1
|
Wang T, Wang X, Liu S, Li M, Wan K, Zheng J, Liao K, Wang J, Zou K, Wang L, Xu H, Lei W, Chen G, Li W. Transcription Factor-Based Gene Therapy Enables Functional Repair of Rat Following Chronic Ischemic Stroke. CNS Neurosci Ther 2025; 31:e70448. [PMID: 40401537 PMCID: PMC12096174 DOI: 10.1111/cns.70448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/21/2025] [Accepted: 05/07/2025] [Indexed: 05/23/2025] Open
Abstract
OBJECTIVE In vivo transcription factor (TF) -mediated gene therapy through astrocyte-to-neuron (AtN) conversion has shown therapeutic effects on rodent and non-human primate cortical ischemic injury in the subacute phase. However, in the clinic, subcortical regions including striatum as well as white matter are vulnerable regions of stroke, with millions of patients beyond subacute phase. In this study, we investigate whether TF-mediated AtN conversion therapy can be extended to treat chronic-phase ischemic stroke involving subcortical regions (e.g., striatum) and white matter, beyond cortical injuries. METHODS Rat middle cerebral artery occlusion (MCAO)-like models were established to induce broad ischemic injuries including cortical and striatal regions. Then multiple rounds of TF-mediated gene therapy treatments through adeno-associated virus (AAV) system to cover the large-scaled infarct areas were conducted in the chronic phase of the stroke models. Magnetic resonance imaging (MRI), [18F] FDG-PET/CT, behavioral tests, immunohistochemistry and bulk-RNA seq were applied to evaluate the AtN conversion, tissue repair and functional recovery. RESULTS Our results revealed that administrated in the chronic phase of ischemic stroke, TF-mediated gene therapy can efficiently regenerate new neurons in both cortical and striatal regions, and promote tissue repair in both grey and white matter. Compared with single round of AAV administration, multiple rounds of treatment regenerated more neurons and led to a significant functional recovery. CONCLUSIONS Our study demonstrates that TF-mediated gene therapy has a broad therapeutic time window and can be applied multiple rounds to treat severe ischemic stroke, making it an attractive therapeutic intervention in the chronic phase after stroke, when current approaches are largely ineffective.
Collapse
Affiliation(s)
- Tao Wang
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Xu Wang
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Shanggong Liu
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Menglei Li
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Kaiying Wan
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Jiajun Zheng
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Kai Liao
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non‐Human Primate Research, GHM Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Jinyu Wang
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Kaiming Zou
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
| | - Lu Wang
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non‐Human Primate Research, GHM Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Hao Xu
- Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non‐Human Primate Research, GHM Institute of CNS RegenerationJinan UniversityGuangzhouChina
| | - Wenliang Lei
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsJinan UniversityGuangzhouChina
- Department of Nuclear Medicine and PET/CT‐MRI CenterThe First Affiliated Hospital of Jinan University & Institute of Molecular and Functional Imaging, Jinan UniversityGuangzhouChina
| | - Gong Chen
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsJinan UniversityGuangzhouChina
- Department of Nuclear Medicine and PET/CT‐MRI CenterThe First Affiliated Hospital of Jinan University & Institute of Molecular and Functional Imaging, Jinan UniversityGuangzhouChina
| | - Wen Li
- Guangdong‐Hong Kong‐Macau Institute of CNS Regeneration (GHMICR)Jinan UniversityGuangzhouChina
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative DrugsJinan UniversityGuangzhouChina
- Department of Nuclear Medicine and PET/CT‐MRI CenterThe First Affiliated Hospital of Jinan University & Institute of Molecular and Functional Imaging, Jinan UniversityGuangzhouChina
| |
Collapse
|
2
|
Papetti AV, Jin M, Ma Z, Stillitano AC, Jiang P. Chimeric brain models: Unlocking insights into human neural development, aging, diseases, and cell therapies. Neuron 2025:S0896-6273(25)00256-9. [PMID: 40300597 DOI: 10.1016/j.neuron.2025.03.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/07/2025] [Accepted: 03/31/2025] [Indexed: 05/01/2025]
Abstract
Human-rodent chimeric brain models serve as a unique platform for investigating the pathophysiology of human cells within a living brain environment. These models are established by transplanting human tissue- or human pluripotent stem cell (hPSC)-derived macroglial, microglial, or neuronal lineage cells, as well as cerebral organoids, into the brains of host animals. This approach has opened new avenues for exploring human brain development, disease mechanisms, and regenerative processes. Here, we highlight recent advancements in using chimeric models to study human neural development, aging, and disease. Additionally, we explore the potential applications of these models for studying human glial cell-replacement therapies, studying in vivo human glial-to-neuron reprogramming, and harnessing single-cell omics and advanced functional assays to uncover detailed insights into human neurobiology. Finally, we discuss strategies to enhance the precision and translational relevance of these models, expanding their impact in stem cell and neuroscience research.
Collapse
Affiliation(s)
- Ava V Papetti
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Mengmeng Jin
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Ziyuan Ma
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Alessandro C Stillitano
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA
| | - Peng Jiang
- Department of Cell Biology and Neuroscience, Rutgers University-New Brunswick, Piscataway, NJ 08854, USA.
| |
Collapse
|
3
|
Zhang T, Zhou X, Li M, Zuo C, Gao W. Comprehensive analysis of bioinformatics identification TST, SQOR and NRDC is mitochondria-related biomarkers of ischemic cerebral apoplexy. Sci Rep 2025; 15:8892. [PMID: 40087350 PMCID: PMC11909143 DOI: 10.1038/s41598-025-85957-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 01/07/2025] [Indexed: 03/17/2025] Open
Abstract
Mitochondria are an important organelle affecting the occurrence and development of ischemic stroke (IS). Although the role of mitochondria in IS has been paid attention to, the relevant biomarkers have not been identified, and the targeted treatment is still lacking. To further solve these problems, in this study, we combined and standardized GSE16561 and GSE58294 datasets in the GEO database as the test set, and GSE22255 as the training set. The mitochondria-related gene set was obtained from MitoCarta3.0 for study. R language was used to screen differentially expressed genes (DEGs), and weighted gene co-expression network analysis (WGCNA) was used to obtain the intersection genes. KEGG pathway enrichment and gene ontology (GO) analysis were performed. LASSO and Logistic methods were used to determine the diagnostic markers of mitochondrial-related IS. The correlation between it and the main immune cells and immune-related factors was analysed. The ROC curve was used to determine the diagnostic efficacy. The protein co-expression network, transcription factor, and miRNA prediction, and drug prediction by cMAP were performed against these markers. Molecular docking tested the binding energy. Transmission electron microscopy was used to observe the structure and morphology of mitochondria in cerebral microvascular endothelial cells of MCAO/R rats. Biochemical kits were used to detect the levels of MDA and SOD in blood and tissues. Elisa was used to detect the changes of the above targets in blood and tissues. In this study, a total of 38 intersection genes were obtained, and a diagnostic model composed of 3 genes (TST, SQOR, NRDC) was further established. KEGG and GO analysis showed that these genes were highly related to immunity and were involved in the immune activities related to neutrophils, CD4+T cells, CD8+T cells, and macrophages. In addition, a total of 42 proteins, 601 transcription factors, and 99 miRNAs related to TST, SQOR, and NRDC were predicted, and the interaction map was constructed. cMAP predicted 5 potential small molecule drugs and molecular docking suggested that W.13 had the best binding energy. In MCAO/R rats, the mitochondria in BMECs were severely damaged and fragmented, accompanied by a decrease in SOD activity and an increase in MDA level. In addition, the levels of TST and SQOR in blood and tissues were increased, and the levels of NRDC were decreased. Our study provides new insights into further understanding of IS from a mitochondrial perspective.
Collapse
Affiliation(s)
- Tianci Zhang
- Hebei University of Chinese medicine, Hebei Key Laboratory of Chinese Medicine Research On Cardio-cerebrovasc, 050091, Shijiazhuang, Hebei, China
| | - Xiaohong Zhou
- Hebei University of Chinese medicine, Hebei Key Laboratory of Chinese Medicine Research On Cardio-cerebrovasc, 050091, Shijiazhuang, Hebei, China
| | - Meng Li
- Hebei University of Chinese medicine, Hebei Key Laboratory of Chinese Medicine Research On Cardio-cerebrovasc, 050091, Shijiazhuang, Hebei, China
| | - Chunyue Zuo
- Hebei University of Chinese medicine, Hebei Key Laboratory of Chinese Medicine Research On Cardio-cerebrovasc, 050091, Shijiazhuang, Hebei, China
| | - Weijuan Gao
- Hebei University of Chinese medicine, Hebei Key Laboratory of Chinese Medicine Research On Cardio-cerebrovasc, 050091, Shijiazhuang, Hebei, China.
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Liu S, Xu X, Omari-Siaw E, Yu J, Deng W. Progress of reprogramming astrocytes into neuron. Mol Cell Neurosci 2024; 130:103947. [PMID: 38862082 DOI: 10.1016/j.mcn.2024.103947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 06/13/2024] Open
Abstract
As the main players in the central nervous system (CNS), neurons dominate most life activities. However, after accidental trauma or neurodegenerative diseases, neurons are unable to regenerate themselves. The loss of this important role can seriously affect the quality of life of patients, ranging from movement disorders to disability and even death. There is no suitable treatment to prevent or reverse this process. Therefore, the regeneration of neurons after loss has been a major clinical problem and the key to treatment. Replacing the lost neurons by transdifferentiation of other cells is the only viable approach. Although much progress has been made in stem cell therapy, ethical issues, immune rejection, and limited cell sources still hinder its clinical application. In recent years, somatic cell reprogramming technology has brought a new dawn. Among them, astrocytes, as endogenously abundant cells homologous to neurons, have good potential and application value for reprogramming into neurons, having been reprogrammed into neurons in vitro and in vivo in a variety of ways.
Collapse
Affiliation(s)
- Sitong Liu
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China
| | - Ximing Xu
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China
| | - Emmanuel Omari-Siaw
- Department of Pharmaceutical Science, Kumasi Technical University, PO Box 854, Kumasi, Ashanti, Ghana
| | - Jiangnan Yu
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China.
| | - Wenwen Deng
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China.
| |
Collapse
|
7
|
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
|
8
|
Cheng X, Nareddula S, Gao HC, Chen Y, Xiao T, Nadew YY, Xu F, Edens PA, Quinn CJ, Kimbrough A, Huang F, Chubykin AA. Impaired Experience-Dependent Theta Oscillation Synchronization and Inter-Areal Synaptic Connectivity in the Visual Cortex of Fmr1 KO Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.23.601989. [PMID: 39211264 PMCID: PMC11360911 DOI: 10.1101/2024.07.23.601989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Fragile X syndrome (FX) is the most prevalent inheritable form of autism spectrum disorder (ASD), characterized by hypersensitivity, difficulty in habituating to new sensory stimuli, and intellectual disability. Individuals with FX often experience visual perception and learning deficits. Visual experience leads to the emergence of the familiarity-evoked theta band oscillations in the primary visual cortex (V1) and the lateromedial area (LM) of mice. These theta oscillations in V1 and LM are synchronized with each other, providing a mechanism of sensory multi-areal binding. However, how this multi-areal binding and the corresponding theta oscillations are altered in FX is not known. Using iDISCO whole brain clearing with light-sheet microscopy, we quantified immediate early gene Fos expression in V1 and LM, identifying deficits in experience-dependent neural activity in FX mice. We performed simultaneous in vivo recordings with silicon probes in V1 and LM of awake mice and channelrhodopsin-2-assisted circuit mapping (CRACM) in acute brain slices to examine the neural activity and strength of long-range synaptic connections between V1 and LM in both wildtype (WT) and Fmr1 knockout (KO) mice, the model of FX, before and after visual experience. Our findings reveal synchronized familiarity-evoked theta oscillations in V1 and LM, the increased strength of V1→LM functional and synaptic connections, which correlated with the corresponding changes of presynaptic short-term plasticity in WT mice. The LM oscillations were attenuated in FX mice and correlated with impaired functional and synaptic connectivity and short-term plasticity in the feedforward (FF) V1→LM and feedback (FB) LM→V1 pathways. Finally, using 4Pi single-molecule localization microscopy (SMLM) in thick brain tissue, we identified experience-dependent changes in the density and shape of dendritic spines in layer 5 pyramidal cells of WT mice, which correlated with the functional synaptic measurements. Interestingly, there was an increased dendritic spine density and length in naïve FX mice that failed to respond to experience. Our study provides the first comprehensive characterization of the role of visual experience in triggering inter-areal neural synchrony and shaping synaptic connectivity in WT and FX mice.
Collapse
|
9
|
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
|
10
|
Tang Y, Gervais C, Moffitt R, Nareddula S, Zimmermann M, Nadew YY, Quinn CJ, Saldarriaga V, Edens P, Chubykin AA. Visual experience induces 4-8 Hz synchrony between V1 and higher-order visual areas. Cell Rep 2023; 42:113482. [PMID: 37999977 PMCID: PMC10790627 DOI: 10.1016/j.celrep.2023.113482] [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: 04/04/2023] [Revised: 09/20/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Visual perceptual experience induces persistent 4-8 Hz oscillations in the mouse primary visual cortex (V1), encoding visual familiarity. Recent studies suggest that higher-order visual areas (HVAs) are functionally specialized and segregated into information streams processing distinct visual features. However, whether visual memories are processed and stored within the distinct streams is not understood. We report here that V1 and lateromedial (LM), but not V1 and anterolateral, become more phase synchronized in 4-8 Hz after the entrainment of visual stimulus that maximally induces responses in LM. Directed information analysis reveals changes in the top-down functional connectivity between V1 and HVAs. Optogenetic inactivation of LM reduces post-stimulus oscillation peaks in V1 and impairs visual discrimination behavior. Our results demonstrate that 4-8 Hz familiarity-evoked oscillations are specific for the distinct visual features and are present in the corresponding HVAs, where they may be used for the inter-areal communication with V1 during memory-related behaviors.
Collapse
Affiliation(s)
- Yu Tang
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
| | - Catherine Gervais
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
| | - Rylann Moffitt
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
| | - Sanghamitra Nareddula
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
| | - Michael Zimmermann
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
| | - Yididiya Y Nadew
- Department of Computer Sciences, Iowa State University, Ames, IA 50011, USA
| | | | - Violeta Saldarriaga
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
| | - Paige Edens
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
| | - Alexander A Chubykin
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
11
|
Tan S, Wang D, Fu Y, Zheng H, Liu Y, Lu B. Targeted clearance of mitochondria by an autophagy-tethering compound (ATTEC) and its potential therapeutic effects. Sci Bull (Beijing) 2023; 68:3013-3026. [PMID: 37940449 DOI: 10.1016/j.scib.2023.10.021] [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: 06/27/2023] [Revised: 08/19/2023] [Accepted: 10/24/2023] [Indexed: 11/10/2023]
Abstract
Increased mitochondrial damage plays a critical role in many neurodegeneration-related diseases such as Parkinson's disease (PD) and Down syndrome (DS). Thus, enhancement of mitochondrial degradation by small molecule compounds may provide promising new strategies to tackle these diseases. Here, we explored the strategy to induce clearance of mitochondria by targeting them to the autophagy machinery by autophagy-tethering compounds (ATTECs). We provided the proof-of-concept evidence demonstrating that the bifunctional compound (mT1) binding to both the outer mitochondrial membrane protein TSPO and the autophagosome protein LC3B simultaneously may enhance the engulfment of damaged mitochondria by autophagosomes and subsequent autophagic degradation of them. In addition, preliminary experiments suggest that mT1 attenuated disease-relevant phenotypes in both a PD cellular model and a DS organoid model. Taken together, we demonstrate the possibility of degrading mitochondria by bifunctional ATTECs, which confirms the capability of degrading organelles by ATTECs and provides potential new strategies in the intervention of mitochondria-related disorders.
Collapse
Affiliation(s)
- Shuixia Tan
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Da Wang
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, State Key Laboratory of Reproductive Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Yuhua Fu
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Huiwen Zheng
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yan Liu
- Institute for Stem Cell and Neural Regeneration, School of Pharmacy, State Key Laboratory of Reproductive Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China.
| | - Boxun Lu
- Neurology Department at Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, School of Life Sciences, Fudan University, Shanghai 200438, China.
| |
Collapse
|
12
|
Huang P, Duan W, Ruan C, Wang L, Hosea R, Wu Z, Zeng J, Wu S, Kasim V. NeuroD1-GPX4 signaling leads to ferroptosis resistance in hepatocellular carcinoma. PLoS Genet 2023; 19:e1011098. [PMID: 38134213 PMCID: PMC10773945 DOI: 10.1371/journal.pgen.1011098] [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: 06/13/2023] [Revised: 01/08/2024] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Cell death resistance is a hallmark of tumor cells that drives tumorigenesis and drug resistance. Targeting cell death resistance-related genes to sensitize tumor cells and decrease their cell death threshold has attracted attention as a potential antitumor therapeutic strategy. However, the underlying mechanism is not fully understood. Recent studies have reported that NeuroD1, first discovered as a neurodifferentiation factor, is upregulated in various tumor cells and plays a crucial role in tumorigenesis. However, its involvement in tumor cell death resistance remains unknown. Here, we found that NeuroD1 was highly expressed in hepatocellular carcinoma (HCC) cells and was associated with tumor cell death resistance. We revealed that NeuroD1 enhanced HCC cell resistance to ferroptosis, a type of cell death caused by aberrant redox homeostasis that induces lipid peroxide accumulation, leading to increased HCC cell viability. NeuroD1 binds to the promoter of glutathione peroxidase 4 (GPX4), a key reductant that suppresses ferroptosis by reducing lipid peroxide, and activates its transcriptional activity, resulting in decreased lipid peroxide and ferroptosis. Subsequently, we showed that NeuroD1/GPX4-mediated ferroptosis resistance was crucial for HCC cell tumorigenic potential. These findings not only identify NeuroD1 as a regulator of tumor cell ferroptosis resistance but also reveal a novel molecular mechanism underlying the oncogenic function of NeuroD1. Furthermore, our findings suggest the potential of targeting NeuroD1 in antitumor therapy.
Collapse
Affiliation(s)
- Ping Huang
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, China
| | - Wei Duan
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, China
| | - Cao Ruan
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, China
| | - Lingxian Wang
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, China
| | - Rendy Hosea
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, China
| | - Zheng Wu
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, China
| | - Jianting Zeng
- Department of Hepatobiliary and Pancreatic Oncology, Chongqing University Cancer Hospital, Chongqing University, Chongqing, China
| | - Shourong Wu
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing, China
| | - Vivi Kasim
- The Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing, China
| |
Collapse
|
13
|
Cheng X, Tang Y, Vidyadhara D, Li BZ, Zimmerman M, Pak A, Nareddula S, Edens PA, Chandra SS, Chubykin AA. Impaired pre-synaptic plasticity and visual responses in auxilin-knockout mice. iScience 2023; 26:107842. [PMID: 37766983 PMCID: PMC10520332 DOI: 10.1016/j.isci.2023.107842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 08/06/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Auxilin (DNAJC6/PARK19), an endocytic co-chaperone, is essential for maintaining homeostasis in the readily releasable pool (RRP) by aiding clathrin-mediated uncoating of synaptic vesicles. Its loss-of-function mutations, observed in familial Parkinson's disease (PD), lead to basal ganglia motor deficits and cortical dysfunction. We discovered that auxilin-knockout (Aux-KO) mice exhibited impaired pre-synaptic plasticity in layer 4 to layer 2/3 pyramidal cell synapses in the primary visual cortex (V1), including reduced short-term facilitation and depression. Computational modeling revealed increased RRP refilling during short repetitive stimulation, which diminished during prolonged stimulation. Silicon probe recordings in V1 of Aux-KO mice demonstrated disrupted visual cortical circuit responses, including reduced orientation selectivity, compromised visual mismatch negativity, and shorter visual familiarity-evoked theta oscillations. Pupillometry analysis revealed an impaired optokinetic response. Auxilin-dependent pre-synaptic endocytosis dysfunction was associated with deficits in pre-synaptic plasticity, visual cortical functions, and eye movement prodromally or at the early stage of motor symptoms.
Collapse
Affiliation(s)
- Xi Cheng
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
| | - Yu Tang
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
| | - D.J. Vidyadhara
- Department of Neurology, Yale University, CT, USA
- Department of Neuroscience, Yale University, CT, USA
| | - Ben-Zheng Li
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Electrical Engineering, University of Colorado, Denver, Denver, CO, USA
| | - Michael Zimmerman
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
- Department of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Alexandr Pak
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
| | - Sanghamitra Nareddula
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
| | - Paige Alyssa Edens
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
| | - Sreeganga S. Chandra
- Department of Neurology, Yale University, CT, USA
- Department of Neuroscience, Yale University, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University, CT, USA
| | - Alexander A. Chubykin
- Department of Biological Sciences, Purdue Institute for Integrative Neuroscience, Purdue Autism Research Center, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
14
|
Talifu Z, Liu JY, Pan YZ, Ke H, Zhang CJ, Xu X, Gao F, Yu Y, Du LJ, Li JJ. In vivo astrocyte-to-neuron reprogramming for central nervous system regeneration: a narrative review. Neural Regen Res 2023; 18:750-755. [PMID: 36204831 PMCID: PMC9700087 DOI: 10.4103/1673-5374.353482] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The inability of damaged neurons to regenerate within the mature central nervous system (CNS) is a significant neuroscientific challenge. Astrocytes are an essential component of the CNS and participate in many physiological processes including blood-brain barrier formation, axon growth regulation, neuronal support, and higher cognitive functions such as memory. Recent reprogramming studies have confirmed that astrocytes in the mature CNS can be transformed into functional neurons. Building on in vitro work, many studies have demonstrated that astrocytes can be transformed into neurons in different disease models to replace damaged or lost cells. However, many findings in this field are controversial, as the source of new neurons has been questioned. This review summarizes progress in reprogramming astrocytes into neurons in vivo in animal models of spinal cord injury, brain injury, Huntington's disease, Parkinson's disease, Alzheimer's disease, and other neurodegenerative conditions.
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; School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, Shandong Province, China
| | - Jia-Yi Liu
- 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, China
| | - Yun-Zhu 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; School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, Shandong Province, China
| | - Han Ke
- 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, China
| | - Chun-Jia Zhang
- 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, 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, 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, China
| | - Yan Yu
- School of Rehabilitation, Capital Medical University; 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, China
| | - Liang-Jie 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, China
| | - Jian-Jun 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; School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, Shandong Province, China
| |
Collapse
|
15
|
Sun C, Chen S. Disease-causing mutations in genes encoding transcription factors critical for photoreceptor development. Front Mol Neurosci 2023; 16:1134839. [PMID: 37181651 PMCID: PMC10172487 DOI: 10.3389/fnmol.2023.1134839] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 04/04/2023] [Indexed: 05/16/2023] Open
Abstract
Photoreceptor development of the vertebrate visual system is controlled by a complex transcription regulatory network. OTX2 is expressed in the mitotic retinal progenitor cells (RPCs) and controls photoreceptor genesis. CRX that is activated by OTX2 is expressed in photoreceptor precursors after cell cycle exit. NEUROD1 is also present in photoreceptor precursors that are ready to specify into rod and cone photoreceptor subtypes. NRL is required for the rod fate and regulates downstream rod-specific genes including the orphan nuclear receptor NR2E3 which further activates rod-specific genes and simultaneously represses cone-specific genes. Cone subtype specification is also regulated by the interplay of several transcription factors such as THRB and RXRG. Mutations in these key transcription factors are responsible for ocular defects at birth such as microphthalmia and inherited photoreceptor diseases such as Leber congenital amaurosis (LCA), retinitis pigmentosa (RP) and allied dystrophies. In particular, many mutations are inherited in an autosomal dominant fashion, including the majority of missense mutations in CRX and NRL. In this review, we describe the spectrum of photoreceptor defects that are associated with mutations in the above-mentioned transcription factors, and summarize the current knowledge of molecular mechanisms underlying the pathogenic mutations. At last, we deliberate the outstanding gaps in our understanding of the genotype-phenotype correlations and outline avenues for future research of the treatment strategies.
Collapse
Affiliation(s)
- Chi Sun
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis, St. Louis, MO, United States
- *Correspondence: Chi Sun,
| | - Shiming Chen
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis, St. Louis, MO, United States
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
16
|
Clark IH, Roman A, Fellows E, Radha S, Var SR, Roushdy Z, Borer SM, Johnson S, Chen O, Borgida JS, Steevens A, Shetty A, Strell P, Low WC, Grande AW. Cell Reprogramming for Regeneration and Repair of the Nervous System. Biomedicines 2022; 10:2598. [PMID: 36289861 PMCID: PMC9599606 DOI: 10.3390/biomedicines10102598] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/24/2022] [Accepted: 10/11/2022] [Indexed: 11/25/2022] Open
Abstract
A persistent barrier to the cure and treatment of neurological diseases is the limited ability of the central and peripheral nervous systems to undergo neuroregeneration and repair. Recent efforts have turned to regeneration of various cell types through cellular reprogramming of native cells as a promising therapy to replenish lost or diminished cell populations in various neurological diseases. This review provides an in-depth analysis of the current viral vectors, genes of interest, and target cellular populations that have been studied, as well as the challenges and future directions of these novel therapies. Furthermore, the mechanisms by which cellular reprogramming could be optimized as treatment in neurological diseases and a review of the most recent cellular reprogramming in vitro and in vivo studies will also be discussed.
Collapse
Affiliation(s)
- Isaac H. Clark
- Department of Biomedical Engineering, Biomedical Engineering Graduate Program, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Alex Roman
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Neuroscience, Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Emily Fellows
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Swathi Radha
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Susanna R. Var
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Zachary Roushdy
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Samuel M. Borer
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Samantha Johnson
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Olivia Chen
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jacob S. Borgida
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Aleta Steevens
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Anala Shetty
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
- Molecular, Cell, Developmental Biology & Genetics Graduate Program, University of Minnesota, Minneapolis, MN 55455, USA
| | - Phoebe Strell
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
- Comparative and Molecular Sciences Graduate Program, University of Minnesota, Minneapolis, MN 55455, USA
| | - Walter C. Low
- Department of Biomedical Engineering, Biomedical Engineering Graduate Program, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Neuroscience, Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
- Molecular, Cell, Developmental Biology & Genetics Graduate Program, University of Minnesota, Minneapolis, MN 55455, USA
- Comparative and Molecular Sciences Graduate Program, University of Minnesota, Minneapolis, MN 55455, USA
| | - Andrew W. Grande
- Department of Biomedical Engineering, Biomedical Engineering Graduate Program, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Neuroscience, Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
17
|
Xu D, Zhong LT, Cheng HY, Wang ZQ, Chen XM, Feng AY, Chen WY, Chen G, Xu Y. Overexpressing NeuroD1 reprograms Müller cells into various types of retinal neurons. Neural Regen Res 2022; 18:1124-1131. [PMID: 36255002 PMCID: PMC9827787 DOI: 10.4103/1673-5374.355818] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The onset of retinal degenerative disease is often associated with neuronal loss. Therefore, how to regenerate new neurons to restore vision is an important issue. NeuroD1 is a neural transcription factor with the ability to reprogram brain astrocytes into neurons in vivo. Here, we demonstrate that in adult mice, NeuroD1 can reprogram Müller cells, the principal glial cell type in the retina, to become retinal neurons. Most strikingly, ectopic expression of NeuroD1 using two different viral vectors converted Müller cells into different cell types. Specifically, AAV7m8 GFAP681::GFP-ND1 converted Müller cells into inner retinal neurons, including amacrine cells and ganglion cells. In contrast, AAV9 GFAP104::ND1-GFP converted Müller cells into outer retinal neurons such as photoreceptors and horizontal cells, with higher conversion efficiency. Furthermore, we demonstrate that Müller cell conversion induced by AAV9 GFAP104::ND1-GFP displayed clear dose- and time-dependence. These results indicate that Müller cells in adult mice are highly plastic and can be reprogrammed into various subtypes of retinal neurons.
Collapse
Affiliation(s)
- Di Xu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Key Laboratory of CNS Regeneration (Ministry of Education), Jinan University, Guangzhou, Guangdong Province, China
| | - Li-Ting Zhong
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Key Laboratory of CNS Regeneration (Ministry of Education), Jinan University, Guangzhou, Guangdong Province, China
| | - Hai-Yang Cheng
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Key Laboratory of CNS Regeneration (Ministry of Education), Jinan University, Guangzhou, Guangdong Province, China
| | - Zeng-Qiang Wang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Key Laboratory of CNS Regeneration (Ministry of Education), Jinan University, Guangzhou, Guangdong Province, China
| | - Xiong-Min Chen
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Key Laboratory of CNS Regeneration (Ministry of Education), Jinan University, Guangzhou, Guangdong Province, China
| | - Ai-Ying Feng
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Key Laboratory of CNS Regeneration (Ministry of Education), Jinan University, Guangzhou, Guangdong Province, China
| | - Wei-Yi Chen
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Key Laboratory of CNS Regeneration (Ministry of Education), Jinan University, Guangzhou, Guangdong Province, China
| | - Gong Chen
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Key Laboratory of CNS Regeneration (Ministry of Education), Jinan University, Guangzhou, Guangdong Province, China,Correspondence to: Ying Xu, ; Gong Chen, .
| | - Ying Xu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Key Laboratory of CNS Regeneration (Ministry of Education), Jinan University, Guangzhou, Guangdong Province, China,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China,Correspondence to: Ying Xu, ; Gong Chen, .
| |
Collapse
|
18
|
The 2021 yearbook of Neurorestoratology. JOURNAL OF NEURORESTORATOLOGY 2022. [DOI: 10.1016/j.jnrt.2022.100008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
19
|
Wang LL, Zhang CL. In vivo glia-to-neuron conversion: pitfalls and solutions. Dev Neurobiol 2022; 82:367-374. [PMID: 35535734 PMCID: PMC9337910 DOI: 10.1002/dneu.22880] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/15/2022] [Accepted: 05/05/2022] [Indexed: 11/08/2022]
Abstract
Neuron loss and disruption of neural circuits are associated with many neurological conditions. A key question is how to rebuild neural circuits for functional improvements. In vivo glia-to-neuron (GtN) conversion emerges as a potential solution for regeneration-based therapeutics. This approach takes advantage of the regenerative ability of resident glial cells to produce new neurons through cell fate reprogramming. Significant progress has been made over the years in this emerging field. However, inappropriate analysis often leads to misleading conclusions that create confusion and hype. In this perspective, we point out the most salient pitfalls associated with some recent studies and provide solutions to prevent them in the future. The goal is to foster healthy development of this promising field and lay a solid cellular foundation for future regeneration-based medicine.
Collapse
Affiliation(s)
- Lei-Lei Wang
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Chun-Li Zhang
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
20
|
Ma NX, Puls B, Chen G. Transcriptomic analyses of NeuroD1-mediated astrocyte-to-neuron conversion. Dev Neurobiol 2022; 82:375-391. [PMID: 35606902 PMCID: PMC9540770 DOI: 10.1002/dneu.22882] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 12/30/2022]
Abstract
Ectopic expression of a single neural transcription factor NeuroD1 can reprogram reactive glial cells into functional neurons both in vitro and in vivo, but the underlying mechanisms are not well understood yet. Here, we used RNA-sequencing technology to capture the transcriptomic changes at different time points during the reprogramming process. We found that following NeuroD1 overexpression, astroglial genes (ACTG1, ALDH1A3, EMP1, CLDN6, SOX21) were significantly downregulated, whereas neuronal genes (DCX, RBFOX3/NeuN, CUX2, RELN, SNAP25) were significantly upregulated. NeuroD family members (NeuroD1/2/6) and signaling pathways (Wnt, MAPK, cAMP) as well as neurotransmitter receptors (acetylcholine, somatostatin, dopamine) were also significantly upregulated. Gene co-expression analysis identified many central genes among the NeuroD1-interacting network, including CABP7, KIAA1456, SSTR2, GADD45G, LRRTM2, and INSM1. Compared to chemical conversion, we found that NeuroD1 acted as a strong driving force and triggered fast transcriptomic changes during astrocyte-to-neuron conversion process. Together, this study reveals many important downstream targets of NeuroD1 such as HES6, BHLHE22, INSM1, CHRNA1/3, CABP7, and SSTR2, which may play critical roles during the transcriptomic landscape shift from a glial profile to a neuronal profile.
Collapse
Affiliation(s)
- Ning-Xin Ma
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Brendan Puls
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Gong Chen
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA.,GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China
| |
Collapse
|
21
|
Leib D, Chen YH, Monteys AM, Davidson BL. Limited astrocyte-to-neuron conversion in the mouse brain using NeuroD1 overexpression. Mol Ther 2022; 30:982-986. [PMID: 35123657 PMCID: PMC8899704 DOI: 10.1016/j.ymthe.2022.01.028] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/15/2022] [Accepted: 01/18/2022] [Indexed: 10/19/2022] Open
Affiliation(s)
- David Leib
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yong Hong Chen
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alex Mas Monteys
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA,Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Beverly L. Davidson
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA,Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA,Corresponding author: Beverly L. Davidson, Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| |
Collapse
|
22
|
Abstract
Cellular identity is established through complex layers of genetic regulation, forged over a developmental lifetime. An expanding molecular toolbox is allowing us to manipulate these gene regulatory networks in specific cell types in vivo. In principle, if we found the right molecular tricks, we could rewrite cell identity and harness the rich repertoire of possible cellular functions and attributes. Recent work suggests that this rewriting of cell identity is not only possible, but that newly induced cells can mitigate disease phenotypes in animal models of major human diseases. So, is the sky the limit, or do we need to keep our feet on the ground? This Spotlight synthesises key concepts emerging from recent efforts to reprogramme cellular identity in vivo. We provide our perspectives on recent controversies in the field of glia-to-neuron reprogramming and identify important gaps in our understanding that present barriers to progress.
Collapse
Affiliation(s)
- Sydney Leaman
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE1 1UL, UK.,MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK
| | - Nicolás Marichal
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE1 1UL, UK
| | - Benedikt Berninger
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE1 1UL, UK.,MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, UK.,Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz 55128, Germany.,The Francis Crick Institute, London NW1 1AT, UK
| |
Collapse
|