| |
Collapse
|
46
|
Chudakova DA, Samoilova EM, Chekhonin VP, Baklaushev VP. Improving Efficiency of Direct Pro-Neural Reprogramming: Much-Needed Aid for Neuroregeneration in Spinal Cord Injury. Cells 2023; 12:2499. [PMID: 37887343 PMCID: PMC10605572 DOI: 10.3390/cells12202499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/13/2023] [Accepted: 10/18/2023] [Indexed: 10/28/2023] Open
Abstract
Spinal cord injury (SCI) is a medical condition affecting ~2.5-4 million people worldwide. The conventional therapy for SCI fails to restore the lost spinal cord functions; thus, novel therapies are needed. Recent breakthroughs in stem cell biology and cell reprogramming revolutionized the field. Of them, the use of neural progenitor cells (NPCs) directly reprogrammed from non-neuronal somatic cells without transitioning through a pluripotent state is a particularly attractive strategy. This allows to "scale up" NPCs in vitro and, via their transplantation to the lesion area, partially compensate for the limited regenerative plasticity of the adult spinal cord in humans. As recently demonstrated in non-human primates, implanted NPCs contribute to the functional improvement of the spinal cord after injury, and works in other animal models of SCI also confirm their therapeutic value. However, direct reprogramming still remains a challenge in many aspects; one of them is low efficiency, which prevents it from finding its place in clinics yet. In this review, we describe new insights that recent works brought to the field, such as novel targets (mitochondria, nucleoli, G-quadruplexes, and others), tools, and approaches (mechanotransduction and electrical stimulation) for direct pro-neural reprogramming, including potential ones yet to be tested.
Collapse
Affiliation(s)
- Daria A. Chudakova
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
| | - Ekaterina M. Samoilova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center of Specialised Medical Care and Medical Technologies FMBA of Russia, 115682 Moscow, Russia
| | - Vladimir P. Chekhonin
- Department of Medical Nanobiotechnology of Medical and Biological Faculty, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, 117997 Moscow, Russia
| | - Vladimir P. Baklaushev
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center of Specialised Medical Care and Medical Technologies FMBA of Russia, 115682 Moscow, Russia
- Department of Medical Nanobiotechnology of Medical and Biological Faculty, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, 117997 Moscow, Russia
| |
Collapse
|
47
|
Tan R, Haseloff RF, Mo Y, Zhang J. In situ neuronal regeneration: the potential of astrocytes. LIFE MEDICINE 2023; 2:lnad036. [PMID: 39872896 PMCID: PMC11749267 DOI: 10.1093/lifemedi/lnad036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 01/06/2024] [Indexed: 01/30/2025]
Affiliation(s)
- Rongbang Tan
- Affiliated Hospital of Guangdong Medical University & Zhanjiang Key Laboratory of Zebrafish Model for Development and Disease, Guangdong Medical University, Zhanjiang 524001, China
| | - Reiner F Haseloff
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin 13125, Germany
| | - Yuqian Mo
- School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Jingjing Zhang
- Affiliated Hospital of Guangdong Medical University & Zhanjiang Key Laboratory of Zebrafish Model for Development and Disease, Guangdong Medical University, Zhanjiang 524001, China
| |
Collapse
|
48
|
Tai W, Zhang CL. In vivo cell fate reprogramming for spinal cord repair. Curr Opin Genet Dev 2023; 82:102090. [PMID: 37506560 PMCID: PMC11025462 DOI: 10.1016/j.gde.2023.102090] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 06/07/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023]
Abstract
Spinal cord injury (SCI) can lead to the loss of motor, sensory, or autonomic function due to neuronal death. Unfortunately, the adult mammalian spinal cord has limited intrinsic regenerative capacity, making it difficult to rebuild the neural circuits necessary for functional recovery. However, recent evidence suggests that in vivo fate reprogramming of resident cells that are normally non-neurogenic can generate new neurons. This process also improves the pathological microenvironment, and the new neurons can integrate into the local neural network, resulting in better functional outcomes in SCI animal models. In this concise review, we focus on recent advances while also discussing the challenges, pitfalls, and opportunities in the field of in vivo cell fate reprogramming for spinal cord repair.
Collapse
Affiliation(s)
- Wenjiao Tai
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chun-Li Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
49
|
Fang YM, Chen WC, Zheng WJ, Yang YS, Zhang Y, Chen XL, Pei MQ, Lin S, He HF. A cutting-edge strategy for spinal cord injury treatment: resident cellular transdifferentiation. Front Cell Neurosci 2023; 17:1237641. [PMID: 37711511 PMCID: PMC10498389 DOI: 10.3389/fncel.2023.1237641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Spinal cord injury causes varying degrees of motor and sensory function loss. However, there are no effective treatments for spinal cord repair following an injury. Moreover, significant preclinical advances in bioengineering and regenerative medicine have not yet been translated into effective clinical therapies. The spinal cord's poor regenerative capacity makes repairing damaged and lost neurons a critical treatment step. Reprogramming-based neuronal transdifferentiation has recently shown great potential in repair and plasticity, as it can convert mature somatic cells into functional neurons for spinal cord injury repair in vitro and in vivo, effectively halting the progression of spinal cord injury and promoting functional improvement. However, the mechanisms of the neuronal transdifferentiation and the induced neuronal subtypes are not yet well understood. This review analyzes the mechanisms of resident cellular transdifferentiation based on a review of the relevant recent literature, describes different molecular approaches to obtain different neuronal subtypes, discusses the current challenges and improvement methods, and provides new ideas for exploring therapeutic approaches for spinal cord injury.
Collapse
Affiliation(s)
- Yu-Ming Fang
- Department of Anaesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wei-Can Chen
- Department of Anaesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wan-Jing Zheng
- Department of Anaesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yu-Shen Yang
- Department of Anaesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yan Zhang
- Department of Anaesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xin-Li Chen
- Department of Anaesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Meng-Qin Pei
- Department of Anaesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Neuroendocrinology Group, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - He-Fan He
- Department of Anaesthesiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
50
|
Li Z, Qi Y, Sun L, Li Z, Chen S, Zhang Y, Ma Y, Han J, Wang Z, Zhang Y, Geng H, Huang B, Wang J, Li G, Li X, Wu S, Ni S. Three-dimensional nanofibrous sponges with aligned architecture and controlled hierarchy regulate neural stem cell fate for spinal cord regeneration. Theranostics 2023; 13:4762-4780. [PMID: 37771775 PMCID: PMC10526661 DOI: 10.7150/thno.87288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/12/2023] [Indexed: 09/30/2023] Open
Abstract
Background: Spinal cord injury (SCI) induces neuronal death and disrupts the nerve fiber bundles, which leads to severe neurological dysfunction and even permanent paralysis. A strategy combining biomimetic nanomaterial scaffolds with neural stem cell (NSC) transplantation holds promise for SCI treatment. Methods: Innovative three-dimensional (3D) nanofibrous sponges (NSs) were designed and developed by a combination of directional electrospinning and subsequent gas-foaming treatment. Immunofluorescence, mRNA sequencing, magnetic resonance imaging, electrophysiological analysis, and behavioral tests were used to investigate the in vitro and in vivo regenerative effects of the 3D NSs. Results: The generated 3D NSs exhibited uniaxially aligned nano-architecture and highly controllable hierarchical structure with super-high porosity (99%), outstanding hydrophilicity, and reasonable mechanical performance. They facilitated cell infiltration, induced cell alignment, promoted neuronal differentiation of NSCs, and enhanced their maturation mediated through cellular adhesion molecule pathways. In vivo, the NSC-seeded 3D NSs efficiently promoted axon reinnervation and remyelination in a rat SCI model, with new "neural relays" developing across the lesion gap. These histological changes were associated with regain of function, including increasing the neurological motor scores of SCI rats, from approximately 2 to 16 (out of 21), and decreasing the sensing time in the tape test from 140 s to 36 s. Additionally, the scaffolds led to restoration of ascending and descending electrophysiological signalling. Conclusion: The as-fabricated 3D NSs effectively regulate NSC fates, and an advanced combination of 3D NS design and transplanted NSCs enables their use as an ideal tissue-engineered scaffold for SCI repair.
Collapse
Affiliation(s)
- Zhiwei Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Ye Qi
- College of Textiles & Clothing, Qingdao University, Qingdao, China
| | - Lei Sun
- Department of Endocrinology, Qilu Hospital of Shandong University and Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, Shandong, 250012, China
| | - Zheng Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Shaojuan Chen
- College of Textiles & Clothing, Qingdao University, Qingdao, China
| | - Yuqi Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yuan Ma
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jinming Han
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Zide Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yulin Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Huimin Geng
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Shaohua Wu
- College of Textiles & Clothing, Qingdao University, Qingdao, China
| | - Shilei Ni
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| |
Collapse
|